201
|
Li M, Xie F, Wang L, Zhu G, Qi LW, Jiang S. Celastrol: An Update on Its Hepatoprotective Properties and the Linked Molecular Mechanisms. Front Pharmacol 2022; 13:857956. [PMID: 35444532 PMCID: PMC9013942 DOI: 10.3389/fphar.2022.857956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
The liver plays an important role in glucose and lipid homeostasis, drug metabolism, and bile synthesis. Metabolic disorder and inflammation synergistically contribute to the pathogenesis of numerous liver diseases, such as metabolic-associated fatty liver disease (MAFLD), liver injury, and liver cancer. Celastrol, a triterpene derived from Tripterygium wilfordii Hook.f., has been extensively studied in metabolic and inflammatory diseases during the last several decades. Here we comprehensively review the pharmacological activities and the underlying mechanisms of celastrol in the prevention and treatment of liver diseases including MAFLD, liver injury, and liver cancer. In addition, we also discuss the importance of novel methodologies and perspectives for the drug development of celastrol. Although celastrol has been claimed as a promising agent against several metabolic diseases, both preclinical and clinical studies are highly required to accelerate the clinical transformation of celastrol in treating different liver illness. It is foreseeable that celastrol-derived therapeutics is evolving in the field of liver ailments.
Collapse
Affiliation(s)
- Mengzhen Li
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, China
| | - Faren Xie
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Lian-Wen Qi
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, China
| | - Shujun Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
202
|
Sabirin F, Lim SM, Neoh CF, Ramasamy K. Hepatoprotection of Probiotics Against Non-Alcoholic Fatty Liver Disease in vivo: A Systematic Review. Front Nutr 2022; 9:844374. [PMID: 35479741 PMCID: PMC9035816 DOI: 10.3389/fnut.2022.844374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/03/2022] [Indexed: 12/20/2022] Open
Abstract
Probiotic supplements have been increasingly reported for their usefulness in delaying the development and progression of non-alcoholic fatty liver disease (NAFLD). Literature on the impact of probiotics on NAFLD covered various aspects of the disease. This study was undertaken to systematically review in vivo findings on hepatoprotection of probiotics against NAFLD. The literature search was performed through Cochrane, PubMed/MEDLINE, Embase, and Web of Science databases. Interventions of known probiotics in NAFLD-induced animal model with at least one measurable NAFLD-related parameter were included. The data were extracted by all authors independently. Quality assessment was conducted using the Systematic Review Center for Laboratory animal Experimentation (SYRCLE's) Risk of Bias (RoB) tool. P-values of measures were compared inter- and intra-study for each parameter. Forty-four probiotic-based studies of NAFLD-induced rodents were shortlisted. The majority of the studies were presented with low/unclear risk of bias. Probiotics improved the histopathology of NAFLD rodents (primary outcome). Most of the probiotic-supplemented NAFLD rodents were presented with mixed effects on serum liver enzymes but with improved hepatic and serum lipid profiles (including increased serum high-density lipoprotein cholesterol). The findings were generally accompanied by downregulation of hepatic lipogenic, oxidative, and inflammatory signallings. Probiotics were found to modulate gut microbiota composition and its products, and intestinal permeability. Probiotics also resulted in better glycaemic control and reduced liver weight. Altogether, the present qualitative appraisals strongly implied the hepatoprotective potential of probiotics against NAFLD in vivo.
Collapse
Affiliation(s)
- Faezah Sabirin
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Malaysia
- Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Malaysia
| | - Chin Fen Neoh
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Malaysia
- *Correspondence: Kalavathy Ramasamy
| |
Collapse
|
203
|
Sun CY, Zheng ZL, Chen CW, Lu BW, Liu D. Targeting Gut Microbiota With Natural Polysaccharides: Effective Interventions Against High-Fat Diet-Induced Metabolic Diseases. Front Microbiol 2022; 13:859206. [PMID: 35369480 PMCID: PMC8965082 DOI: 10.3389/fmicb.2022.859206] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Unhealthy diet, in particular high-fat diet (HFD) intake, can cause the development of several metabolic disorders, including obesity, hyperlipidemia, type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and metabolic syndrome (MetS). These popular metabolic diseases reduce the quality of life, and induce premature death worldwide. Evidence is accumulating that the gut microbiota is inextricably associated with HFD-induced metabolic disorders, and dietary intervention of gut microbiota is an effective therapeutic strategy for these metabolic dysfunctions. Polysaccharides are polymeric carbohydrate macromolecules and sources of fermentable dietary fiber that exhibit biological activities in the prevention and treatment of HFD-induced metabolic diseases. Of note, natural polysaccharides are among the most potent modulators of the gut microbiota composition. However, the prebiotics-like effects of polysaccharides in treating HFD-induced metabolic diseases remain elusive. In this review, we introduce the critical role of gut microbiota human health and HFD-induced metabolic disorders. Importantly, we review current knowledge about the role of natural polysaccharides in improving HFD-induced metabolic diseases by regulating gut microbiota.
Collapse
Affiliation(s)
- Chao-Yue Sun
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | | | - Cun-Wu Chen
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Bao-Wei Lu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Dong Liu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| |
Collapse
|
204
|
Zeybel M, Arif M, Li X, Altay O, Yang H, Shi M, Akyildiz M, Saglam B, Gonenli MG, Yigit B, Ulukan B, Ural D, Shoaie S, Turkez H, Nielsen J, Zhang C, Uhlén M, Borén J, Mardinoglu A. Multiomics Analysis Reveals the Impact of Microbiota on Host Metabolism in Hepatic Steatosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104373. [PMID: 35128832 PMCID: PMC9008426 DOI: 10.1002/advs.202104373] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Indexed: 05/03/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex disease involving alterations in multiple biological processes regulated by the interactions between obesity, genetic background, and environmental factors including the microbiome. To decipher hepatic steatosis (HS) pathogenesis by excluding critical confounding factors including genetic variants and diabetes, 56 heterogenous MAFLD patients are characterized by generating multiomics data including oral and gut metagenomics as well as plasma metabolomics and inflammatory proteomics data. The dysbiosis in the oral and gut microbiome is explored and the host-microbiome interactions based on global metabolic and inflammatory processes are revealed. These multiomics data are integrated using the biological network and HS's key features are identified using multiomics data. HS is finally predicted using these key features and findings are validated in a follow-up cohort, where 22 subjects with varying degree of HS are characterized.
Collapse
Affiliation(s)
- Mujdat Zeybel
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
- NIHR Nottingham Biomedical Research CentreNottingham University Hospitals NHS Trust & University of NottinghamNottinghamNG5 1PBUK
- Nottingham Digestive Diseases CentreSchool of MedicineUniversity of NottinghamNottinghamNG7 2UHUK
| | - Muhammad Arif
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
- Present address:
Laboratory of Cardiovascular Physiology and Tissue Injury and Section on Fibrotic DisordersNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMD20852USA
| | - Xiangyu Li
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
| | - Ozlem Altay
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
| | - Hong Yang
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
| | - Mengnan Shi
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
| | - Murat Akyildiz
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
| | - Burcin Saglam
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
| | - Mehmet Gokhan Gonenli
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
| | - Buket Yigit
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
| | - Burge Ulukan
- Department of Gastroenterology and HepatologySchool of MedicineKoç UniversityIstanbul34010Turkey
| | - Dilek Ural
- School of MedicineKoç UniversityIstanbul34010Turkey
| | - Saeed Shoaie
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonSE1 9RTUK
| | - Hasan Turkez
- Department of Medical BiologyFaculty of MedicineAtatürk UniversityErzurum25240Turkey
| | - Jens Nielsen
- Department of Biology and Biological EngineeringChalmers University of TechnologyGothenburgSE‐41296Sweden
| | - Cheng Zhang
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
- Key Laboratory of Advanced Drug Preparation TechnologiesMinistry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenan Province450001China
| | - Mathias Uhlén
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
| | - Jan Borén
- Department of Molecular and Clinical MedicineUniversity of Gothenburg and Sahlgrenska University Hospital GothenburgGothenburgSE‐41345Sweden
| | - Adil Mardinoglu
- Science for Life LaboratoryKTH – Royal Institute of TechnologyStockholmSE‐17121Sweden
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonSE1 9RTUK
| |
Collapse
|
205
|
Liu L, Fu Q, Li T, Shao K, Zhu X, Cong Y, Zhao X. Gut microbiota and butyrate contribute to nonalcoholic fatty liver disease in premenopause due to estrogen deficiency. PLoS One 2022; 17:e0262855. [PMID: 35108315 PMCID: PMC8809533 DOI: 10.1371/journal.pone.0262855] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) in postmenopausal women has increased significantly. Estrogen plays a very important role in NAFLD, but whether NAFLD in premenopausal women was caused by estrogen deficiency was unknown. Thus, it is of great clinical significance to explore the mechanism of NAFLD in premenopausal women. Gut microbiota and its metabolites short chain fatty acids (SCFA) have been shown to play important roles in the development of NAFLD. In this study, we investigated the impact of gut microbiota and SCFA in NAFLD patients and mice caused by estrogen deficiency. We showed that premenopause NAFLD patients had much lower estrogen levels. Estrogen deficient mice, due to ovariectomy (OVX), suffered more severe liver steatosis with an elevated body weight, abdominal fat weight, serum triglycerides and deterioration in hepatic steatosis. Altered gut microbiota composition and decreased butyrate content were found in NAFLD patients and in OVX mice. Furthermore, fecal microbiota transplantation (FMT) or supplementing with butyrate alleviated NAFLD in OVX mice. The production of antimicrobial peptides (AMP) Reg3ɣ, β-defensins and the expression of intestinal epithelial tight junction, including ZO-1 and Occluding-5, were decreased in the OVX mice compared to control mice. Upregulation of PPAR-ɣ and VLDLR, downregulation of PPAR-ɑ indicated that OVX mice suffered from abnormal lipid metabolism. These data indicate that changes in the gut microbiota and SCFA caused by estrogen reduction, together with a disorder in AMP production and lipid metabolism, promote NAFLD, thus provide SCFAs derived from microbiota as new therapeutic targets for the clinical prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Limin Liu
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Qingsong Fu
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- * E-mail: (XZ); (QF)
| | - Tiantian Li
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Kai Shao
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xing Zhu
- Department of Pathology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Xiaoyun Zhao
- Department of Medical Experiment Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- * E-mail: (XZ); (QF)
| |
Collapse
|
206
|
Mo L, Zhao GL, Li XF, He N, Xiao XL, Xu HX, Yu YG. Biocatalytical Acyl-Modification of Puerarin: Shape Gut Microbiota Profile and Improve Short Chain Fatty Acids Production in Rats. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:44-50. [PMID: 34822099 DOI: 10.1007/s11130-021-00936-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
Gut microbiota balance and metabolites have become a potentially mechanism in maintaining health. The specific aim of this study was to compare the modulation of puerarin and puerarin acid esters on gut microbial composition and metabolites. Male mice were fed a control diet or diets supplemented with puerarin, puerarin propanoate ester, puerarin hexanoate ester, puerarin myristate ester for 24 h, respectively. The result revealed that puerarin acid esters with different chain lengths showed different activities to create more own impacted bacterial. Puerarin propanoate and puerarin hexanoate ester significantly improved the diversity of microbiota and promoted the relative abundance of beneficial gut microbiota such as Lactobacillus, Barnesiella, Clostridium IV, Prevotella. Additionally, the puerarin propanoate ester group showed the capacity to deliver specific propionic acid to the colon. But esters with medium-long chain lengths had more opportunity to alter gut microbiota for enhancing the short chain fatty acids production. As a whole, puerarin acid esters with different chain lengths supplements shaped different gut microbial and short chain fatty acids metabolism, which could improve human health.
Collapse
Affiliation(s)
- Lan Mo
- School of Food Sciences and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Wushan Road 381, Guangzhou, 510641, China
| | - Guang-Lei Zhao
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Xiao-Feng Li
- School of Food Sciences and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Wushan Road 381, Guangzhou, 510641, China.
| | - Ning He
- The Laboratory of Advanced Design and Manufacturing for Precision Biomedical Devices, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Xing-Long Xiao
- School of Food Sciences and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Wushan Road 381, Guangzhou, 510641, China
| | - Hai-Xia Xu
- School of Food Sciences and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Wushan Road 381, Guangzhou, 510641, China
| | - Yi-Gang Yu
- School of Food Sciences and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Wushan Road 381, Guangzhou, 510641, China.
| |
Collapse
|
207
|
Zhang S, Tun HM, Zhang D, Chau HT, Huang FY, Kwok H, Wong DKH, Mak LY, Yuen MF, Seto WK. Alleviation of Hepatic Steatosis: Dithizone-Related Gut Microbiome Restoration During Paneth Cell Dysfunction. Front Microbiol 2022; 13:813783. [PMID: 35283810 PMCID: PMC8914291 DOI: 10.3389/fmicb.2022.813783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), the world’s most common chronic liver disease, is increasingly linked to gut dysbiosis. Paneth cells secrete antimicrobial peptides that regulate the gut microbiome, but their role in the pathogenesis of NAFLD remains unclear. Here, we determine the changes in NAFLD development and gut microbial composition and function via the injection of dithizone that can pharmacologically deplete the granules of Paneth cells. Eight-week-old C57BL/6J male mice (n = 31) were given a high-fat diet (HFD) or standard control diet for 12 weeks. Dithizone (10 mg/kg) was intravenously injected every 3 weeks during the period of diet feeding. Metagenomic DNA was extracted from fecal samples for PacBio Single-Molecule Real-Time sequencing to identify changes in microbial composition and predicted function. We observed dithizone-treated HFD mice, when compared to non-treated HFD mice, to have significant reductions in hepatic triglyceride content (28.98 vs. 53.52 mg/g, p = 0.0419); plasma insulin level (2.18 vs. 6.63 ng/ml, p = 0.0079); and relative mRNA levels of fatty acid synthase (0.52 vs. 1.57, p = 0.0428) and stearoyl-CoA desaturase-1 (0.43 vs. 1.20, p = 0.0121). Bacterial taxonomic profiling found dithizone-treated HFD mice, when compared to non-treated HFD mice, had a lower Firmicutes/Bacteroidetes ratio (2.53 vs. 5.26, p = 0.0541); a higher relative abundance of Bacteroides ASV21 and ASV42 (1.04 vs. 0.22%, p = 0.0277 and 0.96 vs. 0.09%, p = 0.0213); and a reduction in microbes belonging to Firmicutes (all p < 0.05). Bacteroides species correlated positively with predicted microbial functions such as L-methionine (r = 0.54, p = 0.0019) and tetrahydrofolate (r = 0.52, p = 0.0029) biosynthesis. Collectively, dithizone treatment was associated with alleviation in the severity of liver steatosis in HFD mice, possibly through gut microbiome modulation involving the increase in Bacteroides, suggesting microbiome-targeted therapies may have a role in the treatment of NAFLD.
Collapse
Affiliation(s)
- Saisai Zhang
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hein M. Tun
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Dengwei Zhang
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hau-Tak Chau
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fung-Yu Huang
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hin Kwok
- Centre for PanorOmic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Danny Ka-Ho Wong
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lung-Yi Mak
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Man-Fung Yuen
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Man-Fung Yuen,
| | - Wai-Kay Seto
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Wai-Kay Seto,
| |
Collapse
|
208
|
Gut Microbiome in Non-Alcoholic Fatty Liver Disease: From Mechanisms to Therapeutic Role. Biomedicines 2022; 10:biomedicines10030550. [PMID: 35327352 PMCID: PMC8945462 DOI: 10.3390/biomedicines10030550] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered to be a significant health threat globally, and has attracted growing concern in the research field of liver diseases. NAFLD comprises multifarious fatty degenerative disorders in the liver, including simple steatosis, steatohepatitis and fibrosis. The fundamental pathophysiology of NAFLD is complex and multifactor-driven. In addition to viruses, metabolic syndrome and alcohol, evidence has recently indicated that the microbiome is related to the development and progression of NAFLD. In this review, we summarize the possible microbiota-based therapeutic approaches and highlight the importance of establishing the diagnosis of NAFLD through the different spectra of the disease via the gut–liver axis.
Collapse
|
209
|
Song Q, Zhang X. The Role of Gut–Liver Axis in Gut Microbiome Dysbiosis Associated NAFLD and NAFLD-HCC. Biomedicines 2022; 10:biomedicines10030524. [PMID: 35327326 PMCID: PMC8945287 DOI: 10.3390/biomedicines10030524] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered as one of the most prevalent chronic liver diseases worldwide due to the rapidly rising prevalence of obesity and metabolic syndrome. As a hepatic manifestation of metabolic disease, NAFLD begins with hepatic fat accumulation and progresses to hepatic inflammation, termed as non-alcoholic steatohepatitis (NASH), hepatic fibrosis/cirrhosis, and finally leading to NAFLD-related hepatocellular carcinoma (NAFLD-HCC). Accumulating evidence showed that the gut microbiome plays a vital role in the initiation and progression of NAFLD through the gut–liver axis. The gut–liver axis is the mutual communication between gut and liver comprising the portal circulation, bile duct, and systematic circulation. The gut microbiome dysbiosis contributes to NAFLD development by dysregulating the gut–liver axis, leading to increased intestinal permeability and unrestrained transfer of microbial metabolites into the liver. In this review, we systematically summarized the up-to-date information of gut microbiome dysbiosis and metabolomic changes along the stages of steatosis, NASH, fibrosis, and NAFLD-HCC. The components and functions of the gut–liver axis and its association with NAFLD were then discussed. In addition, we highlighted current knowledge of gut microbiome-based treatment strategies targeting the gut–liver axis for preventing NAFLD and its associated HCC.
Collapse
Affiliation(s)
- Qian Song
- Department of Medicine and Therapeutics, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-3763-6102
| |
Collapse
|
210
|
Yue S, Shan B, Peng C, Tan C, Wang Q, Gong J. Theabrownin-targeted regulation of intestinal microorganisms to improve glucose and lipid metabolism in Goto-Kakizaki rats. Food Funct 2022; 13:1921-1940. [PMID: 35088787 DOI: 10.1039/d1fo03374c] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetes is a disease that is characterized by a disturbance of glucose metabolism. Theabrownin (TB) is one of the most active and abundant pigments in Pu-erh tea, and it is a brown pigment with multiple aromatic rings and attached residues of polysaccharides and proteins. TB has been shown to be hypolipidemic and displays fasting blood glucose (FBG)-lowering properties in rats fed a high-fat diet, but the underlying mechanism has not been elucidated. This study aimed to determine the effect of TB in treating diabetes and explore the underlying mechanism of action of intestinal microbes by using Goto-Kakizaki (GK) rats. Diabetic GK rats were treated up to 8 weeks with TB (GK-TB). Following treatment, the body weight, triglyceride (TG) content, fasting blood glucose (FBG) content, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) were significantly lower in the GK-TB group than in the GK control group (P < 0.05). Meanwhile, the circulating adiponectin (ADPN), leptin, and glucokinase levels in the serum of the GK-TB group were significantly higher than those in the GK group, while there was little difference in hepatic lipase (HL) and hormone-sensitive triglyceride lipase (HSL) enzyme activities (P > 0.05). Furthermore, with the extension of treatment time, the number of unique intestinal microorganisms in GK rats greatly increased and an interaction among intestinal microorganisms was observed. The Firmicutes/Bacteroides ratio was decreased significantly, and the composition of Actinobacteria and Proteobacteria was increased. The use of multiple omics technologies showed that TB is involved in the targeted regulation of the core characteristic intestinal flora including Bacteroides thetaiotaomicron (BT), Lactobacillus murinus (LM), Parabacteroides distasonis (PD), and Bacteroides_acidifaciens (BA) which improved the glucose and lipid metabolism of GK rats via the AMP-activated protein kinase signaling pathway, insulin signaling pathway, bile secretion and glycerophospholipid metabolism. Intragastric administration of BT, LM, PD, or BA led to a significantly reduced HOMA-IR in GK rats. Furthermore, BT significantly reduced serum lipid TG and total cholesterol (TC) and BA significantly reduced the serum lipid TC and low-density lipoprotein (LDL). PD significantly reduced serum LDL, while the effect of LM was not significant. However, LM and PD significantly increased the content of ADPN in serum. Taken together, our results indicated that the effect of TB on diabetic rats mainly depends on the targeted regulation of intestinal microorganisms and that TB is a functional food component with great potential to treat or prevent diabetes.
Collapse
Affiliation(s)
- Suijuan Yue
- College of Food Science and Technology, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| | - Bo Shan
- College of Food Science and Technology, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| | - Chunxiu Peng
- College of Horticulture and Landscape, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| | - Chao Tan
- College of Food Science and Technology, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| | - Qiuping Wang
- College of Food Science and Technology, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| | - Jiashun Gong
- College of Food Science and Technology, Yunnan Agricultural University, Heilong Tan, Kunming, Yunnan 650201, China.
| |
Collapse
|
211
|
Abstract
Metabolic (dysfunction) associated fatty liver disease (MAFLD), previously known as non-alcoholic fatty liver disease, is the most common cause of chronic liver disease worldwide. Many risk factors contribute to the pathogenesis of MAFLD with metabolic dysregulation being the final arbiter of its development and progression. MAFLD poses a substantial economic burden to societies, which based on current trends is expected to increase over time. Numerous studies have addressed various aspects of MAFLD from its risk associations to its economic and social burden and clinical diagnosis and management, as well as the molecular mechanisms linking MAFLD to end-stage liver disease and hepatocellular carcinoma. This review summarizes current understanding of the pathogenesis of MAFLD and related diseases, particularly liver cancer. Potential therapeutic agents for MAFLD and diagnostic biomarkers are discussed.
Collapse
|
212
|
Dysbiotic Gut Bacteria in Obesity: An Overview of the Metabolic Mechanisms and Therapeutic Perspectives of Next-Generation Probiotics. Microorganisms 2022; 10:microorganisms10020452. [PMID: 35208906 PMCID: PMC8877435 DOI: 10.3390/microorganisms10020452] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity, a worldwide health concern with a constantly rising prevalence, is a multifactorial chronic disease associated with a wide range of physiological disruptions, including energy imbalance, central appetite and food reward dysregulation, and hormonal alterations and gut dysbiosis. The gut microbiome is a well-recognized factor in the pathophysiology of obesity, and its influence on host physiology has been extensively investigated over the last decade. This review highlights the mechanisms by which gut dysbiosis can contribute to the pathophysiology of obesity. In particular, we discuss gut microbiota’s contribution to host energy homeostatic changes, low-grade inflammation, and regulation of fat deposition and bile acid metabolism via bacterial metabolites, such as short-chain fatty acids, and bacterial components, such as lipopolysaccharides, among others. Finally, therapeutic strategies based on next-generation probiotics aiming to re-shape the intestinal microbiota and reverse metabolic alterations associated with obesity are described.
Collapse
|
213
|
Zhang T, Gao G, Sakandar HA, Kwok LY, Sun Z. Gut Dysbiosis in Pancreatic Diseases: A Causative Factor and a Novel Therapeutic Target. Front Nutr 2022; 9:814269. [PMID: 35242797 PMCID: PMC8885515 DOI: 10.3389/fnut.2022.814269] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic-related disorders such as pancreatitis, pancreatic cancer, and type 1 diabetes mellitus (T1DM) impose a substantial challenge to human health and wellbeing. Even though our understanding of the initiation and progression of pancreatic diseases has broadened over time, no effective therapeutics is yet available for these disorders. Mounting evidence suggests that gut dysbiosis is closely related to human health and disease, and pancreatic diseases are no exception. Now much effort is under way to explore the correlation and eventually potential causation between the gut microbiome and the course of pancreatic diseases, as well as to develop novel preventive and/or therapeutic strategies of targeted microbiome modulation by probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT) for these multifactorial disorders. Attempts to dissect the intestinal microbial landscape and its metabolic profile might enable deep insight into a holistic picture of these complex conditions. This article aims to review the subtle yet intimate nexus loop between the gut microbiome and pancreatic diseases, with a particular focus on current evidence supporting the feasibility of preventing and controlling pancreatic diseases via microbiome-based therapeutics and therapies.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Guangqi Gao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Hafiz Arbab Sakandar
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
- *Correspondence: Zhihong Sun
| |
Collapse
|
214
|
Han L, Sun R, Wang Y, Luo J, Peng X. Soybean diacylglycerol regulates lipid metabolism in D-galactose-induced aging rats by altering gut microbiota and gene expression of colonic epithelial cells. Food Funct 2022; 13:1437-1446. [PMID: 35048932 DOI: 10.1039/d1fo04140a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lipid metabolism is closely related to the health of aging bodies and its disorder often leads to cardiovascular diseases and chronic diseases. Dietary fat is one of the important sources of body fat, which affects the body's lipid metabolism. However, how dietary fat affects lipid metabolism in aging bodies has not been reported. Thus, the effects of soybean diacylglycerol (DAG) on lipid metabolism in D-galactose-induced aging rats were investigated by detecting the serum biochemical indexes, hepatocyte morphology, gut microbiota changes, and gene expression of colonic epithelial cells. The results showed that DAG alleviated the lipid metabolism disorders, and the hepatocyte morphology of aging rats fed DAG was normal. 16S rDNA analysis showed that DAG restored Eisenbergiella and Veillonella that were missing in aging rats. The relative abundances of Romboutsia and Ruminococcus_2 decreased and the relative abundance of Lachnospiraceae NK4A136 group increased significantly with the influence of DAG (P < 0.05). Gene expression profiles showed that the gene expression of colon epithelial cells was altered by DAG and DAG downregulated the genes Lipe and Fabp4 related to the lipolysis of adipocytes. In conclusion, DAG regulated the lipid metabolism of aging rats by regulating gut microbiota and gene expression of colonic epithelial cells.
Collapse
Affiliation(s)
- Lulu Han
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| | - Rongrong Sun
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| | - Yong Wang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
215
|
Zhao E, Tait C, Minacapelli CD, Catalano C, Rustgi VK. Circadian Rhythms, the Gut Microbiome, and Metabolic Disorders. GASTRO HEP ADVANCES 2022; 1:93-105. [PMID: 39129932 PMCID: PMC11307590 DOI: 10.1016/j.gastha.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/15/2021] [Indexed: 08/13/2024]
Abstract
The circadian clock and gut microbiome play integral roles in preserving metabolic homeostasis. Circadian rhythms represent an endogenous time-keeping system that regulates cell and organ functions and synchronizes physiology with external cues to establish metabolic homeostasis. A variety of functions throughout the gastrointestinal tract and liver are under circadian control, including nutrient transport, processing, and detoxification. The gut microbiota also plays an essential role in host metabolism, regulating processes such as digestion, inflammatory modulation, and bile acid metabolism. Both the circadian clock and the gut microbiota influence each other in a reciprocal fashion, as gut dysbiosis can precipitate circadian asynchrony, and vice-versa. Disruption of either system impacts homeostasis in a bidirectional manner and can contribute to metabolic dysfunction. Evidence suggests such disruptions can lead to the development of metabolic diseases, including obesity, diabetes, nonalcoholic fatty liver disease, cirrhosis, and hepatocellular carcinoma. This review will provide a basic overview of the circadian and gut microbial systems, how they are intertwined, and their impact on the liver and gastrointestinal tract and in the development of metabolic disease. Particular areas of discussion include epigenetic regulation of circadian pathways as well as a mechanistic overview of microbial dysbiosis. In addition, therapeutic targets of these systems, including dietary modifications, behavioral modifications, and microbial-directed therapies, will be explored.
Collapse
Affiliation(s)
- Eric Zhao
- Department of Internal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Christopher Tait
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Center for Liver Diseases and Masses, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, New Jersey
| | - Carlos D. Minacapelli
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Center for Liver Diseases and Masses, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, New Jersey
| | - Carolyn Catalano
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Center for Liver Diseases and Masses, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, New Jersey
| | - Vinod K. Rustgi
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Center for Liver Diseases and Masses, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, New Jersey
| |
Collapse
|
216
|
Martínez-Montoro JI, Kuchay MS, Balaguer-Román A, Martínez-Sánchez MA, Frutos MD, Fernández-García JC, Ramos-Molina B. Gut microbiota and related metabolites in the pathogenesis of nonalcoholic steatohepatitis and its resolution after bariatric surgery. Obes Rev 2022; 23:e13367. [PMID: 34729904 DOI: 10.1111/obr.13367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in parallel with the rising prevalence of obesity, leading to major health and socioeconomic consequences. To date, the most effective therapeutic approach for NAFLD is weight loss. Accordingly, bariatric surgery (BS), which produces marked reductions in body weight, is associated with significant histopathological improvements in advanced stages of NAFLD, such as nonalcoholic steatohepatitis (NASH) and liver fibrosis. BS is also associated with substantial taxonomical and functional alterations in gut microbiota, which are believed to play a significant role in metabolic improvement after BS. Interestingly, gut microbiota and related metabolites may be implicated in the pathogenesis of NAFLD through diverse mechanisms, including specific microbiome signatures, short chain fatty acid production or the modulation of one-carbon metabolism. Moreover, emerging evidence highlights the potential association between gut microbiota changes after BS and NASH resolution. In this review, we summarize the current knowledge on the relationship between NAFLD severity and gut microbiota, as well as the role of the gut microbiome and related metabolites in NAFLD improvement after BS.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Malaga (IBIMA), Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mohammad Shafi Kuchay
- Division of Endocrinology and Diabetes, Medanta - The Medicity Hospital, Gurugram, Haryana, India
| | - Andrés Balaguer-Román
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain.,Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | | | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - José Carlos Fernández-García
- Department of Endocrinology and Nutrition, Regional University Hospital of Malaga, Institute of Biomedical Research in Malaga (IBIMA), Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Bruno Ramos-Molina
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
217
|
Liang Q, Zhao Q, Hao X, Wang J, Ma C, Xi X, Kang W. The Effect of Flammulina velutipes Polysaccharide on Immunization Analyzed by Intestinal Flora and Proteomics. Front Nutr 2022; 9:841230. [PMID: 35155543 PMCID: PMC8832141 DOI: 10.3389/fnut.2022.841230] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Proteomics and intestinal flora were used to determine the mechanism of immune modulatory effects of Flammulina velutipes polysaccharide on immunosuppressed mice. The results showed that compared with the model group, F. velutipes polysaccharide could increase thymus and spleen indices and improve thymus tissue structure in mice; IL-2 and IL-4 contents were significantly increased and IL-6 and TNF-α contents were significantly decreased; serum acid phosphatase (ACP), lactate dehydrogenase (LDH) and total antioxidant capacity (T-AOC) activities were increased (P < 0.05); in the liver, superoxide dismutase (SOD) and catalase (CAT) activities were increased (P < 0.001), while malondialdehyde (MDA) content was decreased (P < 0.001). Proteomics discovered that F. velutipes polysaccharides may exert immune modulatory effects by participating in signaling pathways such as immune diseases, transport and catabolism, phagosomes and influenza A, regulating the immune-related proteins Transferrin receptor protein 1 (TFRC) and Radical S-adenosyl methionine domain-containing protein 2 (RSAD2), etc. Gut microbial studies showed that F. velutipes polysaccharides could increase the abundance of intestinal flora and improve the flora structure. Compared to the model group, the content of short-chain fatty acids (SCFAs) and the relative abundance of SCFA-producers Bacteroides and Alloprevotella were increased in the F. velutipes polysaccharide administration group, while Lachnospiraceae_NK4A136_group and f_Lachnospiraceae_Unclassified decreased in relative abundance. Thus, F. velutipes polysaccharide may play an immunomodulatory role by regulating the intestinal environment and improving the balance of flora.
Collapse
Affiliation(s)
- Qiongxin Liang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Qingchun Zhao
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Xuting Hao
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Jinmei Wang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng, China
- *Correspondence: Changyang Ma
| | - Xuefeng Xi
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- College of Physical Education, Henan University, Kaifeng, China
- Xuefeng Xi
| | - Wenyi Kang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng, China
- Wenyi Kang
| |
Collapse
|
218
|
Simas AM, Kramer CD, Genco CA. Diet-Induced Non-alcoholic Fatty Liver Disease and Associated Gut Dysbiosis Are Exacerbated by Oral Infection. FRONTIERS IN ORAL HEALTH 2022; 2:784448. [PMID: 35141703 PMCID: PMC8820505 DOI: 10.3389/froh.2021.784448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence indicates that chronic inflammation due to periodontal disease is associated with progression of non-alcoholic fatty liver disease (NAFLD) caused by a Western diet. NAFLD has also been associated with oral infection with the etiological agent of periodontal disease, Porphyromonas gingivalis. P. gingivalis oral infection has been shown to induce cardiometabolic disease features including hepatic lipid accumulation while also leading to dysbiosis of the gut microbiome. However, the impact of P. gingivalis infection on the gut microbiota of mice with diet-induced NAFLD and the potential for those changes to mediate NAFLD progression has yet to be determined. In the current study, we have demonstrated that P. gingivalis infection induced sustained alterations of the gut microbiota composition and predicted functions, which was associated with the promotion of NAFLD in steatotic mice. Reduced abundance of short-chain fatty acid-producing microbiota was observed after both acute and chronic P. gingivalis infection. Collectively, our findings demonstrate that P. gingivalis infection produces a persistent change in the gut microbiota composition and predicted functions that promotes steatosis and metabolic disease.
Collapse
Affiliation(s)
- Alexandra M. Simas
- Gerald J. and Dorothy R. Friedman School of Nutrition and Science Policy, Graduate Program in Biochemical and Molecular Nutrition, Tufts University, Boston, MA, United States
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Carolyn D. Kramer
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Caroline A. Genco
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
- *Correspondence: Caroline A. Genco
| |
Collapse
|
219
|
Zhang J, Ma X, Fan D. Ginsenoside CK ameliorates hepatic lipid accumulation via activating the LKB1/AMPK pathway in vitro and in vivo. Food Funct 2022; 13:1153-1167. [PMID: 35018944 DOI: 10.1039/d1fo03026d] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a metabolic liver disease with a complex etiology, and is considered as one of the main causes of hepatocellular carcinoma (HCC). The incidence of NAFLD has presented an increasing trend annually as a result of disequilibrium in the dietary structure. However, no specific treatment has been approved for clinical therapy in NAFLD. Ginsenoside CK has been investigated given its various pharmacological activities, but its effects against NAFLD and the underlying mechanism are still unclear. In this study, fructose was used to simulate hepatic fatty degeneration in vivo, while palmitic acid (PA) and oleic acid (OA) were applied to induce lipid accumulation in vitro. The level of lipid accumulation in hepatic tissue and HepG2 cells was evaluated by Oil Red O staining. Detection of serum and liver biomarkers, western blotting, and real-time qPCR were conducted to assess the degree of hepatic steatosis. Our results indicated that ginsenoside CK could decrease the lipid deposition in HepG2 cells, retard the increase of body weight of fructose-fed mice, alleviate the lipid accumulation in serum and hepatic tissue and improve the hepatic inflammation and injury. Mechanically, ginsenoside CK modulated the expression of factors correlated with lipid synthesis and metabolism in vitro and in vivo via activating the phosphorylation of LKB1 and AMPK. Compound C, an inhibitor of AMPK, partially abrogated the beneficial effects of ginsenoside CK in HepG2 cells. In summary, ginsenoside CK acts as a LKB1/AMPK agonist to regulate the lipid metabolism and interfere with the progression of NAFLD.
Collapse
Affiliation(s)
- Jingjing Zhang
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China. .,Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.,Biotech. & Biomed. Research Institute, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Xiaoxuan Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China. .,Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.,Biotech. & Biomed. Research Institute, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China. .,Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.,Biotech. & Biomed. Research Institute, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| |
Collapse
|
220
|
Chen S, Yang M, Wang R, Fan X, Tang T, Li P, Zhou X, Qi K. Suppression of high-fat-diet-induced obesity in mice by dietary folic acid supplementation is linked to changes in gut microbiota. Eur J Nutr 2022; 61:2015-2031. [PMID: 34993642 DOI: 10.1007/s00394-021-02769-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE To investigate whether the effects of dietary folic acid supplementation on body weight gain are mediated by gut microbiota in obesity. METHODS Male C57 BL/6J conventional (CV) and germ-free (GF) mice both aged three to four weeks were fed a high-fat diet (HD), folic acid-deficient HD (FD-HD), folic acid-supplement HD (FS-HD) and a normal-fat diet (ND) for 25 weeks. Faecal microbiota were analyzed by 16S rRNA high-throughput sequencing, and the mRNA expression of genes was determined by the real-time RT-PCR. Short-chain fatty acids (SCFAs) in faeces and plasma were measured using gas chromatography-mass spectrometry. RESULTS In CV mice, HD-induced body weight gain was inhibited by FS-HD, accompanied by declined energy intake, smaller white adipocyte size, and less whitening of brown adipose tissue. Meanwhile, the HD-induced disturbance in the expression of fat and energy metabolism-associated genes (Fas, Atgl, Hsl, Ppar-α, adiponectin, resistin, Ucp2, etc.) in epididymal fat was diminished, and the dysbiosis in faecal microbiota was lessened, by FS-HD. However, in GF mice with HD feeding, dietary folic acid supplementation had almost no effect on body weight gain and the expression of fat- and energy-associated genes. Faecal or plasma SCFA concentrations in CV and GF mice were not altered by either FD-HD or FS-HD feeding. CONCLUSION Dietary folic acid supplementation differently affected body weight gain and associated genes' expression under HD feeding between CV and GF mice, suggesting that gut bacteria might partially share the responsibility for beneficial effects of dietary folate on obesity.
Collapse
Affiliation(s)
- Si Chen
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Mengyi Yang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Rui Wang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Xiuqin Fan
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Tiantian Tang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Ping Li
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Xinhui Zhou
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China
| | - Kemin Qi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institutue, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-li-shi Road, Beijing, 100045, China.
| |
Collapse
|
221
|
He Q, Zhang Y, Ma D, Zhang W, Zhang H. Lactobacillus casei Zhang exerts anti-obesity effect to obese glut1 and gut-specific-glut1 knockout mice via gut microbiota modulation mediated different metagenomic pathways. Eur J Nutr 2022; 61:2003-2014. [PMID: 34984487 DOI: 10.1007/s00394-021-02764-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Obesity is a major risk factor for various metabolic diseases, including metabolic syndrome and type-2 diabetes. Glucose transporter 1 (GLUT1) impairment has been proposed as a mechanism of fat accumulation and glucose tolerance. Our aims were to determine the role of intestinal epithelial glut1 activity in obesity and the mechanism of anti-obesity effect of Lactobacillus casei Zhang (LCZ) intervention in the absence of gut villi-specific glut1 expression. METHODS This study compared the body weight, intestinal microbiota perturbations, fat mass accumulation, and glucose tolerance (by oral glucose tolerance test) between high-fat diet fed villi-specific glut1 knockout (KO) mice and control mice (glut1 flox/flox) with/without LCZ intervention. The intestinal microbiota was evaluated by metagenomic sequencing. RESULTS Our results showed that villi-specific glut1 KO mice had more fat deposition at the premetaphase stage, impaired glucose tolerance, and obvious alterations in gut microbiota compared to control mice. Probiotic administration significantly lowered the body weight, the weights of mesenteric and perirenal white adipose tissues (WAT), and mediated gut microbiota modulation in both types of KO and control mice. The species Barnesiella intestinihominis and Faecalibaculum rodentium might contribute to fasting fat mass accumulation associated with gut-specific glut1 inactivation, while the probiotic-mediated anti-obesity effect was linked to members of the Bacteroides genera, Odoribacter genera and Alistipes finegoldii. CONCLUSION Our study demonstrated that abrogating gut epithelial GLUT1 activity affected the gut microbiota, fat mass accumulation, and glucose tolerance; and LCZ administration reduced fat mass accumulation and central obesity.
Collapse
Affiliation(s)
- Qiuwen He
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Yong Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing (USTB), Beijing, China
| | - Da Ma
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Weiqin Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China. .,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
222
|
Bandopadhyay P, Ganguly D. Gut dysbiosis and metabolic diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:153-174. [DOI: 10.1016/bs.pmbts.2022.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
223
|
Forssten SD, Ouwehand AC. Contribution of the Microbiota to Healthy Aging. COMPREHENSIVE GUT MICROBIOTA 2022:69-84. [DOI: 10.1016/b978-0-12-819265-8.00059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
224
|
Schupack DA, Mars RAT, Voelker DH, Abeykoon JP, Kashyap PC. The promise of the gut microbiome as part of individualized treatment strategies. Nat Rev Gastroenterol Hepatol 2022; 19:7-25. [PMID: 34453142 PMCID: PMC8712374 DOI: 10.1038/s41575-021-00499-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Variability in disease presentation, progression and treatment response has been a central challenge in medicine. Although variability in host factors and genetics are important, it has become evident that the gut microbiome, with its vast genetic and metabolic diversity, must be considered in moving towards individualized treatment. In this Review, we discuss six broad disease groups: infectious disease, cancer, metabolic disease, cardiovascular disease, autoimmune or inflammatory disease, and allergic and atopic diseases. We highlight current knowledge on the gut microbiome in disease pathogenesis and prognosis, efficacy, and treatment-related adverse events and its promise for stratifying existing treatments and as a source of novel therapies. The Review is not meant to be comprehensive for each disease state but rather highlights the potential implications of the microbiome as a tool to individualize treatment strategies in clinical practice. Although early, the outlook is optimistic but challenges need to be overcome before clinical implementation, including improved understanding of underlying mechanisms, longitudinal studies with multiple data layers reflecting gut microbiome and host response, standardized approaches to testing and reporting, and validation in larger cohorts. Given progress in the microbiome field with concurrent basic and clinical studies, the microbiome will likely become an integral part of clinical care within the next decade.
Collapse
Affiliation(s)
- Daniel A Schupack
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Ruben A T Mars
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Dayne H Voelker
- Division of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jithma P Abeykoon
- Division of Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
225
|
Li M, Rajani C, Zheng X, Jia W. The microbial metabolome in metabolic-associated fatty liver disease. J Gastroenterol Hepatol 2022; 37:15-23. [PMID: 34850445 DOI: 10.1111/jgh.15746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022]
Abstract
Metabolism-associated fatty liver disease (MAFLD) is defined as the presence of excess fat in the liver in the absence of excess alcohol consumption and metabolic dysfunction. It has also been described as the hepatic manifestation of metabolic syndrome. The incidence of MAFLD has been reported to be 43-60% in diabetics, ~90% in patients with hyperlipidemia, and 91% in morbidly obese patients. Risk factors that have been associated with the development of MAFLD include male gender, increasing age, obesity, insulin resistance, diabetes, and hyperlipidemia. All of these risk factors have been linked to alterations of the gut microbiota, that is, gut dysbiosis. MAFLD can progress to non-alcoholic steatohepatitis with the presence of inflammation and ballooning, which can deteriorate into cirrhosis, MAFLD-related hepatocellular carcinoma, and liver failure. In this review, we will be focused on the role of the gut microbial metabolome in the development, progression, and potential treatment of MAFLD.
Collapse
Affiliation(s)
- Mengci Li
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cynthia Rajani
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Xiaojiao Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| |
Collapse
|
226
|
Rashed ZE, Grasselli E, Khalifeh H, Canesi L, Demori I. Brown-Algae Polysaccharides as Active Constituents against Nonalcoholic Fatty Liver Disease. PLANTA MEDICA 2022; 88:9-19. [PMID: 33142346 DOI: 10.1055/a-1273-3159] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nonalcoholic fatty liver disease is a metabolic disorder characterized by lipid overloading in hepatocytes that can progress pathogenically and even end in hepatocellular carcinoma. Nonalcoholic fatty liver disease pharmacological treatment is still limited by unwanted side effects, whereas the use of food components with therapeutic potential is advisable. The culinary use of marine algae is traditional for some populations and reviving worldwide, with promising health outcomes due to the large number of bioactive compounds found in seaweeds. The present review focuses on brown-algae polysaccharides, particularly fucoidan, alginate, and laminarin, and summarizes the experimental evidence of their potential effects against nonalcoholic fatty liver disease onset and progression. In vitro and in vivo studies demonstrate that brown-algae polysaccharides exert beneficial actions on satiety feeling, caloric intake, fat absorption, and modulation of the gut microbiota, which could account for indirect effects on energy and lipid homeostasis, thus diminishing the fat overload in the liver. Specific effects against nonalcoholic fatty liver disease pathogenesis and worsening are also described and sustained by the antioxidant, anti-inflammatory, and antisteatotic properties of brown-algae polysaccharides. Further studies are required to clarify the mechanism of action of brown-algae polysaccharides on liver cells, to determine the composition and bioavailability of brown-algae polysaccharides present in different algal sources and to probe the clinical availability of these compounds in the form of algal foods, food supplements, and regulated therapeutics.
Collapse
Affiliation(s)
- Zeinab El Rashed
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
- Rammal Rammal Laboratory (ATAC group), Faculty of Sciences I, Lebanese University, Beirut, Lebanon
| | - Elena Grasselli
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Hala Khalifeh
- Rammal Rammal Laboratory (ATAC group), Faculty of Sciences I, Lebanese University, Beirut, Lebanon
| | - Laura Canesi
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Ilaria Demori
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| |
Collapse
|
227
|
Wang F, Zou J, Xu H, Huang W, Zhang X, Wei Z, Li X, Liu Y, Zou J, Liu F, Zhu H, Yi H, Guan J, Yin S. Effects of Chronic Intermittent Hypoxia and Chronic Sleep Fragmentation on Gut Microbiome, Serum Metabolome, Liver and Adipose Tissue Morphology. Front Endocrinol (Lausanne) 2022; 13:820939. [PMID: 35178032 PMCID: PMC8846366 DOI: 10.3389/fendo.2022.820939] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/06/2022] [Indexed: 12/29/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) and chronic sleep fragmentation (CSF) are two cardinal pathological features of obstructive sleep apnea (OSA). Dietary obesity is a crucial risk intermediator for OSA and metabolic disorders. Gut microbiota affect hepatic and adipose tissue morphology under conditions of CIH or CSF through downstream metabolites. However, the exact relationship is unclear. Herein, chow and high-fat diet (HFD)-fed mice were subjected to CIH or CSF for 10 weeks each and compared to normoxia (NM) or normal sleep (NS) controls. 16S rRNA amplicon sequencing, untargeted liquid chromatography-tandem mass spectrometry, and histological assessment of liver and adipose tissues were used to investigate the correlations between the microbiome, metabolome, and lipid metabolism under CIH or CSF condition. Our results demonstrated that CIH and CSF regulate the abundance of intestinal microbes (such as Akkermansia mucinphila, Clostridium spp., Lactococcus spp., and Bifidobacterium spp.) and functional metabolites, such as tryptophan, free fatty acids, branched amino acids, and bile acids, which influence adipose tissue and hepatic lipid metabolism, and the level of lipid deposition in tissues and peripheral blood. In conclusion, CIH and CSF adversely affect fecal microbiota composition and function, and host metabolism; these findings provide new insight into the independent and synergistic effects of CIH, CSF, and HFD on lipid disorders.
Collapse
Affiliation(s)
- Fan Wang
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Juanjuan Zou
- Department of Otorhinolaryngology and National Health Commission (NHC) Key Laboratory of Otorhinolaryngology, Shandong University Affiliated Qilu Hospital, Jinan, China
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Huajun Xu, ; Jian Guan, ; Shankai Yin,
| | - Weijun Huang
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaoman Zhang
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Zhicheng Wei
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xinyi Li
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yupu Liu
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jianyin Zou
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Feng Liu
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Huaming Zhu
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hongliang Yi
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jian Guan
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Huajun Xu, ; Jian Guan, ; Shankai Yin,
| | - Shankai Yin
- Department of Otolaryngology-Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing and Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Huajun Xu, ; Jian Guan, ; Shankai Yin,
| |
Collapse
|
228
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
229
|
Understanding the Role of the Gut Microbiome and Microbial Metabolites in Non-Alcoholic Fatty Liver Disease: Current Evidence and Perspectives. Biomolecules 2021; 12:biom12010056. [PMID: 35053205 PMCID: PMC8774162 DOI: 10.3390/biom12010056] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. NAFLD begins as a relatively benign hepatic steatosis which can evolve to non-alcoholic steatohepatitis (NASH); the risk of cirrhosis and hepatocellular carcinoma (HCC) increases when fibrosis is present. NAFLD represents a complex process implicating numerous factors—genetic, metabolic, and dietary—intertwined in a multi-hit etiopathogenetic model. Recent data have highlighted the role of gut dysbiosis, which may render the bowel more permeable, leading to increased free fatty acid absorption, bacterial migration, and a parallel release of toxic bacterial products, lipopolysaccharide (LPS), and proinflammatory cytokines that initiate and sustain inflammation. Although gut dysbiosis is present in each disease stage, there is currently no single microbial signature to distinguish or predict which patients will evolve from NAFLD to NASH and HCC. Using 16S rRNA sequencing, the majority of patients with NAFLD/NASH exhibit increased numbers of Bacteroidetes and differences in the presence of Firmicutes, resulting in a decreased F/B ratio in most studies. They also present an increased proportion of species belonging to Clostridium, Anaerobacter, Streptococcus, Escherichia, and Lactobacillus, whereas Oscillibacter, Flavonifaractor, Odoribacter, and Alistipes spp. are less prominent. In comparison to healthy controls, patients with NASH show a higher abundance of Proteobacteria, Enterobacteriaceae, and Escherichia spp., while Faecalibacterium prausnitzii and Akkermansia muciniphila are diminished. Children with NAFLD/NASH have a decreased proportion of Oscillospira spp. accompanied by an elevated proportion of Dorea, Blautia, Prevotella copri, and Ruminococcus spp. Gut microbiota composition may vary between population groups and different stages of NAFLD, making any conclusive or causative claims about gut microbiota profiles in NAFLD patients challenging. Moreover, various metabolites may be involved in the pathogenesis of NAFLD, such as short-chain fatty acids, lipopolysaccharide, bile acids, choline and trimethylamine-N-oxide, and ammonia. In this review, we summarize the role of the gut microbiome and metabolites in NAFLD pathogenesis, and we discuss potential preventive and therapeutic interventions related to the gut microbiome, such as the administration of probiotics, prebiotics, synbiotics, antibiotics, and bacteriophages, as well as the contribution of bariatric surgery and fecal microbiota transplantation in the therapeutic armamentarium against NAFLD. Larger and longer-term prospective studies, including well-defined cohorts as well as a multi-omics approach, are required to better identify the associations between the gut microbiome, microbial metabolites, and NAFLD occurrence and progression.
Collapse
|
230
|
Metabolic Associated Fatty Liver Disease in Children-From Atomistic to Holistic. Biomedicines 2021; 9:biomedicines9121866. [PMID: 34944682 PMCID: PMC8698557 DOI: 10.3390/biomedicines9121866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease has become the most common chronic liver disease in children due to the alarmingly increasing incidence of pediatric obesity. It is well-documented that MAFLD prevalence is directly related to an incremental increase in BMI. The multiple hits theory was designed for providing insights regarding the pathogenesis of steatohepatitis and fibrosis in MAFLD. Recent evidence suggested that the microbiome is a crucial contributor in the pathogenesis of MAFLD. Aside from obesity, the most common risk factors for pediatric MAFLD include male gender, low-birth weight, family history of obesity, MAFLD, insulin resistance, type 2 diabetes mellitus, obstructive sleep apnea, and polycystic ovarium syndrome. Usually, pediatric patients with MAFLD have nonspecific symptoms consisting of fatigue, malaise, or diffuse abdominal pain. A wide spectrum of biomarkers was proposed for the diagnosis of MAFLD and NASH, as well as for quantifying the degree of fibrosis, but liver biopsy remains the key diagnostic and staging tool. Nevertheless, elastography-based methods present promising results in this age group as potential non-invasive replacers for liver biopsy. Despite the lack of current guidelines regarding MAFLD treatment in children, lifestyle intervention was proven to be crucial in the management of these patients.
Collapse
|
231
|
Jiang Y, Chowdhury S, Xu BH, Meybodi MA, Damiris K, Devalaraju S, Pyrsopoulos N. Nonalcoholic fatty liver disease is associated with worse intestinal complications in patients hospitalized for Clostridioides difficile infection. World J Hepatol 2021; 13:1777-1790. [PMID: 34904045 PMCID: PMC8637681 DOI: 10.4254/wjh.v13.i11.1777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/08/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease with increasing prevalence worldwide. Clostridioides difficile infection (CDI) remains the most common cause of nosocomial diarrhea in developed countries. AIM To assess the impact of NAFLD on the outcomes of hospitalized patients with CDI. METHODS This study was a retrospective cohort study. The Nationwide Inpatient Sample database was used to identify a total of 7239 adults admitted as inpatients with a primary diagnosis of CDI and coexisting NAFLD diagnosis from 2010 to 2014 using ICD-9 codes. Patients with CDI and coexisting NAFLD were compared to those with CDI and coexisting alcoholic liver disease (ALD) and viral liver disease (VLD), individually. Primary outcomes included mortality, length of stay, and total hospitalization charges. Secondary outcomes were in-hospital complications. Multivariate regression was used for outcome analysis after adjusting for possible confounders. RESULTS CDI with NAFLD was independently associated with lower rates of acute respiratory failure (2.7% vs 4.2%, P < 0.01; 2.7% vs 4.2%, P < 0.05), shorter length of stay (days) (5.75 ± 0.16 vs 6.77 ± 0.15, P < 0.001; 5.75 ± 0.16 vs 6.84 ± 0.23, P <0.001), and lower hospitalization charges (dollars) (38150.34 ± 1757.01 vs 46326.72 ± 1809.82, P < 0.001; 38150.34 ± 1757.01 vs 44641.74 ± 1660.66, P < 0.001) when compared to CDI with VLD and CDI with ALD, respectively. CDI with NAFLD was associated with a lower rate of acute kidney injury (13.0% vs 17.2%, P < 0.01), but a higher rate of intestinal perforation (P < 0.01) when compared to VLD. A lower rate of mortality (0.8% vs 2.7%, P < 0.05) but a higher rate of intestinal obstruction (4.6% vs 2.2%, P = 0.001) was also observed when comparing CDI with NAFLD to ALD. CONCLUSION Hospitalized CDI patients with NAFLD had more intestinal complications compared to CDI patients with VLD and ALD. Gut microbiota dysbiosis may contribute to the pathogenesis of intestinal complications.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, United States
| | - Salil Chowdhury
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, United States
| | - Bing-Hong Xu
- Liver Center and Center for Asian Health, RWJBH-Saint Barnabas Medical Center, Florham Park, NJ 07932, United States
| | - Mohamad Aghaie Meybodi
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, United States
| | - Konstantinos Damiris
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, United States
| | - Samanthika Devalaraju
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, United States
| | - Nikolaos Pyrsopoulos
- Department of Medicine, Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07101, United States.
| |
Collapse
|
232
|
Shen B, Wang J, Guo Y, Gu T, Shen Z, Zhou C, Li B, Xu X, Li F, Zhang Q, Cai X, Dong H, Lu L. Dextran Sulfate Sodium Salt-Induced Colitis Aggravates Gut Microbiota Dysbiosis and Liver Injury in Mice With Non-alcoholic Steatohepatitis. Front Microbiol 2021; 12:756299. [PMID: 34795650 PMCID: PMC8593467 DOI: 10.3389/fmicb.2021.756299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Objective: Inflammatory bowel disease (IBD) is characterized by gut microbiota dysbiosis, which is also frequently observed in patients with non-alcoholic fatty liver disease. Whether gut microbiota dysbiosis in IBD patients promotes the development of non-alcoholic steatohepatitis (NASH) remains unclear. We aimed to explore the role of gut microbiota dysbiosis in the development of NASH in mice with dextran sulfate sodium salt (DSS) induced colitis. Design: Dextran sulfate sodium salt was used to induce colitis, and high fat (HF), in combination with a high-fructose diet, was used to induce NASH in C57BL/6J male mice. Mice were treated with (1%) DSS to induce colitis in cycles, and each cycle consisted of 7 days of DSS administration followed by a 10-day interval. The cycles were repeated throughout the experimental period of 19 weeks. Pathological alterations in colitis and NASH were validated by hematoxylin and eosin (H&E), oil red O, Sirius red staining, and immunofluorescence. Gut microbiota was examined by 16S rRNA sequencing, and gene expression profiles of hepatic non-parenchymal cells (NPCs) were detected by RNA sequencing. Results: Dextran sulfate sodium salt administration enhanced the disruption of the gut-vascular barrier and aggravated hepatic inflammation and fibrosis in mice with NASH. DSS-induced colitis was accompanied by gut microbiota dysbiosis, characterized by alteration in the core microbiota composition. Compared with the HF group, the abundance of p_Proteobacteria and g_Bacteroides increased, while that of f_S24-7 decreased in the DSS + HF mice. Specifically, gut microbiota dysbiosis was characterized by enrichment of lipopolysaccharide producing bacteria and decreased abundance of short-chain fatty acid-producing bacteria. Gene expression analysis of liver NPCs indicated that compared with the HF group, genes related to both inflammatory response and angiocrine signaling were altered in the DSS + HF group. The expression levels of inflammation-related and vascular development genes correlated significantly with the abundance of p_Proteobacteria, g_Bacteroides, or f_S24-7 in the gut microbiota, implying that gut microbiota dysbiosis induced by DSS might aggravate hepatic inflammation and fibrosis by altering the gene expression in NPCs. Conclusion: Dextran sulfate sodium salt-induced colitis may promote the progression of liver inflammation and fibrosis by inducing microbiota dysbiosis, which triggers an inflammatory response and disrupts angiocrine signaling in liver NPCs. The abundance of gut microbiota was associated with expression levels of inflammation-related genes in liver NPCs and may serve as a potential marker for the progression of NASH.
Collapse
Affiliation(s)
- Bo Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuecheng Guo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Gu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyang Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Binghang Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianjun Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobo Cai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Dong
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
233
|
He LH, Yao DH, Wang LY, Zhang L, Bai XL. Gut Microbiome-Mediated Alteration of Immunity, Inflammation, and Metabolism Involved in the Regulation of Non-alcoholic Fatty Liver Disease. Front Microbiol 2021; 12:761836. [PMID: 34795655 PMCID: PMC8593644 DOI: 10.3389/fmicb.2021.761836] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of end-stage liver disease, leading to a rapidly growing global public health burden. The term “gut microbiome (GM)” refers to the approximately 100 trillion microbial cells that inhabit the host’s gastrointestinal tract. There is increasing evidence that GM is involved in the pathogenesis of NAFLD and may be a potential target for intervention. To explore GM-based strategies for precise diagnosis and treatment of NAFLD, great efforts have been made to develop a comprehensive and in-depth understanding of the host–microbe interaction. This review evaluates this interaction critically, mainly considering the intricate regulation of the metabolism, immunity, and inflammatory status during the evolution of the disease pathogenesis, revealing roles for the GM in NAFLD by examining advances in potential mechanisms, diagnostics, and modulation strategies. Synopsis: Considering the intricate metabolic and immune/inflammatory homeostasis regulation, we evaluate the latest understanding of the host–microbe interaction and reveal roles for the gastrointestinal microbiome in NAFLD. Strategies targeting the gastrointestinal microbiome for the diagnosis and treatment of NAFLD are proposed.
Collapse
Affiliation(s)
- Li-Hong He
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Dun-Han Yao
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ling-Yun Wang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
234
|
Yang Y, Jiang X, Pandol SJ, Han YP, Zheng X. Green Plant Pigment, Chlorophyllin, Ameliorates Non-alcoholic Fatty Liver Diseases (NAFLDs) Through Modulating Gut Microbiome in Mice. Front Physiol 2021; 12:739174. [PMID: 34764881 PMCID: PMC8576288 DOI: 10.3389/fphys.2021.739174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Non-alcoholic fatty liver diseases (NAFLDs) along with metabolic syndrome and Type-2 diabetes (T2D) are increasingly prevalent worldwide. Without an effective resolution, simple hepatic steatosis may lead to non-alcoholic steatohepatitis (NASH), characterized by hepatocyte damage, chronic inflammation, necrosis, fatty degeneration, and cirrhosis. The gut microbiome is vital for metabolic homeostasis. Conversely, dysbiosis contributes to metabolic diseases including NAFLD. Specifically, diet composition is critical for the enterotype of gut microbiota. We reasoned that green pigment rich in vegetables may modulate the gut microbiome for metabolic homeostasis. In this study, C57BL/6 mice under a high fat diet (HFD) were treated with sodium copper chlorophyllin (CHL), a water-soluble derivative of chlorophyll, in drinking water. After 28 weeks of HFD feeding, liver steatosis was established accompanied by gut microbiota dysbiosis, intestinal impairment, endotoxemia, systemic inflammation, and insulin resistance. Administration of CHL effectively alleviated systemic and intestinal inflammation and maintained tight junction in the intestinal barrier. CHL rebalanced gut microbiota in the mice under high fat feeding and attenuated hepatic steatosis, insulin resistance, dyslipidemia, and reduced body weight. Fecal flora transplants from the CHL-treated mice ameliorated steatosis as well. Thus, dietary green pigment or the administration of CHL may maintain gut eubiosis and intestinal integrity to attenuate systemic inflammation and relieve NASH.
Collapse
Affiliation(s)
- You Yang
- Center for Diabetes and Metabolism Research, Division of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China.,The College of Life Sciences, Sichuan University, Chengdu, China
| | - Xile Jiang
- Department of Nutrition, West China College of Medicine, Sichuan University, Chengdu, China
| | | | - Yuan-Ping Han
- The College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiaofeng Zheng
- Center for Diabetes and Metabolism Research, Division of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
235
|
Wu Y, Li Q, Liu J, Liu Y, Xu Y, Zhang R, Yu Y, Wang Y, Yang C. Integrating Serum Metabolome and Gut Microbiome to Evaluate the Benefits of Lauric Acid on Lipopolysaccharide- Challenged Broilers. Front Immunol 2021; 12:759323. [PMID: 34721434 PMCID: PMC8554146 DOI: 10.3389/fimmu.2021.759323] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
Lauric acid (LA) is a crucial medium-chain fatty acid (MCFA) that has many beneficial effects on humans and animals. This study aimed to investigate the effects of LA on the intestinal barrier, immune functions, serum metabolism, and gut microbiota of broilers under lipopolysaccharide (LPS) challenge. A total of 384 one-day-old broilers were randomly divided into four groups, and fed with a basal diet, or a basal diet supplemented with 75 mg/kg antibiotic (ANT), or a basal diet supplemented with 1000 mg/kg LA. After 42 days of feeding, three groups were intraperitoneally injected with 0.5 mg/kg Escherichia coli- derived LPS (LPS, ANT+LPS and LA+LPS groups) for three consecutive days, and the control (CON) group was injected with the same volume of saline. Then, the birds were sacrificed. Results showed that LA pretreatment significantly alleviated the weight loss and intestinal mucosal injuries caused by LPS challenge. LA enhanced immune functions and inhibited inflammatory responses by upregulating the concentrations of immunoglobulins (IgA, IgM, and IgY), decreasing IL-6 and increasing IL-4 and IL-10. Metabolomics analysis revealed a significant difference of serum metabolites by LA pretreatment. Twenty-seven serum metabolic biomarkers were identified and mostly belong to lipids. LA also markedly modulated the pathway for sphingolipid metabolism, suggesting its ability to regulate lipid metabolism. Moreover,16S rRNA analysis showed that LA inhibited LPS-induced gut dysbiosis by altering cecal microbial composition (reducing Escherichia-Shigella, Barnesiella and Alistipes, and increasing Lactobacillus and Bacteroides), and modulating the production of volatile fatty acids (VFAs). Pearson’s correlation assays showed that alterations in serum metabolism and gut microbiota were strongly correlated to the immune factors; there were also strong correlations between serum metabolites and microbiota composition. The results highlight the potential of LA as a dietary supplement to combat bacterial LPS challenge in animal production and to promote food safety.
Collapse
Affiliation(s)
- Yanping Wu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Qing Li
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Jinsong Liu
- Institute of Animal Health Products, Zhejiang Vegamax Biotechnology Co., Ltd., Anji, China
| | - Yulan Liu
- Institute of Animal Health Products, Zhejiang Vegamax Biotechnology Co., Ltd., Anji, China
| | - Yinglei Xu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Ruiqiang Zhang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Yang Yu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Yongxia Wang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Caimei Yang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| |
Collapse
|
236
|
Yaskolka Meir A, Rinott E, Tsaban G, Zelicha H, Kaplan A, Rosen P, Shelef I, Youngster I, Shalev A, Blüher M, Ceglarek U, Stumvoll M, Tuohy K, Diotallevi C, Vrhovsek U, Hu F, Stampfer M, Shai I. Effect of green-Mediterranean diet on intrahepatic fat: the DIRECT PLUS randomised controlled trial. Gut 2021; 70:2085-2095. [PMID: 33461965 PMCID: PMC8515100 DOI: 10.1136/gutjnl-2020-323106] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/23/2020] [Accepted: 11/28/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To examine the effectiveness of green-Mediterranean (MED) diet, further restricted in red/processed meat, and enriched with green plants and polyphenols on non-alcoholic fatty liver disease (NAFLD), reflected by intrahepatic fat (IHF) loss. DESIGN For the DIRECT-PLUS 18-month randomized clinical trial, we assigned 294 participants with abdominal obesity/dyslipidaemia into healthy dietary guidelines (HDG), MED and green-MED weight-loss diet groups, all accompanied by physical activity. Both isocaloric MED groups consumed 28 g/day walnuts (+440 mg/day polyphenols provided). The green-MED group further consumed green tea (3-4 cups/day) and Mankai (a Wolffia globosa aquatic plant strain; 100 g/day frozen cubes) green shake (+1240 mg/day total polyphenols provided). IHF% 18-month changes were quantified continuously by proton magnetic resonance spectroscopy (MRS). RESULTS Participants (age=51 years; 88% men; body mass index=31.3 kg/m2; median IHF%=6.6%; mean=10.2%; 62% with NAFLD) had 89.8% 18-month retention-rate, and 78% had eligible follow-up MRS. Overall, NAFLD prevalence declined to: 54.8% (HDG), 47.9% (MED) and 31.5% (green-MED), p=0.012 between groups. Despite similar moderate weight-loss in both MED groups, green-MED group achieved almost double IHF% loss (-38.9% proportionally), as compared with MED (-19.6% proportionally; p=0.035 weight loss adjusted) and HDG (-12.2% proportionally; p<0.001). After 18 months, both MED groups had significantly higher total plasma polyphenol levels versus HDG, with higher detection of Naringenin and 2-5-dihydroxybenzoic-acid in green-MED. Greater IHF% loss was independently associated with increased Mankai and walnuts intake, decreased red/processed meat consumption, improved serum folate and adipokines/lipids biomarkers, changes in microbiome composition (beta-diversity) and specific bacteria (p<0.05 for all). CONCLUSION The new suggested strategy of green-Mediterranean diet, amplified with green plant-based proteins/polyphenols as Mankai, green tea, and walnuts, and restricted in red/processed meat can double IHF loss than other healthy nutritional strategies and reduce NAFLD in half. TRIAL REGISTRATION NUMBER NCT03020186.
Collapse
Affiliation(s)
- Anat Yaskolka Meir
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ehud Rinott
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gal Tsaban
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel,Division of Cardiology, Soroka Medical Center, Beer Sheva, Israel
| | - Hila Zelicha
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Kaplan
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Philip Rosen
- Division of Diagnostic and Interventional Imaging, Soroka University Medical Center, Beer Sheva, Israel
| | - Ilan Shelef
- Division of Diagnostic and Interventional Imaging, Soroka University Medical Center, Beer Sheva, Israel
| | - Ilan Youngster
- Pediatric Division and Center for Microbiome Research, Shamir Medical Center, Be’er Ya’akov, Israel
| | - Aryeh Shalev
- Division of Cardiology, Soroka Medical Center, Beer Sheva, Israel
| | - Matthias Blüher
- Medical Department III – Endocrinology, Nephrology, Rheumatology, Leipzig University, Leipzig, Germany
| | - Uta Ceglarek
- Institute for Laboratory Medicine, Leipzig University, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III – Endocrinology, Nephrology, Rheumatology, Leipzig University, Leipzig, Germany
| | - Kieran Tuohy
- Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michelle All'Adige, Italy
| | - Camilla Diotallevi
- Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michelle All'Adige, Italy,Faculty of Science and Technology, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Urska Vrhovsek
- Faculty of Science and Technology, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Frank Hu
- Department of Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Meir Stampfer
- Department of Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Iris Shai
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel .,Department of Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
237
|
Moore TM, Terrazas A, Strumwasser AR, Lin AJ, Zhu X, Anand ATS, Nguyen CQ, Stiles L, Norheim F, Lang JM, Hui ST, Turcotte LP, Zhou Z. Effect of voluntary exercise upon the metabolic syndrome and gut microbiome composition in mice. Physiol Rep 2021; 9:e15068. [PMID: 34755487 PMCID: PMC8578881 DOI: 10.14814/phy2.15068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022] Open
Abstract
The metabolic syndrome is a cluster of conditions that increase an individual's risk of developing diseases. Being physically active throughout life is known to reduce the prevalence and onset of some aspects of the metabolic syndrome. Furthermore, previous studies have demonstrated that an individual's gut microbiome composition has a large influence on several aspects of the metabolic syndrome. However, the mechanism(s) by which physical activity may improve metabolic health are not well understood. We sought to determine if endurance exercise is sufficient to prevent or ameliorate the development of the metabolic syndrome and its associated diseases. We also analyzed the impact of physical activity under metabolic syndrome progression upon the gut microbiome composition. Utilizing whole-body low-density lipoprotein receptor (LDLR) knockout mice on a "Western Diet," we show that long-term exercise acts favorably upon glucose tolerance, adiposity, and liver lipids. Exercise increased mitochondrial abundance in skeletal muscle but did not reduce liver fibrosis, aortic lesion area, or plasma lipids. Lastly, we observed several changes in gut bacteria and their novel associations with metabolic parameters of clinical importance. Altogether, our results indicate that exercise can ameliorate some aspects of the metabolic syndrome progression and alter the gut microbiome composition.
Collapse
Affiliation(s)
- Timothy M. Moore
- Division of CardiologyDepartment of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Anthony Terrazas
- Division of CardiologyDepartment of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Alexander R. Strumwasser
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Amanda J. Lin
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Xiaopeng Zhu
- Division of Pediatric EndocrinologyDepartment of Pediatrics UCLA Children's Discovery and Innovation InstituteDepartment of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
- Present address:
Department of Endocrinology and Metabolism. Zhongshan HospitalFudan UniversityShanghaiP.R.China
| | - Akshay T. S. Anand
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Christina Q. Nguyen
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Frode Norheim
- Department of Human GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- Present address:
Department of NutritionFaculty of MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jennifer M. Lang
- Department of Human GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Simon T. Hui
- Division of CardiologyDepartment of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Lorraine P. Turcotte
- Department of Biological SciencesDana & David Dornsife College of Letters, Arts, and SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and HypertensionUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
238
|
Gu X, Lu Q, Zhang C, Tang Z, Chu L. Clinical Application and Progress of Fecal Microbiota Transplantation in Liver Diseases: A Review. Semin Liver Dis 2021; 41:495-506. [PMID: 34261137 PMCID: PMC8492191 DOI: 10.1055/s-0041-1732319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The human gut harbors a dense and highly diverse microbiota of approximately 1,000 bacterial species. The interaction between the host and gut bacteria strongly influences human health. Numerous evidence suggest that intestinal flora imbalance is closely associated with the development and treatment of liver diseases, including acute liver injury and chronic liver diseases (cirrhosis, autoimmune liver disease, and fatty liver). Therefore, regulating the gut microbiota is expected to be a new method for the adjuvant treatment of liver diseases. Fecal microbiota transplantation (FMT) is defined as the transplantation of gut microbiota from healthy donors to sick patients via the upper or lower gastrointestinal route to restore the normal intestinal balance. In this study, we briefly review the current research on the gut microbiota and its link to liver diseases and then summarize the evidence to elucidate the clinical application and development of FMT in liver disease treatment.
Collapse
Affiliation(s)
- Xinpei Gu
- Department of Human Anatomy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Qin Lu
- Department of Prescription Science, School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chengcheng Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhewei Tang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Address for correspondence Liuxi Chu, PhD Institute of Child Development and Education, School of Biological Sciences and Medical Engineering, Southeast UniversityNanjing - 210096China
| | - Liuxi Chu
- Institute of Child Development and Education, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
239
|
Logan IE, Shulzhenko N, Sharpton TJ, Bobe G, Liu K, Nuss S, Jones ML, Miranda CL, Vasquez-Perez S, Pennington JM, Leonard SW, Choi J, Wu W, Gurung M, Kim JP, Lowry MB, Morgun A, Maier CS, Stevens JF, Gombart AF. Xanthohumol Requires the Intestinal Microbiota to Improve Glucose Metabolism in Diet-Induced Obese Mice. Mol Nutr Food Res 2021; 65:e2100389. [PMID: 34496124 PMCID: PMC8571065 DOI: 10.1002/mnfr.202100389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/27/2021] [Indexed: 12/14/2022]
Abstract
SCOPE The polyphenol xanthohumol (XN) improves dysfunctional glucose and lipid metabolism in diet-induced obesity animal models. Because XN changes intestinal microbiota composition, the study hypothesizes that XN requires the microbiota to mediate its benefits. METHODS AND RESULTS To test the hypothesis, the study feeds conventional and germ-free male Swiss Webster mice either a low-fat diet (LFD, 10% fat derived calories), a high-fat diet (HFD, 60% fat derived calories), or a high-fat diet supplemented with XN at 60 mg kg-1 body weight per day (HXN) for 10 weeks, and measure parameters of glucose and lipid metabolism. In conventional mice, the study discovers XN supplementation decreases plasma insulin concentrations and improves Homeostatic Model Assessment of Insulin Resistance (HOMA-IR). In germ-free mice, XN supplementation fails to improve these outcomes. Fecal sample 16S rRNA gene sequencing analysis suggests XN supplementation changes microbial composition and dramatically alters the predicted functional capacity of the intestinal microbiota. Furthermore, the intestinal microbiota metabolizes XN into bioactive compounds, including dihydroxanthohumol (DXN), an anti-obesogenic compound with improved bioavailability. CONCLUSION XN requires the intestinal microbiota to mediate its benefits, which involves complex diet-host-microbiota interactions with changes in both microbial composition and functional capacity. The study results warrant future metagenomic studies which will provide insight into complex microbe-microbe interactions and diet-host-microbiota interactions.
Collapse
Affiliation(s)
- Isabelle E Logan
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | | | - Thomas J Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA
- Department of Statistics, Oregon State University, Corvallis, OR, 97331, USA
| | - Gerd Bobe
- Department of Animal Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Kitty Liu
- Department of Biochemistry and Biophysics, Corvallis, OR, 97331, USA
| | - Stephanie Nuss
- Carlson College of Veterinary Medicine, Corvallis, OR, 97331, USA
| | - Megan L Jones
- Department of Biochemistry and Biophysics, Corvallis, OR, 97331, USA
| | - Cristobal L Miranda
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | | | - Jamie M Pennington
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Scott W Leonard
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Wenbin Wu
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Manoj Gurung
- Carlson College of Veterinary Medicine, Corvallis, OR, 97331, USA
| | - Joyce P Kim
- Department of Biochemistry and Biophysics, Corvallis, OR, 97331, USA
| | - Malcolm B Lowry
- Department of Microbiology, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Andrey Morgun
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, 97331, USA
| | - Jan F Stevens
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Adrian F Gombart
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
240
|
Zhang X, Shi L, Chen R, Zhao Y, Ren D, Yang X. Chlorogenic acid inhibits trimethylamine- N-oxide formation and remodels intestinal microbiota to alleviate liver dysfunction in high L-carnitine feeding mice. Food Funct 2021; 12:10500-10511. [PMID: 34558577 DOI: 10.1039/d1fo01778k] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
High L-carnitine ingestion has been shown to cause liver injury, mechanically due to an elevated circulating level of trimethylamine-N-oxide (TMAO), a gut microbiota-derived metabolite from L-carnitine. This study aimed to investigate whether chlorogenic acid (CGA), a health-promoting polyphenol, could inhibit TMAO formation and thereafter might prevent L-carnitine-induced liver injury in mice. Feeding of mice with 3% L-carnitine in drinking water increased the serum and urinary levels of TMAO (p < 0.01 vs. Normal), whereas the serum and urinary TMAO formation was sharply reduced by CGA administration (p < 0.01). At the phylum level, CGA inhibited the L-carnitine-induced increase in the abundance of Firmicutes and Proteobacteria, while it promoted Bacteroidetes. At the genus level, CGA notably increased the abundance of Akkermansia and Bacteroides, but reduced the population of Erysipelatoclostridium, Faecalibaculum and Erysipelotrichaceae in high L-carnitine feeding mice. Meanwhile, CGA caused strong inhibition against the increase of liver injury markers (i.e. AST, ALT and ALP), hepatic inflammatory cytokines (i.e. IL-1, IL-6, TNF-α and TNF-β) and dyslipidemia (i.e. TC, TG, LDL-C and HDL-C) in L-carnitine-fed mice (p < 0.05). These findings suggest that CGA holds great potential to alleviate liver dysfunction induced by high L-carnitine ingestion. The beneficial effect might be attributed to the protection against TMAO formation and the improvement of the health-promoting gut microbiota, as well as the antioxidant and anti-inflammatory properties of CGA.
Collapse
Affiliation(s)
- Xiangnan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Lin Shi
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Rui Chen
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
241
|
Won SM, Seo MJ, Kwon MJ, Park KW, Yoon JH. Oral Administration of Latilactobacillus sakei ADM14 Improves Lipid Metabolism and Fecal Microbiota Profile Associated With Metabolic Dysfunction in a High-Fat Diet Mouse Model. Front Microbiol 2021; 12:746601. [PMID: 34690997 PMCID: PMC8527011 DOI: 10.3389/fmicb.2021.746601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Effects of Latilactobacillus sakei ADM14 on changes in lipid metabolism and fecal microbiota composition were studied in high-fat diet (HFD) mouse model. The mice were divided into three groups: normal diet (ND), high-fat diet (HD), and HFD plus L. sakei ADM14 (HDA). Oral administration of L. sakei ADM14 daily for 10weeks decreased body weight gain, fat tissue mass, and liver weight in mice and reduced the size of histologically stained liver adipocytes. In addition, serum total cholesterol, triglycerides, and blood glucose decreased significantly. Latilactobacillus sakei ADM14 regulated the expression of genes related to lipid metabolism in epididymal adipose tissue and liver and induced changes in the composition of fecal microbiota, thereby improving energy harvests and changing metabolic disorder-related taxa. A significant decrease (p<0.05) in the Firmicutes to Bacteroidetes ratio was found in the HDA group compared to the HD group, particularly due to the difference in the relative abundance of the Bacteroidetes between the two groups over 10weeks. Differences in proportions of some taxa reported to have correlation with obesity were also found between HD and HDA groups. These results suggest that L. sakei ADM14 can have a positive effect on metabolic disorders such as obesity and fatty liver through effective regulation of host lipid metabolism and gut microbiota.
Collapse
Affiliation(s)
- Sung-Min Won
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Min Ju Seo
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Min Ju Kwon
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Kye Won Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Jung-Hoon Yoon
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
242
|
Ji Y, Lee H, Kaura S, Yip J, Sun H, Guan L, Han W, Ding Y. Effect of Bariatric Surgery on Metabolic Diseases and Underlying Mechanisms. Biomolecules 2021; 11:1582. [PMID: 34827579 PMCID: PMC8615605 DOI: 10.3390/biom11111582] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Obesity is a highly prevalent public health concern, attributed to multifactorial causes and limited in treatment options. Several comorbidities are closely associated with obesity such as the development of type 2 diabetes mellitus (T2DM), cardiovascular and cerebrovascular diseases, and nonalcoholic fatty liver disease (NAFLD). Bariatric surgery, which can be delivered in multiple forms, has been remarked as an effective treatment to decrease the prevalence of obesity and its associated comorbidities. The different types of bariatric surgery create a variety of new pathways for food to metabolize in the body and truncate the stomach's caliber. As a result, only a small quantity of food is tolerated, and the body mass index noticeably decreases. This review describes the improvements of obesity and its comorbidities following bariatric surgery and their mechanism of improvement. Additionally, endocrine function improvements after bariatric surgery, which contributes to the patients' health improvement, are described, including the role of glucagon-like peptide-1 (GLP-1), fibroblast growth factors 19 and 21 (FGF-19, FGF-21), and pancreatic peptide YY (PYY). Lastly, some of the complications of bariatric surgery, including osteoporosis, iron deficiency/anemia, and diarrhea, as well as their potential mechanisms, are described.
Collapse
Affiliation(s)
- Yu Ji
- Department of General Surgery, Beijing Luhe Clinical Institute, Capital Medical University, Beijing 101149, China;
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.L.); (S.K.); (L.G.); (Y.D.)
- John D. Dingell VA Medical Center, 4646 John R Street (11R), Detroit, MI 48201, USA
| | - Hangil Lee
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.L.); (S.K.); (L.G.); (Y.D.)
| | - Shawn Kaura
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.L.); (S.K.); (L.G.); (Y.D.)
| | - James Yip
- Department of General Surgery, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Hao Sun
- Central Laboratory, Beijing Luhe Clinical Institute, Capital Medical University, Beijing 101149, China;
| | - Longfei Guan
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.L.); (S.K.); (L.G.); (Y.D.)
- John D. Dingell VA Medical Center, 4646 John R Street (11R), Detroit, MI 48201, USA
- Department of General Surgery, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Wei Han
- Department of General Surgery, Beijing Luhe Clinical Institute, Capital Medical University, Beijing 101149, China;
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.L.); (S.K.); (L.G.); (Y.D.)
| |
Collapse
|
243
|
Houron C, Ciocan D, Trainel N, Mercier-Nomé F, Hugot C, Spatz M, Perlemuter G, Cassard AM. Gut Microbiota Reshaped by Pectin Treatment Improves Liver Steatosis in Obese Mice. Nutrients 2021; 13:3725. [PMID: 34835981 PMCID: PMC8621973 DOI: 10.3390/nu13113725] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Pectin, a soluble fiber, improves non-alcoholic fatty-liver disease (NAFLD), but its mechanisms are unclear. We aimed to investigate the role of pectin-induced changes in intestinal microbiota (IM) in NAFLD. We recovered the IM from mice fed a high-fat diet, treated or not with pectin, to perform a fecal microbiota transfer (FMT). Mice fed a high-fat diet, which induces NAFLD, were treated with pectin or received a fecal microbiota transfer (FMT) from mice treated with pectin before (preventive FMT) or after (curative FMT) being fed a high-fat diet. Pectin prevented the development of NAFLD, induced browning of adipose tissue, and modified the IM without increasing the abundance of proteobacteria. Preventive FMT also induced browning of white adipose tissue but did not improve liver steatosis, in contrast to curative FMT, which induced an improvement in steatosis. This was associated with an increase in the concentration of short-chain fatty acids (SCFAs), in contrast to preventive FMT, which induced an increase in the concentration of branched SCFAs. Overall, we show that the effect of pectin may be partially mediated by gut bacteria.
Collapse
Affiliation(s)
- Camille Houron
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
| | - Dragos Ciocan
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
- AP-HP, Hepato-Gastroenterology and Nutrition, Hôpital Antoine-Béclère, 92140 Clamart, France
| | - Nicolas Trainel
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
| | - Françoise Mercier-Nomé
- Université Paris-Saclay, Inserm, CNRS, Institut Paris Saclay d’Innovation Thérapeutique, 5 rue J.B. Clément, 92296 Châtenay-Malabry, France;
| | - Cindy Hugot
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
| | - Madeleine Spatz
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
| | - Gabriel Perlemuter
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
- AP-HP, Hepato-Gastroenterology and Nutrition, Hôpital Antoine-Béclère, 92140 Clamart, France
| | - Anne-Marie Cassard
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, 32 rue des carnets, 92140 Clamart, France; (C.H.); (D.C.); (N.T.); (C.H.); (M.S.); (G.P.)
| |
Collapse
|
244
|
Li W, Deng M, Gong J, Zhang X, Ge S, Zhao L. Sodium Acetate Inhibit TGF-β1-Induced Activation of Hepatic Stellate Cells by Restoring AMPK or c-Jun Signaling. Front Nutr 2021; 8:729583. [PMID: 34660662 PMCID: PMC8515000 DOI: 10.3389/fnut.2021.729583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are crucial gut microbial metabolites that play a major role in the occurrence and development of hepatic fibrosis (HF). However, the effect of SCFAs on hepatic stellate cells (HSCs), the major pro-fibrogenic cells, is yet undefined. In this study, the effects of three major SCFAs (acetate, propionate, and butyrate) were assessed on the activation of HSCs. LX2 cells were activated with TGF-β1 and treated with sodium acetate (NaA), sodium propionate (NaP), or sodium butyrate (NaB). SCFA treatment significantly reduced the protein levels of α-SMA and the phosphorylation of Smad2 and decreased the mRNA expression of Acta2/Col1a1/Fn in cells compared to the TGF-β1 treatment. Among the three SCFAs, NaA revealed the best efficacy at alleviating TGF-β1-induced LX2 cell activation. Additionally, acetate accumulated in the cells, and G protein-coupled receptor (GPR) 43 silencing did not have any impact on the inhibition of LX2 cell activation by NaA. These findings indicated that NaA enters into the cells to inhibit LX2 cell activation independent of GPR43. The results of phosphokinase array kit and Western blot indicated that NaA increased the AMP-activated protein kinase (AMPK) activation and reduced the phosphorylation of c-Jun in cultured LX2 cells, and siRNA-peroxisome proliferator-activated receptor (PPAR) -γ abolished the inhibitory effects of NaA against TGF-β1-induced LX2 cell activation. In conclusion, this study showed that NaA inhibited LX2 cell activation by activating the AMPK/PPARγ and blocking the c-Jun signaling pathways. Thus, SCFAs might represent a novel and viable approach for alleviating HF.
Collapse
Affiliation(s)
- Weiwei Li
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Mingjuan Deng
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Jiahui Gong
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoying Zhang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
| | - Shaoyang Ge
- Hebei Engineering Research Center of Animal Product, Sanhe, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
245
|
Shi J, Yang Y, Xu W, Cai H, Wu J, Long J, Cai Q, Zheng W, Flynn CR, Shu XO, Yu D. Sex-Specific Associations between Gut Microbiome and Non-Alcoholic Fatty Liver Disease among Urban Chinese Adults. Microorganisms 2021; 9:microorganisms9102118. [PMID: 34683439 PMCID: PMC8537656 DOI: 10.3390/microorganisms9102118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has been linked to altered gut microbiome; however, evidence from large population-based studies is limited. We compared gut microbiome profiles of 188 male and 233 female NAFLD cases with 571 male and 567 female controls from two longitudinal studies of urban Chinese adults. History of NAFLD was assessed during surveys administered in 2004-2017. Microbiota were assessed using 16S rRNA sequencing of stool samples collected in 2015-2018. Associations of NAFLD with microbiome diversity and composition were evaluated by generalized linear or logistic regression models. Compared with controls, male cases had lower microbial α-diversity, higher abundance of genera Dialister and Streptococcus and Bifidobacterium species, lower abundance of genus Phascolarctobacterium, and lower prevalence of taxa including order RF39 (all p < 0.05). In contrast, female cases had higher α-diversity, higher abundance of genus Butyricimonas and a family of order Clostridiales, lower abundance of Dialister and Bifidobacterium species, and higher prevalence of RF39. Significant NAFLD-sex interactions were found for α-diversity and above taxa (all false discovery rate < 0.1). In conclusion, we observed sex-specific gut microbiome features related to history of NAFLD. Further studies are needed to validate our findings and evaluate the health effects of NAFLD-related gut microbiota.
Collapse
Affiliation(s)
- Jiajun Shi
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Yaohua Yang
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Wanghong Xu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai 200433, China;
| | - Hui Cai
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Jie Wu
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Jirong Long
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Qiuyin Cai
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Wei Zheng
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Charles R. Flynn
- Department of Surgery, Division of General Surgery, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA;
| | - Xiao-Ou Shu
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
| | - Danxia Yu
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Suite 600, Nashville, TN 37232, USA; (J.S.); (Y.Y.); (H.C.); (J.W.); (J.L.); (Q.C.); (W.Z.); (X.-O.S.)
- Correspondence: ; Tel.: +1-615-936-7389; Fax: +1-615-936-8291
| |
Collapse
|
246
|
Park E, Jeong JJ, Won SM, Sharma SP, Gebru YA, Ganesan R, Gupta H, Suk KT, Kim DJ. Gut Microbiota-Related Cellular and Molecular Mechanisms in the Progression of Nonalcoholic Fatty Liver Disease. Cells 2021; 10:cells10102634. [PMID: 34685614 PMCID: PMC8534099 DOI: 10.3390/cells10102634] [Citation(s) in RCA: 12] [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: 08/27/2021] [Revised: 09/25/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common and increasing liver diseases worldwide. NAFLD is a term that involves a variety of conditions such as fatty liver, steatohepatitis, or fibrosis. Gut microbiota and its products have been extensively studied because of a close relation between NAFLD and microbiota in pathogenesis. In the progression of NAFLD, various microbiota-related molecular and cellular mechanisms, including dysbiosis, leaky bowel, endotoxin, bile acids enterohepatic circulation, metabolites, or alcohol-producing microbiota, are involved. Currently, diagnosis and treatment techniques using these mechanisms are being developed. In this review, we will introduce the microbiota-related mechanisms in the progression of NAFLD and future directions will be discussed.
Collapse
|
247
|
Flores‐Guerrero JL, Post A, van Dijk PR, Connelly MA, Garcia E, Navis G, Bakker SJL, Dullaart RPF. Circulating trimethylamine-N-oxide is associated with all-cause mortality in subjects with nonalcoholic fatty liver disease. Liver Int 2021; 41:2371-2382. [PMID: 33993608 PMCID: PMC8518486 DOI: 10.1111/liv.14963] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Trimethylamine-N-oxide (TMAO), a gut microbiota-liver metabolite, has been associated with cardiometabolic disease. However, whether TMAO is associated with nonalcoholic fatty liver disease (NAFLD) and NAFLD-related health outcomes remains unclear. We aimed to investigate the association of TMAO with NAFLD and to assess the extent to which the association of TMAO with all-cause mortality is dependent on the presence of NAFLD in the general population. METHODS We included 5292 participants enrolled in the Prevention of Renal and Vascular End-stage Disease (PREVEND) cohort study. Cox proportional-hazards regression analyses were performed to study the association of TMAO with all-cause mortality in subjects with and without a fatty liver index (FLI) ≥60, which was used as a proxy of NAFLD. RESULTS During a median follow-up of 8.2 years, 307 subjects died, of whom 133 were classified with NAFLD. TMAO was positively and independently associated with baseline FLI (Std β 0.08, 95% CI 0.05, 0.11, P < .001). Higher TMAO was associated with increased risk of all-cause mortality in subjects with NAFLD, in crude analysis (hazard ratio [HR] per 1 SD, 2.55, 95% CI 1.60, 4.05, P < .001) and after full adjustment (adj HR 1.90, 95% CI 1.18, 3.04, P = .008). Such an association was not present in subjects without NAFLD (crude HR 1.14, 95% CI 0.81, 1.71, P = .39; adj HR 0.95, 95% CI 0.65, 1.39, P = .78). CONCLUSION This prospective study revealed that plasma concentrations of TMAO were associated with all-cause mortality in subjects with NAFLD, independently of traditional risk factors.
Collapse
Affiliation(s)
- Jose L. Flores‐Guerrero
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Adrian Post
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Peter R. van Dijk
- Department of Internal MedicineDivision of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | | | - Erwin Garcia
- Laboratory Corporation of America Holdings (Labcorp)MorrisvilleNCUSA
| | - Gerjan Navis
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Stephan J. L. Bakker
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Robin P. F. Dullaart
- Department of Internal MedicineDivision of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
248
|
Jiang J, Xiong J, Ni J, Chen C, Wang K. Live Combined B. subtilis and E. faecium Alleviate Liver Inflammation, Improve Intestinal Barrier Function, and Modulate Gut Microbiota in Mice with Non-Alcoholic Fatty Liver Disease. Med Sci Monit 2021; 27:e931143. [PMID: 34482357 PMCID: PMC8428156 DOI: 10.12659/msm.931143] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a chronic, progressive liver disease with an increasing incidence rate. This study investigated the protective effects of live combined Bacillus subtilis and Enterococcus faecium (LCBE) on NAFLD, and its possible mechanisms. Material/Methods Five-week-old C57BL/6 mice were randomly divided into 3 groups: chow, HFD, and HFD+LCBE groups. The levels of serum biochemical markers, glucose tolerance, insulin, the inflammatory cytokines IL-1β, IL-6, and TNF-α, LPS, and histological staining were measured using commercial kits. qPCR was used to examine the mRNA expression levels of inflammatory cytokines in the liver. Western blotting was used to determine the protein levels of TLR4, NF-κB p65, PPAR-α, and CPT-1 in the liver, and occludin and Claudin1 in the intestine. The intestinal flora of the mice was analyzed by high-throughput sequencing of the V3–V4 region of 16S rDNA. Results LCBE significantly lowered the body weight, liver/body weight ratio, and serum glucose level, and increased the serum insulin level in NAFLD mice. In addition, LCBE treatment improved the liver function and lipid profile, decreased the levels of LPS and inflammatory cytokines, and downregulated the expression of TLR4 and NF-κB p65. Moreover, LCBE enhanced the intestinal barrier function by increasing the expression of occludin and Claudin1. Furthermore, LCBE modulated the composition of the gut microbiota by reducing the Firmicutes to Bacteroidetes ratio, and the proportion of inflammation-related and LPS-producing bacteria, thus re-arranging the structure of the gut microbiota. Conclusions LCBE protects against NAFLD by alleviating inflammation, restoring the intestinal barrier, and modulating gut microbiota composition.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Jie Xiong
- Department of Gastroenterology and Hepatology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Jianbo Ni
- Department of Gastroenterology and Hepatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Congying Chen
- Department of Gastroenterology and Hepatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Kezhou Wang
- Department of Pathology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
249
|
Lécuyer E, Le Roy T, Gestin A, Lacombe A, Philippe C, Ponnaiah M, Huré JB, Fradet M, Ichou F, Boudebbouze S, Huby T, Gautier E, Rhimi M, Maguin E, Kapel N, Gérard P, Venteclef N, Garlatti M, Chassaing B, Lesnik P. Tolerogenic Dendritic Cells Shape a Transmissible Gut Microbiota That Protects From Metabolic Diseases. Diabetes 2021; 70:2067-2080. [PMID: 34078628 PMCID: PMC8576430 DOI: 10.2337/db20-1177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/26/2021] [Indexed: 11/13/2022]
Abstract
Excess chronic contact between microbial motifs and intestinal immune cells is known to trigger a low-grade inflammation involved in many pathologies such as obesity and diabetes. The important skewing of intestinal adaptive immunity in the context of diet-induced obesity (DIO) is well described, but how dendritic cells (DCs) participate in these changes is still poorly documented. To address this question, we challenged transgenic mice with enhanced DC life span and immunogenicity (DChBcl-2 mice) with a high-fat diet. Those mice display resistance to DIO and metabolic alterations. The DIO-resistant phenotype is associated with healthier parameters of intestinal barrier function and lower intestinal inflammation. DChBcl-2 DIO-resistant mice demonstrate a particular increase in tolerogenic DC numbers and function, which is associated with strong intestinal IgA, T helper 17, and regulatory T-cell immune responses. Microbiota composition and function analyses reveal that the DChBcl-2 mice microbiota is characterized by lower immunogenicity and an enhanced butyrate production. Cohousing experiments and fecal microbial transplantations are sufficient to transfer the DIO resistance status to wild-type mice, demonstrating that maintenance of DCs' tolerogenic ability sustains a microbiota able to drive DIO resistance. The tolerogenic function of DCs is revealed as a new potent target in metabolic disease management.
Collapse
Affiliation(s)
- Emelyne Lécuyer
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
| | - Tiphaine Le Roy
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
- Sorbonne/INSERM, Nutrition et obésités: approches systémiques (nutriOmics), Hôpital Pitié- Salpêtrière, Paris, France
| | - Aurélie Gestin
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Amélie Lacombe
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Catherine Philippe
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Maharajah Ponnaiah
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Baptiste Huré
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
| | - Magali Fradet
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Farid Ichou
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Samira Boudebbouze
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Thierry Huby
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Emmanuel Gautier
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Moez Rhimi
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Emmanuelle Maguin
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM UMRS 1139, Université de Paris, Paris, France
| | - Philippe Gérard
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nicolas Venteclef
- INSERM, Cordeliers Research Centre, Immunity and Metabolism of Diabetes (IMMEDIAB), Université de Paris, Paris, France
| | - Michèle Garlatti
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
| | - Benoit Chassaing
- Neuroscience Institute and Institute for Biomedical Sciences, Georgia State University, Atlanta, GA
- INSERM, U1016, Team "Mucosal microbiota in chronic inflammatory diseases," Paris, France
| | - Philippe Lesnik
- INSERM, UMRS 1166 Institute of Cardiometabolism and Nutrition, Sorbonne Université, Paris, France
- Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
250
|
Li X, Jiang L, Xia Q, Zeng X, Wang W, Pan D, Wu Z. Effects of novel flavonoid-enriched yogurt on the diversity of intestinal microbiota in mice. Braz J Microbiol 2021; 52:2287-2298. [PMID: 34449069 DOI: 10.1007/s42770-021-00598-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022] Open
Abstract
Soy isoflavone glycoside cannot be effectively absorbed by the human intestinal tract, but probiotics with related hydrolases can transform it into aglycone to promote its absorption. In this study, a novel flavonoid-enriched yogurt was developed using an isolated β-glucosidase-producing strain (Lactiplantibacillus plantarum GY). The flavonoid aglycone-enhanced yogurt was fed to ICR mice for 21 days, and its effects were observed. The yogurt can affect the gut microbial diversity of mice, especially increasing the abundance of Parasutterella, the Bacteroidales S24-7 group, and Phascolarctobacterium in the intestinal tract of mice. Meanwhile, the ratio of Bacteroidetes/Firmicutes in the intestinal tract of mice fed with the flavonoid aglycone-enriched yogurt increased. The difference in the content of butyric acid between the L-GY + IS and the control groups was significant (P < 0.05). Therefore, milk fermentation with β-glucosidase-producing strains is a promising approach for developing flavonoid glycoside-enriched yogurt products.
Collapse
Affiliation(s)
- Xiefei Li
- State Key Laboratory for Managing Biotic and Chemical Threats To the Quality and Safety of Agro-Products, Ningbo University, 315211, Ningbo, Zhejiang, People's Republic of China.,Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Lan Jiang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Qiang Xia
- State Key Laboratory for Managing Biotic and Chemical Threats To the Quality and Safety of Agro-Products, Ningbo University, 315211, Ningbo, Zhejiang, People's Republic of China.,Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Xiaoqun Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats To the Quality and Safety of Agro-Products, Ningbo University, 315211, Ningbo, Zhejiang, People's Republic of China.,Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Weijun Wang
- Zhejiang Yiming Food Company, Wenzhou, Zhejiang, People's Republic of China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats To the Quality and Safety of Agro-Products, Ningbo University, 315211, Ningbo, Zhejiang, People's Republic of China. .,Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China. .,National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, Jiangxi, 330022, People's Republic of China.
| | - Zhen Wu
- State Key Laboratory for Managing Biotic and Chemical Threats To the Quality and Safety of Agro-Products, Ningbo University, 315211, Ningbo, Zhejiang, People's Republic of China. .,Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Ningbo University, Ningbo, Zhejiang, People's Republic of China.
| |
Collapse
|