1
|
Zha A, Qi M, Deng Y, Li H, Wang N, Wang C, Liao S, Wan D, Xiong X, Liao P, Wang J, Yin Y, Tan B. Gut Bifidobacterium pseudocatenulatum protects against fat deposition by enhancing secondary bile acid biosynthesis. IMETA 2024; 3:e261. [PMID: 39742294 PMCID: PMC11683477 DOI: 10.1002/imt2.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
Gut microbiome is crucial for lipid metabolism in humans and animals. However, how specific gut microbiota and their associated metabolites impact fat deposition remains unclear. In this study, we demonstrated that the colonic microbiome of lean and obese pigs differentially contributes to fat deposition, as evidenced by colonic microbiota transplantation experiments. Notably, the higher abundance of Bifidobacterium pseudocatenulatum was significantly associated with lower backfat thickness in lean pigs. Microbial-derived lithocholic acid (LCA) species were also significantly enriched in lean pigs and positively correlated with the abundance of B. pseudocatenulatum. In a high-fat diet (HFD)-fed mice model, administration of live B. pseudocatenulatum decreased fat deposition and enhances colonic secondary bile acid biosynthesis. Importantly, pharmacological inhibition of the bile salt hydrolase (BSH), which mediates secondary bile acid biosynthesis, impaired the anti-fat deposition effect of B. pseudocatenulatum in antibiotic-pretreated, HFD-fed mice. Furthermore, dietary LCA also decreased fat deposition in HFD-fed rats and obese pig models. These findings provide mechanistic insights into the anti-fat deposition role of B. pseudocatenulatum and identify BSH as a potential target for preventing excessive fat deposition in humans and animals.
Collapse
Affiliation(s)
- Andong Zha
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- School of Basic Medical Science, Central South UniversityChangshaChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Yuankun Deng
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Hao Li
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Nan Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Chengming Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Simeng Liao
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Peng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Yulong Yin
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Bi'e Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| |
Collapse
|
2
|
Singh V, Mahra K, Jung D, Shin JH. Gut Microbes in Polycystic Ovary Syndrome and Associated Comorbidities; Type 2 Diabetes, Non-Alcoholic Fatty Liver Disease (NAFLD), Cardiovascular Disease (CVD), and the Potential of Microbial Therapeutics. Probiotics Antimicrob Proteins 2024; 16:1744-1761. [PMID: 38647957 DOI: 10.1007/s12602-024-10262-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2024] [Indexed: 04/25/2024]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine anomalies among females of reproductive age, highlighted by hyperandrogenism. PCOS is multifactorial as it can be associated with obesity, insulin resistance, low-grade chronic inflammation, and dyslipidemia. PCOS also leads to dysbiosis by lowering microbial diversity and beneficial microbes, such as Faecalibacterium, Roseburia, Akkermenisa, and Bifidobacterium, and by causing a higher load of opportunistic pathogens, such as Escherichia/Shigella, Fusobacterium, Bilophila, and Sutterella. Wherein, butyrate producers and Akkermansia participate in the glucose uptake by inducing glucagon-like peptide-1 (GLP-1) and glucose metabolism, respectively. The abovementioned gut microbes also maintain the gut barrier function and glucose homeostasis by releasing metabolites such as short-chain fatty acids (SCFAs) and Amuc_1100 protein. In addition, PCOS-associated gut is found to be higher in gut-microbial enzyme β-glucuronidase, causing the de-glucuronidation of conjugated androgen, making it susceptible to reabsorption by entero-hepatic circulation, leading to a higher level of androgen in the circulatory system. Overall, in PCOS, such dysbiosis increases the gut permeability and LPS in the systemic circulation, trimethylamine N-oxide (TMAO) in the circulatory system, chronic inflammation in the adipose tissue and liver, and oxidative stress and lipid accumulation in the liver. Thus, in women with PCOS, dysbiosis can promote the progression and severity of type 2 diabetes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVD). To alleviate such PCOS-associated complications, microbial therapeutics (probiotics and fecal microbiome transplantation) can be used without any side effects, unlike in the case of hormonal therapy. Therefore, this study sought to understand the mechanistic significance of gut microbes in PCOS and associated comorbidities, along with the role of microbial therapeutics that can ease the life of PCOS-affected women.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Kanika Mahra
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - DaRyung Jung
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
3
|
Wu Q, Li W, Kwok LY, Lv H, Sun J, Sun Z. Regional variation and adaptive evolution in Bifidobacterium pseudocatenulatum: Insights into genomic and functional diversity in human gut. Food Res Int 2024; 192:114840. [PMID: 39147525 DOI: 10.1016/j.foodres.2024.114840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Bifidobacterium pseudocatenulatum is a prevalent gut microbe in humans of all ages and plays a crucial role in host health. However, its adaptive evolutionary characteristics remain poorly understood. This study analyzed the genome of 247 B. pseudocatenulatum isolates from Chinese, Vietnamese, Japanese and other region populations using population genomics and functional genomics. Our findings revealed high genetic heterogeneity and regional clustering within B. pseudocatenulatum isolates. Significant differences were observed in genome characteristics, phylogeny, and functional genes. Specifically, Chinese and Vietnamese isolates exhibited a higher abundance of genes involved in the metabolism of plant-derived carbohydrates (GH13, GH43, and GH5 enzyme families), aligning with the predominantly vegetable-, wheat- and fruit-based diets of these populations. Additionally, we found widespread transmission of antibiotic resistance genes (tetO and tetW) through mobile genetic elements, such as genomic islands (GIs), resulting in substantial intra-regional differences. Our findings highlight distinct adaptive evolution in B. pseudocatenulatum driven by gene specialization, possibly in response to regional variations in diet and lifestyle. This study sheds light on bifidobacteria colonization mechanisms in the host gut. IMPORTANCE: Gut microbiota, as a key link in the gut-brain axis, helps to maintain the health of the organism, among which, Bifidobacterium pseudocatenulatum (B. pseudocatenulatum) is an important constituent member of the gut microbiota, which plays an important role in maintaining the balance of gut microbiota. The probiotic properties of B. pseudocatenulatum have been widely elaborated, and in order to excavate its evolutionary features at the genomic level, here we focused on the genetic background and evolutionary mechanism of the B. pseudocatenulatum genomes isolated from the intestinal tracts of different populations. Ultimately, based on the phylogenetic tree, we found that B. pseudocatenulatum has high genetic diversity and regional clustering phenomenon, in which plant-derived carbohydrate metabolism genes (GH13, GH43, GH5) showed significant regional differences, and this genetic differentiation drove the adaptive evolution, which likely shaped by diet and lifestyle.
Collapse
Affiliation(s)
- Qiong Wu
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Weicheng Li
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Collaborative Innovative Center for Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Huimin Lv
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Jiaqi Sun
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China; Collaborative Innovative Center for Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China.
| |
Collapse
|
4
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Huurman RH, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Diet-Induced Obesity in Mice Affects the Maternal Gut Microbiota and Immune Response in Mid-Pregnancy. Int J Mol Sci 2024; 25:9076. [PMID: 39201761 PMCID: PMC11354285 DOI: 10.3390/ijms25169076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/10/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Maternal obesity during pregnancy is associated with adverse pregnancy outcomes. This might be due to undesired obesity-induced changes in the maternal gut microbiota and related changes in the maternal immune adaptations during pregnancy. The current study examines how obesity affects gut microbiota and immunity in pregnant obese and lean mice during mid-pregnancy (gestational day 12 (GD12)). C57BL/6 mice were fed a high-fat diet or low-fat diet from 8 weeks before mating and during pregnancy. At GD12, we analyzed the gut microbiota composition in the feces and immune responses in the intestine (Peyer's patches, mesenteric lymph nodes) and the peripheral circulation (spleen and peripheral blood). Maternal obesity reduced beneficial bacteria (e.g., Bifidobacterium and Akkermansia) and changed intestinal and peripheral immune responses (e.g., dendritic cells, Th1/Th2/Th17/Treg axis, monocytes). Numerous correlations were found between obesity-associated bacterial genera and intestinal/peripheral immune anomalies. This study shows that maternal obesity impacts the abundance of specific bacterial gut genera as compared to lean mice and deranges maternal intestinal immune responses that subsequently change peripheral maternal immune responses in mid-pregnancy. Our findings underscore the opportunities for early intervention strategies targeting maternal obesity, ideally starting in the periconceptional period, to mitigate these obesity-related pregnancy effects.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Romy H. Huurman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Régine P. M. Steegers-Theunissen
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
5
|
Zhang LN, Tan JT, Ng HY, Liao YS, Zhang RQ, Chan KH, Hung IFN, Lam TTY, Cheung KS. Baseline Gut Microbiota Was Associated with Long-Term Immune Response at One Year Following Three Doses of BNT162b2. Vaccines (Basel) 2024; 12:916. [PMID: 39204040 PMCID: PMC11359560 DOI: 10.3390/vaccines12080916] [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: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND This study explored neutralizing IgG antibody levels against COVID-19 decline over time post-vaccination. We conducted this prospective cohort study to investigate the function of gut microbiota in the host immune response following three doses of BNT162b2. METHODS Subjects who received three doses of BNT162b2 were recruited from three centers in Hong Kong. Blood samples were obtained before the first dose and at the one-year timepoint for IgG ELISA to determine the level of neutralizing antibody (NAb). The primary outcome was a high immune response (NAb > 600 AU/mL). We performed shotgun DNA metagenomic sequencing on baseline fecal samples to identify bacterial species and metabolic pathways associated with high immune response using linear discriminant analysis effect size analysis. RESULTS A total of 125 subjects were recruited (median age: 52 years [IQR: 46.2-59.0]; male: 43 [34.4%]), and 20 were regarded as low responders at the one-year timepoint. Streptococcus parasanguinis (log10LDA score = 2.38, p = 0.003; relative abundance of 2.97 × 10-5 vs. 0.03%, p = 0.001), Bacteroides stercoris (log10LDA score = 4.29, p = 0.024; relative abundance of 0.14% vs. 2.40%, p = 0.014) and Haemophilus parainfluenzae (log10LDA score = 2.15, p = 0.022; relative abundance of 0.01% vs. 0, p = 0.010) were enriched in low responders. Bifidobacterium pseudocatenulatum (log10LDA score = 2.99, p = 0.048; relative abundance of 0.09% vs. 0.36%, p = 0.049) and Clostridium leptum (log10LDA score = 2.38, p = 0.014; relative abundance of 1.2 × 10-5% vs. 0, p = 0.044) were enriched in high responders. S. parasanguinis was negatively correlated with the superpathway of pyrimidine ribonucleotides de novo biosynthesis (log10LDA score = 2.63), which contributes to inflammation and antibody production. H. parainfluenzae was positively correlated with pathways related to anti-inflammatory processes, including the superpathway of histidine, purine, and pyrimidine biosynthesis (log10LDA score = 2.14). CONCLUSION Among three-dose BNT162b2 recipients, S. parasanguinis, B. stercoris and H. parainfluenzae were associated with poorer immunogenicity at one year, while B. pseudocatenulatum and C. leptum was associated with a better response.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Jing-Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Ho-Yu Ng
- School of Clinical Medicine, The University of Hong Kong, Hong Kong, China;
| | - Yun-Shi Liao
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, China; (Y.-S.L.); (T.T.-Y.L.)
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong, China
| | - Rui-Qi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Kwok-Hung Chan
- Department of Microbiology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China;
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, China; (Y.-S.L.); (T.T.-Y.L.)
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| |
Collapse
|
6
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Obesity and diet independently affect maternal immunity, maternal gut microbiota and pregnancy outcome in mice. Front Immunol 2024; 15:1376583. [PMID: 39072322 PMCID: PMC11272480 DOI: 10.3389/fimmu.2024.1376583] [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: 01/25/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Maternal obesity poses risks for both mother and offspring during pregnancy, with underlying mechanisms remaining largely unexplored. Obesity is associated with microbial gut dysbiosis and low-grade inflammation, and also the diet has a major impact on these parameters. This study aimed to investigate how maternal obesity and diet contribute to changes in immune responses, exploring potential associations with gut microbiota dysbiosis and adverse pregnancy outcomes in mice. Methods Before mating, C57BL/6 mice were assigned to either a high-fat-diet (HFD) or low-fat-diet (LFD) to obtain obese (n=17) and lean (n=10) mice. To distinguish between the effects of obesity and diet, 7 obese mice were switched from the HFD to the LFD from day 7 until day 18 of pregnancy ("switch group"), which was the endpoint of the study. T helper (Th) cell subsets were studied in the spleen, mesenteric lymph nodes (MLN) and Peyer's patches (PP), while monocyte subsets and activation status were determined in maternal blood (flow cytometry). Feces were collected before and during pregnancy (day 7,14,18) for microbiota analysis (16S rRNA sequencing). Pregnancy outcome included determination of fetal and placental weight. Results Obesity increased splenic Th1 and regulatory T cells, MLN Th1 and PP Th17 cells and enhanced IFN-γ and IL-17A production by splenic Th cells upon ex vivo stimulation. Switching diet decreased splenic and PP Th2 cells and classical monocytes, increased intermediate monocytes and activation of intermediate/nonclassical monocytes. Obesity and diet independently induced changes in the gut microbiota. Various bacterial genera were increased or decreased by obesity or the diet switch. These changes correlated with the immunological changes. Fetal weight was lower in the obese than the lean group, while placental weight was lower in the switch than the obese group. Discussion This study demonstrates that obesity and diet independently impact peripheral and intestinal immune responses at the end of pregnancy. Simultaneously, both factors affect specific bacterial gut genera and lead to reduced fetal or placental weight. Our data suggest that switching diet during pregnancy to improve maternal health is not advisable and it supports pre/probiotic treatment of maternal obesity-induced gut dysbiosis to improve maternal immune responses and pregnancy outcome.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Kochumon S, Malik MZ, Sindhu S, Arefanian H, Jacob T, Bahman F, Nizam R, Hasan A, Thomas R, Al-Rashed F, Shenouda S, Wilson A, Albeloushi S, Almansour N, Alhamar G, Al Madhoun A, Alzaid F, Thanaraj TA, Koistinen HA, Tuomilehto J, Al-Mulla F, Ahmad R. Gut Dysbiosis Shaped by Cocoa Butter-Based Sucrose-Free HFD Leads to Steatohepatitis, and Insulin Resistance in Mice. Nutrients 2024; 16:1929. [PMID: 38931284 PMCID: PMC11207001 DOI: 10.3390/nu16121929] [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: 04/15/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND High-fat diets cause gut dysbiosis and promote triglyceride accumulation, obesity, gut permeability changes, inflammation, and insulin resistance. Both cocoa butter and fish oil are considered to be a part of healthy diets. However, their differential effects on gut microbiome perturbations in mice fed high concentrations of these fats, in the absence of sucrose, remains to be elucidated. The aim of the study was to test whether the sucrose-free cocoa butter-based high-fat diet (C-HFD) feeding in mice leads to gut dysbiosis that associates with a pathologic phenotype marked by hepatic steatosis, low-grade inflammation, perturbed glucose homeostasis, and insulin resistance, compared with control mice fed the fish oil based high-fat diet (F-HFD). RESULTS C57BL/6 mice (5-6 mice/group) were fed two types of high fat diets (C-HFD and F-HFD) for 24 weeks. No significant difference was found in the liver weight or total body weight between the two groups. The 16S rRNA sequencing of gut bacterial samples displayed gut dysbiosis in C-HFD group, with differentially-altered microbial diversity or relative abundances. Bacteroidetes, Firmicutes, and Proteobacteria were highly abundant in C-HFD group, while the Verrucomicrobia, Saccharibacteria (TM7), Actinobacteria, and Tenericutes were more abundant in F-HFD group. Other taxa in C-HFD group included the Bacteroides, Odoribacter, Sutterella, Firmicutes bacterium (AF12), Anaeroplasma, Roseburia, and Parabacteroides distasonis. An increased Firmicutes/Bacteroidetes (F/B) ratio in C-HFD group, compared with F-HFD group, indicated the gut dysbiosis. These gut bacterial changes in C-HFD group had predicted associations with fatty liver disease and with lipogenic, inflammatory, glucose metabolic, and insulin signaling pathways. Consistent with its microbiome shift, the C-HFD group showed hepatic inflammation and steatosis, high fasting blood glucose, insulin resistance, increased hepatic de novo lipogenesis (Acetyl CoA carboxylases 1 (Acaca), Fatty acid synthase (Fasn), Stearoyl-CoA desaturase-1 (Scd1), Elongation of long-chain fatty acids family member 6 (Elovl6), Peroxisome proliferator-activated receptor-gamma (Pparg) and cholesterol synthesis (β-(hydroxy β-methylglutaryl-CoA reductase (Hmgcr). Non-significant differences were observed regarding fatty acid uptake (Cluster of differentiation 36 (CD36), Fatty acid binding protein-1 (Fabp1) and efflux (ATP-binding cassette G1 (Abcg1), Microsomal TG transfer protein (Mttp) in C-HFD group, compared with F-HFD group. The C-HFD group also displayed increased gene expression of inflammatory markers including Tumor necrosis factor alpha (Tnfa), C-C motif chemokine ligand 2 (Ccl2), and Interleukin-12 (Il12), as well as a tendency for liver fibrosis. CONCLUSION These findings suggest that the sucrose-free C-HFD feeding in mice induces gut dysbiosis which associates with liver inflammation, steatosis, glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Shihab Kochumon
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Md. Zubbair Malik
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Sardar Sindhu
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Hossein Arefanian
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Texy Jacob
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatemah Bahman
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheeba Nizam
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Amal Hasan
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Reeby Thomas
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatema Al-Rashed
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Steve Shenouda
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ajit Wilson
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Shaima Albeloushi
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Nourah Almansour
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ghadeer Alhamar
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ashraf Al Madhoun
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fawaz Alzaid
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Thangavel Alphonse Thanaraj
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Heikki A. Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland;
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
| | - Fahd Al-Mulla
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheed Ahmad
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| |
Collapse
|
8
|
Chen Y, Xie C, Lei Y, Ye D, Wang L, Xiong F, Wu H, He Q, Zhou H, Li L, Xing J, Wang C, Zheng M. Theabrownin from Qingzhuan tea prevents high-fat diet-induced MASLD via regulating intestinal microbiota. Biomed Pharmacother 2024; 174:116582. [PMID: 38642504 DOI: 10.1016/j.biopha.2024.116582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
The aim of this study was to investigate whether the therapeutic effect of theabrownin extracted from Qingzhuan tea (QTB) on metabolic dysfunction-associated steatosis liver disease (MASLD) is related to the regulation of intestinal microbiota and its metabolite short-chain fatty acids (SCFAs). Mice were divided into four groups and received normal diet (ND), high-fat diet (HFD) and HFD+QTB (180, 360 mg/kg) for 8 weeks. The results showed that QTB significantly reduced the body weight of HFD mice, ameliorated liver lipid and dyslipidemia, and increased the level of intestinal SCFAs in HFD mice. The results of 16 S rRNA showed that the relative abundance of Bacteroides, Blautia and Lachnoclostridium and their main metabolites acetate and propionate were significantly increased after QTB intervention. The relative abundance of Colidextribacter, Faecalibaculum and Lactobacillus was significantly reduced. QTB can also significantly up-regulate the expression of ATGL, PPARα, FFAR2 and FFAR3, and inhibit the expression of LXRα, SREBP-1c, FAS and HMGCR genes. This makes it possible to act as a prebiotic to prevent MASLD.
Collapse
Affiliation(s)
- Yong Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Chen Xie
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China; Obstetrics and Gynecology of the Second Affiliated Hospital of Hubei University of Science and Technology, Xianning 437100, China
| | - Yining Lei
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Dan Ye
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Le Wang
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Fang Xiong
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Hui Wu
- Xianning Public Inspection Center of Hubei Province, Xianning 437100, China
| | - Qiang He
- Xianning Public Inspection Center of Hubei Province, Xianning 437100, China
| | - Hongfu Zhou
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Ling Li
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Jun Xing
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Cai Wang
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China
| | - Min Zheng
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Hubei Industrial Technology Research Institute of Intelligent Health, Xianning 437100, China.
| |
Collapse
|
9
|
Zeng Q, Qi Z, He X, Luo C, Wen J, Wei J, Yue F, Zhao X, Wei H, Chen T. Bifidobacterium pseudocatenulatum NCU-08 ameliorated senescence via modulation of the AMPK/Sirt1 signaling pathway and gut microbiota in mice. Food Funct 2024; 15:4095-4108. [PMID: 38563760 DOI: 10.1039/d3fo04575g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Aging is a degenerative disease in which organisms and neurological functions decline. Emerging research has underscored the vital role of the gut microbiota in age-related processes. However, the identification of aging-associated core microbiota remains limited. In this investigation, we isolated a strain of B. pseudocatenulatum NCU-08 from the feces of centenarians and assessed its impact on aging using a mouse model induced by D-gal. Our study revealed the exceptional probiotic attributes of B. pseudocatenulatum NCU-08. Administration of B. pseudocatenulatum NCU-08 significantly ameliorated age-related memory impairment, motor dysfunction, and anxiety-like behaviors in aging mice (p < 0.01). Moreover, tissue staining analysis demonstrated that B. pseudocatenulatum NCU-08 reduced the intensity of SA-β-gal-positive in the hippocampus of aging mice. It also reversed pathological damage and structural abnormalities in brain and intestinal tissue. B. pseudocatenulatum NCU-08 inhibited neuroinflammation induced by TLR4/NF-κB (p < 0.01) and preserved the blood-brain barrier integrity by activating the AMPK/Sirt1 pathway (p < 0.05). Furthermore, it mitigated neuronal apoptosis and oxidative stress by upregulating the PI3K/AKT signaling pathway (p < 0.01) and enhancing the activities of antioxidant enzymes, including GSH-Px (p < 0.01), SOD (p < 0.01), and CAT (p < 0.01). Besides, analysis of 16S rRNA sequencing data demonstrated that treatment with B. pseudocatenulatum NCU-08 restored intestinal microbiota homeostasis after senescence. It enhanced the abundance of beneficial bacteria while suppressing the growth of pathogenic microorganisms. In summary, our study unveiled that this novel strain of B. pseudocatenulatum NCU-08 exerts anti-aging effects through regulating the AMPK/Sirt1 pathway and intestinal microbiota. It holds promise as a functional food for promoting anti-aging effects and offers a novel approach to address aging and associated metabolic disorders.
Collapse
Affiliation(s)
- Qingwei Zeng
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, P. R. China
| | - Zhanghua Qi
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
| | - Xia He
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
| | - Chuanlin Luo
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
| | - Jianing Wen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
| | - Fenfang Yue
- School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Xuanqi Zhao
- School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Hong Wei
- The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhong Shan Er Lu, Guangzhou 510080, P. R. China.
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, P. R. China
| |
Collapse
|
10
|
Li B, Zhang B, Zhang F, Liu X, Zhang Y, Peng W, Teng D, Mao R, Yang N, Hao Y, Wang J. Interaction between Dietary Lactoferrin and Gut Microbiota in Host Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7596-7606. [PMID: 38557058 DOI: 10.1021/acs.jafc.3c09050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The gut microbiota are known to play an important role in host health and disease. Alterations in the gut microbiota composition can disrupt the stability of the gut ecosystem, which may result in noncommunicable chronic diseases (NCCDs). Remodeling the gut microbiota through personalized nutrition is a novel therapeutic avenue for both disease control and prevention. However, whether there are commonly used gut microbiota-targeted diets and how gut microbiota-diet interactions combat NCCDs and improve health remain questions to be addressed. Lactoferrin (LF), which is broadly used in dietary supplements, acts not only as an antimicrobial in the defense against enteropathogenic bacteria but also as a prebiotic to propagate certain probiotics. Thus, LF-induced gut microbiota alterations can be harnessed to induce changes in host physiology, and the underpinnings of their relationships and mechanisms are beginning to unravel in studies involving humans and animal models.
Collapse
Affiliation(s)
- Bing Li
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Bo Zhang
- International Joint Research Laboratory for Biomedical Nanomaterials of Henan, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Fuli Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Xiaomeng Liu
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Yunxia Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Weifeng Peng
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Da Teng
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Ruoyu Mao
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Na Yang
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Ya Hao
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Jianhua Wang
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| |
Collapse
|
11
|
Beckers KF, Flanagan JP, Sones JL. Microbiome and pregnancy: focus on microbial dysbiosis coupled with maternal obesity. Int J Obes (Lond) 2024; 48:439-448. [PMID: 38145995 PMCID: PMC10978494 DOI: 10.1038/s41366-023-01438-7] [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: 08/25/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
Obesity is becoming a worldwide pandemic with over one billion people affected. Of women in the United States, who are of childbearing age, two-thirds of them are considered overweight/obese. Offspring of women with obesity have a greater likelihood of developing cardiometabolic disease later in life, therefore making obesity a transgenerational issue. Emerging topics such as maternal microbial dysbiosis with altered levels of bacterial phyla and maternal obesity programming offspring cardiometabolic disease are a novel area of research discussed in this review. In the authors' opinion, beneficial therapeutics will be developed from knowledge of bacterial-host interactions at the most specific level possible. Although there is an abundance of obesity-related microbiome research, it is not concise, readily available, nor easy to interpret at this time. This review details the current knowledge regarding the relationship between obesity and the gut microbiome, with an emphasis on maternal obesity.
Collapse
Affiliation(s)
- Kalie F Beckers
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Juliet P Flanagan
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jenny L Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
- Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, USA.
| |
Collapse
|
12
|
Aref M, FaragAllah EM, Goda NIA, Abu-Alghayth MH, Abomughaid MM, Mahboub HH, Alwutayd KM, Elsherbini HA. Chia seeds ameliorate cardiac disease risk factors via alleviating oxidative stress and inflammation in rats fed high-fat diet. Sci Rep 2024; 14:2940. [PMID: 38316807 PMCID: PMC10844609 DOI: 10.1038/s41598-023-41370-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/25/2023] [Indexed: 02/07/2024] Open
Abstract
Obesity upsurges the risk of developing cardiovascular disease, primarily heart failure and coronary heart disease. Chia seeds have a high concentration of dietary fiber and increased concentrations of anti-inflammatoryand antioxidant compounds. They are used for weight loss plus enhancing blood glucose and lipid profile. The current perspective was commenced to examine the protective influence of chia seeds ingestion on cardiovascular disease risk factors in high-fat diet-fed rats. Forty male albino rats (with an initial body weight of 180-200 g) were used in this study. Rats were randomly and equally divided into 4 groups: Group I was the control group and group II was a control group with chia seeds supplementation. Group III was a high-fat diet group (HFD) that received HFD for 10 weeks and group IV was fed on HFD plus chia seeds for 10 weeks. In all groups Echocardiographic measurements were performed, initial and final BMI, serum glucose, AC/TC ratio, lipid profile, insulin (with a computed HOMA-IR), creatinine phosphokinase-muscle/brain (CPK-MB), CRP, and cardiac troponin I (cTnI) and MAP were estimated. Whole heart weight (WHW) was calculated, and then WHW/body weight (BW) ratio was estimated. Eventually, a histopathological picture of cardiac tissues was performed to assess the changes in the structure of the heart under Haematoxylin and Eosin and Crossmon's trichrome stain. Ingestion of a high diet for 10 weeks induced a clear elevation in BMI, AC/ TC, insulin resistance, hyperlipidemia, CRP, CPK-MB, and cTnI in all HFD groups. Moreover, there was a significant increase in MAP, left ventricular end diastolic diameter (LVEDD), and left ventricular end systolic diameter (LVESD). Furthermore, histological cardiac examination showed structural alteration of the normal structure of the heart tissue with an increase in collagen deposition. Also, the Bcl-2 expression in the heart muscle was significantly lower, but Bax expression was significantly higher. Chia seeds ingestion combined with HFD noticeably ameliorated the previously-recorded biochemical biomarkers, hemodynamic and echocardiography measures, and histopathological changes. Outcomes of this report reveal that obesity is a hazard factor for cardiovascular disease and chia seeds could be a good candidate for cardiovascular system protection.
Collapse
Affiliation(s)
- Mohamed Aref
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Sharkia, Egypt
| | | | - Nehal I A Goda
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Mohammed H Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, Al Nakhil, 67714, Bisha, Saudi Arabia
| | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, Al Nakhil, 67714, Bisha, Saudi Arabia
| | - Heba H Mahboub
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, Egypt.
| | - Khairiah Mubarak Alwutayd
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Hadeel A Elsherbini
- Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
13
|
Van Syoc EP, Damani J, DiMattia Z, Ganda E, Rogers CJ. The Effects of Bifidobacterium Probiotic Supplementation on Blood Glucose: A Systematic Review and Meta-Analysis of Animal Models and Clinical Evidence. Adv Nutr 2024; 15:100137. [PMID: 37923223 PMCID: PMC10831893 DOI: 10.1016/j.advnut.2023.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/21/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Probiotic supplementation is a potential therapeutic for metabolic diseases, including obesity, metabolic syndrome (MetS), and type 2 diabetes (T2D), but most studies deliver multiple species of bacteria in addition to prebiotics or oral pharmaceuticals. This may contribute to conflicting evidence in existing meta-analyses of probiotics in these populations and warrants a systematic review of the literature to assess the contribution of a single probiotic genus to better understand the contribution of individual probiotics to modulate blood glucose. We conducted a systematic review and meta-analysis of animal studies and human randomized controlled trials (RCTs) to assess the effects of Bifidobacterium (BF) probiotic supplementation on markers of glycemia. In a meta-analysis of 6 RCTs, BF supplementation had no effect on fasting blood glucose {FBG; mean difference [MD] = -1.99 mg/dL [95% confidence interval (CI): -4.84, 0.86], P = 0.13}, and there were no subgroup differences between subjects with elevated FBG concentrations and normoglycemia. However, BF supplementation reduced FBG concentrations in a meta-analysis comprised of studies utilizing animal models of obesity, MetS, or T2D [n = 16; MD = -36.11 mg/dL (CI: -49.04, -23.18), P < 0.0001]. Translational gaps from animal to human trials include paucity of research in female animals, BF supplementation in subjects that were normoglycemic, and lack of methodologic reporting regarding probiotic viability and stability. More research is necessary to assess the effects of BF supplementation in human subjects with elevated FBG concentrations. Overall, there was consistent evidence of the efficacy of BF probiotics to reduce elevated FBG concentrations in animal models but not clinical trials, suggesting that BF alone may have minimal effects on glycemic control, may be more effective when combined with multiple probiotic species, or may be more effective in conditions of hyperglycemia rather than elevated FBG concentrations.
Collapse
Affiliation(s)
- Emily P Van Syoc
- Dual-Title Ph.D Program in Integrative & Biomedical Physiology and Clinical & Translational Science, The Pennsylvania State University, University Park, PA, United States; Department of Animal Science, The Pennsylvania State University, University Park, PA, United States; The One Health Microbiome Center, The Pennsylvania State University, University Park, PA, United States
| | - Janhavi Damani
- The Intercollege Graduate Degree Program in Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Zachary DiMattia
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Erika Ganda
- Department of Animal Science, The Pennsylvania State University, University Park, PA, United States; The One Health Microbiome Center, The Pennsylvania State University, University Park, PA, United States
| | - Connie J Rogers
- Department of Nutritional Sciences, University of Georgia, Athens, GA, United States.
| |
Collapse
|
14
|
Sun QY, Wang XY, Huang ZP, Song J, Zheng ED, Gong FH, Huang XW. Depletion of gut microbiota facilitates fibroblast growth factor 21-mediated protection against acute pancreatitis in diabetic mice. World J Diabetes 2023; 14:1824-1838. [PMID: 38222783 PMCID: PMC10784798 DOI: 10.4239/wjd.v14.i12.1824] [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: 08/22/2023] [Revised: 09/28/2023] [Accepted: 11/25/2023] [Indexed: 12/14/2023] Open
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21), primarily secreted by the pancreas, liver, and adipose tissues, plays a pivotal role in regulating glucose and lipid metabolism. Acute pancreatitis (AP) is a common inflammatory disease with specific clinical manifestations. Many patients with diabetes present with concurrent inflammatory symptoms. Diabetes exacerbates intestinal permeability and intestinal inflammation, thus leading to the progression to AP. Our previous study indicated that FGF21 significantly attenuated susceptibility to AP in mice. AIM To investigate the potential protective role of FGF21 against AP in diabetic mice. METHODS In the present study, a mouse model of AP was established in diabetic (db)/db diabetic mice through ceruletide injections. Thereafter, the protective effects of recombinant FGF21 protein against AP were evaluated, with an emphasis on examining serum amylase (AMS) levels and pancreatic and intestinal inflammatory cytokines [interleukin (IL)-6, tumor necrosis factor-alpha (TNF-), and intestinal IL-1β]. Additionally, the impact of this treatment on the histopathologic changes of the pancreas and small intestinal was examined to elucidate the role of FGF21 in diabetic mice with AP. An antibiotic (Abx) cocktail was administered in combination with FGF21 therapy to investigate whether the effect of FGF21 on AP in diabetic mice with AP was mediated through the modulation of the gut microbiota. Subsequently, the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt), a bioinformatics software package, was used to predict different pathways between the groups and to explore the potential mechanisms by which the gut microbiota influenced the protective effect of FGF21. RESULTS The results indicated that FGF21 notably diminished the levels of serum AMS (944.5 ± 15.9 vs 1732 ± 83.9, P < 0.01) and inflammatory factors including IL-6 (0.2400 ± 0.55 vs 1.233 ± 0.053, P < 0.01), TNF- (0.7067 ± 0.22 vs 1.433 ± 0.051, P < 0.01), and IL-1β (1.377 ± 0.069 vs 0.3328 ± 0.02542, P < 0.01) in diabetic mice with AP. Moreover, notable signs of recovery were observed in the pancreatic structure of the mice. The histologic evidence of inflammation in the small intestine, including edema and villous damage, was significantly alleviated. FGF21 also significantly altered the composition of the gut microbiota, reestablishing the Bacteroidetes/Firmicutes ratio. Upon treatment with an Abx cocktail to deplete the gut microbiota, the FGF21 + Abx group showed lower levels of serum AMS (0.9328 ± 0.075 vs 0.2249 ± 0.023, P < 0.01) and inflammatory factors (1.083 ± 0.12 vs 0.2799 ± 0.032, p < 0.01) than the FGF21 group. Furthermore, the FGF21 + Abx group exhibited diminished injury to the pancreatic and small intestinal tissues, accompanied by a significant decrease in blood glucose levels (17.50 ± 1.1 vs 9.817 ± 0.69 mmol/L, P < 0.001). These findings underscored the superior protective effects of the combination therapy involving an Abx cocktail with FGF21 over the FGF21 treatment alone in diabetic mice with AP. The gut microbiota composition across different groups was further characterized, and a differential expression analysis of gene functions was undertaken using the PICRUSt2 prediction method. These findings suggested that FGF21 could potentially confer therapeutic effects on diabetic mice with AP by modulating the sulfate reduction I pathway and the superpathway of n-acetylceramide degradation in the gut microbiota. CONCLUSION This study reveals the potential of FGF21 in improving pancreatic and intestinal damage recovery, reducing blood glucose levels, and reshaping gut microbiota composition in diabetic mice with AP. Notably, the protective effects of FGF21 are augmented when combined with the Abx cocktail.
Collapse
Affiliation(s)
- Qi-Yan Sun
- School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
- Zhejiang Medical Products Service Center, Hangzhou 310012, Zhejiang Province, China
| | - Xu-Ye Wang
- School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Zu-Pin Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, Zhejiang Province, China
| | - Jing Song
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, Zhejiang Province, China
| | - En-Dong Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, Zhejiang Province, China
| | - Fang-Hua Gong
- School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, Zhejiang Province, China
| | - Xiao-Wang Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, Zhejiang Province, China
| |
Collapse
|
15
|
Luo J, Wu YX, Ma LT, Zhang MY, Li FT, Cai YY, Zheng F, Yue H. Immunomodulatory effect of Liangyi paste on the gut microbiota of mice. J Pharm Biomed Anal 2023; 236:115706. [PMID: 37738734 DOI: 10.1016/j.jpba.2023.115706] [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: 07/19/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/24/2023]
Abstract
Liangyi paste (LY) is a traditional Chinese medicine made from a mixture of Ginseng and Rehmanniae radix praeparata. The present study aimed to investigate the effects of LY on gut microbiota diversity in immunocompromised mice. The chemical composition of LY extract was analyzed using UPLC-Q-Orbitrap-MS/MS, and the differences in the structure and diversity of the intestinal microbiota of LY extract were examined using 16S rRNA. In this study, identified and analyzed 66 compounds from the LY. These compounds included 11 iridoids, 6 oligosaccharides, 21 protopanaxtriols, 23 protopanaxadiols, 2 OLE, 1 Ionone and 2 phenylethanoside, using advanced UPLC-Q-Orbitrap-MS/MS technology. Through the use of 16S rRNA analysis, the study found that LY significantly increased the relative abundance of the Firmicutes phylum in immunocompromised mice, while decreasing the abundance of the Proteobacteria and Actinobacteria phyla. At the genus level, LY significantly increased the relative abundance of beneficial bacteria such as Clostridium_sensu_stricto_l, Lactobacillus, and Limosilactobacillus in immunocompromised mice. Conversely, the paste extract decreased the relative abundance of harmful bacteria such as Enterococcus and Escherichia Shigella in immunocompromised mice. These findings highlight the potential of LY to serve as a natural dietary supplement for enhancing gut microbiota diversity and promoting gut health. The identification of numerous compounds within the paste extract demonstrates its complexity and potential as a source for further research and development. Additionally, the LY extract exerted a significant influence on both nucleotide and amino acid metabolism. To sum up, the findings suggest that the LY extract has the potential to modulate the structure and diversity of gut microbiota, as well as promote metabolic balance in immunocompromised mice.
Collapse
Affiliation(s)
- Jing Luo
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Yong-Xi Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Li-Ting Ma
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Mei-Yu Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Fang-Tong Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Yong-Yu Cai
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Fei Zheng
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Hao Yue
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| |
Collapse
|
16
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
17
|
Pongen YL, Thirumurugan D, Ramasubburayan R, Prakash S. Harnessing actinobacteria potential for cancer prevention and treatment. Microb Pathog 2023; 183:106324. [PMID: 37633504 DOI: 10.1016/j.micpath.2023.106324] [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: 07/19/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Actinobacteria are gram-positive bacteria with high G:C ratio in their genetic makeup. They have been noted and studied for their capacity to produce bioactive substances with a range of uses in human health, and they also exhibit a unique property of adapting to extreme environments quite well. Actinobacteria may play an essential role in cancer prevention and treatment due to their synthesis of anticancer compounds, as indicated by recent studies. The aim of this review is to give a summary of what is currently known about the connection between actinobacteria and different types of cancer. This paper delineates the diverse array of actinobacterial bioactive compounds possessing anticancer properties, elucidates their mechanisms of action and explores potential applications in cancer treatment. Furthermore, this review highlights how the microbiome influences the onset and progression of cancer, as well as the discussing the potential benefits that actinobacteria may bring in terms of controlling the microbiome and contributing to the regulation of the tumour microenvironment to cure or prevent cancer.
Collapse
Affiliation(s)
- Yimtar L Pongen
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur - 603 203, Chengalpattu District, Tamil Nadu, India
| | - Durairaj Thirumurugan
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur - 603 203, Chengalpattu District, Tamil Nadu, India.
| | - Ramasamy Ramasubburayan
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai - 600 077, Tamil Nadu, India
| | - Santhiyagu Prakash
- Marine Biotechnology Laboratory, Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, (OMR Campus), Tamilnadu Dr. J. Jayalalithaa Fisheries University, Vaniyanchavadi, Chennai - 603 103, Tamil Nadu, India.
| |
Collapse
|
18
|
Li Y, Han L, Liu J, Kang L, Zhao L, Cui K. Yeast Peptides Improve the Intestinal Barrier Function and Alleviate Weaning Stress by Changing the Intestinal Microflora Structure of Weaned Lambs. Microorganisms 2023; 11:2472. [PMID: 37894129 PMCID: PMC10608930 DOI: 10.3390/microorganisms11102472] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Early weaning stress in lambs leads to decreased feed intake, damage to intestinal morphology, changes in the microbial flora structure, and subsequent complications. Yeast peptides are antimicrobial peptides with anti-inflammatory, antioxidant, and bacteriostasis effects. To study the effects of yeast peptides on relieving weaning stress in lambs, 54 lambs were randomly divided into three groups: ewe-reared (ER), yeast-peptide-treated (AP), and early-weaned (EW) lambs. The body weight and dry matter intake did not significantly differ among all groups. After weaning, the daily gain and feed conversion rate decreased significantly (p < 0.01), but AP showed an upward trend. In the EW group, immunoglobulin (Ig) levels changed significantly post-weaning (IgG decreased; IgA and IgM increased); the villi shortened, the crypt depth increased, and the villi height/crypt depth decreased (p < 0.001). The abundance and diversity of microflora among all groups were not significantly different. A column coordinate analysis showed significant differences in the intestinal microbial structure between the AP and EW groups. Lactobacillus, Aeriscardovia, Ruminosaceae_UCG-014, and Catenisphaera may play key roles in alleviating weaning stress in lambs. Our study provides new clues for alleviating weaning stress in lambs by describing the influence of yeast peptides on the intestinal microflora during weaning.
Collapse
Affiliation(s)
- Yanjun Li
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China;
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China; (L.H.); (J.L.); (L.K.)
| | - Lulu Han
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China; (L.H.); (J.L.); (L.K.)
| | - Jie Liu
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China; (L.H.); (J.L.); (L.K.)
| | - Lingyun Kang
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China; (L.H.); (J.L.); (L.K.)
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Kai Cui
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China; (L.H.); (J.L.); (L.K.)
| |
Collapse
|
19
|
Pepe RB, Lottenberg AM, Fujiwara CTH, Beyruti M, Cintra DE, Machado RM, Rodrigues A, Jensen NSO, Caldas APS, Fernandes AE, Rossoni C, Mattos F, Motarelli JHF, Bressan J, Saldanha J, Beda LMM, Lavrador MSF, Del Bosco M, Cruz P, Correia PE, Maximino P, Pereira S, Faria SL, Piovacari SMF. Position statement on nutrition therapy for overweight and obesity: nutrition department of the Brazilian association for the study of obesity and metabolic syndrome (ABESO-2022). Diabetol Metab Syndr 2023; 15:124. [PMID: 37296485 PMCID: PMC10251611 DOI: 10.1186/s13098-023-01037-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a chronic disease resulting from multifactorial causes mainly related to lifestyle (sedentary lifestyle, inadequate eating habits) and to other conditions such as genetic, hereditary, psychological, cultural, and ethnic factors. The weight loss process is slow and complex, and involves lifestyle changes with an emphasis on nutritional therapy, physical activity practice, psychological interventions, and pharmacological or surgical treatment. Because the management of obesity is a long-term process, it is essential that the nutritional treatment contributes to the maintenance of the individual's global health. The main diet-related causes associated with excess weight are the high consumption of ultraprocessed foods, which are high in fats, sugars, and have high energy density; increased portion sizes; and low intake of fruits, vegetables, and grains. In addition, some situations negatively interfere with the weight loss process, such as fad diets that involve the belief in superfoods, the use of teas and phytotherapics, or even the avoidance of certain food groups, as has currently been the case for foods that are sources of carbohydrates. Individuals with obesity are often exposed to fad diets and, on a recurring basis, adhere to proposals with promises of quick solutions, which are not supported by the scientific literature. The adoption of a dietary pattern combining foods such as grains, lean meats, low-fat dairy, fruits, and vegetables, associated with an energy deficit, is the nutritional treatment recommended by the main international guidelines. Moreover, an emphasis on behavioral aspects including motivational interviewing and the encouragement for the individual to develop skills will contribute to achieve and maintain a healthy weight. Therefore, this Position Statement was prepared based on the analysis of the main randomized controlled studies and meta-analyses that tested different nutrition interventions for weight loss. Topics in the frontier of knowledge such as gut microbiota, inflammation, and nutritional genomics, as well as the processes involved in weight regain, were included in this document. This Position Statement was prepared by the Nutrition Department of the Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), with the collaboration of dietitians from research and clinical fields with an emphasis on strategies for weight loss.
Collapse
Affiliation(s)
- Renata Bressan Pepe
- Grupo de Obesidade e Sindrome Metabolica, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP Brazil
| | - Ana Maria Lottenberg
- Laboratório de Lipides (LIM10), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP Brazil
- Nutrition Department of the Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), Rua Mato Grosso 306 – cj 1711, Sao Paulo, SP 01239-040 Brazil
| | - Clarissa Tamie Hiwatashi Fujiwara
- Grupo de Obesidade e Sindrome Metabolica, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP Brazil
| | - Mônica Beyruti
- Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), São Paulo, SP Brazil
| | - Dennys Esper Cintra
- Centro de Estudos em Lipídios e Nutrigenômica – CELN – University of Campinas, Campinas, SP Brazil
| | - Roberta Marcondes Machado
- Liga Acadêmica de Controle de Diabetes do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP Brazil
| | - Alessandra Rodrigues
- Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), São Paulo, SP Brazil
| | - Natália Sanchez Oliveira Jensen
- Liga Acadêmica de Controle de Diabetes do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP Brazil
| | | | - Ariana Ester Fernandes
- Grupo de Obesidade e Sindrome Metabolica, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP Brazil
| | - Carina Rossoni
- Instituto de Saúde Ambiental, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Fernanda Mattos
- Programa de Obesidade e Cirurgia Bariátrica do Hospital Universitário Clementino Fraga Filho da UFRJ, Rio de Janeiro, RJ Brazil
| | - João Henrique Fabiano Motarelli
- Núcleo de Estudos e Extensão em Comportamento Alimentar e Obesidade (NEPOCA) da Universidade de São Paulo - FMRP/USP, Ribeirão Preto, Brazil
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG Brazil
| | | | - Lis Mie Masuzawa Beda
- Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), São Paulo, SP Brazil
| | - Maria Sílvia Ferrari Lavrador
- Liga Acadêmica de Controle de Diabetes do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP Brazil
| | - Mariana Del Bosco
- Brazilian Association for the Study of Obesity and Metabolic Syndrome (ABESO), São Paulo, SP Brazil
| | - Patrícia Cruz
- Grupo de Obesidade e Sindrome Metabolica, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP Brazil
| | | | - Priscila Maximino
- Instituto PENSI - Fundação José Luiz Egydio Setúbal, Instituto Pensi, Fundação José Luiz Egydio Setúbal, Hospital Infantil Sabará, São Paulo, SP Brazil
| | - Silvia Pereira
- Núcleo de Saúde Alimentar da Sociedade Brasileira de Cirurgia Bariátrica e Metabólica, São Paulo, Brazil
| | | | | |
Collapse
|
20
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
21
|
Interaction between gut microbiota and sex hormones and their relation to sexual dimorphism in metabolic diseases. Biol Sex Differ 2023; 14:4. [PMID: 36750874 PMCID: PMC9903633 DOI: 10.1186/s13293-023-00490-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Metabolic diseases, such as obesity, metabolic syndrome (MetS) and type 2 diabetes (T2D), are now a widespread pandemic in the developed world. These pathologies show sex differences in their development and prevalence, and sex steroids, mainly estrogen and testosterone, are thought to play a prominent role in this sexual dimorphism. The influence of sex hormones on these pathologies is not only reflected in differences between men and women, but also between women themselves, depending on the hormonal changes associated with the menopause. The observed sex differences in gut microbiota composition have led to multiple studies highlighting the interaction between steroid hormones and the gut microbiota and its influence on metabolic diseases, ultimately pointing to a new therapy for these diseases based on the manipulation of the gut microbiota. This review aims to shed light on the role of sexual hormones in sex differences in the development and prevalence of metabolic diseases, focusing on obesity, MetS and T2D. We focus also the interaction between sex hormones and the gut microbiota, and in particular the role of microbiota in aspects such as gut barrier integrity, inflammatory status, and the gut-brain axis, given the relevance of these factors in the development of metabolic diseases.
Collapse
|
22
|
Pezzino S, Sofia M, Faletra G, Mazzone C, Litrico G, La Greca G, Latteri S. Gut-Liver Axis and Non-Alcoholic Fatty Liver Disease: A Vicious Circle of Dysfunctions Orchestrated by the Gut Microbiome. BIOLOGY 2022; 11:1622. [PMID: 36358323 PMCID: PMC9687983 DOI: 10.3390/biology11111622] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 09/24/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent, multifactorial, and poorly understood liver disease with an increasing incidence worldwide. NAFLD is typically asymptomatic and coupled with other symptoms of metabolic syndrome. The prevalence of NAFLD is rising in tandem with the prevalence of obesity. In the Western hemisphere, NAFLD is one of the most prevalent causes of liver disease and liver transplantation. Recent research suggests that gut microbiome dysbiosis may play a significant role in the pathogenesis of NAFLD by dysregulating the gut-liver axis. The so-called "gut-liver axis" refers to the communication and feedback loop between the digestive system and the liver. Several pathological mechanisms characterized the alteration of the gut-liver axis, such as the impairment of the gut barrier and the increase of the intestinal permeability which result in endotoxemia and inflammation, and changes in bile acid profiles and metabolite levels produced by the gut microbiome. This review will explore the role of gut-liver axis disruption, mediated by gut microbiome dysbiosis, on NAFLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| |
Collapse
|
23
|
Drey E, Kok CR, Hutkins R. Role of Bifidobacterium pseudocatenulatum in Degradation and Consumption of Xylan-Derived Carbohydrates. Appl Environ Microbiol 2022; 88:e0129922. [PMID: 36200766 PMCID: PMC9599329 DOI: 10.1128/aem.01299-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Xylans, a family of xylose-based polysaccharides, are dietary fibers resistant to digestion. They therefore reach the large intestine intact; there, they are utilized by members of the gut microbiota. They are initially broken down by primary degraders that utilize extracellular xylanases to cleave xylan into smaller oligomers. The resulting xylooligosaccharides (XOS) can either be further metabolized directly by primary degraders or cross-feed secondary consumers, including Bifidobacterium. While several Bifidobacterium species have metabolic systems for XOS, most grow poorly on longer-chain XOS and xylan substrates. In this study, we isolated strains of Bifidobacterium pseudocatenulatum and observed that some, including B. pseudocatenulatum ED02, displayed growth on XOS with a high degree of polymerization (DP) and straight-chain xylan, suggesting a primary degrader phenotype that is rare in Bifidobacterium. In silico analyses revealed that only the genomes of these xylan-fermenting (xylan+) strains contained an extracellular GH10 endo-β-1.4 xylanase, a key enzyme for primary degradation of xylan. The presence of an extracellular xylanase was confirmed by the appearance of xylan hydrolysis products in cell-free supernatants. Extracellular xylanolytic activity was only detected in xylan+ strains, as indicated by the production of XOS fragments with a DP of 2 to 6, identified by thin-layer chromatography (TLC) and high-performance liquid chromatography (HPLC). Additionally, in vitro fecal fermentations revealed that strains with a xylan+ phenotype can persist with xylan supplementation. These results indicate that xylan+ B. pseudocatenulatum strains may have a competitive advantage in the complex environment of the gastrointestinal tract, due to their ability to act as primary degraders of xylan through extracellular enzymatic degradation. IMPORTANCE The beneficial health effects of dietary fiber are now well established. Moreover, low fiber consumption is associated with increased risks of metabolic and systemic diseases. This so-called "fiber gap" also has a profound impact on the composition of the gut microbiome, leading to a disrupted or dysbiotic microbiota. Therefore, understanding the mechanisms by which keystone bacterial species in the gut utilize xylans and other dietary fibers may provide a basis for developing strategies to restore gut microbiome function. The results described here provide biochemical and genetic evidence for primary xylan utilization by human-derived Bifidobacterium pseudocatenulatum and show also that cooperative utilization of xylans occurs among other members of this species.
Collapse
Affiliation(s)
- Elizabeth Drey
- Department of Food Science and Technology, Food Innovation Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| | - Car Reen Kok
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Complex Biosystems, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| | - Robert Hutkins
- Department of Food Science and Technology, Food Innovation Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska—Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
24
|
Cheng L, Wei Y, Xu L, Peng L, Wang Y, Wei X. Gut Microbiota Differentially Mediated by Qingmao Tea and Qingzhuan Tea Alleviated High-Fat-Induced Obesity and Associated Metabolic Disorders: The Impact of Microbial Fermentation. Foods 2022; 11:3210. [PMCID: PMC9601715 DOI: 10.3390/foods11203210] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although dark tea is a unique microbial-fermented tea with a high reputation for having an antiobesity effect, little is known about the effect of microbial fermentation on tea leaves’ antiobesity properties. This study compared the antiobesity effects of microbial-fermented Qingzhuan tea (QZT) and unfermented Qingmao tea (QMT), providing insight into their underlying mechanisms associated with gut microbiota. Our results indicated that the supplementation of QMT extract (QMTe) and QZT extract (QZTe) displayed similar antiobesity effects in high-fat diet (HFD)-fed mice, but the hypolipidemic effect of QZTe was significantly stronger than that of QMTe. The microbiomic analysis indicated that QZTe was more effective than QMTe at regulating HFD-caused gut microbiota dysbiosis. Akkermansiaceae and Bifidobacteriaceae, which have negative correlations with obesity, were enhanced notably by QZTe, whereas Faecalibaculum and Erysipelotrichaceae, which are positively correlated with obesity, were decreased dramatically by QMTe and QZTe. A Tax4Fun analysis of QMTe/QZTe-mediated gut microbiota revealed that QMTe supplementation drastically reversed the HFD-induced upregulation of glycolysis and energy metabolism, whereas QZTe supplementation significantly restored the HFD-caused downregulation of pyruvate metabolism. Our findings suggested that microbial fermentation showed a limited effect on tea leaves’ antiobesity, but enhanced their hypolipidemic activity, and QZT could attenuate obesity and associated metabolic disorders by favorably modulating gut microbiota.
Collapse
Affiliation(s)
- Lizeng Cheng
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lurong Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lanlan Peng
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yuanfeng Wang
- College of Life Sciences, Shanghai Normal University, 100 Guilin Road, Shanghai 200234, China
- Correspondence: (Y.W.); (X.W.); Tel.: +86-18616184495 (Y.W.); +86-021-34208533 (X.W.)
| | - Xinlin Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- Correspondence: (Y.W.); (X.W.); Tel.: +86-18616184495 (Y.W.); +86-021-34208533 (X.W.)
| |
Collapse
|
25
|
Moreira GV, Araujo LCC, Murata GM, Matos SL, Carvalho CRO. Kombucha tea improves glucose tolerance and reduces hepatic steatosis in obese mice. Biomed Pharmacother 2022; 155:113660. [PMID: 36095960 DOI: 10.1016/j.biopha.2022.113660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), often associated with obesity, is becoming one of the most common liver diseases worldwide. It is estimated to affect one billion individuals and may be present in approximately 25% of the population globally. NAFLD is viewed as a hepatic manifestation of metabolic syndrome, with humans and animal models presenting dyslipidemia, hypertension, and diabetes. The gut-liver axis has been considered the main pathogenesis branch for NAFLD development. Considering that foods or beverages could modulate the gastrointestinal tract, immune system, energy homeostasis regulation, and even the gut-liver axis, we conducted an exploratory study to analyze the effects of kombucha probiotic on hepatic steatosis, glucose tolerance, and hepatic enzymes involved in carbohydrate and fat metabolism using a pre-clinical model. The diet-induced obese mice presented glucose intolerance, hyperinsulinemia, hepatic steatosis, increased collagen fiber deposition in liver vascular spaces, and upregulated TNF-alpha and SREBP-1 gene expression. Mice receiving the kombucha supplement displayed improved glucose tolerance, reduced hyperinsulinemia, decreased citrate synthase and phosphofructokinase-1 enzyme activities, downregulated G-protein-coupled bile acid receptor, also known as TGR5, and farnesol X receptor gene expression, and attenuated steatosis and hepatic collagen fiber deposition. The improvement in glucose tolerance was accompanied by the recovery of acute insulin-induced liver AKT serine phosphorylation. Thus, it is possible to conclude that this probiotic drink has a beneficial effect in reducing the metabolic alterations associated with diet-induced obesity. This probiotic beverage deserves an extension of studies to confirm or refute its potentially beneficial effects.
Collapse
Affiliation(s)
- Gabriela V Moreira
- University of São Paulo, Department of Physiology and Biophysics, Institute of Biological Science, São Paulo 05508-900, Brazil
| | - Layanne C C Araujo
- University of São Paulo, Department of Physiology and Biophysics, Institute of Biological Science, São Paulo 05508-900, Brazil
| | - Gilson M Murata
- University of São Paulo, Department of Medicine, School of Medicine, São Paulo 01246-903, Brazil
| | - Sandro L Matos
- University of São Paulo, Department of Physiology and Biophysics, Institute of Biological Science, São Paulo 05508-900, Brazil
| | - Carla R O Carvalho
- University of São Paulo, Department of Physiology and Biophysics, Institute of Biological Science, São Paulo 05508-900, Brazil.
| |
Collapse
|
26
|
Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease. Foods 2022; 11:foods11172703. [PMID: 36076888 PMCID: PMC9455821 DOI: 10.3390/foods11172703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Diet and lifestyle are crucial factors that influence the susceptibility of humans to nonalcoholic fatty liver disease (NAFLD). Personalized diet patterns chronically affect the composition and activity of microbiota in the human gut; consequently, nutrition-related dysbiosis exacerbates NAFLD via the gut–liver axis. Recent advances in diagnostic technology for gut microbes and microbiota-derived metabolites have led to advances in the diagnosis, treatment, and prognosis of NAFLD. Microbiota-derived metabolites, including tryptophan, short-chain fatty acid, fat, fructose, or bile acid, regulate the pathophysiology of NAFLD. The microbiota metabolize nutrients, and metabolites are closely related to the development of NAFLD. In this review, we discuss the influence of nutrients, gut microbes, their corresponding metabolites, and metabolism in the pathogenesis of NAFLD.
Collapse
|
27
|
Effects of Probiotic Supplementation during Pregnancy on the Future Maternal Risk of Metabolic Syndrome. Int J Mol Sci 2022; 23:ijms23158253. [PMID: 35897822 PMCID: PMC9330652 DOI: 10.3390/ijms23158253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics are live microorganisms that induce health benefits in the host. Taking probiotics is generally safe and well tolerated by pregnant women and their children. Consumption of probiotics can result in both prophylactic and therapeutic effects. In healthy adult humans, the gut microbiome is stable at the level of the dominant taxa: Bacteroidetes, Firmicutes and Actinobacteria, and has a higher presence of Verrucomicrobia. During pregnancy, an increase in the number of Proteobacteria and Actinobacteria phyla and a decrease in the beneficial species Roseburia intestinalis and Faecalibacterium prausnitzii are observed. Pregnancy is a "window" to the mother's future health. The aim of this paper is to review studies assessing the potentially beneficial effects of probiotics in preventing the development of diseases that appear during pregnancy, which are currently considered as risk factors for the development of metabolic syndrome, and consequently, reducing the risk of developing maternal metabolic syndrome in the future. The use of probiotics in gestational diabetes mellitus, preeclampsia and excessive gestational weight gain is reviewed. Probiotics are a relatively new intervention that can prevent the development of these disorders during pregnancy, and thus, would reduce the risk of metabolic syndrome resulting from these disorders in the mother's future.
Collapse
|
28
|
Saha P, Bose D, Stebliankin V, Cickovski T, Seth RK, Porter DE, Brooks BW, Mathee K, Narasimhan G, Colwell R, Scott GI, Chatterjee S. Prior exposure to microcystin alters host gut resistome and is associated with dysregulated immune homeostasis in translatable mouse models. Sci Rep 2022; 12:11516. [PMID: 35799048 PMCID: PMC9262933 DOI: 10.1038/s41598-022-15708-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/28/2022] [Indexed: 11/09/2022] Open
Abstract
A strong association between exposure to the common harmful algal bloom toxin microcystin and the altered host gut microbiome has been shown. We tested the hypothesis that prior exposure to the cyanotoxin microcystin-LR may alter the host resistome. We show that the mice exposed to microcystin-LR had an altered microbiome signature that harbored antibiotic resistance genes. Host resistome genotypes such as mefA, msrD, mel, ant6, and tet40 increased in diversity and relative abundance following microcystin-LR exposure. Interestingly, the increased abundance of these genes was traced to resistance to common antibiotics such as tetracycline, macrolides, glycopeptide, and aminoglycosides, crucial for modern-day treatment of several diseases. Increased abundance of these genes was positively associated with increased expression of PD1, a T-cell homeostasis marker, and pleiotropic inflammatory cytokine IL-6 with a concomitant negative association with immunosurveillance markers IL-7 and TLR2. Microcystin-LR exposure also caused decreased TLR2, TLR4, and REG3G expressions, increased immunosenescence, and higher systemic levels of IL-6 in both wild-type and humanized mice. In conclusion, the results show a first-ever characterization of the host resistome following microcystin-LR exposure and its connection to host immune status and antimicrobial resistance that can be crucial to understand treatment options with antibiotics in microcystin-exposed subjects in clinical settings.
Collapse
Affiliation(s)
- Punnag Saha
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
- NIEHS Center for Oceans and Human Health and Climate Change Interactions, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
- Columbia VA Medical Center, Columbia, SC, 29209, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
- NIEHS Center for Oceans and Human Health and Climate Change Interactions, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
- Columbia VA Medical Center, Columbia, SC, 29209, USA
| | - Vitalii Stebliankin
- Knight Foundation School of Computing & Information Sciences, Florida International University, Miami, FL, 33199, USA
| | - Trevor Cickovski
- Knight Foundation School of Computing & Information Sciences, Florida International University, Miami, FL, 33199, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
- Columbia VA Medical Center, Columbia, SC, 29209, USA
| | - Dwayne E Porter
- NIEHS Center for Oceans and Human Health and Climate Change Interactions, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
| | - Bryan W Brooks
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA
| | - Giri Narasimhan
- Knight Foundation School of Computing & Information Sciences, Florida International University, Miami, FL, 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA
| | - Rita Colwell
- CosmosID Inc, Germantown, MD, 20874, USA
- University of Maryland Institute for Advanced Computer Studies, University of Maryland, College Park, MD, 20742, USA
- Maryland Pathogen Research Institute, University of Maryland, College Park, MD, 20742, USA
| | - Geoff I Scott
- NIEHS Center for Oceans and Human Health and Climate Change Interactions, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA.
- NIEHS Center for Oceans and Human Health and Climate Change Interactions, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, 29208, USA.
- Columbia VA Medical Center, Columbia, SC, 29209, USA.
| |
Collapse
|
29
|
Olmo R, Wetzels SU, Armanhi JSL, Arruda P, Berg G, Cernava T, Cotter PD, Araujo SC, de Souza RSC, Ferrocino I, Frisvad JC, Georgalaki M, Hansen HH, Kazou M, Kiran GS, Kostic T, Krauss-Etschmann S, Kriaa A, Lange L, Maguin E, Mitter B, Nielsen MO, Olivares M, Quijada NM, Romaní-Pérez M, Sanz Y, Schloter M, Schmitt-Kopplin P, Seaton SC, Selvin J, Sessitsch A, Wang M, Zwirzitz B, Selberherr E, Wagner M. Microbiome Research as an Effective Driver of Success Stories in Agrifood Systems – A Selection of Case Studies. Front Microbiol 2022; 13:834622. [PMID: 35903477 PMCID: PMC9315449 DOI: 10.3389/fmicb.2022.834622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Increasing knowledge of the microbiome has led to significant advancements in the agrifood system. Case studies based on microbiome applications have been reported worldwide and, in this review, we have selected 14 success stories that showcase the importance of microbiome research in advancing the agrifood system. The selected case studies describe products, methodologies, applications, tools, and processes that created an economic and societal impact. Additionally, they cover a broad range of fields within the agrifood chain: the management of diseases and putative pathogens; the use of microorganism as soil fertilizers and plant strengtheners; the investigation of the microbial dynamics occurring during food fermentation; the presence of microorganisms and/or genes associated with hazards for animal and human health (e.g., mycotoxins, spoilage agents, or pathogens) in feeds, foods, and their processing environments; applications to improve HACCP systems; and the identification of novel probiotics and prebiotics to improve the animal gut microbiome or to prevent chronic non-communicable diseases in humans (e.g., obesity complications). The microbiomes of soil, plants, and animals are pivotal for ensuring human and environmental health and this review highlights the impact that microbiome applications have with this regard.
Collapse
Affiliation(s)
- Rocío Olmo
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
- *Correspondence: Rocío Olmo,
| | - Stefanie Urimare Wetzels
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Jaderson Silveira Leite Armanhi
- Symbiomics Microbiome Solutions, Florianópolis, Brazil
- Genomics for Climate Change Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Paulo Arruda
- Genomics for Climate Change Research Center, Universidade Estadual de Campinas, Campinas, Brazil
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, Brazil
- Departamento de Genética e Evolução, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gabriele Berg
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Tomislav Cernava
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Paul D. Cotter
- Food Bioscience, Teagasc Food Research Centre Moorepark, Fermoy, Ireland
- APC Microbiome Ireland and VistaMilk, Cork, Ireland
| | - Solon Cordeiro Araujo
- SCA, Consultoria em Microbiologia Agrícola, Campinas, Brazil
- Brazil National Association of Inoculant Producers and Importers (ANPII), Campinas, Brazil
| | - Rafael Soares Correa de Souza
- Symbiomics Microbiome Solutions, Florianópolis, Brazil
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, Brazil
| | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Science, University of Torino, Torino, Italy
| | - Jens C. Frisvad
- Department of Biotechnology and Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marina Georgalaki
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | - Hanne Helene Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maria Kazou
- Laboratory of Dairy Research, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | | | - Tanja Kostic
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Susanne Krauss-Etschmann
- Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute for Experimental Medicine, Christian Albrechts University, Kiel, Germany
| | - Aicha Kriaa
- Microbiota Interaction With Human and Animal Team (MIHA), Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Lene Lange
- BioEconomy, Research & Advisory, Copenhagen, Denmark
| | - Emmanuelle Maguin
- Microbiota Interaction With Human and Animal Team (MIHA), Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Birgit Mitter
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Mette Olaf Nielsen
- Department of Animal Science, Faculty of Technical Sciences, Aarhus University, Tjele, Denmark
| | - Marta Olivares
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Narciso Martín Quijada
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Marina Romaní-Pérez
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Michael Schloter
- Research Unit Comparative Microbiome Analysis, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | - Joseph Selvin
- School of Life Sciences, Pondicherry University, Puducherry, India
| | - Angela Sessitsch
- Bioresources Unit, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Tulln, Austria
| | - Mengcen Wang
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, China
| | - Benjamin Zwirzitz
- Institute of Food Science, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Martin Wagner
- FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
30
|
Kim YT, Kim CH, Kwon JG, Cho JH, Shin YS, Kim HB, Lee JH. In vivo Trial of Bifidobacterium longum Revealed the Complex Network Correlations Between Gut Microbiota and Health Promotional Effects. Front Microbiol 2022; 13:886934. [PMID: 35783421 PMCID: PMC9247516 DOI: 10.3389/fmicb.2022.886934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Complete genome sequence analysis of Bifidobacterium longum subsp. longum BCBL-583 isolated from a Korean female fecal sample showed no virulence factor or antibiotic resistance gene, suggesting human safety. In addition, this strain has oxygen and heat tolerance genes for food processing, and cholesterol reduction and mucin adhesion-related genes were also found. For in vivo evaluations, a high fat diet (HFD) mouse model was used, showing that BCBL-583 administration to the model (HFD-583) reduced the total cholesterol and LDL-cholesterol in the blood and decreased pro-inflammatory cytokines but increased anti-inflammatory cytokines, substantiating its cholesterol reduction and anti-inflammation activities. Subsequent microbiome analysis of the fecal samples from the HFD mouse model revealed that BCBL-583 administration changed the composition of gut microbiota. After 9 weeks feeding of bifidobacteria, Firmicutes, Actinobacteria, and Bacteroidetes increased, but Proteobacteria maintained in the HFD mouse models. Further comparative species-level compositional analysis revealed the inhibitions of cholesterol reduction-related Eubacterium coprostanoligenes and obesity-related Lactococcus by the supplementation of B. longum BCBL-583, suggesting its possible cholesterol reduction and anti-obesity activities. The correlation analysis of HFD-583 between the gut microbiota compositional change and cholesterol/immune response showed that Verrucomicrobia, Firmicutes, Actinobacteria, and Bacteroidetes may play an important role in cholesterol reduction and anti-inflammation. However, correlation analysis of Proteobacteria showed the reverse correlation in HFD-583. Interestingly, the correlation analysis of B. longum ATCC 15707 administration to HFD model showed similar patterns of cholesterol but different in immune response patterns. Therefore, this correlation analysis suggests that the microbial composition and inflammatory cytokine/total-cholesterol may be closely related in the administration of BCBL-583 in the HFD mice group. Consequently, BCBL-583 could be a good probiotic strain for gut health promotion through gut microbiota modulation.
Collapse
Affiliation(s)
- You-Tae Kim
- Department of Food and Animal Biotechnology, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Chul-Hong Kim
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin, South Korea
- Food Research Center, Binggrae Co., Ltd., Namyangju, South Korea
| | - Joon-Gi Kwon
- Department of Food and Animal Biotechnology, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Jae Hyoung Cho
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Young-Sup Shin
- Food Research Center, Binggrae Co., Ltd., Namyangju, South Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
- Hyeun Bum Kim,
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
- *Correspondence: Ju-Hoon Lee,
| |
Collapse
|
31
|
The Comparative Analysis of Genomic Diversity and Genes Involved in Carbohydrate Metabolism of Eighty-Eight Bifidobacterium pseudocatenulatum Isolates from Different Niches of China. Nutrients 2022; 14:nu14112347. [PMID: 35684146 PMCID: PMC9183100 DOI: 10.3390/nu14112347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/03/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
Eighty-eight Bifidobacterium pseudocatenulatum strains, which were isolated from human, chicken and cow fecal samples from different niches of China, were compared genomically in this study to evaluate their diversity. It was found that B. pseudocatenulatum displayed a closed pan-genome, including abundant glycoside hydrolase families of the carbohydrate active enzyme (CAZy). A total of 30 kinds of glycoside hydrolases (GHs), 14 kinds of glycosyl transferases (GTs), 13 kinds of carbohydrate-binding modules (CBMs), 6 kinds of carbohydrate-esterases (CEs), and 2 kinds of auxiliary activities (AAs) gene families were identified across the genomes of the 88 B. pseudocatenulatum strains. Specifically, this showed that significant differences were also present in the number of 10 carbohydrate-active enzyme gene families (GT51, GH13_32, GH26, GH42, GH121, GH3, AA3, CBM46, CE2, and CE6) among the strains derived from the hosts of different age groups, particularly between strains from infants and those from other human age groups. Twelve different individuals of B. pseudocatenulatum from four main clusters were selected for further study to reveal the genetic diversity of carbohydrate metabolism-related genes within the same phylogenetics. The animal experiment showed that 3 weeks of oral administration and 1 week after cessation of administration of these strains did not markedly alter the serum routine inflammatory indicators in mice. Furthermore, the administration of these strains did not significantly cause adverse changes in the gut microbiota, as indicated by the α- and β-diversity indexes, relative to the control group (normal diet). Beyond that, FAHBZ9L5 significantly increased the abundance of B. pseudocatenulatum after 3 weeks and significantly increased the abundance of acetic acid and butyric acid in the host’s intestinal tract 3 and 4 weeks after the first administration, respectively, compared with the control group. Corresponding to this, comparative genomic analyses of 12 B. pseudocatenulatum suggest that FAHBZ9L5-specific genes were rich in ABC transporters and carbohydrate esterase. Combining the results of comparative genomics analyses and animal experiment, it is suggested that the strains containing certain gene clusters contribute to another competitive growth advantage of B. pseudocatenulatum, which facilitates its intestinal carbohydrate metabolism in a host.
Collapse
|
32
|
Singh V, Park YJ, Lee G, Unno T, Shin JH. Dietary regulations for microbiota dysbiosis among post-menopausal women with type 2 diabetes. Crit Rev Food Sci Nutr 2022; 63:9961-9976. [PMID: 35635755 DOI: 10.1080/10408398.2022.2076651] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Type 2 diabetes (T2D) and T2D-associated comorbidities, such as obesity, are serious universally prevalent health issues among post-menopausal women. Menopause is an unavoidable condition characterized by the depletion of estrogen, a gonadotropic hormone responsible for secondary sexual characteristics in women. In addition to sexual dimorphism, estrogen also participates in glucose-lipid homeostasis, and estrogen depletion is associated with insulin resistance in the female body. Estrogen level in the gut also regulates the microbiota composition, and even conjugated estrogen is actively metabolized by the estrobolome to maintain insulin levels. Moreover, post-menopausal gut microbiota is different from the pre-menopausal gut microbiota, as it is less diverse and lacks the mucolytic Akkermansia and short-chain fatty acid (SCFA) producers such as Faecalibacterium and Roseburia. Through various metabolites (SCFAs, secondary bile acid, and serotonin), the gut microbiota plays a significant role in regulating glucose homeostasis, oxidative stress, and T2D-associated pro-inflammatory cytokines (IL-1, IL-6). While gut dysbiosis is common among post-menopausal women, dietary interventions such as probiotics, prebiotics, and synbiotics can ease post-menopausal gut dysbiosis. The objective of this review is to understand the relationship between post-menopausal gut dysbiosis and T2D-associated factors. Additionally, the study also provided dietary recommendations to avoid T2D progression among post-menopausal women.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Yeong-Jun Park
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Tatsuya Unno
- Department of Biotechnology, Jeju National University, Jeju, South Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
33
|
Yang H, Jin X, Cheng T, Shan G, Lu C, Gu J, Zhan C, Xu F, Ge D. Comprehensive Genome-Scale Analysis of Esophageal Carcinoma With Esophageal Tissue-Resident Micro-Environment Discrepancy. Front Microbiol 2022; 13:859352. [PMID: 35586863 PMCID: PMC9108775 DOI: 10.3389/fmicb.2022.859352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022] Open
Abstract
To figure out the molecular mechanism in the esophageal squamous carcinoma (ESCC) with the discrepancy in the tissue-resident microbiota, we selected clinical features, RNA sequences, and transcriptomes of ESCC patients from The Cancer Genome Atlas (TCGA) website and detailed tissue-resident microbiota information from The Cancer Microbiome Atlas (n = 60) and explored the infiltration condition of particular microbiota in each sample. We classified the tissue-resident micro-environment of ESCC into two clusters (A and B) and built a predictive classifier model. Cluster A has a higher proportion of certain tissue-resident microbiota with comparatively better survival, while Cluster B has a lower proportion of certain tissue-resident microbiota with comparatively worse survival. We showed traits of gene and clinicopathology in the esophageal tissue-resident micro-environment (ETM) phenotypes. By comparing the two clusters’ molecular signatures, we find that the two clusters have obvious differences in gene expression and mutation, which lead to pathway expression discrepancy. Several pathways are closely related to tumorigenesis. Our results may demonstrate a synthesis of the infiltration pattern of the esophageal tissue-resident micro-environment in ESCC. We reveal the mechanism of esophageal tissue-resident microbiota discrepancy in ESCC, which may contribute to therapy progress for patients with ESCC.
Collapse
|
34
|
Zheng H, Ji H, Fan K, Xu H, Huang Y, Zheng Y, Xu Q, Li C, Zhao L, Li Y, Gao H. Targeting Gut Microbiota and Host Metabolism with Dendrobium officinale Dietary Fiber to Prevent Obesity and Improve Glucose Homeostasis in Diet-Induced Obese Mice. Mol Nutr Food Res 2022; 66:e2100772. [PMID: 35225418 DOI: 10.1002/mnfr.202100772] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/29/2021] [Indexed: 12/26/2022]
Abstract
SCOPE Obesity is becoming a major public health problem due to excess dietary fat intake. Dendrobium officinale (D. officinale) is a medicine food homology plant and exerts multiple health-promoting effects. However, its antiobesity effects and the potential mechanisms remain unclear. METHODS AND RESULTS High-fat diet (HFD)-fed mice are administered D. officinale dietary fiber (DODF) daily by gavage for 11 weeks. The results show that treatment with DODF alleviates obesity, liver steatosis, inflammation, and oxidant stress in HFD-induced obese mice. Improved glucose homeostasis in obese mice after DODF treatment is achieved by enhancing insulin pathway and hepatic glycogen synthesis. DODF restructures the gut microbiota in obese mice by decreasing the relative abundance of Bilophila and increasing the relative abundances of Akkermansia, Bifidobacterium, and Muribaculum. Also, DODF reshapes the metabolic phenotype of obese mice as indicated by up-regulating energy metabolism, increasing acetate and taurine, and reducing serum low density/very low density lipoproteins (LDL/VLDL). These beneficial effects are partly transferred by FMT, implying the gut microbiota as a target for the protective effect of DODF on obesity-related symptoms. CONCLUSION The results suggest that DODF can be used as a novel prebiotics to maintain the gut microbial homeostasis and improve metabolic health, preventing obesity and related metabolic syndrome.
Collapse
Affiliation(s)
- Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.,Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Hui Ji
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Kai Fan
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hangying Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yinli Huang
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Yafei Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qingqing Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen Li
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
35
|
Zhang J, Yu H, Zhong H, Wang Q, Tang J, Shen F, Cai H, Liu T, Feng F, Zhao M. Dietary emulsifier glycerol monodecanoate affects gut microbiota contributing to regulating lipid metabolism, insulin sensitivity and inflammation. Food Funct 2022; 13:8804-8817. [DOI: 10.1039/d2fo01689c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glycerol monodecanoate (GMD) is a medium-chain monoacylglycerol that possesses emulsifying and antibacterial properties. Common emulsifiers carboxymethylcellulose and polysorbate-80 have been reported to cause intestinal microbiota dysbiosis and metabolic disturbances. While...
Collapse
|
36
|
Bloom PP, Tapper EB, Young VB, Lok AS. Microbiome therapeutics for hepatic encephalopathy. J Hepatol 2021; 75:1452-1464. [PMID: 34453966 PMCID: PMC10471317 DOI: 10.1016/j.jhep.2021.08.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/20/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Hepatic encephalopathy (HE) is a complication of cirrhosis characterised by neuropsychiatric and motor dysfunction. Microbiota-host interactions play an important role in HE pathogenesis. Therapies targeting microbial community composition and function have been explored for the treatment of HE. Prebiotics, probiotics and faecal microbiota transplant (FMT) have been used with the aim of increasing the abundance of potentially beneficial taxa, while antibiotics have been used to decrease the abundance of potentially harmful taxa. Other microbiome therapeutics, including postbiotics and absorbents, have been used to target microbial products. Microbiome-targeted therapies for HE have had some success, notably lactulose and rifaximin, with probiotics and FMT also showing promise. However, there remain several challenges to the effective application of microbiome therapeutics in HE, including the resilience of the microbiome to sustainable change and unpredictable clinical outcomes from microbiota alterations. Future work in this space should focus on rigorous trial design, microbiome therapy selection, and a personalised approach to HE.
Collapse
Affiliation(s)
- Patricia P Bloom
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, USA.
| | - Elliot B Tapper
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, USA
| | - Vincent B Young
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, USA; Department of Microbiology and Immunology, University of Michigan, USA
| | - Anna S Lok
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, USA
| |
Collapse
|
37
|
Cheng M, Ge X, Zhong C, Fu R, Ning K, Xu S. Micro-coevolution of host genetics with gut microbiome in three Chinese ethnic groups. J Genet Genomics 2021; 48:972-983. [PMID: 34562635 DOI: 10.1016/j.jgg.2021.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022]
Abstract
Understanding the micro-coevolution of the human gut microbiome with host genetics is challenging but essential in both evolutionary and medical studies. To gain insight into the interactions between host genetic variation and the gut microbiome, we analyzed both the human genome and gut microbiome collected from a cohort of 190 students in the same boarding college and representing 3 ethnic groups, Uyghur, Kazakh, and Han Chinese. We found that differences in gut microbiome were greater between genetically distinct ethnic groups than those genetically closely related ones in taxonomic composition, functional composition, enterotype stratification, and microbiome genetic differentiation. We also observed considerable correlations between host genetic variants and the abundance of a subset of gut microbial species. Notably, interactions between gut microbiome species and host genetic variants might have coordinated effects on specific human phenotypes. Bacteroides ovatus, previously reported to modulate intestinal immunity, is significantly correlated with the host genetic variant rs12899811 (meta-P = 5.55 × 10-5), which regulates the VPS33B expression in the colon, acting as a tumor suppressor of colorectal cancer. These results advance our understanding of the micro-coevolution of the human gut microbiome and their interactive effects with host genetic variation on phenotypic diversity.
Collapse
Affiliation(s)
- Mingyue Cheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xueling Ge
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chaofang Zhong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ruiqing Fu
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Shuhua Xu
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China; Human Phenome Institute, Fudan University, Shanghai 201203, China.
| |
Collapse
|
38
|
He LH, Yao DH, Wang LY, Zhang L, Bai XL. Gut Microbiome-Mediated Alteration of Immunity, Inflammation, and Metabolism Involved in the Regulation of Non-alcoholic Fatty Liver Disease. Front Microbiol 2021; 12:761836. [PMID: 34795655 PMCID: PMC8593644 DOI: 10.3389/fmicb.2021.761836] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of end-stage liver disease, leading to a rapidly growing global public health burden. The term “gut microbiome (GM)” refers to the approximately 100 trillion microbial cells that inhabit the host’s gastrointestinal tract. There is increasing evidence that GM is involved in the pathogenesis of NAFLD and may be a potential target for intervention. To explore GM-based strategies for precise diagnosis and treatment of NAFLD, great efforts have been made to develop a comprehensive and in-depth understanding of the host–microbe interaction. This review evaluates this interaction critically, mainly considering the intricate regulation of the metabolism, immunity, and inflammatory status during the evolution of the disease pathogenesis, revealing roles for the GM in NAFLD by examining advances in potential mechanisms, diagnostics, and modulation strategies. Synopsis: Considering the intricate metabolic and immune/inflammatory homeostasis regulation, we evaluate the latest understanding of the host–microbe interaction and reveal roles for the gastrointestinal microbiome in NAFLD. Strategies targeting the gastrointestinal microbiome for the diagnosis and treatment of NAFLD are proposed.
Collapse
Affiliation(s)
- Li-Hong He
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Dun-Han Yao
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ling-Yun Wang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Di DS, Li C, Dai Y, Wei MH, Wang SS, Song WJ, Zhou HL, Cui Y, Zhang RY, Huang Q, Wang Q. Integrative Analysis of LGR5/6 Gene Variants, Gut Microbiota Composition and Osteoporosis Risk in Elderly Population. Front Microbiol 2021; 12:765008. [PMID: 34795657 PMCID: PMC8593465 DOI: 10.3389/fmicb.2021.765008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/13/2021] [Indexed: 11/25/2022] Open
Abstract
Objective: This study aimed to explore the relationships between the common variants of R-spondin/Wnt signaling genes, gut microbiota composition, and osteoporosis (OP) risk in elderly Chinese Han population. Design: Dual-energy X-ray absorptiometry was used to obtain the OP-associated measurements at multiple skeleton sites among all 1,168 participants. Genotyping data was obtained by using the next-generation sequencing in the discovery stage (n = 400, 228 OP patients) and SNPscan technology in the replication stage (n = 768, 356 OP patients). Bioinformatic analysis was performed to provide more evidence for the genotype-OP associations. The 16S ribosomal RNA gene high-throughput sequencing technology was adopted to explore OP-associated gut microbiota variations. Results: The genetic variants of rs10920362 in the LGR6 gene (P-FDR = 1.19 × 10–6) and rs11178860 in the LGR5 gene (P-FDR = 1.51 × 10–4) were found to associate with OP risk significantly. Several microbial taxa were associated with the BMDs and T-scores at multiple skeleton sites. The associations between rs10920362 and BMD-associated microbiota maintained significance after adjusting confounders. The rs10920362 CT/TT genotype associated with a decreased relative abundance of Actinobacteria (β = −1.32, P < 0.001), Bifidobacteriaceae (β = −1.70, P < 0.001), and Bifidobacterium (β = −1.70, P < 0.001) compared to the CC genotype. Conclusion: Our findings suggested that the variants loci of LGR6 may be associate with OP pathogenesis via gut microbiota modifications. The relationship between host genetics and gut microbiome provides new perspectives about OP prevention and treatment.
Collapse
Affiliation(s)
- Dong-Sheng Di
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can Li
- Department of Cancer Prevention and Control, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu Dai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mu-Hong Wei
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Shan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Jing Song
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao-Long Zhou
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Cui
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ru-Yi Zhang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Huang
- Department of Rehabilitation Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Chung The H, Nguyen Ngoc Minh C, Tran Thi Hong C, Nguyen Thi Nguyen T, Pike LJ, Zellmer C, Pham Duc T, Tran TA, Ha Thanh T, Van MP, Thwaites GE, Rabaa MA, Hall LJ, Baker S. Exploring the Genomic Diversity and Antimicrobial Susceptibility of Bifidobacterium pseudocatenulatum in a Vietnamese Population. Microbiol Spectr 2021; 9:e0052621. [PMID: 34523984 PMCID: PMC8557894 DOI: 10.1128/spectrum.00526-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/13/2021] [Indexed: 01/29/2023] Open
Abstract
Bifidobacterium pseudocatenulatum is a member of the human gut microbiota, and specific variants of B. pseudocatenulatum have been associated with health benefits such as improving gut integrity and reducing inflammatory responses. Here, we aimed to assess the genomic diversity and predicted metabolic profiles of B. pseudocatenulatum cells found colonizing the gut of healthy Vietnamese adults and children. We found that the population of B. pseudocatenulatum from each individual was distinct and highly diverse, with intraclonal variation attributed largely to a gain or loss of carbohydrate-utilizing enzymes. The B. pseudocatenulatum genomes were enriched with glycosyl hydrolases predicted to target plant-based nondigestible carbohydrates (GH13, GH43) but not host-derived glycans. Notably, the exopolysaccharide biosynthesis region from organisms isolated from healthy children showed extensive genetic diversity and was subject to a high degree of genetic modification. Antimicrobial susceptibility profiling revealed that the Vietnamese B. pseudocatenulatum cells were uniformly susceptible to beta-lactams but exhibited variable resistance to azithromycin, tetracycline, ciprofloxacin, and metronidazole. The genomic presence of ermX and tet variants conferred resistance against azithromycin and tetracycline, respectively; ciprofloxacin resistance was associated with a mutation(s) in the quinolone resistance-determining region (GyrA, S115, and/or D119). Our work provides the first detailed genomic and antimicrobial resistance characterization of B. pseudocatenulatum found in the Vietnamese population, which can be exploited for the rational design of probiotics. IMPORTANCE Bifidobacterium pseudocatenulatum is a beneficial member of the human gut microbiota. The organism can modulate inflammation and has probiotic potential, but its characteristics are largely strain dependent and associated with distinct genomic and biochemical features. Population-specific beneficial microbes represent a promising avenue for the development of potential probiotics, as they may exhibit a more suitable profile in the target population. This study investigates the underexplored diversity of B. pseudocatenulatum in Vietnam and provides more understanding of its genomic diversity, metabolic potential, and antimicrobial susceptibility. Such data from indigenous populations are essential for selecting probiotic candidates that can be accelerated into further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Hao Chung The
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | | | | | - Lindsay J. Pike
- The Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Caroline Zellmer
- University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Trung Pham Duc
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Tuan-Anh Tran
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Tuyen Ha Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Minh Pham Van
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Guy E. Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, Oxford University, Oxford, United Kingdom
| | - Maia A. Rabaa
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, Oxford University, Oxford, United Kingdom
| | - Lindsay J. Hall
- Quadram Institute Biosciences, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Stephen Baker
- The Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
- University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
41
|
Gawlik A, Salonen A, Jian C, Yanover C, Antosz A, Shmoish M, Wasniewska M, Bereket A, Wudy SA, Hartmann MF, Thivel D, Matusik P, Weghuber D, Hochberg Z. Personalized approach to childhood obesity: Lessons from gut microbiota and omics studies. Narrative review and insights from the 29th European childhood obesity congress. Pediatr Obes 2021; 16:e12835. [PMID: 34296826 DOI: 10.1111/ijpo.12835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 06/20/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
The traditional approach to childhood obesity prevention and treatment should fit most patients, but misdiagnosis and treatment failure could be observed in some cases that lie away from average as part of individual variation or misclassification. Here, we reflect on the contributions that high-throughput technologies such as next-generation sequencing, mass spectrometry-based metabolomics and microbiome analysis make towards a personalized medicine approach to childhood obesity. We hypothesize that diagnosing a child as someone with obesity captures only part of the phenotype; and that metabolomics, genomics, transcriptomics and analyses of the gut microbiome, could add precision to the term "obese," providing novel corresponding biomarkers. Identifying a cluster -omic signature in a given child can thus facilitate the development of personalized prognostic, diagnostic, and therapeutic approaches. It can also be applied to the monitoring of symptoms/signs evolution, treatment choices and efficacy, predisposition to drug-related side effects and potential relapse. This article is a narrative review of the literature and summary of the main observations, conclusions and perspectives raised during the annual meeting of the European Childhood Obesity Group. Authors discuss some recent advances and future perspectives on utilizing a systems approach to understanding and managing childhood obesity in the context of the existing omics data.
Collapse
Affiliation(s)
- Aneta Gawlik
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ching Jian
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Chen Yanover
- Healthcare Informatics, IBM Research-Haifa, Haifa, Israel
| | - Aleksandra Antosz
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Michael Shmoish
- Bioinformatics Knowledge Unit, The Lokey Centre, Technion - Israel Institute of Technology, Haifa, Israel
| | - Malgorzata Wasniewska
- Department of Human Pathology in Adulthood and Childhood, University of Messina, Messina, Italy
| | - Abdullah Bereket
- School of Medicine, Department of Paediatric Endocrinology, Marmara University, Istanbul, Turkey
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Division of Paediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - David Thivel
- University Clermont Auvergne, UFR Medicine, Clermont-Ferrand, France
| | - Pawel Matusik
- Department of Paediatrics and Paediatric Endocrinology, Faculty of Medical Sciences, Medical University of Silesia, Katowice, Poland
| | - Daniel Weghuber
- Department of Paediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Ze'ev Hochberg
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
42
|
Fabersani E, Marquez A, Russo M, Ross R, Torres S, Fontana C, Puglisi E, Medina R, Gauffin-Cano P. Lactic Acid Bacteria Strains Differently Modulate Gut Microbiota and Metabolic and Immunological Parameters in High-Fat Diet-Fed Mice. Front Nutr 2021; 8:718564. [PMID: 34568404 PMCID: PMC8458958 DOI: 10.3389/fnut.2021.718564] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Dietary strategies, including the use of probiotics as preventive agents that modulate the gut microbiota and regulate the function of adipose tissue, are suitable tools for the prevention or amelioration of obesity and its comorbidities. We aimed to evaluate the effect of lactic acid bacteria (LAB) with different adipo- and immuno-modulatory capacities on metabolic and immunological parameters and intestinal composition microbiota in high-fat-diet-induced in mice fed a high-fat diet Methods: Balb/c weaning male mice were fed a standard (SD) or high-fat diet (HFD) with or without supplementation with Limosilactobacillus fermentum CRL1446 (CRL1446), Lactococcus lactis CRL1434 (CRL1434), or Lacticaseibacillus casei CRL431 (CRL431) for 45 days. Biochemical and immunological parameters, white-adipose tissue histology, gut microbiota composition, and ex vivo cellular functionality (adipocytes and macrophages) were evaluated in SD and HFD mice. Results: CRL1446 and CRL1434 administration, unlike CRL431, induced significant changes in the body and adipose tissue weights and the size of adipocytes. Also, these strains caused a decrease in plasmatic glucose, cholesterol, triglycerides, leptin, TNF-α, IL-6 levels, and an increase of IL-10. The CRL1446 and CRL1434 obese adipocyte in ex vivo functionality assays showed, after LPS stimulus, a reduction in leptin secretion compared to obese control, while with CRL431, no change was observed. In macrophages from obese mice fed with CRL1446 and CRL1434, after LPS stimulus, lower levels of MCP-1, TNF-α, IL-6 compared to obese control were observed. In contrast, CRL431 did not induce modification of cytokine values. Regarding gut microbiota, all strain administration caused a decrease in Firmicutes/Bacteroidetes index and diversity. As well as, related to genus results, all strains increased, mainly the genera Alistipes, Dorea, Barnesiella, and Clostridium XIVa. CRL1446 induced a higher increase in the Lactobacillus genus during the study period. Conclusions: The tested probiotic strains differentially modulated the intestinal microbiota and metabolic/immunological parameters in high-fat-diet-induced obese mice. These results suggest that CRL1446 and CRL1434 strains could be used as adjuvant probiotics strains for nutritional treatment to obesity and overweight. At the same time, the CRL431 strain could be more beneficial in pathologies that require regulation of the immune system.
Collapse
Affiliation(s)
- Emanuel Fabersani
- Facultad de Agronomía y Zootecnia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Antonela Marquez
- Centro de Referencia para Lactobacilos -CONICET, Tucumán, Argentina
| | - Matías Russo
- Centro de Referencia para Lactobacilos -CONICET, Tucumán, Argentina
| | - Romina Ross
- Instituto de Biotecnología Farmacéutica y Alimentaria -CONICET, Tucumán, Argentina
- Facultad Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino, Tucumán, Argentina
| | - Sebastián Torres
- Instituto de Bioprospección y Fisiología Vegetal -CONICET, Tucumán, Argentina
| | - Cecilia Fontana
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Edoardo Puglisi
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Roxana Medina
- Facultad de Agronomía y Zootecnia, Universidad Nacional de Tucumán, Tucumán, Argentina
- Centro de Referencia para Lactobacilos -CONICET, Tucumán, Argentina
| | - Paola Gauffin-Cano
- Centro de Referencia para Lactobacilos -CONICET, Tucumán, Argentina
- Facultad Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino, Tucumán, Argentina
| |
Collapse
|
43
|
He N, Wang S, Lv Z, Zhao W, Li S. Low molecular weight chitosan oligosaccharides (LMW-COSs) prevent obesity-related metabolic abnormalities in association with the modification of gut microbiota in high-fat diet (HFD)-fed mice. Food Funct 2021; 11:9947-9959. [PMID: 33108433 DOI: 10.1039/d0fo01871f] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, the two enzymatic low molecular weight chitosan oligosaccharides (LMW-COSs), LMW-COS-H and LMW-COS-L, were prepared with average MWs of 879.6 Da and 360.9 Da, respectively. Compared to LMW-COS-L, the LMW-COS-H was more effective in improving high-fat diet (HFD)-induced metabolic abnormalities, such as obesity, hyperlipidemia, low-grade inflammation and insulin resistance. The subsequent analysis of gut microbiota showed that the supplement of LMW-COSs caused overall structural and genus/species-specific changes in the gut microbiota, which were significantly correlated with the metabolic parameters. Specifically, both of the LMW-COSs significantly decreased the relative abundance of inflammatory bacteria such as Erysipelatoclostridium and Alistipes, whereas that of the beneficial intestinal bacteria (such as Akkermansia and Gammaproteobacteria) increased significantly. This study suggested that there were potential prebiotic functions of LMW-COSs in HFD fed mice, which regulated the dysfunctional gut microbiota, alleviated low-grade inflammation and maintained the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Ningning He
- College of Basic Medicine, Qingdao University, 266071, Qingdao, China.
| | | | | | | | | |
Collapse
|
44
|
Castro-Rodríguez DC, Reyes-Castro LA, Vega CC, Rodríguez-González GL, Yáñez-Fernández J, Zambrano E. Leuconostoc mesenteroides subsp. mesenteroides SD23 Prevents Metabolic Dysfunction Associated with High-Fat Diet-Induced Obesity in Male Mice. Probiotics Antimicrob Proteins 2021; 12:505-516. [PMID: 31129870 DOI: 10.1007/s12602-019-09556-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
High-fat diet (HFD) consumption induces obesity and increases blood glucose, insulin resistance, and metabolic disorders. Recent studies suggest that probiotics might be a novel approach to counteract these effects in the treatment of obesity. Here, we evaluated the effect of Leuconostoc mesenteroides subsp. mesenteroides SD23 on obesity-related metabolic dysfunction. In the present study, mice were randomly divided into four dietary groups: standard diet (C), HFD (OB), standard diet with L. mesenteroides SD23 (CP), and HFD with L. mesenteroides SD23 (OBP). Diets were maintained for 14 weeks. Animal weight was monitored and biochemical and histological analyses were performed after intervention. OB showed metabolic dysfunction, and increased the number of larger adipocytes compared to C. OB induced liver tumor necrosis factor-α (TNF-α) expression, increased cholesterol, leptin, and glucose levels compared to C. OBP reduced body weight, glucose, cholesterol, and leptin levels and improved glucose tolerance compared to OB. OBP also reduced liver steatosis, the number of larger adipocytes in adipose tissue, and reduced the villus height in the small intestine. OBP decreased expression of TNF-α and increased expression of IL-10 in liver. The parameters evaluated in the CP were similar to the C. This study provides novel evidence that dietary intervention with L. mesenteroides SD23 improves metabolic dysfunction related to obesity in HFD-fed mice.
Collapse
Affiliation(s)
- Diana C Castro-Rodríguez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Bioprocess Department, Unidad Profesional Interdisciplinaria de Biotecnología (UPIBI), Instituto Politécnico Nacional (IPN), Mexico City, Mexico.,CONACyT-Cátedras, Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis A Reyes-Castro
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Claudia C Vega
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jorge Yáñez-Fernández
- Bioprocess Department, Unidad Profesional Interdisciplinaria de Biotecnología (UPIBI), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
45
|
Rodríguez-Daza MC, Pulido-Mateos EC, Lupien-Meilleur J, Guyonnet D, Desjardins Y, Roy D. Polyphenol-Mediated Gut Microbiota Modulation: Toward Prebiotics and Further. Front Nutr 2021; 8:689456. [PMID: 34268328 PMCID: PMC8276758 DOI: 10.3389/fnut.2021.689456] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The genome of gut microbes encodes a collection of enzymes whose metabolic functions contribute to the bioavailability and bioactivity of unabsorbed (poly)phenols. Datasets from high throughput sequencing, metabolome measurements, and other omics have expanded the understanding of the different modes of actions by which (poly)phenols modulate the microbiome conferring health benefits to the host. Progress have been made to identify direct prebiotic effects of (poly)phenols; albeit up to date, these compounds are not recognized as prebiotics sensu stricto. Interestingly, certain probiotics strains have an enzymatic repertoire, such as tannase, α-L-rhamnosidase, and phenolic acid reductase, involved in the transformation of different (poly)phenols into bioactive phenolic metabolites. In vivo studies have demonstrated that these (poly)phenol-transforming bacteria thrive when provided with phenolic substrates. However, other taxonomically distinct gut symbionts of which a phenolic-metabolizing activity has not been demonstrated are still significantly promoted by (poly)phenols. This is the case of Akkermansia muciniphila, a so-called antiobesity bacterium, which responds positively to (poly)phenols and may be partially responsible for the health benefits formerly attributed to these molecules. We surmise that (poly)phenols broad antimicrobial action free ecological niches occupied by competing bacteria, thereby allowing the bloom of beneficial gut bacteria. This review explores the capacity of (poly)phenols to promote beneficial gut bacteria through their direct and collaborative bacterial utilization and their inhibitory action on potential pathogenic species. We propose the term duplibiotic, to describe an unabsorbed substrate modulating the gut microbiota by both antimicrobial and prebiotic modes of action. (Poly)phenol duplibiotic effect could participate in blunting metabolic disturbance and gut dysbiosis, positioning these compounds as dietary strategies with therapeutic potential.
Collapse
Affiliation(s)
- Maria Carolina Rodríguez-Daza
- Faculty of Agriculture and Food Sciences, Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada.,Department of Food Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Elena C Pulido-Mateos
- Faculty of Agriculture and Food Sciences, Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada.,Department of Food Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Joseph Lupien-Meilleur
- Faculty of Agriculture and Food Sciences, Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada.,Department of Food Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Denis Guyonnet
- Diana Nova, Symrise Nutrition, Clichy-la-Garenne, France
| | - Yves Desjardins
- Faculty of Agriculture and Food Sciences, Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada.,Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Denis Roy
- Faculty of Agriculture and Food Sciences, Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, QC, Canada.,Department of Food Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| |
Collapse
|
46
|
Diet-Regulating Microbiota and Host Immune System in Liver Disease. Int J Mol Sci 2021; 22:ijms22126326. [PMID: 34199182 PMCID: PMC8231888 DOI: 10.3390/ijms22126326] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota has been known to modulate the immune responses in chronic liver diseases. Recent evidence suggests that effects of dietary foods on health care and human diseases are related to both the immune reaction and the microbiome. The gut-microbiome and intestinal immune system play a central role in the control of bacterial translocation-induced liver disease. Dysbiosis, small intestinal bacterial overgrowth, translocation, endotoxemia, and the direct effects of metabolites are the main events in the gut-liver axis, and immune responses act on every pathways of chronic liver disease. Microbiome-derived metabolites or bacteria themselves regulate immune cell functions such as recognition or activation of receptors, the control of gene expression by epigenetic change, activation of immune cells, and the integration of cellular metabolism. Here, we reviewed recent reports about the immunologic role of gut microbiotas in liver disease, highlighting the role of diet in chronic liver disease.
Collapse
|
47
|
Ashaolu TJ, Fernández-Tomé S. Gut mucosal and adipose tissues as health targets of the immunomodulatory mechanisms of probiotics. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.04.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
48
|
Abstract
PURPOSE OF REVIEW An increasing body of evidence suggests that the gut microbiome influences the pathogenesis of insulin resistance and type 2 diabetes (T2D). In this review, we will discuss the latest findings regarding the mechanisms linking the gut microbiome and microbial metabolites with T2D and therapeutic approaches based on the gut microbiota for the prevention and treatment of T2D. RECENT FINDINGS Alterations in the gut microbial composition are associated with the risk of T2D. The gut microbiota can metabolize dietary- and host-derived factors to produce numerous microbial metabolites, which are involved in metabolic processes modulating nutrition and energy harvest, gut barrier function, systemic inflammation, and glucose metabolism. Microbial metabolites are important mediators of microbial-host crosstalk impacting host glucose metabolism. Furthermore, microbiome-based interventions may have beneficial effects on glycemic control. Future research is required to develop personalized T2D therapy based on microbial composition and/or metabolites.
Collapse
|
49
|
Zhou MX, Tian X, Wu ZQ, Li K, Li ZJ. Fuzhuan brick tea supplemented with areca nuts: Effects on serum and gut microbiota in mice. J Food Biochem 2021; 45:e13737. [PMID: 33876445 DOI: 10.1111/jfbc.13737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 11/28/2022]
Abstract
Areca nut and Fuzhuan brick tea, a type of natural plant products, have obvious effects of fat reduction and weight loss; however, there is no report on their synergistic effect. This study investigated the effects of Fuzhuan brick tea supplemented with different concentrations of areca nut (5% (LAF), 10% (MAF), and 20% (HAF)) on serum and gut microbiota in Kunming (KM) mice. The results showed that Fuzhuan brick tea supplemented with areca nuts (AFTs) could reduce weight, prevent the accumulation of fat, inhibit the increase in the levels of serum triglyceride, total cholesterol, low-density lipoprotein cholesterol, blood glucose, free fatty acid, insulin, and total bile acid, alleviate the decrease in high-density lipoprotein cholesterol level, and regulate the composition of gut microbiota by high-fat diet intervention. The HAF group with 20% areca nut content showed the best effect. These results could provide a novel approach to prevent obesity and hyperlipidemia. PRACTICAL APPLICATIONS: Consumption of areca nut and tea is widespread in Asia and other regions. As a controversial raw material, the damage due to areca nut to oral mucosa health has often aroused public concern and heated discussion; however, its medicinal value has been confirmed in terms of its pharmacological effects in various aspects. Fuzhuan brick tea, a type of traditional postfermented dark tea, has been confirmed to exert effects of antiobesity. Therefore, the areca nut and Fuzhuan brick tea, as a type of natural plant products, have obvious effects of fat reduction and weight loss; however, their synergistic effect has not been reported. To our knowledge, this study is the first to explore the effects of the Fuzhuan brick tea supplemented with areca nuts (AFTs) on serum and gut microbiota in mice. On the premise of exerting their beneficial effects (especially in terms of easing food stagnation and eliminating indigestion) and reducing their toxic and side effects, the effects of AFTs on health were further clarified, which could provide a novel direction for the development and utilization of areca nut. Moreover, our research would increase public understanding of areca nut and provide guidance to the Fuzhuan brick tea processing industry.
Collapse
Affiliation(s)
- Ming-Xi Zhou
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xing Tian
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Zhong-Qin Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ke Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zong-Jun Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
50
|
Identification of New Potential Biotherapeutics from Human Gut Microbiota-Derived Bacteria. Microorganisms 2021; 9:microorganisms9030565. [PMID: 33803291 PMCID: PMC7998412 DOI: 10.3390/microorganisms9030565] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
The role of the gut microbiota in health and disease is well recognized and the microbiota dysbiosis observed in many chronic diseases became a new therapeutic target. The challenge is to get a better insight into the functionality of commensal bacteria and to use this knowledge to select live biotherapeutics as new preventive or therapeutic products. In this study, we set up a screening approach to evaluate the functional capacities of a set of 21 strains isolated from the gut microbiota of neonates and adults. For this purpose, we selected key biological processes involved in the microbiome-host symbiosis and known to impact the host physiology i.e., the production of short-chain fatty acids and the ability to strengthen an epithelial barrier (Caco-2), to induce the release of the anti-inflammatory IL-10 cytokine after co-culture with human immune cells (PBMC) or to increase GLP-1 production from STC-1 endocrine cell line. This strategy highlighted fifteen strains exhibiting beneficial activities among which seven strains combined several of them. Interestingly, this work revealed for the first time a high prevalence of potential health-promoting functions among intestinal commensal strains and identified several appealing novel candidates for the management of chronic diseases, notably obesity and inflammatory bowel diseases.
Collapse
|