1
|
Liu J, Lu L, Song H, Liu S, Liu G, Lou B, Shi W. Effects of triclosan on lipid metabolism and underlying mechanisms in the cyprinid fish Squalidus argentatus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175627. [PMID: 39168348 DOI: 10.1016/j.scitotenv.2024.175627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
The ubiquitous presence of the disinfectant triclosan (TCS) has raised global concerns regarding its potential threat to aquatic organisms. However, the effects of TCS on lipid metabolism in fish and its underlying mechanisms remain unclear. This study investigated the effect of environmentally relevant levels of TCS on the lipid metabolism in the cyprinid fish Squalidus argentatus. Our results showed that the lipid metabolism in the cyprinid fish S. argentatus was perturbed by 28-day exposure to TCS, as evidenced by higher levels of lipid accumulation in both the liver and blood. To elucidate the mechanisms underlying toxicity, we evaluated oxidative stress, inflammatory status, and lipase activity in the liver. Our findings indicated increased ROS-specific fluorescence intensity, superoxide dismutase (SOD) activity, and malondialdehyde (MDA) content in the livers of S. argentatus exposed to TCS, suggesting oxidative damage. Additionally, TCS treatment induced the production of proinflammatory cytokines in the liver of S. argentatus exposed to TCS, which suppressed hepatic lipase activity. Intestinal tissue morphology, inflammation, and blood lipopolysaccharide (LPS) levels were also examined. Significant increases in goblet cell count and MDA levels were observed in the intestinal tract. After 28 days of TCS exposure, the serum LPS levels were significantly elevated. 16S rRNA sequencing was conducted to analyze the effects of TCS on the diversity and composition of the intestinal microbiota. Transcriptomic analysis was performed to reveal global molecular alterations following TCS exposure. In conclusion, our results indicate that TCS may disrupt the lipid metabolism in S. argentatus by (i) inducing hepatic oxidative stress and inflammation, which suppress lipoprotein lipase activity, (ii) affecting the production of beneficial metabolites and endotoxins by dysregulating gut microbiota composition, and (iii) altering the expression levels of lipid metabolism-related pathways.
Collapse
Affiliation(s)
- Jindian Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Lingzheng Lu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hongjian Song
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Shuai Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Bao Lou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Gribsholt SB, Szépligeti SK, Sørensen HT, Mueller NT, Karagas MR, Ehrenstein V. Prenatal and Early-Life Anti-Infectives and Obesity at Age 7 Years. Pharmacoepidemiol Drug Saf 2024; 33:e70055. [PMID: 39533505 DOI: 10.1002/pds.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE To examine associations of prenatal and early-life anti-infective exposures with obesity at 7 years. METHODS In this nationwide, registry-based, prevalence study, we included all children with an anthropometric assessment at age 7 years from the Children's Database and linked their data with Danish population-based registries from 2001 to 2018. We defined exposure to anti-infectives (anti-bacterials, anti-virals, and anti-fungals) by outpatient dispensings or by infection diagnoses at hospital encounters. The earliest date defined the exposure timing category: prenatal (-9 months- < 0 months), infancy (0- < 2 years), and early childhood (2- < 5 years). We computed prevalence ratios (aPRs) for associations of anti-infective exposure with obesity prevalence at 7 years of age, adjusting for maternal and perinatal factors. RESULTS We included 460 363 children (51% boys). Prevalence of obesity at 7 years of age was 38% higher (aPR = 1.38, 95% confidence interval (CI): 1.27-1.49) among children exposed to any anti-infective, 21% higher (aPR = 1.21, 95% CI: 1.12-1.31) among children exposed to anti-infectives in infancy, and 14% higher (aPR = 1.14, 95% CI: 1.03-1.26) among children exposed to anti-infectives in early childhood. Exposure to anti-bacterials was associated with obesity in a similar time-dependent pattern [prenatal: aPR = 1.39 (95% CI: 1.29-1.50), infancy: aPR = 1.21 (95% CI: 1.12-1.30), and early childhood: aPR = 1.14 (95% CI: 1.03-1.25)]. For anti-virals and anti-fungals, exposure during infancy and early childhood was associated with larger aPRs than prenatal exposure. Furthermore, obesity prevalence increased monotonically with number of the anti-infective prescriptions. CONCLUSION These findings suggest that prenatal and early-life exposure to anti-infectives increases the risk of childhood obesity and that the magnitude of the associations depends on anti-infective type, timing, and dose.
Collapse
Affiliation(s)
- Sigrid Bjerge Gribsholt
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Noel T Mueller
- Department Pediatrics Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, Aurora, Colorado, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Vera Ehrenstein
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
- Center for Population Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
3
|
Muheyati D, Han J, Lv M, Jielili M, Jing Z, Zaibibuli K, Aisike G, Aihemaiti A, Yu Y, Kaliszewski CR. Composition of gut microbiota in obese and normal-weight Uygur adults and its association with adipocyte-related factors. Sci Rep 2024; 14:24649. [PMID: 39428421 PMCID: PMC11491455 DOI: 10.1038/s41598-024-76351-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Obesity is a serious global health issue. Emerging evidence indicates that the gut microbiota may contribute to the development of obesity, possibly by instigating inflammatory processes. The objective of this research is to conduct a comparative analysis of the gut microbiota composition in obese and normal-weight Uygur adults, while examining the associations with adipocyte-related factors and dietary variables. According to the inclusion and exclusion criteria, twenty-seven Uygur adults with obesity and twenty Uygur adults with normal-weight were recruited from a local community. Anthropometric measurements and blood samples were collected. Gut microbiota composition was analyzed using 16 S rRNA gene sequencing. Adipocyte-related factors were measured using enzyme-linked immunosorbent assay (ELISA). Statistical analyses were performed to compare the gut microbiota composition between the two groups and to identify correlations between gut microbiota and adipocyte-related factors. Compared with the normal-weight group, the obese group exhibited a marked reduction in both diversity and richness of the gut microbiota, alongside a decrease in Ruminococcaceae_UCG_014, Coprococcus_2, and Parabacteroides, and an increase in Megamonas and Lachnoclostridium, implying a potential link to the development of obesity. Individuals with obesity were found to have higher Leptin (LEP), Interleukin-6 (IL-6), and C-reactive protein (CRP) than normal-weight individuals. Obese Uygur adults exhibited a gut microbiota characterized by diminished diversity and richness relative to normal-weight individuals. Parabacteroides, Megamonas, and Lachnoclostridium may play an important role in the development of obesity in Uygur population. Underlying mechanisms need further investigation.
Collapse
Affiliation(s)
- Dina Muheyati
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jia Han
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China.
| | - Meixia Lv
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Muhairemu Jielili
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Zhao Jing
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Kaibinuer Zaibibuli
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Guliqiekeran Aisike
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Ayinuer Aihemaiti
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yalu Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | | |
Collapse
|
4
|
Yu T, Luo L, Xue J, Tang W, Wu X, Yang F. Gut microbiota-NLRP3 inflammasome crosstalk in metabolic dysfunction-associated steatotic liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102458. [PMID: 39233138 DOI: 10.1016/j.clinre.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease associated with metabolic dysfunction, ranging from hepatic steatosis with or without mild inflammation to nonalcoholic steatohepatitis, which can rapidly progress to liver fibrosis and even liver cancer. In 2023, after several rounds of Delphi surveys, a new consensus recommended renaming NAFLD as metabolic dysfunction-associated steatotic liver disease (MASLD). Ninety-nine percent of NAFLD patients meet the new MASLD criteria related to metabolic cardiovascular risk factors under the "multiple parallel hits" of lipotoxicity, insulin resistance (IR), a proinflammatory diet, and an intestinal microbiota disorder, and previous research on NAFLD remains valid. The NLRP3 inflammasome, a well-known member of the pattern recognition receptor (PRR) family, can be activated by danger signals transmitted by pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), as well as cytokines involved in immune and inflammatory responses. The activation of the NLRP3 inflammasome pathway by MASLD triggers the production of the inflammatory cytokines IL-1β and IL-18. In MASLD, while changes in the composition and metabolites of the intestinal microbiota occur, the disrupted intestinal microbiota can also generate the inflammatory cytokines IL-1β and IL-18 by damaging the intestinal barrier, negatively regulating the liver on the gut-liver axis, and further aggravating MASLD. Therefore, modulating the gut-microbiota-liver axis through the NLRP3 inflammasome may emerge as a novel therapeutic approach for MASLD patients. In this article, we review the evidence regarding the functions of the NLRP3 inflammasome and the intestinal microbiota in MASLD, as well as their interactions in this disease.
Collapse
Affiliation(s)
- Tingting Yu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan 430000, PR China
| | - Lei Luo
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China
| | - Juan Xue
- Department of Gastroenterology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan 430015, PR China
| | - Wenqian Tang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China
| | - Xiaojie Wu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan 430000, PR China
| | - Fan Yang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China.
| |
Collapse
|
5
|
Wu D, An R, Wang D, Jiang L, Huang L, Lu T, Xu W, Xu J, Zhang J. Regulatory Effects of Maternal Intake of Microbial-Derived Antioxidants on Colonization of Microbiota in Breastmilk and That of Intestinal Microbiota in Offspring. Animals (Basel) 2024; 14:2582. [PMID: 39272367 PMCID: PMC11394528 DOI: 10.3390/ani14172582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
In this study, sixteen Sprague Dawley (SD) female rats and eight SD male rats were co-housed to mate. Pregnant SD female rats were fed with a control diet or an MA diet. Breast milk, maternal ileum, and intestinal samples of the offspring were collected at the day of birth and ten days afterwards. The results showed that the impact of MA was more obvious on the microbiota of mature milk (p = 0.066) than on that of colostrum. In addition, MA additive did not significantly affect maternal ileal microbiota, but affected offsprings' colonic microbiota significantly ten days after birth (p = 0.035). From the day of giving birth to ten days afterwards, in addition to the increase in microbial richness and diversity, at genus level, the dominant bacteria of breastmilk changed from Pseudomonas veronii to Bacillus and Lactococcus. Different from breastmilk microbiota, ten days after giving birth, the maternal ileal microbiota and the offsprings' intestinal microbiota were dominated by Lactobacillus. Instead of ileal microbiota, offsprings' colonic microbiota is a key action site of maternal MA additive. Therefore, the current findings have significant implications for the development of maternal feed aimed at modulating the intestinal microbiota of offspring, ultimately leading to improved health outcomes for both mothers and their offspring.
Collapse
Affiliation(s)
- Dangjin Wu
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ran An
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Di Wang
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Luoxin Jiang
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liu Huang
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tenghui Lu
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weina Xu
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- USJ-Kong Hon Academy for Cellular Nutrition, University of Saint Joseph, Macao 999078, China
| | - Jianxiong Xu
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- USJ-Kong Hon Academy for Cellular Nutrition, University of Saint Joseph, Macao 999078, China
| | - Jing Zhang
- Shanghai Key Laboratory of Veterinary and Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- USJ-Kong Hon Academy for Cellular Nutrition, University of Saint Joseph, Macao 999078, China
| |
Collapse
|
6
|
Ponce-España E, Cruz-Chamorro I, Santos-Sánchez G, Álvarez-López AI, Fernández-Santos JM, Pedroche J, Millán-Linares MC, Bejarano I, Lardone PJ, Carrillo-Vico A. Anti-obesogenic effect of lupin-derived protein hydrolysate through modulation of adiposopathy, insulin resistance and gut dysbiosis in a diet-induced obese mouse. Biomed Pharmacother 2024; 178:117198. [PMID: 39059351 DOI: 10.1016/j.biopha.2024.117198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
The prevalence of obesity is increasingly widespread, resembling a global epidemic. Lifestyle changes, such as consumption of high-energy-dense diets and physical inactivity, are major contributors to obesity. Common features of this metabolic pathology involve an imbalance in lipid and glucose homeostasis including dyslipidemia, insulin resistance and adipose tissue dysfunction. Moreover, the importance of the gut microbiota in the development and susceptibility to obesity has recently been highlighted. In recent years, new strategies based on the use of functional foods, in particular bioactive peptides, have been proposed to counteract obesity outcomes. In this context, the present study examines the effects of a lupin protein hydrolysate (LPH) on obesity, dyslipidemia and gut dysbiosis in mice fed a high-fat diet (HFD). After 12 weeks of LPH treatment, mice gained less weight and showed decreased adipose dysfunction compared to the HFD-fed group. HFD-induced dyslipidemia (increased triglycerides, cholesterol and LDL concentration) and insulin resistance were both counteracted by LPH consumption. Discriminant analysis differentially distributed LPH-treated mice compared to non-treated mice. HFD reduced gut ecological parameters, promoted the blooming of deleterious taxa and reduced the abundance of commensal members. Some of these changes were corrected in the LPH group. Finally, correlation analysis suggested that changes in this microbial population could be responsible for the improvement in obesity outcomes. In conclusion, this is the first study to show the effect of LPH on improving weight gain, adiposopathy and gut dysbiosis in the context of diet-induced obesity, pointing to the therapeutic potential of bioactive peptides in metabolic diseases.
Collapse
Affiliation(s)
- Eduardo Ponce-España
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - José María Fernández-Santos
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Justo Pedroche
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra Utrera Km 1, Seville 41013, Spain
| | | | - Ignacio Bejarano
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville 41013, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville 41009, Spain.
| |
Collapse
|
7
|
Tian Y, Rimal B, Bisanz JE, Gui W, Wolfe TM, Koo I, Murray IA, Nettleford SK, Yokoyama S, Dong F, Koshkin S, Prabhu KS, Turnbaugh PJ, Walk ST, Perdew GH, Patterson AD. Effects of Early Life Exposures to the Aryl Hydrocarbon Receptor Ligand TCDF on Gut Microbiota and Host Metabolic Homeostasis in C57BL/6J Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:87005. [PMID: 39140734 PMCID: PMC11323762 DOI: 10.1289/ehp13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 04/30/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Exposure to persistent organic pollutants (POPs) and disruptions in the gastrointestinal microbiota have been positively correlated with a predisposition to factors such as obesity, metabolic syndrome, and type 2 diabetes; however, it is unclear how the microbiome contributes to this relationship. OBJECTIVE This study aimed to explore the association between early life exposure to a potent aryl hydrocarbon receptor (AHR) agonist and persistent disruptions in the microbiota, leading to impaired metabolic homeostasis later in life. METHODS This study used metagenomics, nuclear magnetic resonance (NMR)- and mass spectrometry (MS)-based metabolomics, and biochemical assays to analyze the gut microbiome composition and function, as well as the physiological and metabolic effects of early life exposure to 2,3,7,8-tetrachlorodibenzofuran (TCDF) in conventional, germ-free (GF), and Ahr-null mice. The impact of TCDF on Akkermansia muciniphila (A. muciniphila) in vitro was assessed using optical density (OD 600), flow cytometry, transcriptomics, and MS-based metabolomics. RESULTS TCDF-exposed mice exhibited lower abundances of A. muciniphila, lower levels of cecal short-chain fatty acids (SCFAs) and indole-3-lactic acid (ILA), as well as lower levels of the gut hormones glucagon-like peptide 1 (GLP-1) and peptide YY (PYY), findings suggestive of disruption in the gut microbiome community structure and function. Importantly, microbial and metabolic phenotypes associated with early life POP exposure were transferable to GF recipients in the absence of POP carry-over. In addition, AHR-independent interactions between POPs and the microbiota were observed, and they were significantly associated with growth, physiology, gene expression, and metabolic activity outcomes of A. muciniphila, supporting suppressed activity along the ILA pathway. CONCLUSIONS These data obtained in a mouse model point to the complex effects of POPs on the host and microbiota, providing strong evidence that early life, short-term, and self-limiting POP exposure can adversely impact the microbiome, with effects persisting into later life with associated health implications. https://doi.org/10.1289/EHP13356.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - Bipin Rimal
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Jordan E. Bisanz
- Department of Biochemistry and Molecular Biology, Penn State, University Park, Pennsylvania, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Wei Gui
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - Trenton M. Wolfe
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Shaneice K. Nettleford
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Shigetoshi Yokoyama
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Fangcong Dong
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Sergei Koshkin
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - K. Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Peter J. Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub–San Francisco, San Francisco, California, USA
| | - Seth T. Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| |
Collapse
|
8
|
Wang S, Fu Z, Chen W, Wu S, Ke S, Tu J, Wei B. Saccharina Japonica Polysaccharides Suppress High-Fat Diet-Induced Obesity and Modulate Gut Microbiota Composition and Function. Chem Biodivers 2024; 21:e202401088. [PMID: 38856108 DOI: 10.1002/cbdv.202401088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Recent studies have highlighted the potential of Saccharina japonica Polysaccharides (SJPs) in alleviating high-fat diet (HFD)-induced obesity by regulating gut microbiota, which warrants further exploration to elucidate the underlying structure-activity relationship. In this study, five polysaccharide fractions (Sj-T, Sj-T-1, Sj-T-2, Sj-T-3, and Sj-T-4) with different structure characteristics were prepared from S. japonica, and their effects on HFD-induced obesity and gut microbiota composition were investigated using C57BL/6J mice. The results revealed that oral administration of Sj-T considerably suppressed HFD-induced obesity, glucose metabolic dysfunction, and other disordered symptoms. While, Sj-T-2, which has the lowest molecular weight, was the most effective in alleviating HFD-induced obesity and had the second-best effect on improving HFD-induced impaired glucose tolerance among the five SJPs. Supplementation with SJPs significantly modulated HFD-induced gut microbiota dysbiosis both at the phylum and species levels, such as enriching Desulfobacterota and Actinobacteriota, while suppressing the abundance of Bacteroidota. Sj-T also dramatically restored the gut microbiota composition by modulating the abundance of many crucial gut bacterial taxa, including s_Bacteroides_acidifaciens, s_Lachnospiraceae _bacterium, and g_Lachnospiraceae_NK4A136_group. Besides, SJPs also dramatically altered the function of gut microbiota, including many carbohydrate-metabolism enzymes. This study highlights the potential of SJPs in preventing obesity and restoring intestinal homeostasis in obese individuals.
Collapse
Affiliation(s)
- Sijia Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, CA-90024, USA
| | - Zixi Fu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Weibing Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Sitong Wu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Songze Ke
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jianfeng Tu
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, PR China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Moganshan Research Institute at Deqing County Zhejiang University of Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
9
|
Nuszkiewicz J, Kukulska-Pawluczuk B, Piec K, Jarek DJ, Motolko K, Szewczyk-Golec K, Woźniak A. Intersecting Pathways: The Role of Metabolic Dysregulation, Gastrointestinal Microbiome, and Inflammation in Acute Ischemic Stroke Pathogenesis and Outcomes. J Clin Med 2024; 13:4258. [PMID: 39064298 PMCID: PMC11278353 DOI: 10.3390/jcm13144258] [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: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of mortality and long-term disability worldwide, driven by complex and multifaceted etiological factors. Metabolic dysregulation, gastrointestinal microbiome alterations, and systemic inflammation are emerging as significant contributors to AIS pathogenesis. This review addresses the critical need to understand how these factors interact to influence AIS risk and outcomes. We aim to elucidate the roles of dysregulated adipokines in obesity, the impact of gut microbiota disruptions, and the neuroinflammatory cascade initiated by lipopolysaccharides (LPS) in AIS. Dysregulated adipokines in obesity exacerbate inflammatory responses, increasing AIS risk and severity. Disruptions in the gut microbiota and subsequent LPS-induced neuroinflammation further link systemic inflammation to AIS. Advances in neuroimaging and biomarker development have improved diagnostic precision. Here, we highlight the need for a multifaceted approach to AIS management, integrating metabolic, microbiota, and inflammatory insights. Potential therapeutic strategies targeting these pathways could significantly improve AIS prevention and treatment. Future research should focus on further elucidating these pathways and developing targeted interventions to mitigate the impacts of metabolic dysregulation, microbiome imbalances, and inflammation on AIS.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Beata Kukulska-Pawluczuk
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Katarzyna Piec
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Karina Motolko
- Student Research Club of Neurology, Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| |
Collapse
|
10
|
Visuthranukul C, Sriswasdi S, Tepaamorndech S, Chamni S, Leelahavanichkul A, Joyjinda Y, Aksornkitti V, Chomtho S. Enhancing gut microbiota and microbial function with inulin supplementation in children with obesity. Int J Obes (Lond) 2024:10.1038/s41366-024-01590-8. [PMID: 39033197 DOI: 10.1038/s41366-024-01590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND OBJECTIVES Gut dysbiosis that resulted from the alteration between host-microbe interaction might worsen obesity-induced systemic inflammation. Gut microbiota manipulation by supplementation of prebiotic inulin may reverse metabolic abnormalities and improve obesity. This study aimed to determine whether inulin supplementation improved intestinal microbiota and microbial functional pathways in children with obesity. METHODS Children with obesity whose BMI above median + 2SDs were recruited to a randomized, double-blinded placebo-controlled study. The participants aged 7-15 years were assigned to inulin supplement extracted from Thai Jerusalem artichoke (intervention), maltodextrin (placebo), and dietary fiber advice groups. All participants received similar monthly conventional advice and follow-up for 6 months. Fecal samples were collected for gut microbiome analysis using 16S rRNA sequencing. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States was performed to infer microbial functional pathways. RESULTS One hundred and forty-three children with available taxonomic and functional pathway abundance profiles were evaluated. A significant increase in alpha-diversity was observed in the inulin group. Inulin supplementation substantially enhanced Bifidobacterium, Blautia, Megasphaera, and several butyrate-producing bacteria, including Agathobacter, Eubacterium coprostanoligenes, and Subdoligranulum, compared to the other groups. The inulin group showed a significant difference in functional pathways of proteasome and riboflavin metabolism. These changes correlated with clinical and metabolic outcomes exclusively in the inulin group. CONCLUSIONS Inulin supplementation significantly promoted gut bacterial diversity and improved gut microbiota dysbiosis in children with obesity. The modulation of functional pathways by inulin suggests its potential to establish beneficial interactions between the gut microbiota and host physiology. Inulin supplementation could be a strategic treatment to restore the balance of intestinal microbiota and regulate their functions in childhood obesity.
Collapse
Affiliation(s)
- Chonnikant Visuthranukul
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Sira Sriswasdi
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Surapun Tepaamorndech
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Center of Excellent in Natural Products and Nanoparticles (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Inflammation and Immunology Research Unit (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yutthana Joyjinda
- WHO-CC for Research and Training on Viral Zoonoses, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Thai Red Cross Emerging Infection Diseases-Health Science Center, Bangkok, 10330, Thailand
| | - Vitavat Aksornkitti
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sirinuch Chomtho
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
11
|
Zhou J, Hao J, Zhong Z, Yang J, Lv T, Zhao B, Lin H, Chi J, Guo H. Fecal Microbiota Transplantation in Mice Exerts a Protective Effect Against Doxorubicin-Induced Cardiac Toxicity by Regulating Nrf2-Mediated Cardiac Mitochondrial Fission and Fusion. Antioxid Redox Signal 2024; 41:1-23. [PMID: 37756370 DOI: 10.1089/ars.2023.0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Aims: The relationship between the gut microbiota and cardiovascular system has been increasingly clarified. Fecal microbiota transplantation (FMT), used to improve gut microbiota, has been applied clinically for disease treatment and has great potential in combating doxorubicin (DOX)-induced cardiotoxicity. However, the application of FMT in the cardiovascular field and its molecular mechanisms are poorly understood. Results: During DOX-induced stress, FMT alters the gut microbiota and serum metabolites, leading to a reduction in cardiac injury. Correlation analysis indicated a close association between serum metabolite indole-3-propionic acid (IPA) and cardiac function. FMT and IPA achieve this by facilitating the translocation of Nfe2l2 (Nrf2) from the cytoplasm to the nucleus, thereby activating the expression of antioxidant molecules, reducing reactive oxygen species production, and inhibiting excessive mitochondrial fission. Consequently, mitochondrial function is preserved, leading to the mitigation of cardiac injury under DOX-induced stress. Innovation: FMT has the ability to modify the composition of the gut microbiota, providing not only protection to the intestinal mucosa but also influencing the generation of serum metabolites and regulating the Nrf2 gene to modulate the balance of cardiac mitochondrial fission and fusion. This study comprehensively demonstrates the efficacy of FMT in countering DOX-induced myocardial damage and elucidates the pathways linking the microbiota and the heart. Conclusion: FMT alters the gut microbiota and serum metabolites of recipient mice, promoting nuclear translocation of Nrf2 and subsequent activation of downstream antioxidant molecule expression, while inhibiting excessive mitochondrial fission to preserve cardiac integrity. Correlation analysis highlights IPA as a key contributor among differentially regulated metabolites.
Collapse
Affiliation(s)
- Jiedong Zhou
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jinjin Hao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zuoquan Zhong
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Juntao Yang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Tingting Lv
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Bingjie Zhao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- Department of Cardiovascular, The Affiliated Lihuili Hospital of Ningbo University, Healthy Science Center, Ningbo University, Ningbo, China
| | - Jufang Chi
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, China
| | - Hangyuan Guo
- School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
12
|
Joshua Ashaolu T, Joshua Olatunji O, Can Karaca A, Lee CC, Mahdi Jafari S. Anti-obesity and anti-diabetic bioactive peptides: A comprehensive review of their sources, properties, and techno-functional challenges. Food Res Int 2024; 187:114427. [PMID: 38763677 DOI: 10.1016/j.foodres.2024.114427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/21/2024]
Abstract
The scourge of obesity arising from obesogens and poor dieting still ravages our planet as half of the global population may be overweight and obese by 2035. This metabolic disorder is intertwined with type 2 diabetes (T2D), both of which warrant alternative therapeutic options other than clinically approved drugs like orlistat with their tendency of abuse and side effects. In this review, we comprehensively describe the global obesity problem and its connection to T2D. Obesity, overconsumption of fats, the mechanism of fat digestion, obesogenic gut microbiota, inhibition of fat digestion, and natural anti-obesity compounds are discussed. Similar discussions are made for diabetes with regard to glucose regulation, the diabetic gut microbiota, and insulinotropic compounds. The sources and production of anti-obesity bioactive peptides (AOBPs) and anti-diabetic bioactive peptides (ADBPs) are also described while explaining their structure-function relationships, gastrointestinal behaviors, and action mechanisms. Finally, the techno-functional applications of AOBPs and ADBPs are highlighted.
Collapse
Affiliation(s)
- Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang 550000, Vietnam; Faculty of Medicine, Duy Tan University, Da Nang 550000, Vietnam.
| | | | - Asli Can Karaca
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, 34469 Istanbul, Turkey.
| | - Chi-Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey.
| | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran; Halal Research Center of IRI, Iran Food and Drug Administration, Ministry of Health and Medical Education, Tehran, Iran.
| |
Collapse
|
13
|
Zhu Y, Yuan Y, Si H, Li S, Zhao F, Mu R, Lin Z, Wang X, Qiu Q, Xu C, Ji L, Li Z. Lipidomic and transcriptomic profiles provide new insights into the triacylglycerol and glucose handling capacities of the Arctic fox. Front Vet Sci 2024; 11:1388532. [PMID: 38988981 PMCID: PMC11233799 DOI: 10.3389/fvets.2024.1388532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
The Arctic fox (Vulpes lagopus) is a species indigenous to the Arctic and has developed unique lipid metabolism, but the mechanisms remain unclear. Here, the significantly increased body weight of Arctic foxes was consistent with the significantly increased serum very-low-density lipoprotein (VLDL), and the 40% crude fat diet further increased the Arctic fox body weight. The enhanced body weight gain stems primarily from increased subcutaneous adipose tissue accumulation. The adipose triacylglycerol and phosphatidylethanolamine were significantly greater in Arctic foxes. The adipose fatty-acid synthase content was significantly lower in Arctic foxes, highlighting the main role of exogenous fatty-acids in fat accumulation. Considering the same diet, liver-derived fat dominates adipose expansion in Arctic foxes. Liver transcriptome analysis revealed greater fat and VLDL synthesis in Arctic foxes, consistent with the greater VLDL. Glucose homeostasis wasn't impacted in Arctic foxes. And the free fatty-acids in adipose, which promote insulin resistance, also did not differ between groups. However, the hepatic glycogen was greater in Arctic foxes and transcriptome analysis revealed upregulated glycogen synthesis, improving glucose homeostasis. These results suggest that the superior fat accumulation capacity and distinct characteristics of hepatic and adipose lipid and glucose metabolism facilitate glucose homeostasis and massive fat accumulation in Arctic foxes.
Collapse
Affiliation(s)
- Yuhang Zhu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yuan Yuan
- School of Ecology and Environment, Northwestern Polytechnical University, Xi’an, China
| | - Huazhe Si
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Songze Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Fei Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ruina Mu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zihan Lin
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Xiaoxu Wang
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qiang Qiu
- School of Ecology and Environment, Northwestern Polytechnical University, Xi’an, China
| | - Chao Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Lele Ji
- National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, China
| | - Zhipeng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
14
|
Freitas RGBON, Vasques ACJ, da Rocha Fernandes G, Ribeiro FB, Solar I, Shivappa N, Hébert JR, de Almeida-Pititto B, Geloneze B, Ferreira SRG. Gut bacterial markers involved in association of dietary inflammatory index with visceral adiposity. Nutrition 2024; 122:112371. [PMID: 38430843 DOI: 10.1016/j.nut.2024.112371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE To deepen the understanding of the influence of diet on weight gain and metabolic disturbances, we examined associations between diet-related inflammation and body composition and fecal bacteria abundances in participants of the Nutritionists' Health Study. METHODS Early-life, dietary and clinical data were obtained from 114 women aged ≤45 years. A validated food frequency questionnaire was used to calculate the energy-adjusted dietary inflammatory index (E-DII). Participants' data were compared by E-DII quartiles using ANOVA or Kruskal-Wallis. Associations of DXA-determined body composition with the E-DII were tested by multiple linear regression using DAG-oriented adjustments. Fecal microbiota was analyzed targeting the V4 region of the 16S rRNA gene. Spearman correlation coefficients were used to test linear associations; differential abundance of genera across the E-DII quartiles was assessed by pair-wise comparisons. RESULTS E-DII score was associated with total fat (b=1.80, p<0.001), FMI (b=0.08, p<0.001) and visceral fat (b=1.19, p=0.02), independently of maternal BMI, birth type and breastfeeding. E-DII score was directly correlated to HOMA-IR (r=0.30; p=0.004), C-reactive protein (r=0.29; p=0.003) and to the abundance of Actinomyces, and inversely correlated to the abundance of Eubacterium.xylanophilum.group. Actinomyces were significantly more abundant in the highest (most proinflammatory) E-DII quartile. CONCLUSIONS Association of E-DII with markers of insulin resistance, inflammation, body adiposity and certain gut bacteria are consistent with beneficial effects of anti-inflammatory diet on body composition and metabolic profile. Bacterial markers, such as Actinomyces, could be involved in the association between the dietary inflammation with visceral adiposity. Studies designed to explore how a pro-inflammatory diet affects both central fat deposition and gut microbiota are needed.
Collapse
Affiliation(s)
- Renata G B O N Freitas
- Department of Epidemiology, School of Public Health, University of São Paulo, Brazil; Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil
| | - Ana Carolina J Vasques
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | | | - Francieli B Ribeiro
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Isabela Solar
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; School of Applied Sciences, University of Campinas, São Paulo, Brazil
| | - Nitin Shivappa
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Department of Nutrition, Connecting Health Innovations, Columbia, SC
| | - James R Hébert
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Department of Nutrition, Connecting Health Innovations, Columbia, SC
| | | | - Bruno Geloneze
- Laboratory of Investigation in Metabolism and Diabetes, School of Medical Sciences, University of Campinas, Brazil; Obesity and Comorbidities Research Center, University of Campinas, São Paulo, Brazil
| | | |
Collapse
|
15
|
Melali H, Abdolahi A, Sheikhbahaei E, Vakili K, Mahmoudieh M, Keleidari B, Shahabi S. Impact of Probiotics on Gastrointestinal Function and Metabolic Status After Roux-en-Y Gastric Bypass: A Double-Blind, Randomized Trial. Obes Surg 2024; 34:2033-2041. [PMID: 38653887 DOI: 10.1007/s11695-024-07225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Postoperative changes in gut microbiota may occur in patients undergoing Roux-en-Y gastric bypass surgery. In this study, we evaluate the impact of administering probiotic tablets on the gastrointestinal function and metabolic status of these patients. MATERIALS AND METHODS This double-blinded randomized clinical trial was conducted from 2021 to 2022 on 135 Roux-en-Y surgery candidates. The intervention group underwent the surgical procedure and started receiving probiotic supplements (Familact Co.) 1 week after surgery; the control group received a placebo. The laboratory and anthropometric data were measured and analyzed before and 3 and 6 months after the intervention. GIQLI questionnaire was also used at the beginning and 6 months after the intervention to evaluate GI symptoms. RESULTS We observed significantly reduced BMI in both groups after surgeries (P < 0.001). The levels of FBS and HbA1C were significantly lower in the probiotic group compared to the placebo in 3 months (P = 0.02 and P = 0.001, respectively) and 6 months (P < 0.001 for both) after the intervention. The levels of vitamin B12 increased significantly in the probiotic group (P < 0.001), and the values were substantially higher than the placebo group in 3 and 6 months (P < 0.001), respectively. Analysis of the GIQLI questionnaire before and 6 months after interventions also revealed significant improvement in the GIQLI score in both groups (P < 0.001 for probiotics and P = 0.03 for placebo). CONCLUSION Probiotic supplement administration following RYGB improves patients' vitamin and metabolic profile, as well as GI function, although it cannot significantly affect weight loss.
Collapse
Affiliation(s)
- Hamid Melali
- Isfahan Minimally Invasive Surgery and Obesity Research Center, Department of Surgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alimeh Abdolahi
- Department of General Surgery, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Erfan Sheikhbahaei
- Isfahan Minimally Invasive Surgery and Obesity Research Center, Department of Surgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Iran University of Medical Sciences, Hazrat-E Rasool General Hospital, Masouri St. Niyayesh St. Satarkhan Ave., Tehran, 14665-354, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Mahmoudieh
- Isfahan Minimally Invasive Surgery and Obesity Research Center, Department of Surgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrouz Keleidari
- Isfahan Minimally Invasive Surgery and Obesity Research Center, Department of Surgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahab Shahabi
- Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Iran University of Medical Sciences, Hazrat-E Rasool General Hospital, Masouri St. Niyayesh St. Satarkhan Ave., Tehran, 14665-354, Iran.
| |
Collapse
|
16
|
Sahle Z, Engidaye G, Shenkute Gebreyes D, Adenew B, Abebe TA. Fecal microbiota transplantation and next-generation therapies: A review on targeting dysbiosis in metabolic disorders and beyond. SAGE Open Med 2024; 12:20503121241257486. [PMID: 38826830 PMCID: PMC11143861 DOI: 10.1177/20503121241257486] [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: 09/15/2023] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
The human microbiome, particularly the gut microbiome, has emerged as a central determinant of health and disease. Dysbiosis, an imbalance in the microbial composition of the gut, is associated with a variety of metabolic and other diseases, highlighting the potential for microbiota-targeted treatments. Fecal microbiota transplantation has received considerable attention as a promising therapy to modulate the gut microbiome and restore microbial homeostasis. However, challenges remain, including standardization, safety, and long-term efficacy. This review summarizes current knowledge on fecal microbiota transplantation and describes the next generation therapies targeting microbiome. This review looked at the mechanistic understanding of fecal microbiota transplantation and alternative strategies, elucidating their potential role in improving dysbiosis-associated metabolic disorders, such as obesity, and type 2 diabetes and others. Additionally, this review discussed the growing application of therapies targeting the gut microbiome. Insights from clinical trials, preclinical studies, and emerging technologies provide a comprehensive overview of the evolving landscape of microbiome-based interventions. Through a critical assessment of current advances and prospects, this review aims to highlight the therapeutic potential of targeting gut microbiome and pave the way for innovative approaches in precision medicine and personalized treatments.
Collapse
Affiliation(s)
- Zenawork Sahle
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Getabalew Engidaye
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Demissew Shenkute Gebreyes
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Behailu Adenew
- Department of Medical Laboratory Science, Debre Berhan Compressive Specialized Hospital, Debre Berhan, Ethiopia
| | - Tsegahun Asfaw Abebe
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| |
Collapse
|
17
|
Mei EH, Yao C, Chen YN, Nan SX, Qi SC. Multifunctional role of oral bacteria in the progression of non-alcoholic fatty liver disease. World J Hepatol 2024; 16:688-702. [PMID: 38818294 PMCID: PMC11135273 DOI: 10.4254/wjh.v16.i5.688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 04/07/2024] [Indexed: 05/22/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver disorders of varying severity, ultimately leading to fibrosis. This spectrum primarily consists of NAFL and non-alcoholic steatohepatitis. The pathogenesis of NAFLD is closely associated with disturbances in the gut microbiota and impairment of the intestinal barrier. Non-gut commensal flora, particularly bacteria, play a pivotal role in the progression of NAFLD. Notably, Porphyromonas gingivalis, a principal bacterium involved in periodontitis, is known to facilitate lipid accumulation, augment immune responses, and induce insulin resistance, thereby exacerbating fibrosis in cases of periodontitis-associated NAFLD. The influence of oral microbiota on NAFLD via the "oral-gut-liver" axis is gaining recognition, offering a novel perspective for NAFLD management through microbial imbalance correction. This review endeavors to encapsulate the intricate roles of oral bacteria in NAFLD and explore underlying mechanisms, emphasizing microbial control strategies as a viable therapeutic avenue for NAFLD.
Collapse
Affiliation(s)
- En-Hua Mei
- Shanghai Medical College, Fudan University, Shanghai 200000, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Chao Yao
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Yi-Nan Chen
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Shun-Xue Nan
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Sheng-Cai Qi
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China.
| |
Collapse
|
18
|
Visuthranukul C, Leelahavanichkul A, Tepaamorndech S, Chamni S, Mekangkul E, Chomtho S. Inulin supplementation exhibits increased muscle mass via gut-muscle axis in children with obesity: double evidence from clinical and in vitro studies. Sci Rep 2024; 14:11181. [PMID: 38755201 PMCID: PMC11099025 DOI: 10.1038/s41598-024-61781-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
Gut microbiota manipulation may reverse metabolic abnormalities in obesity. Our previous studies demonstrated that inulin supplementation significantly promoted Bifidobacterium and fat-free mass in obese children. We aimed to study gut-muscle axis from inulin supplementation in these children. In clinical phase, the plasma samples from 46 participants aged 7-15 years, were analyzed for muscle biomarkers before and after 6-month inulin supplementation. In parallel, the plausible mechanism of muscle production via gut-muscle axis was examined using macrophage cell line. Bifidobacterium was cultured in semi-refined medium with inulin used in the clinical phase. Cell-free supernatant was collected and used in lipopolysaccharide (LPS)-induced macrophage cell line to determine inflammatory and anti-inflammatory gene expression. In clinical phase, IL-15 and creatinine/cystatin C ratio significantly increased from baseline to the 6th month. In vitro study showed that metabolites derived from Bifidobacterium capable of utilizing inulin contained the abundance of SCFAs. In the presence of LPS, treatment from Bifidobacterium + inulin downregulated TNF-α, IL-6, IL-1β, and iNOS, but upregulated FIZZ-1 and TGF-β expression. Inulin supplementation promoted the muscle biomarkers in agreement with fat-free mass gain, elucidating by Bifidobacterium metabolites derived from inulin digestion showed in vitro anti-inflammatory activity and decreased systemic pro-inflammation, thus promoting muscle production via gut-muscle axis response.Clinical Trial Registry number: NCT03968003.
Collapse
Affiliation(s)
- Chonnikant Visuthranukul
- Pediatric Nutrition Research Unit, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Pathumwan, Bangkok, 10330, Thailand.
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Inflammation and Immunology Research Unit (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Surapun Tepaamorndech
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Natural Products and Nanoparticles Research Unit (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Eakkarin Mekangkul
- Pediatric Nutrition Research Unit, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Pathumwan, Bangkok, 10330, Thailand
| | - Sirinuch Chomtho
- Pediatric Nutrition Research Unit, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road, Pathumwan, Bangkok, 10330, Thailand
| |
Collapse
|
19
|
Zhang Z, Jia Y, Zhang C, Zhang Z, Jin F, Pan D, Li D, Wu X. Efficacy of epigallocatechin gallate (EGCG) and its underlying mechanism in preventing bisphenol-A-induced metabolic disorders in mice. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134098. [PMID: 38522198 DOI: 10.1016/j.jhazmat.2024.134098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
To investigate the efficacy of epigallocatechin gallate (EGCG) and its underlying mechanism in preventing bisphenol-A-induced metabolic disorders, in this study, a mice model of metabolic disorders induced by BPA was developed to investigate the efficacy and mechanism of EGCG using microbiomes and metabolomics. The results showed that EGCG reduced body weight, liver weight ratio, and triglyceride and total cholesterol levels in mice by decreasing the mRNA expression of genes related to fatty acid synthesis (Elov16) and cholesterol synthesis (CYP4A14) and increasing the mRNA expression of genes related to fatty acid oxidation (Lss) and cholesterol metabolism (Cyp7a1). In addition, EGCG normalized BPA-induced intestinal microbial dysbiosis. Metabolic pathway analysis showed that low-dose EGCG was more effective than high-dose EGCG at affecting the biosynthesis of L-cysteine, glycerophosphorylcholine, and palmitoleic acid. These results provide specific data and a theoretical basis for the risk assessment of BPA and the utilization of EGCG.
Collapse
Affiliation(s)
- Zhaoxian Zhang
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Yaoyi Jia
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Chenghui Zhang
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Zikang Zhang
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Fangsha Jin
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Dandan Pan
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China.
| | - Xiangwei Wu
- Key Laboratory of Agri-food Safety of Anhui Province, College of Resources and Environment, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036 Anhui, PR China.
| |
Collapse
|
20
|
Guo W, Liu S, Khan MZ, Wang J, Chen T, Alugongo GM, Li S, Cao Z. Bovine milk microbiota: Key players, origins, and potential contributions to early-life gut development. J Adv Res 2024; 59:49-64. [PMID: 37423549 PMCID: PMC11081965 DOI: 10.1016/j.jare.2023.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND Bovine milk is a significant substitute for human breast milk and holds great importance in infant nutrition and health. Apart from essential nutrients, bovine milk also contains bioactive compounds, including a microbiota derived from milk itself rather than external sources of contamination. AIM OF REVIEW Recognizing the profound impact of bovine milk microorganisms on future generations, our review focuses on exploring their composition, origins, functions, and applications. KEY SCIENTIFIC CONCEPTS OF REVIEW Some of the primary microorganisms found in bovine milk are also present in human milk. These microorganisms are likely transferred to the mammary gland through two pathways: the entero-mammary pathway and the rumen-mammary pathway. We also elucidated potential mechanisms by which milk microbiota contribute to infant intestinal development. The mechanisms include the enhancing of the intestinal microecological niche, promoting the maturation of immune system, strengthening the intestinal epithelial barrier function, and interacting with milk components (e.g., oligosaccharides) via cross-feeding effect. However, given the limited understanding of bovine milk microbiota, further studies are necessary to validate hypotheses regarding their origins and to explore their functions and potential applications in early intestinal development.
Collapse
Affiliation(s)
- Wenli Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Muhammad Z Khan
- Faculty of Veterinary and Animal Sciences, Department of Animal Breeding and Genetics, The University of Agriculture, Dera Ismail Khan 29220, Pakistan
| | - Jingjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Tianyu Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Gibson M Alugongo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
21
|
Li XM, Lv Q, Chen YJ, Yan LB, Xiong X. Association between childhood obesity and gut microbiota: 16S rRNA gene sequencing-based cohort study. World J Gastroenterol 2024; 30:2249-2257. [PMID: 38690025 PMCID: PMC11056921 DOI: 10.3748/wjg.v30.i16.2249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/18/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND This study aimed to identify characteristic gut genera in obese and normal-weight children (8-12 years old) using 16S rDNA sequencing. The research aimed to provide insights for mechanistic studies and prevention strategies for childhood obesity. Thirty normal-weight and thirty age- and sex-matched obese children were included. Questionnaires and body measurements were collected, and fecal samples underwent 16S rDNA sequencing. Significant differences in body mass index (BMI) and body-fat percentage were observed between the groups. Analysis of gut microbiota diversity revealed lower α-diversity in obese children. Di-fferences in gut microbiota composition were found between the two groups. Prevotella and Firmicutes were more abundant in the obese group, while Bacteroides and Sanguibacteroides were more prevalent in the control group. AIM To identify the characteristic gut genera in obese and normal-weight children (8-12-year-old) using 16S rDNA sequencing, and provide a basis for subsequent mechanistic studies and prevention strategies for childhood obesity. METHODS Thirty each normal-weight, 1:1 matched for age and sex, and obese children, with an obese status from 2020 to 2022, were included in the control and obese groups, respectively. Basic information was collected through questionnaires and body measurements were obtained from both obese and normal-weight children. Fecal samples were collected from both groups and subjected to 16S rDNA sequencing using an Illumina MiSeq sequencing platform for gut microbiota diversity analysis. RESULTS Significant differences in BMI and body-fat percentage were observed between the two groups. The Ace and Chao1 indices were significantly lower in the obese group than those in the control group, whereas differences were not significant in the Shannon and Simpson indices. Kruskal-Wallis tests indicated significant differences in unweighted and weighted UniFrac distances between the gut microbiota of normal-weight and obese children (P < 0.01), suggesting substantial disparities in both the species and quantity of gut microbiota between the two groups. Prevotella, Firmicutes, Bacteroides, and Sanguibacteroides were more abundant in the obese and control groups, respectively. Heatmap results demonstrated significant differences in the gut microbiota composition between obese and normal-weight children. CONCLUSION Obese children exhibited lower α-diversity in their gut microbiota than did the normal-weight children. Significant differences were observed in the composition of gut microbiota between obese and normal-weight children.
Collapse
Affiliation(s)
- Xu-Ming Li
- Laboratory Department, Nanjing Medical University Affiliated Obstetrics and Gynecology Hospital (Nanjing Maternal and Child Health Hospital), Nanjing 210004, Jiangsu Province, China
| | - Qing Lv
- Department of Pediatrics, Shenzhen University General Hospital, Shenzhen 518055, Guangdong Province, China
| | - Ya-Jun Chen
- Department of Inspection Division, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, Jiangsu Province, China
| | - Lu-Biao Yan
- Department of Pediatrics, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, Jiangsu Province, China
| | - Xin Xiong
- Department of Neonatology, Chenzhou First People’s Hospital, Chenzhou 423000, Hunan Province, China
| |
Collapse
|
22
|
Kang P, Wang AZX. Microbiota-gut-brain axis: the mediator of exercise and brain health. PSYCHORADIOLOGY 2024; 4:kkae007. [PMID: 38756477 PMCID: PMC11096970 DOI: 10.1093/psyrad/kkae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024]
Abstract
The brain controls the nerve system, allowing complex emotional and cognitive activities. The microbiota-gut-brain axis is a bidirectional neural, hormonal, and immune signaling pathway that could link the gastrointestinal tract to the brain. Over the past few decades, gut microbiota has been demonstrated to be an essential component of the gastrointestinal tract that plays a crucial role in regulating most functions of various body organs. The effects of the microbiota on the brain occur through the production of neurotransmitters, hormones, and metabolites, regulation of host-produced metabolites, or through the synthesis of metabolites by the microbiota themselves. This affects the host's behavior, mood, attention state, and the brain's food reward system. Meanwhile, there is an intimate association between the gut microbiota and exercise. Exercise can change gut microbiota numerically and qualitatively, which may be partially responsible for the widespread benefits of regular physical activity on human health. Functional magnetic resonance imaging (fMRI) is a non-invasive method to show areas of brain activity enabling the delineation of specific brain regions involved in neurocognitive disorders. Through combining exercise tasks and fMRI techniques, researchers can observe the effects of exercise on higher brain functions. However, exercise's effects on brain health via gut microbiota have been little studied. This article reviews and highlights the connections between these three interactions, which will help us to further understand the positive effects of exercise on brain health and provide new strategies and approaches for the prevention and treatment of brain diseases.
Collapse
Affiliation(s)
- Piao Kang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | |
Collapse
|
23
|
Gao Y, Liu L, Cui Y, Zhang J, Wu X. The causality of gut microbiota on onset and progression of sepsis: a bi-directional Mendelian randomization analysis. Front Immunol 2024; 15:1266579. [PMID: 38698853 PMCID: PMC11063379 DOI: 10.3389/fimmu.2024.1266579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Background Several observational studies have proposed a potential link between gut microbiota and the onset and progression of sepsis. Nevertheless, the causality of gut microbiota and sepsis remains debatable and warrants more comprehensive exploration. Methods We conducted a two-sample Mendelian randomization (MR) analysis to test the causality between gut microbiota and the onset and progression of sepsis. The genome-wide association study (GWAS) summary statistics for 196 bacterial traits were extracted from the MiBioGen consortium, whereas the GWAS summary statistics for sepsis and sepsis-related outcomes came from the UK Biobank. The inverse-variance weighted (IVW) approach was the primary method used to examine the causal association. To complement the IVW method, we utilized four additional MR methods. We performed a series of sensitivity analyses to examine the robustness of the causal estimates. Results We assessed the causality of 196 bacterial traits on sepsis and sepsis-related outcomes. Genus Coprococcus2 [odds ratio (OR) 0.81, 95% confidence interval (CI) (0.69-0.94), p = 0.007] and genus Dialister (OR 0.85, 95% CI 0.74-0.97, p = 0.016) had a protective effect on sepsis, whereas genus Ruminococcaceae UCG011 (OR 1.10, 95% CI 1.01-1.20, p = 0.024) increased the risk of sepsis. When it came to sepsis requiring critical care, genus Anaerostipes (OR 0.49, 95% CI 0.31-0.76, p = 0.002), genus Coprococcus1 (OR 0.65, 95% CI 0.43-1.00, p = 0.049), and genus Lachnospiraceae UCG004 (OR 0.51, 95% CI 0.34-0.77, p = 0.001) emerged as protective factors. Concerning 28-day mortality of sepsis, genus Coprococcus1 (OR 0.67, 95% CI 0.48-0.94, p = 0.020), genus Coprococcus2 (OR 0.48, 95% CI 0.27-0.86, p = 0.013), genus Lachnospiraceae FCS020 (OR 0.70, 95% CI 0.52-0.95, p = 0.023), and genus Victivallis (OR 0.82, 95% CI 0.68-0.99, p = 0.042) presented a protective effect, whereas genus Ruminococcus torques group (OR 1.53, 95% CI 1.00-2.35, p = 0.049), genus Sellimonas (OR 1.25, 95% CI 1.04-1.50, p = 0.019), and genus Terrisporobacter (OR 1.43, 95% CI 1.02-2.02, p = 0.040) presented a harmful effect. Furthermore, genus Coprococcus1 (OR 0.42, 95% CI 0.19-0.92, p = 0.031), genus Coprococcus2 (OR 0.34, 95% CI 0.14-0.83, p = 0.018), and genus Ruminiclostridium6 (OR 0.43, 95% CI 0.22-0.83, p = 0.012) were associated with a lower 28-day mortality of sepsis requiring critical care. Conclusion This MR analysis unveiled a causality between the 21 bacterial traits and sepsis and sepsis-related outcomes. Our findings may help the development of novel microbiota-based therapeutics to decrease the morbidity and mortality of sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Xiuying Wu
- Department of Anesthesia, ShengJing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Fang C, Shen Y, Ma Z, Li Y, Zhang J, Liu C, Ye Y. l-Theanine Prevents Colonic Damage via NF-κB/MAPK Signaling Pathways Induced by a High-Fat Diet in Rats. Mol Nutr Food Res 2024; 68:e2300797. [PMID: 38549456 DOI: 10.1002/mnfr.202300797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/07/2024] [Indexed: 05/12/2024]
Abstract
SCOPE l-Theanine (l-Thea) is an amino acid which is naturally present in tea leaves. It has been associated with possible health advantages, including obesity prevention, but the underlying molecular mechanisms have not been elucidated. METHODS AND RESULTS A multiomics approach is utilized to examine the mechanism by which l-Thea exerts its antiobesity effects. This study reveals that l-Thea administration significantly ameliorates high-fat diet (HFD)-induced obesity in rats by improving body weight and hyperlipidemia. l-Thea mitigates HFD-induced inflammation and reverses hepatic and colonic damage, and intestinal barrier. This research verifies that the preventive effect of l-Thea on obesity in rats induced by an HFD with colitis is accomplished by suppressing the phosphorylation of important proteins in the NF-κB/mitogen-activated protein kinase (MAPK) pathways. Metabolome analysis reveals that l-Thea regulates HFD-induced metabolic disorders, specifically through modulation of steroid hormone biosynthesis. Microbiome analysis reveals that l-Thea mitigates HFD-induced dysbiosis by increasing the relative abundance of obesity-associated probiotic bacteria, including Blautia coccoides and Lactobacillus murinus, while simultaneously suppressing the abundance of pathogenic bacteria. CONCLUSIONS l-Thea alleviates colitis generated by an HFD by restoring the integrity of the intestinal barrier, suppressing inflammation through regulation of MAPK/NF-κB signaling pathways, and enhancing microbial metabolism in colon.
Collapse
Affiliation(s)
- Chunyan Fang
- Institute of Quality Standard and Testing Technology Research, Tea Research Institute, Tea Refining and Innovation Key Laboratory of Sichuan Province, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, PR China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yifeng Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Ziyang Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuchen Li
- College of Horticulture, Tea Refining and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Jingyi Zhang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chen Liu
- College of Horticulture, Tea Refining and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Yulong Ye
- Institute of Quality Standard and Testing Technology Research, Tea Research Institute, Tea Refining and Innovation Key Laboratory of Sichuan Province, Sichuan Academy of Agricultural Sciences, Chengdu, 610066, PR China
| |
Collapse
|
25
|
Zheng Y, Zhang S, Zhang Z, Zhang T, Teng X, Xiao G, Huang S. Isolation of Lactobacillus acidophilus strain and its anti-obesity effect in a diet induced obese murine model. Lett Appl Microbiol 2024; 77:ovae021. [PMID: 38400571 DOI: 10.1093/lambio/ovae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
Intestinal microbiota is a potential determinant of obesity, with probiotic bile salt hydrolase (BSH) as one of the key mechanisms in the anti-obesity effects. In this study, we present a Lactobacillus acidophilus GOLDGUT-LA100 (LA100) with high BSH activity, good gastric acid and bile salt tolerance, and a potential anti-obesity effect. LA100's anti-obesity effects were evaluated in a high-fat diet-induced, obese mouse model. LA100 administration alleviates high-fat diet-induced pathophysiological symptoms, such as body weight gain, high serum glucose and cholesterol level, hepatic lipid accumulation, and adipose inflammation. These results demonstrate concrete anti-obesity benefit in animal models and show promising applications in future clinical studies.
Collapse
Affiliation(s)
- Yanyi Zheng
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Silu Zhang
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Zhizhu Zhang
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | | | - Xin Teng
- Bluepha Co., Ltd, Shenzhen 518000, China
| | - Guoxun Xiao
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Song Huang
- Bluepha Co., Ltd, Shenzhen 518000, China
- Department of Chemical and Biological Engineering, Xiamen University, Xiamen 361102, China
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
26
|
Kelly Souza Silveira B, Mayumi Usuda Prado Rocha D, Stampini Duarte Martino H, Grancieri M, Juste Contin Gomes M, Cuquetto Mantovani H, Bressan J, Hermana Miranda Hermsdorff H. Daily Cashew and Brazil Nut Consumption Modifies Intestinal Health in Overweight Women on Energy-Restricted Intervention: A Randomized Controlled Trial (Brazilian Nuts Study). J Nutr 2024; 154:962-977. [PMID: 38246355 DOI: 10.1016/j.tjnut.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Increased intestinal permeability and dysbiosis are related to obesity. Nuts can provide nutrients and bioactive compounds that modulate gut microbiota and inflammation, enhancing the beneficial effects of weight loss. OBJECTIVES To evaluate the effect of consuming cashew nuts (Anacardium occidentale L.) and Brazil nuts (Bertholletia excelsa H.B.K) on intestinal permeability and microbiota, fecal SCFAs and pH, inflammation, and weight loss in energy restriction condition. METHODS In this 8-week randomized controlled trial, 40 women with overweight or obesity were assigned to energy-restricted groups (-500 kcal/d): control group (free of nuts) or Brazilian nuts group (BN: 30 g of cashew nuts and 15 g of Brazil nuts per day). Permeability was analyzed by the lactulose/mannitol test and the microbiota by sequencing the 16S gene in the V3-V4 regions. Plasma concentrations of inflammatory cytokines (TNF, IL-6, IL-10, IL-8, IL-17A) and C-reactive protein were analyzed. RESULTS In total, 25 women completed the intervention. Both groups lost weight without statistical differences. Lactulose excretion increased only in the control group (P < 0.05). The BN consumption increased fecal propionic acid and potentially beneficial bacteria, such as Ruminococcus, Roseburia, strains NK4A214 and UCG-002 from the Ruminococcaceae family, but also Lachnospiraceae family, Bacteroides, and Lachnoclostridium, when compared to the control group. Changes in intestinal permeability were correlated to a greater reduction in body fat (kg), and IL-8, and increases in Ruminococcus abundance. CONCLUSION Our findings demonstrate a positive impact of BN consumption within an energy-restricted context, linked to the augmentation of potentially beneficial bacteria and pathways associated with body fat reduction. Besides, BN consumption mitigated increased intestinal permeability, although its capacity to diminish permeability or enhance weight loss proved limited. This trial was registered at the Brazilian Registry of Clinical Trials as ReBEC (ID: RBR-3ntxrm).
Collapse
Affiliation(s)
- Brenda Kelly Souza Silveira
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Daniela Mayumi Usuda Prado Rocha
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | | | - Mariana Grancieri
- Experimental Nutrition Laboratory, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Mariana Juste Contin Gomes
- Experimental Nutrition Laboratory, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Hilário Cuquetto Mantovani
- Anaerobic Microbioloy Laboratory, Department of Microbiology, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Josefina Bressan
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Helen Hermana Miranda Hermsdorff
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil.
| |
Collapse
|
27
|
Liu Z, Ma C, Gao H, Huang X, Zhang Y, Liu C, Hou R, Zhang Q, Li Q. A polysaccharide from salviae miltiorrhizae radix inhibits weight gain of mice with high-fat diet via modulating intestinal bacteria. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:479-487. [PMID: 37647505 DOI: 10.1002/jsfa.12948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Obesity, a global chronic disease, has been recognized as a severe risk to health. In our study, a novel polysaccharide named ARS was isolated and purified from aerial part of salviae miltiorrhizae radix. Our aim is to investigate the weight-reducing effect of a polysaccharide from salviae miltiorrhizae radix on mice fed a high-fat diet. RESULTS The novel polysaccharide ARS mainly consisted of glucose and galactose with a molar ratio of 0.59:1.00. We found that treatment with ARS could inhibit weight gain of mice fed a high-fat diet via modulating the intestinal bacteria. Moreover, we surveyed its mechanism in mice, and the gut microbiota sequencing results demonstrated that ARS can reverse or resist high-fat-diet-induced significant weight gain or obesity by increasing the diversity of gut microbiota and optimizing the ratio of Firmicutes to Bacteroidetes. Phylum and species analysis of gut microbiota demonstrated that obesity caused by a high-fat diet was accompanied by significant changes in the microbial communities, but ARS could reverse the disturbance of gut microbiota induced by the high-fat diet to maintain homeostasis. CONCLUSION Overall, our findings suggested a new function of ARS in regulating gut microbiota, which provides a theoretical basis for the development of high-quality ARS functional foods and the application of dietary supplements. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhihai Liu
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
- Department of Microbiology and Immunology, College of Husbandry and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ce Ma
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaoli Huang
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Yaru Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Congmin Liu
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Ranran Hou
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| | - Qidi Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Qiu Li
- College of Chemistry and Pharmaceutical Sciences & Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
28
|
Debnath N, Yadav P, Mehta PK, Gupta P, Kumar D, Kumar A, Gautam V, Yadav AK. Designer probiotics: Opening the new horizon in diagnosis and prevention of human diseases. Biotechnol Bioeng 2024; 121:100-117. [PMID: 37881101 DOI: 10.1002/bit.28574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/19/2023] [Accepted: 09/23/2023] [Indexed: 10/27/2023]
Abstract
Probiotic microorganisms have been used for therapeutic purposes for over a century, and recent advances in biotechnology and genetic engineering have opened up new possibilities for developing therapeutic approaches using indigenous probiotic microorganisms. Diseases are often related to metabolic and immunological factors, which play a critical role in their onset. With the help of advanced genetic tools, probiotics can be modified to produce or secrete important therapeutic peptides directly into mucosal sites, increasing their effectiveness. One potential approach to enhancing human health is through the use of designer probiotics, which possess immunogenic characteristics. These genetically engineered probiotics hold promise in providing novel therapeutic options. In addition to their immunogenic properties, designer probiotics can also be equipped with sensors and genetic circuits, enabling them to detect a range of diseases with remarkable precision. Such capabilities may significantly advance disease diagnosis and management. Furthermore, designer probiotics have the potential to be used in diagnostic applications, offering a less invasive and more cost-effective alternative to conventional diagnostic techniques. This review offers an overview of the different functional aspects of the designer probiotics and their effectiveness on different diseases and also, we have emphasized their limitations and future implications. A comprehensive understanding of these functional attributes may pave the way for new avenues of prevention and the development of effective therapies for a range of diseases.
Collapse
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT), India
| | - Pooja Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT), India
| | - Praveen K Mehta
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT), India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Deepak Kumar
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok K Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir (UT), India
| |
Collapse
|
29
|
Hu S, Zhou J, Hao J, Zhong Z, Wu H, Zhang P, Yang J, Guo H, Chi J. Emodin ameliorates doxorubicin-induced cardiotoxicity by inhibiting ferroptosis through the remodeling of gut microbiota composition. Am J Physiol Cell Physiol 2024; 326:C161-C176. [PMID: 38009195 DOI: 10.1152/ajpcell.00477.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
The relationship between gut microbiota and doxorubicin-induced cardiotoxicity (DIC) is becoming increasingly clear. Emodin (EMO), a naturally occurring anthraquinone, exerts cardioprotective effects and plays a protective role by regulating gut microbiota composition. Therefore, the protective effect of EMO against DIC injury and its underlying mechanisms are worth investigating. In this study, we analyzed the differences in the gut microbiota in recipient mice transplanted with different flora using 16S-rDNA sequencing, analyzed the differences in serum metabolites among groups of mice using a nontargeted gas chromatography-mass spectrometry coupling system, and assessed cardiac function based on cardiac morphological staining, cardiac injury markers, and ferroptosis indicator assays. We found EMO ameliorated DIC and ferroptosis, as evidenced by decreased myocardial fibrosis, cardiomyocyte hypertrophy, and myocardial disorganization; improved ferroptosis indicators; and the maintenance of normal mitochondrial morphology. The protective effect of EMO was eliminated by the scavenging effect of antibiotics on the gut microbiota. Through fecal microbiota transplantation (FMT), we found that EMO restored the gut microbiota disrupted by doxorubicin (DOX) to near-normal levels. This was evidenced by an increased proportion of Bacteroidota and a decreased proportion of Verrucomicrobiota. FMT resulted in changes in the composition of serum metabolites. Mice transplanted with EMO-improved gut microbiota showed better cardiac function and ferroptosis indices; however, these beneficial effects were not observed in Nrf2 (Nfe2l2)-/- mice. Overall, EMO exerted a protective effect against DIC by attenuating ferroptosis, and the above effects occurred by remodeling the composition of gut microbiota perturbed by DOX and required Nrf2 mediation.NEW & NOTEWORTHY This study demonstrated for the first time the protective effect of emodin against DIC and verified by FMT that its cardioprotective effect was achieved by remodeling gut microbiota composition, resulting in attenuation of ferroptosis. Furthermore, we demonstrated that these effects were mediated by the redox-related gene Nrf2.
Collapse
Affiliation(s)
- Songqing Hu
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, People's Republic of China
- School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jiedong Zhou
- School of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Jinjin Hao
- School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zuoquan Zhong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Haowei Wu
- School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Peipei Zhang
- Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Juntao Yang
- School of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Hangyuan Guo
- School of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Jufang Chi
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, People's Republic of China
| |
Collapse
|
30
|
Andersen ML, Gozal D, Pires GN, Tufik S. Exploring the potential relationships among obstructive sleep apnea, erectile dysfunction, and gut microbiota: a narrative review. Sex Med Rev 2023; 12:76-86. [PMID: 37385976 DOI: 10.1093/sxmrev/qead026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 07/01/2023]
Abstract
INTRODUCTION Poor sleep quality is closely associated with comorbidities affecting a multitude of organ systems. Among the sleep disorders in the population, there has recently been an increase in the prevalence of obstructive sleep apnea (OSA), which has particularly affected men. The intermittent hypoxia and sleep fragmentation associated with OSA can result in the manifestation or aggravation of a number of pathophysiologic conditions, including the impairment of reproductive function in men and women. In this context, erectile dysfunction (ED) is of particular concern. Other consequences of OSA are changes in the gastrointestinal microbiota, with the resultant dysbiosis having potentially harmful consequences that promote downstream exacerbation of various comorbidities. OBJECTIVES This narrative review aims to explore the potential relationships among ED, gut microbiota, and OSA. METHODS A search of the relevant literature was performed in the PubMed, Embase, Medline, and Web of Science databases. RESULTS Sleep is important for regulating the body's functions, and sleep deprivation can negatively affect health. OSA can damage organic functions, including reproductive function, and can lead to ED. Restoring the microbiota and improving sleep can help to improve sexual function or reverse ED and enhance other associated conditions mediated through the gut-brain axis relationship. Probiotics and prebiotics can be used as supportive strategies in the prevention and treatment of OSA, as they help to reduce systemic inflammation and improve intestinal barrier function. CONCLUSION A good diet, a healthy lifestyle, and proper bowel function are essential in controlling depression and several other pathologies. Modulating the gut microbiota through probiotics and prebiotics can provide a viable strategy for developing new therapeutic options in treating many conditions. A better understanding of these a priori unrelated phenomena would foster our understanding of the effects of OSA on human fertility and how changes in gut microbiota may play a role.
Collapse
Affiliation(s)
- Monica Levy Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil
| |
Collapse
|
31
|
Chu NHS, He J, Leung KHT, Ma RCW, Lee JYS, Varney J, Chan JCN, Muir JG, Chow E. Higher Short-Chain Fermentable Carbohydrates Are Associated with Lower Body Fat and Higher Insulin Sensitivity in People with Prediabetes. Nutrients 2023; 15:5070. [PMID: 38140329 PMCID: PMC10745595 DOI: 10.3390/nu15245070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The quality of carbohydrates has metabolic consequences in people with prediabetes. However, the causality of short-chain fermentable carbohydrate intakes and metabolic parameters has not been explored in the prediabetic or diabetic population. We investigated associations between different types of carbohydrates, including fermentable oligosaccharides, disaccharides, monosaccharides, polyols (FODMAPs), and polysaccharides (dietary fibre), and body composition and glucose/insulin responses in subjects with prediabetes. In this prospective cross-sectional study, 177 subjects with impaired glucose tolerance (IGT) (mean age: 60 (54-62) years, 41% men) underwent an assessment of body composition and completed six-point oral glucose tolerance tests (OGTT), Homeostatic Model Assessment of Insulin Resistance (HOMA2-IR), insulin sensitivity, detailed 3-day food records, and physical activity questionnaire. Daily habitual FODMAP intake decreased progressively with increasing BMI, ranging from 7.9 (6.2-12.7) g/d in subjects with normal BMI and 6.6 (4.6-9.9) g/d in subjects with overweight to 5.8 (3.8-9.0) g/d in subjects with obesity (p = 0.038). After adjustment for age and gender, galactooligosaccharides (GOSs) were negatively correlated with body fat (Standardised Beta coefficient β = -0.156, p = 0.006) and positively associated with insulin sensitivity (β = 0.243, p = 0.001). This remained significant after adjustment for macronutrients, fibre, and physical activity (p = 0.035 and p = 0.010, respectively). In individuals with IGT, higher dietary GOS intake was associated with lower body fat and higher insulin sensitivity independent of macronutrients and fibre intake, calling for interventional studies to evaluate the effect of FODMAP intake in prediabetes.
Collapse
Affiliation(s)
- Natural H. S. Chu
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Jie He
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Kathy H. T. Leung
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Ronald C. W. Ma
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jimmy Y. S. Lee
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Jane Varney
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Juliana C. N. Chan
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jane G. Muir
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Elaine Chow
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
32
|
Yan M, Man S, Sun B, Ma L, Guo L, Huang L, Gao W. Gut liver brain axis in diseases: the implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:443. [PMID: 38057297 PMCID: PMC10700720 DOI: 10.1038/s41392-023-01673-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023] Open
Abstract
Gut-liver-brain axis is a three-way highway of information interaction system among the gastrointestinal tract, liver, and nervous systems. In the past few decades, breakthrough progress has been made in the gut liver brain axis, mainly through understanding its formation mechanism and increasing treatment strategies. In this review, we discuss various complex networks including barrier permeability, gut hormones, gut microbial metabolites, vagus nerve, neurotransmitters, immunity, brain toxic metabolites, β-amyloid (Aβ) metabolism, and epigenetic regulation in the gut-liver-brain axis. Some therapies containing antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), polyphenols, low FODMAP diet and nanotechnology application regulate the gut liver brain axis. Besides, some special treatments targeting gut-liver axis include farnesoid X receptor (FXR) agonists, takeda G protein-coupled receptor 5 (TGR5) agonists, glucagon-like peptide-1 (GLP-1) receptor antagonists and fibroblast growth factor 19 (FGF19) analogs. Targeting gut-brain axis embraces cognitive behavioral therapy (CBT), antidepressants and tryptophan metabolism-related therapies. Targeting liver-brain axis contains epigenetic regulation and Aβ metabolism-related therapies. In the future, a better understanding of gut-liver-brain axis interactions will promote the development of novel preventative strategies and the discovery of precise therapeutic targets in multiple diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| | - Benyue Sun
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, 300072, Tianjin, China.
| |
Collapse
|
33
|
Lin K, Dong C, Zhao B, Zhou B, Yang L. Glucagon-like peptide-1 receptor agonist regulates fat browning by altering the gut microbiota and ceramide metabolism. MedComm (Beijing) 2023; 4:e416. [PMID: 38020719 PMCID: PMC10661313 DOI: 10.1002/mco2.416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/22/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Studies have shown that antidiabetic drugs can alter the gut microbiota. The hypoglycemic effects of the drugs can be attributed in part to certain species in the gut microbiome that help the drugs work more effectively. In addition, increasing energy expenditure via the induction of adipose tissue browning has become an appealing strategy to treat obesity and associated metabolic complications. Currently, glucagon-like peptide-1 receptor agonist (GLP-1 RA) treatment for metabolic disorders such as obesity and type 2 diabetes has been widely studied. To determine the mechanism of a long-acting GLP-1 RA affects adipose tissue browning and the gut microbiome, we treated high-fat diet mice with GLP-1 RA and demonstrated that the drug can regulate adipose tissue browning. 16S rRNA and untargeted metabolomics assays suggested that it increased the abundance of bacterium Lactobacillus reuteri and decreased serum ceramide levels in mice. L. reuteri was negatively correlated with ceramide. We found that the mechanism of ceramide decline was alkaline ceramidase 2 (Acer2) overexpression. Moreover, L. reuteri can play a therapeutic synergistic role with GLP-1 RA, suggesting that gut microbiota can be used as a part of the treatment of diabetes.
Collapse
Affiliation(s)
- Ke Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Chunyan Dong
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Binyan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
34
|
Carboni J, Basalely A, Singer P, Castellanos L, Sethna CB. Association Between Dietary Fiber Intake and Cardiometabolic Risk Factors in Adolescents in the United States. J Pediatr 2023; 262:113616. [PMID: 37473987 DOI: 10.1016/j.jpeds.2023.113616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE To determine the association between dietary fiber intake and markers of cardiometabolic risk in adolescents, with blood pressure (BP) as the primary outcome of interest and secondary outcome measures including other established markers of childhood cardiometabolic risk, such as obesity, lipids, albuminuria, estimated glomerular filtration rate (eGFR), and uric acid. STUDY DESIGN Dietary fiber intake was assessed by two 24-hour dietary recall interviews, which were averaged and corrected for body weight. Logistic and linear regression models were used to analyze the cross-sectional association between dietary fiber and cardiometabolic markers. Participants aged 13-17 years in the National Health and Nutritional Examination Survey 2009-2018 who completed a 24-hour dietary recall survey were included. Exclusion criteria included pregnancy, small for gestational age status, and history of major health comorbidities. RESULTS In fully adjusted regression models, low dietary fiber intake was significantly associated with greater diastolic blood pressure (β = -13.29; 95% CI, -20.66 to -5.93), body mass index z-score (β = -0.91; 95% CI, -1.47 to -0.34), and uric acid (β = -0.80; 95% CI, -1.44 to -0.16). CONCLUSIONS The association found between low dietary fiber intake and poor childhood cardiometabolic risk markers indicate a need for prospective studies using fiber intake as a dietary intervention in childhood and as a tool for prevention of many chronic conditions.
Collapse
Affiliation(s)
| | - Abby Basalely
- Zucker School of Medicine at Northwell/Hofstra, Uniondale, NY; Division of Pediatric Nephrology, Cohen Children's Medical Center, New Hyde Park, NY; Feinstein Institutes for Medical Research, Manhasset, NY
| | - Pamela Singer
- Zucker School of Medicine at Northwell/Hofstra, Uniondale, NY; Division of Pediatric Nephrology, Cohen Children's Medical Center, New Hyde Park, NY
| | - Laura Castellanos
- Zucker School of Medicine at Northwell/Hofstra, Uniondale, NY; Division of Pediatric Nephrology, Cohen Children's Medical Center, New Hyde Park, NY
| | - Christine B Sethna
- Zucker School of Medicine at Northwell/Hofstra, Uniondale, NY; Division of Pediatric Nephrology, Cohen Children's Medical Center, New Hyde Park, NY; Feinstein Institutes for Medical Research, Manhasset, NY.
| |
Collapse
|
35
|
Sannino DR, Dobson AJ. Acetobacter pomorum in the Drosophila gut microbiota buffers against host metabolic impacts of dietary preservative formula and batch variation in dietary yeast. Appl Environ Microbiol 2023; 89:e0016523. [PMID: 37800920 PMCID: PMC10617557 DOI: 10.1128/aem.00165-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/05/2023] [Indexed: 10/07/2023] Open
Abstract
Gut microbiota are fundamentally important for healthy function in animal hosts. Drosophila melanogaster is a powerful system for understanding host-microbiota interactions, with modulation of the microbiota inducing phenotypic changes that are conserved across animal taxa. Qualitative differences in diet, such as preservatives and dietary yeast batch variation, may affect fly health indirectly via microbiota, and may potentially have hitherto uncharacterized effects directly on the fly. These factors are rarely considered, controlled, and are not standardized among laboratories. Here, we show that the microbiota's impact on fly triacylglyceride (TAG) levels-a commonly-measured metabolic index-depends on both preservatives and yeast, and combinatorial interactions among the three variables. In studies of conventional, axenic, and gnotobiotic flies, we found that microbial impacts were apparent only on specific yeast-by-preservative conditions, with TAG levels determined by a tripartite interaction of the three experimental factors. When comparing axenic and conventional flies, we found that preservatives caused more variance in host TAG than microbiota status, and certain yeast-preservative combinations even reversed effects of microbiota on TAG. Preservatives had major effects in axenic flies, suggesting either direct effects on the fly or indirect effects via media. However, Acetobacter pomorum buffers the fly against this effect, despite the preservatives inhibiting growth, indicating that this bacterium benefits the host in the face of mutual environmental toxicity. Our results suggest that antimicrobial preservatives have major impacts on host TAG, and that microbiota modulates host TAG dependent on the combination of the dietary factors of preservative formula and yeast batch. IMPORTANCE Drosophila melanogaster is a premier model for microbiome science, which has greatly enhanced our understanding of the basic biology of host-microbe interactions. However, often overlooked factors such as dietary composition, including yeast batch variability and preservative formula, may confound data interpretation of experiments within the same lab and lead to different findings when comparing between labs. Our study supports this notion; we find that the microbiota does not alter host TAG levels independently. Rather, TAG is modulated by combinatorial effects of microbiota, yeast batch, and preservative formula. Specific preservatives increase TAG even in germ-free flies, showing that a commonplace procedure in fly husbandry alters metabolic physiology. This work serves as a cautionary tale that fly rearing methodology can mask or drive microbiota-dependent metabolic changes and also cause microbiota-independent changes.
Collapse
Affiliation(s)
- David R. Sannino
- School of Molecular Biosciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Adam J. Dobson
- School of Molecular Biosciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
36
|
Cheng H, Zhang D, Wu J, Liu J, Zhou Y, Tan Y, Feng W, Peng C. Interactions between gut microbiota and polyphenols: A mechanistic and metabolomic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154979. [PMID: 37552899 DOI: 10.1016/j.phymed.2023.154979] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/30/2023] [Accepted: 07/15/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Polyphenols are a class of naturally sourced compounds with widespread distribution and an extensive array of bioactivities. However, due to their complex constituents and weak absorption, a convincing explanation for their remarkable bioactivity remains elusive for a long time. In recent years, interaction with gut microbiota is hypothesized to be a reasonable explanation of the potential mechanisms for natural compounds especially polyphenols. OBJECTIVES This review aims to present a persuasive explanation for the contradiction between the limited bioavailability and the remarkable bioactivities of polyphenols by examining their interactions with gut microbiota. METHODS We assessed literatures published before April 10, 2023, from several databases, including Scopus, PubMed, Google Scholar, and Web of Science. The keywords used include "polyphenols", "gut microbiota", "short-chain fatty acids", "bile acids", "trimethylamine N-oxide", "lipopolysaccharides" "tryptophan", "dopamine", "intestinal barrier", "central nervous system", "lung", "anthocyanin", "proanthocyanidin", "baicalein", "caffeic acid", "curcumin", "epigallocatechin-3-gallate", "ferulic acid", "genistein", "kaempferol", "luteolin", "myricetin", "naringenin", "procyanidins", "protocatechuic acid", "pterostilbene", "quercetin", "resveratrol", etc. RESULTS: The review first demonstrates that polyphenols significantly alter gut microbiota diversity (α- and β-diversity) and the abundance of specific microorganisms. Polyphenols either promote or inhibit microorganisms, with various factors influencing their effects, such as dosage, treatment duration, and chemical structure of polyphenols. Furthermore, the review reveals that polyphenols regulate several gut microbiota metabolites, including short-chain fatty acids, dopamine, trimethylamine N-oxide, bile acids, and lipopolysaccharides. Polyphenols affect these metabolites by altering gut microbiota composition, modifying microbial enzyme activity, and other potential mechanisms. The changed microbial metabolites induced by polyphenols subsequently trigger host responses in various ways, such as acting as intestinal acid-base homeostasis regulators and activating on specific target receptors. Additionally, polyphenols are transformed into microbial derivatives by gut microbiota and these polyphenols' microbial derivatives have many potential advantages (e.g., increased bioactivity, improved absorption). Lastly, the review shows polyphenols maintain intestinal barrier, central nervous system, and lung function homeostasis by regulating gut microbiota. CONCLUSION The interaction between polyphenols and gut microbiota provides a credible explanation for the exceptional bioactivities of polyphenols. This review aids our understanding of the underlying mechanisms behind the bioactivity of polyphenols.
Collapse
Affiliation(s)
- Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Jing Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Juan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Yaochuan Zhou
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yuzhu Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| |
Collapse
|
37
|
Tedjo DI, Wilbrink JA, Boekhorst J, Timmerman HM, Nienhuijs SW, Stronkhorst A, Savelkoul PHM, Masclee AAM, Penders J, Jonkers DMAE. Impact of Sleeve Gastrectomy on Fecal Microbiota in Individuals with Morbid Obesity. Microorganisms 2023; 11:2353. [PMID: 37764197 PMCID: PMC10537490 DOI: 10.3390/microorganisms11092353] [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: 07/03/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The intestinal microbiota plays an important role in the etiology of obesity. Sleeve gastrectomy (SG) is a frequently performed and effective therapy for morbid obesity. OBJECTIVE To investigate the effect of sleeve gastrectomy on the fecal microbiota of individuals with morbid obesity and to examine whether shifts in microbiota composition are associated with markers of inflammation and intestinal barrier function. METHODS Fecal and blood samples of healthy individuals (n = 27) and morbidly obese individuals pre-SG (n = 24), and at 2 months (n = 13) and 6 months post-SG (n = 9) were collected. The 16SrRNA gene was sequenced to assess microbiota composition. Fecal calprotectin, plasma inflammatory markers and intestinal permeability markers (multi-sugar test) were determined. RESULTS Fecal microbiota composition between morbidly obese and lean individuals was significantly different. The fecal microbiota composition changed significantly 2 and 6 months post-SG (p = 0.008) compared to pre-SG but not towards a more lean profile. The post-SG microbiota profile was characterized by an increase in facultative anaerobic bacteria, characteristic for the upper gastrointestinal tract. No correlations were found between inflammatory markers, intestinal permeability and microbial profile changes. CONCLUSIONS Fecal microbiota composition in morbidly obese individuals changed significantly following SG. This change might be explained by functional changes induced by the SG procedure.
Collapse
Affiliation(s)
- Danyta I. Tedjo
- Division Gastroenterology-Hepatology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (D.I.T.); (J.A.W.); (D.M.A.E.J.)
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (P.H.M.S.); (J.P.)
| | - Jennifer A. Wilbrink
- Division Gastroenterology-Hepatology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (D.I.T.); (J.A.W.); (D.M.A.E.J.)
- Department of Gastroenterology, Zuyderland Ziekenhuis, 6162 Sittard-Geleen, The Netherlands
| | - Jos Boekhorst
- NIZO Food Research B.V., 6718 Ede, The Netherlands; (J.B.); (H.M.T.)
| | | | - Simon W. Nienhuijs
- Department of Surgery and Gastroenterology, Catharina Hospital, 5623 Eindhoven, The Netherlands; (S.W.N.); (A.S.)
| | - Arnold Stronkhorst
- Department of Surgery and Gastroenterology, Catharina Hospital, 5623 Eindhoven, The Netherlands; (S.W.N.); (A.S.)
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (P.H.M.S.); (J.P.)
- Department of Medical Microbiology & Infection Control, VU University Medical Center, 1081 Amsterdam, The Netherlands
| | - Ad A. M. Masclee
- Division Gastroenterology-Hepatology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (D.I.T.); (J.A.W.); (D.M.A.E.J.)
| | - John Penders
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (P.H.M.S.); (J.P.)
| | - Daisy M. A. E. Jonkers
- Division Gastroenterology-Hepatology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, 6229 Maastricht, The Netherlands; (D.I.T.); (J.A.W.); (D.M.A.E.J.)
| |
Collapse
|
38
|
de Queiroz Cavalcanti SA, de Almeida LA, Gasparotto J. Effects of a high saturated fatty acid diet on the intestinal microbiota modification and associated impacts on Parkinson's disease development. J Neuroimmunol 2023; 382:578171. [PMID: 37562163 DOI: 10.1016/j.jneuroim.2023.578171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Recent research has focused on the link between diet, intestinal microbiota, and the impact of excessive consumption of saturated fatty acids. Saturated fatty acids, found in animal fats, dairy, and processed foods, contribute to dysbiosis, increase intestinal barrier permeability, chronic low-grade inflammation, oxidative stress, and dysfunction of the blood-brain barrier, affecting the central nervous system. High intake of saturated fatty acids is associated with an increased risk of developing Parkinson's disease (PD). Diets low in saturated fats, rich in fibers, promote microbial diversity, improve gut health, and potentially reduce the risk of neurodegenerative diseases like PD.
Collapse
Affiliation(s)
| | - Leonardo Augusto de Almeida
- Instituto de Ciências Biomédicas - Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, CEP: 37130-001 Alfenas, Minas Gerais, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas - Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, CEP: 37130-001 Alfenas, Minas Gerais, Brazil.
| |
Collapse
|
39
|
Chen YK, Liu TT, Teia FKF, Xie MZ. Exploring the underlying mechanisms of obesity and diabetes and the potential of Traditional Chinese Medicine: an overview of the literature. Front Endocrinol (Lausanne) 2023; 14:1218880. [PMID: 37600709 PMCID: PMC10433171 DOI: 10.3389/fendo.2023.1218880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Obesity and diabetes are closely related metabolic disorders that have become major public health concerns worldwide. Over the past few decades, numerous studies have explored the underlying mechanisms of these disorders and identified various risk factors, including genetics, lifestyle, and dietary habits. Traditional Chinese Medicine (TCM) has been increasingly recognized for its potential to manage obesity and diabetes. Weight loss is difficult to sustain, and several diabetic therapies, such as sulfonylureas, thiazolidinediones, and insulin, might make it harder to lose weight. While lifestyle changes should be the primary approach for people interested in lowering weight, drugs are also worth investigating. Since some of the newer glucose-lowering medications that cause weight loss, such as glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter 2 inhibitors (SGLT2i), are additionally utilized or are under consideration for use as anti-obesity drugs, the frontier between glucose-lowering medication and weight loss drugs appears to be shifting. This review provides an overview of the literature on the underlying mechanisms of obesity and diabetes and the prospect of TCM in their management. We discuss the various TCM interventions, including acupuncture, herbal medicine, and dietary therapy, and their effects on metabolic health. We also highlight the potential of TCM in regulating gut microbiota, reducing inflammation, and improving insulin sensitivity. The findings suggest that TCM may provide a promising approach to preventing and managing obesity and diabetes. However, further well-designed studies are needed to confirm the efficacy and safety of TCM interventions and to elucidate their underlying mechanisms of action.
Collapse
Affiliation(s)
- Yan-kun Chen
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| | - Ting-ting Liu
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| | - Farah Khameis Farag Teia
- Department of Agro-technology, Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Centre for Research, Khartoum, Sudan
| | - Meng-zhou Xie
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation and Medicated Diet and Dietotherapy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
40
|
Gao Y, Tian T. mTOR Signaling Pathway and Gut Microbiota in Various Disorders: Mechanisms and Potential Drugs in Pharmacotherapy. Int J Mol Sci 2023; 24:11811. [PMID: 37511569 PMCID: PMC10380532 DOI: 10.3390/ijms241411811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) integrates multiple intracellular and extracellular upstream signals involved in the regulation of anabolic and catabolic processes in cells and plays a key regulatory role in cell growth and metabolism. The activation of the mTOR signaling pathway has been reported to be associated with a wide range of human diseases. A growing number of in vivo and in vitro studies have demonstrated that gut microbes and their complex metabolites can regulate host metabolic and immune responses through the mTOR pathway and result in disorders of host physiological functions. In this review, we summarize the regulatory mechanisms of gut microbes and mTOR in different diseases and discuss the crosstalk between gut microbes and their metabolites and mTOR in disorders in the gastrointestinal tract, liver, heart, and other organs. We also discuss the promising application of multiple potential drugs that can adjust the gut microbiota and mTOR signaling pathways. Despite the limited findings between gut microbes and mTOR, elucidating their relationship may provide new clues for the prevention and treatment of various diseases.
Collapse
Affiliation(s)
- Yuan Gao
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
41
|
Chen HH, Wu QJ, Zhang TN, Zhao YH. Gut microbiome and serum short-chain fatty acids are associated with responses to chemo- or targeted therapies in Chinese patients with lung cancer. Front Microbiol 2023; 14:1165360. [PMID: 37564290 PMCID: PMC10411610 DOI: 10.3389/fmicb.2023.1165360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/28/2023] [Indexed: 08/12/2023] Open
Abstract
Background The association between gut microbes and short-chain fatty acids (SCFAs) and therapeutic responses of patients with lung cancer (LC) receiving therapy remains unknown. Methods Fecal and serum samples were prospectively collected from patients with LC, classified as responders, if they presented durable clinical benefits, and non-responders, if not. The composition of gut microbes was analyzed using 16S ribosomal DNA sequencing. Serum SCFA concentrations were detected using gas chromatography. Cell proliferation, migration, invasion, cell cycle, and apoptosis assays were performed on isobutyric acid-treated A549 cells. Reverse transcription-quantitative PCR, Western blotting, immunocytochemistry, and immunofluorescence staining experiments have been performed to investigate the expression of associated genes or proteins. Results Non-responders harbored higher microbiome α-diversity but lower β-diversity compared with responders. Compared to the patients with low α-diversity, those with high α-diversity showed significantly shorter progression-free survival. Additionally, β-diversity has also been observed between these two groups. Specifically, Parasutterella, Clostridiaceae, and Prevotella_7 were more abundant among responders, whereas Bacteroides_stercoris and Christensenellaceae_R-7_group were more abundant in non-responders. The serum SCFA (especially acetate and isobutyrate) levels tended to be higher in responders. Isobutyric acid inhibited the proliferation, migration, and invasion of A549 cells by inducing apoptosis and G1/S arrest while upregulating the expression of GPR41, GPR43, and GPR5C and downregulating that of PAR1, and increasing the activity of histone acetyltransferases. Conclusion We revealed the influence of gut microbiota and SCFAs on the therapeutic responses in patients with LC and the anti-tumor effect of isobutyric acid, indicating their potential use as therapeutic targets.
Collapse
Affiliation(s)
- Huan-Huan Chen
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Cui J, Wang J, Wang Y. The role of short-chain fatty acids produced by gut microbiota in the regulation of pre-eclampsia onset. Front Cell Infect Microbiol 2023; 13:1177768. [PMID: 37600950 PMCID: PMC10432828 DOI: 10.3389/fcimb.2023.1177768] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/21/2023] [Indexed: 08/22/2023] Open
Abstract
Background Preeclampsia (PE) is a common pregnancy-related disorder characterized by disrupted maternal-fetal immune tolerance, involving diffuse inflammatory responses and vascular endothelial damage. Alterations in the gut microbiota (GM) during pregnancy can affect intestinal barrier function and immune balance. Aims and purpose This comprehensive review aims to investigate the potential role of short-chain fatty acids (SCFAs), essential metabolites produced by the GM, in the development of PE. The purpose is to examine their impact on colonic peripheral regulatory T (Treg) cells, the pathogenic potential of antigen-specific helper T (Th) cells, and the inflammatory pathways associated with immune homeostasis. Key insights An increasing body of evidence suggests that dysbiosis in the GM can lead to alterations in SCFA levels, which may significantly contribute to the development of PE. SCFAs enhance the number and function of colonic Treg cells, mitigate the pathogenic potential of GM-specific Th cells, and inhibit inflammatory progression, thereby maintaining immune homeostasis. These insights highlight the potential significance of GM dysregulation and SCFAs produced by GM in the pathogenesis of PE. While the exact causes of PE remain elusive, and definitive clinical treatments are lacking, the GM and SCFAs present promising avenues for future clinical applications related to PE, offering a novel approach for prophylaxis and therapy.
Collapse
Affiliation(s)
| | - Jun Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
43
|
Wang H, Zhao T, Liu Z, Danzengquzhen, Cisangzhuoma, Ma J, Li X, Huang X, Li B. The neuromodulatory effects of flavonoids and gut Microbiota through the gut-brain axis. Front Cell Infect Microbiol 2023; 13:1197646. [PMID: 37424784 PMCID: PMC10327292 DOI: 10.3389/fcimb.2023.1197646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/09/2023] [Indexed: 09/10/2023] Open
Abstract
Recent investigations show that dietary consumption of flavonoids could potentially confer neuroprotective effects through a variety of direct and indirect mechanisms. Numerous flavonoids have been shown to cross the BBB and accumulate within the central nervous system (CNS). Some of these compounds purportedly counteract the accumulation and deleterious effects of reactive oxygen species, fostering neuronal survival and proliferation by inhibiting neuroinflammatory and oxidative stress responses. Moreover, several studies suggest that gut microbiota may participate in regulating brain function and host behavior through the production and modulation of bioactive metabolites. Flavonoids may shape gut microbiota composition by acting as carbon substrates to promote the growth of beneficial bacteria that produce these neuroprotective metabolites, consequently antagonizing or suppressing potential pathogens. By influencing the microbiota-gut-brain axis through this selection process, flavonoids may indirectly improve brain health. This review examines the current state of research into the relationship between bioactive flavonoids, gut microbiota, and the gut-brain axis.
Collapse
Affiliation(s)
- Haoran Wang
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Tingting Zhao
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Danzengquzhen
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Cisangzhuoma
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Jinying Ma
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Xin Li
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| |
Collapse
|
44
|
Liu Y, Shuai P, Chen W, Liu Y, Li D. Association between Helicobacter pylori infection and metabolic syndrome and its components. Front Endocrinol (Lausanne) 2023; 14:1188487. [PMID: 37404306 PMCID: PMC10316390 DOI: 10.3389/fendo.2023.1188487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Background and aim The association between Helicobacter pylori (H. pylori) infection and metabolic syndrome (MetS) has been studied previously; however, the results remain controversial, which could be partly due to the different criteria used for defining MetS. We adopted five MetS criteria to provide better understanding of the association between H. pylori infection and MetS. Methods Physical examination data of 100,708 subjects were obtained from January 2014 to December 2018. MetS was defined based on five criteria including: International Diabetes Federation (IDF), The Third Report of the National Cholesterol Education Program Expert Panel, Adult Treatment Panel III (ATP III), Joint Statement of International Multi-Societies (JIS), Chinese Diabetes Society (CDS), and the Guidelines for the Prevention and Treatment of Type 2 Diabetes in China (2017 edition)(CDS DM). Multivariate logistic regression analysis was performed to elucidate the association between H. pylori infection and MetS and its components. Results The prevalence of MetS defined assessed using IDF, ATP III, JIS, CDS and CDS DM criteria was 15.8%, 19.9%, 23.7%, 8.7% and 15.4%, respectively. In males, the prevalence of MetS assessed using the five criteria in H. pylori-positive group was higher than that in negative-group; however, in females, same results were obtained using the three international criteria. In males, the prevalence of all MetS components was found to be higher in the H. pylori-positive group than those in the negative group; however, in females, only the prevalence of dyslipidemia and waist circumferences exhibited significant differences. Multivariate logistic regression analysis revealed that H. pylori infection in males was positively correlated with MetS. Additionally, H. pylori infection was found to be positively correlated with the waist circumference in the general population, and with hypertension and hyperglycemia in males. Conclusions H. pylori infection was found to be positively associated with MetS in males in China.
Collapse
Affiliation(s)
- Ying Liu
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Ping Shuai
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Wanjing Chen
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Yuping Liu
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Dongyu Li
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Sheykhsaran E, Abbasi A, Ebrahimzadeh Leylabadlo H, Sadeghi J, Mehri S, Naeimi Mazraeh F, Feizi H, Bannazadeh Baghi H. Gut microbiota and obesity: an overview of microbiota to microbial-based therapies. Postgrad Med J 2023; 99:384-402. [PMID: 37294712 DOI: 10.1136/postgradmedj-2021-141311] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/15/2022] [Indexed: 12/14/2022]
Abstract
The increasing prevalence of obesity and overweight is a significant public concern throughout the world. Obesity is a complex disorder involving an excessive amount of body fat. It is not just a cosmetic concern. It is a medical challenge that increases the risk of other diseases and health circumstances, such as diabetes, heart disease, high blood pressure and certain cancers. Environmental and genetic factors are involved in obesity as a significant metabolic disorder along with diabetes. Gut microbiota (GM) has a high potential for energy harvesting from the diet. In the current review, we aim to consider the role of GM, gut dysbiosis and significant therapies to treat obesity. Dietary modifications, probiotics, prebiotics, synbiotics compounds, using faecal microbiota transplant, and other microbial-based therapies are the strategies to intervene in obesity reducing improvement. Each of these factors serves through various mechanisms including a variety of receptors and compounds to control body weight. Trial and animal investigations have indicated that GM can affect both sides of the energy-balancing equation; first, as an influencing factor for energy utilisation from the diet and also as an influencing factor that regulates the host genes and energy storage and expenditure. All the investigated articles declare the clear and inevitable role of GM in obesity. Overall, obesity and obesity-relevant metabolic disorders are characterised by specific modifications in the human microbiota's composition and functions. The emerging therapeutic methods display positive and promising effects; however, further research must be done to update and complete existing knowledge.
Collapse
Affiliation(s)
- Elham Sheykhsaran
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Sciences and Technology Research Institute, Faculty of Nutrition Sciences and food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Javid Sadeghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Mehri
- Department of Biochemistry and structural Biology, University of Alabama, Birmingham, Alabama, USA
| | - Fariba Naeimi Mazraeh
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Feizi
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Zhang P, Zhang D, Lai J, Fu Y, Wu L, Huang H, Pan Y, Jiang J, Xi C, Che Z, Song X, Hu S. Characteristics of the gut microbiota in bipolar depressive disorder patients with distinct weight. CNS Neurosci Ther 2023; 29 Suppl 1:74-83. [PMID: 36604186 PMCID: PMC10314097 DOI: 10.1111/cns.14078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/25/2022] [Accepted: 12/17/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Preliminary studies have indicated metabolic dysfunction and gut dysbiosis in patients with bipolar disorder (BD). In this study, we aimed to clarify the impact of the gut microbial composition and function on metabolic dysfunction in BD patients with an acute depressive episode. METHODS Fresh fecal samples were provided from 58 patients with BD depression, including 29 with normal weight (NW) and 29 with overweight/obesity (OW), and 31 healthy controls (HCs). The hypervariable region of 16 S rRNA gene (V3-V4) sequencing was performed using IonS5TMXL platform to evaluate the bacterial communities. Differences of microbial community and correlation to clinical parameters across different groups were analyzed. RESULTS Compared to NW and HCs, the OW group showed a decreased tendency in alpha diversity index. Beta diversity was markedly different among these groups (PERMANOVA: R2 = 0.034, p = 0.01) and was higher in patients versus HCs. A total number of 24 taxa displayed significantly different abundance among OW, NW, and HCs. At the family level, the abundance of three taxa was remarkably increased in NW, one in OW, and one in HCs. At the genus level, five taxa were enriched in OW, eight in NW, and two in HCs. The relative abundance of the genera Megamonas was positively associated with BMI, while Eggerthella was negatively correlated with BMI. Functional prediction analysis revealed the metabolism of cofactors and vitamins and amino acid were highly enriched in OW compared to HCs. In addition, microbial functions involved in "lipid metabolism" were depleted while the "fructose and mannose metabolism" was enriched in OW compared to NW group. CONCLUSIONS Specific bacterial taxa involved in pathways regulating the lipid, energy, and amino acid metabolisms may underlie the weight concerns in depressed BD patients. Potential targeting gut microbial therapy is provided for overweight/obesity patients with BD, which still need further studies in the future.
Collapse
Affiliation(s)
- Peifen Zhang
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- The First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Danhua Zhang
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Mental Disorder's Management in Zhejiang ProvinceHangzhouChina
- Brain Research Institute of Zhejiang UniversityHangzhouChina
- MOE Frontier Science Center for Brain Science & Brain‐Machine IntegrationZhejiang University
| | - Yaoyang Fu
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lingling Wu
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | | | - Yanmeng Pan
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiajun Jiang
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Caixi Xi
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Ziyuan Che
- College of Agriculture & BiotechnologyZhejiang UniversityHangzhouChina
| | - Xueqin Song
- The First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Mental Disorder's Management in Zhejiang ProvinceHangzhouChina
- Brain Research Institute of Zhejiang UniversityHangzhouChina
- MOE Frontier Science Center for Brain Science & Brain‐Machine IntegrationZhejiang University
| |
Collapse
|
47
|
Mohamad Ishak NS, Ikemoto K. Pyrroloquinoline-quinone to reduce fat accumulation and ameliorate obesity progression. Front Mol Biosci 2023; 10:1200025. [PMID: 37214340 PMCID: PMC10196175 DOI: 10.3389/fmolb.2023.1200025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Obesity is a major health concern worldwide, and its prevalence continues to increase in several countries. Pyrroloquinoline quinone (PQQ) is naturally found in some foods and is available as a dietary supplement in its disodium crystal form. The potential health benefits of PQQ have been studied, considering its antioxidant and anti-inflammatory properties. Furthermore, PQQ has been demonstrated to significantly influence the functions of mitochondria, the organelles responsible for energy production within cells, and their dysfunction is associated with various health conditions, including obesity complications. Here, we explore PQQ properties that can be exploited in obesity treatment and highlight the underlying molecular mechanisms. We review animal and cell culture studies demonstrating that PQQ is beneficial for reducing the accumulation of visceral and hepatic fat. In addition to inhibiting lipogenesis, PQQ can increase mitochondria number and function, leading to improved lipid metabolism. Besides diet-induced obesity, PQQ ameliorates programing obesity of the offspring through maternal supplementation and alters gut microbiota, which reduces obesity risk. In obesity progression, PQQ mitigates mitochondrial dysfunction and obesity-associated inflammation, resulting in the amelioration of the progression of obesity co-morbidities, including non-alcoholic fatty liver disease, chronic kidney disease, and Type 2 diabetes. Overall, PQQ has great potential as an anti-obesity and preventive agent for obesity-related complications. Although human studies are still lacking, further investigations to address obesity and associated disorders are still warranted.
Collapse
|
48
|
Mitropoulou G, Stavropoulou E, Vaou N, Tsakris Z, Voidarou C, Tsiotsias A, Tsigalou C, Taban BM, Kourkoutas Y, Bezirtzoglou E. Insights into Antimicrobial and Anti-Inflammatory Applications of Plant Bioactive Compounds. Microorganisms 2023; 11:1156. [PMID: 37317131 DOI: 10.3390/microorganisms11051156] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/16/2023] Open
Abstract
Plants have long been thought to contribute to health promotion due to their fiber and phenolic content, as well as their inherent biological potential. The bioactive derivatives of medicinal plants are a valuable resource in the fight against serious diseases all around the world. The present review focuses on the current state of knowledge on the usage and medicinal applications of plant bioactives. Issues concerning the effect of aromatic plant derivatives on human gut microbiota and their antimicrobial and anti-inflammatory potentials are discussed and worth further exploring.
Collapse
Affiliation(s)
- Gregoria Mitropoulou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Elisavet Stavropoulou
- Department of Infectious Diseases, Centre Hospitalier Universitaire Vaudois (CHUV), 1101 Lausanne, Switzerland
| | - Natalia Vaou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Zacharias Tsakris
- Laboratory of Microbiology, Department of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Chrysa Voidarou
- Department of Agriculture, University of Ioannina, 47132 Arta, Greece
| | - Arsenis Tsiotsias
- Department of Obstetrics, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Birce Mercanoglou Taban
- Dairy Technology Department, Faculty of Agriculture, Veterinary and Agriculture Campus, Ankara University, Diskapi, Ankara 06110, Turkey
| | - Yiannis Kourkoutas
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
49
|
Zhang L, Zhang R, Li L. Effects of Probiotic Supplementation on Exercise and the Underlying Mechanisms. Foods 2023; 12:foods12091787. [PMID: 37174325 PMCID: PMC10178086 DOI: 10.3390/foods12091787] [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: 04/03/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Long-term, high-intensity exercise can trigger stress response pathways in multiple organs, including the heart and lungs, gastrointestinal tract, skeletal muscle, and neuroendocrine system, thus affecting their material and energy metabolism, immunity, oxidative stress, and endocrine function, and reducing exercise function. As a natural, safe, and convenient nutritional supplement, probiotics have been a hot research topic in the field of biomedical health in recent years. Numerous studies have shown that probiotic supplementation improves the health of the body through the gut-brain axis and the gut-muscle axis, and probiotic supplementation may also improve the stress response and motor function of the body. This paper reviews the progress of research on the role of probiotic supplementation in material and energy metabolism, intestinal barrier function, immunity, oxidative stress, neuroendocrine function, and the health status of the body, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Li Zhang
- Department of Physical Education, China University of Mining and Technology, Beijing 100083, China
| | - Ruhao Zhang
- Department of Physical Education, China University of Mining and Technology, Beijing 100083, China
| | - Lu Li
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
50
|
Soltani S, Ashoori M, Dehghani F, Meshkini F, Clayton ZS, Abdollahi S. Effects of probiotic/synbiotic supplementation on body weight in patients with diabetes: a systematic review and meta-analyses of randomized-controlled trials. BMC Endocr Disord 2023; 23:86. [PMID: 37085813 PMCID: PMC10120130 DOI: 10.1186/s12902-023-01338-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 04/10/2023] [Indexed: 04/23/2023] Open
Abstract
OBJECTIVE The aim of the present study was to assess the effect of probiotic/synbiotic supplementation on anthropometric measures in adults with diabetes, independent of body weight. METHODS PubMed, Scopus, Web of Sciences and the Cochrane Library were searched for randomized controlled trials (RCTs) up until December 14, 2022. The effect sizes were pooled using an inverse-variance random-effects model. The methodological quality of studies as well as the quality of evidence was assessed using standard tools. RESULTS Thirty-two RCTs met the established inclusion criteria. Overall, compared with the respective control groups, probiotic/synbiotic supplementation resulted in a significant reduction in body weight (weighted mean difference [WMD]: -0.50 kg; 95% CI: -0.83, -0.17; I2 = 79.8%, n = 27 studies]), body mass index (WMD: -0.24 kg/m2; 95% CI: -0.39, -0.09; I2 = 85.7%, n = 30 studies), and waist circumference (WMD: -0.90 cm; 95% CI: -1.13, -0.52; I2 = 0%, n = 11 studies). However, hip circumference and waist to hip ratio were not significantly improved. CONCLUSIONS Our analysis revealed that probiotic/synbiotic supplementation may assist with weight management in patients with diabetes, especially when consumed at higher doses, in younger adults, and in participants with obesity. However, more studies are needed to elucidate the anti-obesity effects of specific strains of probiotics/synbiotics.
Collapse
Affiliation(s)
- Sepideh Soltani
- Yazd Cardiovascular Research Center, Noncommunicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marziyeh Ashoori
- School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Dehghani
- Department of nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Fatemeh Meshkini
- Department of Biochemistry, School of medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Shima Abdollahi
- Department of Nutrition, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|