101
|
Burakova I, Smirnova Y, Gryaznova M, Syromyatnikov M, Chizhkov P, Popov E, Popov V. The Effect of Short-Term Consumption of Lactic Acid Bacteria on the Gut Microbiota in Obese People. Nutrients 2022; 14:3384. [PMID: 36014890 PMCID: PMC9415828 DOI: 10.3390/nu14163384] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity is a problem of modern health care that causes the occurrence of many concomitant diseases: arterial hypertension, diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. New strategies for the treatment and prevention of obesity are being developed that are based on using probiotics for modulation of the gut microbiota. Our study aimed to evaluate the bacterial composition of the gut of obese patients before and after two weeks of lactic acid bacteria (Lactobacillus acidophilus, Lactiplantibacillus plantarum, Limosilactobacillus fermentum, and Lactobacillus delbrueckii) intake. The results obtained showed an increase in the number of members of the phylum Actinobacteriota in the group taking nutritional supplements, while the number of phylum Bacteroidota decreased in comparison with the control group. There has also been an increase in potentially beneficial groups: Bifidobacterium, Blautia, Eubacterium, Anaerostipes, Lactococcus, Lachnospiraceae ND3007, Streptococcus, Escherichia-Shigella, and Lachnoclostridium. Along with this, a decrease in the genera was demonstrated: Faecalibacterium, Pseudobutyrivibrio, Subdoligranulum, Faecalibacterium, Clostridium sensu stricto 1 and 2, Catenibacterium, Megasphaera, Phascolarctobacterium, and the Oscillospiraceae NK4A214 group, which contribute to the development of various metabolic disorders. Modulation of the gut microbiota by lactic acid bacteria may be one of the ways to treat obesity.
Collapse
Affiliation(s)
- Inna Burakova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Yuliya Smirnova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Mariya Gryaznova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Mikhail Syromyatnikov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Pavel Chizhkov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Evgeny Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Vasily Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| |
Collapse
|
102
|
Delgadinho M, Ginete C, Santos B, Fernandes C, Silva C, Miranda A, de Vasconcelos JN, Brito M. How Hydroxyurea Alters the Gut Microbiome: A Longitudinal Study Involving Angolan Children with Sickle Cell Anemia. Int J Mol Sci 2022; 23:9061. [PMID: 36012325 PMCID: PMC9409137 DOI: 10.3390/ijms23169061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Sickle cell anemia (SCA) is an inherited hematological disorder and a serious global health problem, especially in Sub-Saharan Africa. Although hydroxyurea (HU) is the leading treatment for patients with SCA, its effects on the gut microbiome have not yet been explored. In this context, the aim of this study was to investigate this association by characterizing the gut microbiome of an Angolan SCA pediatric population before and after 6 months of HU treatment. A total of 66 stool samples were obtained and sequenced for the 16S rRNA gene (V3-V4 regions). Significant associations were observed in alpha and beta-diversity, with higher values of species richness for the children naïve for HU. We also noticed that children after HU had higher proportions of several beneficial bacteria, mostly short-chain fatty acids (SCFAs) producing species, such as Blautia luti, Roseburia inulinivorans, Eubacterium halli, Faecalibacterium, Ruminococcus, Lactobacillus rogosae, among others. In addition, before HU there was a higher abundance of Clostridium_g24, which includes C. bolteae and C. clostridioforme, both considered pathogenic. This study provides the first evidence of the HU effect on the gut microbiome and unravels several microorganisms that could be considered candidate biomarkers for disease severity and HU efficacy.
Collapse
Affiliation(s)
- Mariana Delgadinho
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
| | - Catarina Ginete
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
| | - Brígida Santos
- Centro de Investigação em Saúde de Angola (CISA), Hospital Geral do Bengo, Bengo 9999, Angola
- Hospital Pediátrico David Bernardino (HPDB), Luanda 3067, Angola
| | - Carolina Fernandes
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
| | - Carina Silva
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
- Centro de Estatística e Aplicações, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Armandina Miranda
- Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal
| | | | - Miguel Brito
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
- Centro de Investigação em Saúde de Angola (CISA), Hospital Geral do Bengo, Bengo 9999, Angola
| |
Collapse
|
103
|
Memon FU, Yang Y, Zhang G, Leghari IH, Lv F, Wang Y, Laghari F, Khushk FA, Si H. Chicken Gut Microbiota Responses to Dietary Bacillus subtilis Probiotic in the Presence and Absence of Eimeria Infection. Microorganisms 2022; 10:1548. [PMID: 36013966 PMCID: PMC9412415 DOI: 10.3390/microorganisms10081548] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Coccidiosis is a well-known poultry disease that causes the severe destruction of the intestinal tract, resulting in reduced growth performance and immunity, disrupted gut homeostasis and perturbed gut microbiota. Supplementation of probiotics were explored to play a key role in improving growth performance, enhancing innate and adaptive immunity, maintaining gut homeostasis and modulating gut microbiota during enteric infection. This study was therefore designed to investigate the chicken gut whole microbiota responses to Bacillus subtilis (B. subtilis) probiotic feeding in the presence as well as absence of Eimeria infection. For that purpose, 84 newly hatched chicks were assigned into four groups, including (1) non-treated non-challenged control group (CG - ET), (2) non-treated challenged control group (CG + ET), (3) B. subtilis-fed non-challenged group (BS - ET) and (4) B. subtilis-fed challenged group (BS + ET). CG + ET and BS + ET groups were challenged with Eimeria tenella (E. tenella) on 21 day of housing. Our results for Alpha diversity revealed that chickens in both infected groups (CG + ET and BS + ET) had lowest indexes of Ace, Chao 1 and Shannon, while highest indexes of Simpson were found in comparison to non-challenged groups (CG - ET and BS - ET). Firmicutes was the most affected phylum in all experimental groups following Proteobacteria and Bacteroidota, which showed increased abundance in both non-challenged groups, whereas Proteobacteria and Bacteroidota affected both challenged groups. The linear discriminant analysis effect size method (lEfSe) analysis revealed that compared to the CG + ET group, supplementation of probiotic in the presence of Eimeria infection increased the abundance of some commensal genera, included Clostridium sensu stricto 1, Corynebacterium, Enterococcus, Romboutsia, Subdoligranulum, Bacillus, Turicibacter and Weissella, with roles in butyrate production, anti-inflammation, metabolic reactions and the modulation of protective pathways against pathogens. Collectively, these findings evidenced that supplementation of B. subtilis probiotic was positively influenced with commensal genera, thereby alleviating the Eimeria-induced intestinal disruption.
Collapse
Affiliation(s)
- Fareed Uddin Memon
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
- Department of Poultry Husbandry, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tando Jam 70060, Pakistan; (I.H.L.); (F.A.K.)
| | - Yunqiao Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
| | - Geyin Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
| | - Imdad Hussain Leghari
- Department of Poultry Husbandry, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tando Jam 70060, Pakistan; (I.H.L.); (F.A.K.)
| | - Feifei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
| | - Yuhan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
| | - Farooque Laghari
- Department of Animal Production and Environment Control, College of Animal Sciences and Technology, Southeast Agriculture University, Harbin 150030, China;
| | - Farooque Ahmed Khushk
- Department of Poultry Husbandry, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tando Jam 70060, Pakistan; (I.H.L.); (F.A.K.)
| | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (F.U.M.).; (Y.Y.); (G.Z.); (F.L.); (Y.W.)
| |
Collapse
|
104
|
Zhao H, Li Y, Lv P, Huang J, Tai R, Jin X, Wang J, Wang X. Salmonella Phages Affect the Intestinal Barrier in Chicks by Altering the Composition of Early Intestinal Flora: Association With Time of Phage Use. Front Microbiol 2022; 13:947640. [PMID: 35910610 PMCID: PMC9329052 DOI: 10.3389/fmicb.2022.947640] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/15/2022] [Indexed: 01/03/2023] Open
Abstract
Phages show promise in replacing antibiotics to treat or prevent bacterial diseases in the chicken breeding industry. Chicks are easily affected by their environment during early growth. Thus, this study investigated whether oral phages could affect the intestinal barrier function of chicks with a focus on the cecal microbiome. In a two-week trial, forty one-day-old hens were randomly divided into four groups: (1) NC, negative control; (2) Phage 1, 109 PFU phage/day (days 3–5); (3) Phage 2, 109 PFU phage/day (days 8–10); and (4) AMX, 1 mg/mL amoxicillin/day (days 8–10). High-throughput sequencing results of cecal contents showed that oral administration of phages significantly affected microbial community structure and community composition, and increased the relative abundance of Enterococcus. The number of different species in the Phage 1 group was much higher than that in the Phage 2 group, and differences in alpha and beta diversity also indicated that the magnitude of changes in the composition of the cecal microbiota correlated with the time of phage use. Particularly in the first stage of cecal microbiota development, oral administration of bacteriophages targeting Salmonella may cause substantial changes in chicks, as evidenced by the results of the PICRUSt2 software function prediction, reminding us to be cautious about the time of phage use in chicks and to avoid high oral doses of phages during the first stage. Additionally, the Phage 2 samples not only showed a significant increase in the relative abundance of Bifidobacterium and Subdoligranulum, but also improved the intestinal morphology (jejunum) and increased the mRNA expression level of occludin and ZO-1. We concluded that phages do not directly interact with eukaryotic cells. The enhancement of intestinal barrier function by phages in chicks may be related to changes in the intestinal flora induced by phages. This implies that phages may affect intestinal health by regulating the intestinal flora. This study provides new ideas for phage prevention of intestinal bacterial infections and promotes large-scale application of phages in the poultry industry.
Collapse
Affiliation(s)
- Hongze Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yue Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Peilin Lv
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinmei Huang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rong Tai
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiue Jin
- Hubei Provincial Institute of Veterinary Drug Control, Wuhan, China
| | - Jianhua Wang
- Hubei Provincial Institute of Veterinary Drug Control, Wuhan, China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Xiliang Wang,
| |
Collapse
|
105
|
Gong J, He L, Zou Q, Zhao Y, Zhang B, Xia R, Chen B, Cao M, Gong W, Lin L, Lin X, Wang G, Guo M, He J, Xiao C, Chen J. Association of serum 25-hydroxyvitamin D (25(OH)D) levels with the gut microbiota and metabolites in postmenopausal women in China. Microb Cell Fact 2022; 21:137. [PMID: 35820909 PMCID: PMC9275287 DOI: 10.1186/s12934-022-01858-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Vitamin D insufficiency or deficiency is associated with an altered microbiota in older men. However, the relationship between the gut microbiota and 25-hydroxyvitamin D (25(OH)D) levels remains unknown in postmenopausal women. In this study, fecal microbiota profiles for 88 postmenopausal women in the high 25(OH)D (HVD) group (n = 44) and the low 25(OH)D (LVD) group (n = 44) were determined. An integrated 16S rRNA gene sequencing and liquid chromatography–mass spectrometry (LC–MS)-based metabolomics approach was applied to explore the association of serum 25(OH)D levels with the gut microbiota and fecal metabolic phenotype. Adjustments were made using several statistical models for potential confounding variables identified from the literature. Results The results demonstrated that the community diversity estimated by the Observe, Chao1 and ACE indexes was significantly lower in the LVD group than in the HVD group. Additionally, two kinds of characteristic differences in the microflora were analyzed in the HVD group, and ten kinds of characteristic differences in the microflora were analyzed in the LVD group. We observed that some bacteria belonging to the genera Bifidobacterium, Bacillus, F0332 and Gemella, were enriched in the LVD group, as were other genera, including Lachnoclostridium, UC5_1_2E3, Ruminococcus_gnavus_group and un_f_Lachnospiraceae. Christensenellaceae, Eggerthellaceae and Cloacibacillus were enriched in the HVD group. The L-pyroglutamic acid, inosine, and L-homocysteic acid levels were higher in the HVD group and were negatively correlated with the 1,2-benzenedicarboxylic acid and cholic acid metabolic levels. Conclusions These observations provide a better understanding of the relationships between serum 25(OH)D levels and the fecal microbiota and metabolites in postmenopausal women. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01858-6.
Collapse
Affiliation(s)
- Jinhua Gong
- Xiamen Institute of Union Respiratory Health, Xiamen, China
| | - Lina He
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Qinyuan Zou
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Yangyang Zhao
- School of Medicine, Xiamen University, Xiamen, China
| | - Bangzhou Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Rongmu Xia
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Baolong Chen
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Man Cao
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Wenxiu Gong
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Lin Lin
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Xiujuan Lin
- Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Guowei Wang
- Institute of Basic Theories of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Muyun Guo
- Pulmonary and Critical Care Medicine, Anyang District Hospital, Anyang, China
| | - Jianquan He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China. .,Department of Rehabilitation, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Chuanxing Xiao
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China. .,Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China. .,School of Basic Medical Science, Central South University, Changsha, China.
| | - Jian Chen
- Department of Rehabilitation, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
106
|
Gradisteanu Pircalabioru G, Liaw J, Gundogdu O, Corcionivoschi N, Ilie I, Oprea L, Musat M, Chifiriuc MC. Effects of the Lipid Profile, Type 2 Diabetes and Medication on the Metabolic Syndrome—Associated Gut Microbiome. Int J Mol Sci 2022; 23:ijms23147509. [PMID: 35886861 PMCID: PMC9318871 DOI: 10.3390/ijms23147509] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
Metabolic syndrome (MetSyn) is a major health problem affecting approximately 25% of the worldwide population. Since the gut microbiota is highly connected to the host metabolism, several recent studies have emerged to characterize the role of the microbiome in MetSyn development and progression. To this end, our study aimed to identify the microbiome patterns which distinguish MetSyn from type 2 diabetes mellitus (T2DM). We performed 16S rRNA amplicon sequencing on a cohort of 70 individuals among which 40 were MetSyn patients. The microbiome of MetSyn patients was characterised by reduced diversity, loss of butyrate producers (Subdoligranulum, Butyricicoccus, Faecalibacterium prausnitzii) and enrichment in the relative abundance of fungal populations. We also show a link between the gut microbiome and lipid metabolism in MetSyn. Specifically, low-density lipoproteins (LDL) and high-density lipoproteins (HDL) display a positive effect on gut microbial diversity. When interrogating the signature of gut microbiota in a subgroup of patients harbouring both MetSyn and T2DM conditions, we observed a significant increase in taxa such as Bacteroides, Clostridiales, and Erysipelotrichaceae. This preliminary study shows for the first time that T2DM brings unique signatures of gut microbiota in MetSyn patients. We also highlight the impact of metformin treatment on the gut microbiota. Metformin administration was linked to changes in Prevotellaceae, Rickenellaceae, and Clostridiales. Further research focusing on the microbiome-metabolome patterns is needed to clarify the exact association of various gut microbial communities with the progression of T2DM and the occurrence of various complications in MetSyn patients.
Collapse
Affiliation(s)
| | - Janie Liaw
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
- Correspondence: (G.G.P.); (O.G.)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT9 5PX, UK;
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine—King Michael I of Romania, 300645 Timisoara, Romania
| | | | - Luciana Oprea
- National Institute of Endocrinology C.I. Parhon, 011863 Bucharest, Romania; (L.O.); (M.M.)
| | - Madalina Musat
- National Institute of Endocrinology C.I. Parhon, 011863 Bucharest, Romania; (L.O.); (M.M.)
- Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania;
- Romanian Academy, 010071 Bucharest, Romania
| |
Collapse
|
107
|
Gravina AG, Romeo M, Pellegrino R, Tuccillo C, Federico A, Loguercio C. Just Drink a Glass of Water? Effects of Bicarbonate-Sulfate-Calcium-Magnesium Water on the Gut-Liver Axis. Front Pharmacol 2022; 13:869446. [PMID: 35837275 PMCID: PMC9274271 DOI: 10.3389/fphar.2022.869446] [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: 02/14/2022] [Accepted: 05/16/2022] [Indexed: 12/18/2022] Open
Abstract
Background and Aim: Fonte Essenziale® water is a bicarbonate-sulfate-calcium-magnesium water, low in sodium, recognized by the Italian health care system in hydropinotherapy and hepatobiliary dyspepsia therapy. We wanted to explore its effects on the gut-liver axis and microbiota in non-alcoholic fatty liver disease patients. Patients and Methods: We considered enrollment for 70 patients, of which four were excluded. We finally enrolled 55 patients with ultrasound-documented steatosis (SPs+) and 11 patients without it (SPs-). They then drank 400 ml of water for 6 months in the morning on an empty stomach. Routine hematochemical and metabolic parameters, oxidative stress parameters, gastrointestinal hormone levels, and fecal parameters of the gut microbiota were evaluated at three different assessment times, at baseline (T0), after 6 months (T6), and after a further 6 months of water washout (T12). We lost, in follow-up, 4 (T6) and 22 (T12) patients. Results: Between T0-T6, we observed a significant Futuin A and Selenoprotein A decrease and a GLP-1 and PYY increase in SPs+ and the same for Futuin A and GLP-1 in SPs-. Effects were lost at T12. In SPs+, between T0-T12 and T6-12, a significant reduction in Blautia was observed; between T0-T12, a reduction of Collinsella unc. was observed; and between T0-T12 and T6-12, an increase in Subdoligranulum and Dorea was observed. None of the bacterial strains we analyzed varied significantly in the SPs- population. Conclusion: These results indicate beneficial effects of water on gastrointestinal hormones and hence on the gut-liver axis in the period in which subjects drank water both in SPs- and in SPs+.
Collapse
|
108
|
Abstract
The gut microbiota is now considered as one of the key elements contributing to the regulation of host health. Virtually all our body sites are colonised by microbes suggesting different types of crosstalk with our organs. Because of the development of molecular tools and techniques (ie, metagenomic, metabolomic, lipidomic, metatranscriptomic), the complex interactions occurring between the host and the different microorganisms are progressively being deciphered. Nowadays, gut microbiota deviations are linked with many diseases including obesity, type 2 diabetes, hepatic steatosis, intestinal bowel diseases (IBDs) and several types of cancer. Thus, suggesting that various pathways involved in immunity, energy, lipid and glucose metabolism are affected.In this review, specific attention is given to provide a critical evaluation of the current understanding in this field. Numerous molecular mechanisms explaining how gut bacteria might be causally linked with the protection or the onset of diseases are discussed. We examine well-established metabolites (ie, short-chain fatty acids, bile acids, trimethylamine N-oxide) and extend this to more recently identified molecular actors (ie, endocannabinoids, bioactive lipids, phenolic-derived compounds, advanced glycation end products and enterosynes) and their specific receptors such as peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ), aryl hydrocarbon receptor (AhR), and G protein-coupled receptors (ie, GPR41, GPR43, GPR119, Takeda G protein-coupled receptor 5).Altogether, understanding the complexity and the molecular aspects linking gut microbes to health will help to set the basis for novel therapies that are already being developed.
Collapse
Affiliation(s)
- Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| |
Collapse
|
109
|
Han DS, Wu WK, Liu PY, Yang YT, Hsu HC, Kuo CH, Wu MS, Wang TG. Differences in the gut microbiome and reduced fecal butyrate in elders with low skeletal muscle mass. Clin Nutr 2022; 41:1491-1500. [DOI: 10.1016/j.clnu.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
|
110
|
Abstract
The gut microbiota is now considered as one of the key elements contributing to the regulation of host health. Virtually all our body sites are colonised by microbes suggesting different types of crosstalk with our organs. Because of the development of molecular tools and techniques (ie, metagenomic, metabolomic, lipidomic, metatranscriptomic), the complex interactions occurring between the host and the different microorganisms are progressively being deciphered. Nowadays, gut microbiota deviations are linked with many diseases including obesity, type 2 diabetes, hepatic steatosis, intestinal bowel diseases (IBDs) and several types of cancer. Thus, suggesting that various pathways involved in immunity, energy, lipid and glucose metabolism are affected.In this review, specific attention is given to provide a critical evaluation of the current understanding in this field. Numerous molecular mechanisms explaining how gut bacteria might be causally linked with the protection or the onset of diseases are discussed. We examine well-established metabolites (ie, short-chain fatty acids, bile acids, trimethylamine N-oxide) and extend this to more recently identified molecular actors (ie, endocannabinoids, bioactive lipids, phenolic-derived compounds, advanced glycation end products and enterosynes) and their specific receptors such as peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ), aryl hydrocarbon receptor (AhR), and G protein-coupled receptors (ie, GPR41, GPR43, GPR119, Takeda G protein-coupled receptor 5).Altogether, understanding the complexity and the molecular aspects linking gut microbes to health will help to set the basis for novel therapies that are already being developed.
Collapse
Affiliation(s)
- Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Brussels, Belgium
| |
Collapse
|
111
|
Gutiérrez-Repiso C, Garrido-Sánchez L, Alcaide-Torres J, Cornejo-Pareja I, Ocaña-Wilhelmi L, García-Fuentes E, Moreno-Indias I, Tinahones FJ. Predictive Role of Gut Microbiota in Weight Loss Achievement after Bariatric Surgery. J Am Coll Surg 2022; 234:861-871. [PMID: 35426398 DOI: 10.1097/xcs.0000000000000145] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Bariatric surgery induces changes in gut microbiota that have been suggested to contribute to weight loss and metabolic improvement. However, whether preoperative gut microbiota composition could predict response to bariatric surgery has not yet been elucidated. STUDY DESIGN Seventy-six patients who underwent sleeve gastrectomy were classified according to the percentage of excess weight loss (%EWL) 1 year after surgery in the responder group: >50%EWL (n=50) and the nonresponder group: <50%EWL (n=26). Patients were evaluated before surgery, and 3 months and 1 year after surgery. Gut microbiota composition was analyzed before surgery (n=76) and 3 months after bariatric surgery (n=40). RESULTS Diversity analysis did not show differences between groups before surgery or 3 months after surgery. Before surgery, there were differences in the abundance of members belonging to Bacteroidetes and Firmicutes phyla (nonresponder group: enriched in Bacteroidaceae, Bacteroides, Bacteroides uniformis, Alistipes finegoldii, Alistipes alistipes, Dorea formicigenerans, and Ruminococcus gnavus. Responder group: enriched in Peptostreptococcaceae, Gemmiger, Gemiger formicilis, Barnesiella, Prevotellaceae, and Prevotella; linear discriminant analysis >2; p < 0.05). Prevotella-to-Bacteroides ratio was significantly lower in the nonresponder group compared to the responder group (p = 0.048). After surgery, the responder group showed an enrichment in taxa that have been shown to have beneficial effects on host metabolism. Before surgery, PICRUSt analysis showed an enrichment in pathways involved in the biosynthesis components of the O-antigen polysaccharideunits in lipopolysaccharides in the nonresponder group. CONCLUSIONS Preoperative gut microbiota could have an impact on bariatric surgery outcomes. Prevotella-to-Bacteroides ratio could be used as a predictive tool for weight loss trajectory. Early after surgery, patients who experienced successful weight loss showed an enrichment in taxa related to beneficial effects on host metabolism.
Collapse
Affiliation(s)
- Carolina Gutiérrez-Repiso
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Garrido-Sánchez
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Alcaide-Torres
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Cornejo-Pareja
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Ocaña-Wilhelmi
- Unidad de Gestión Clínica de Cirugía General y del Aparato Digestivo (Ocaña-Wilhelmi), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología (Ocaña-Wilhelmi), Universidad de Málaga, Málaga, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo (García-Fuentes), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) (García-Fuentes), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Moreno-Indias
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J Tinahones
- From the Unidad de Gestión Clínica de Endocrinología y Nutrición (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn) (Gutiérrez-Repiso, Garrido-Sánchez, Alcaide-Torres, Cornejo-Pareja, Moreno-Indias, Tinahones), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina y Dermatología (Tinahones), Universidad de Málaga, Málaga, Spain
| |
Collapse
|
112
|
Montanari C, Ceccarani C, Corsello A, Zuvadelli J, Ottaviano E, Dei Cas M, Banderali G, Zuccotti G, Borghi E, Verduci E. Glycomacropeptide Safety and Its Effect on Gut Microbiota in Patients with Phenylketonuria: A Pilot Study. Nutrients 2022; 14:1883. [PMID: 35565850 PMCID: PMC9104775 DOI: 10.3390/nu14091883] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 12/07/2022] Open
Abstract
Glycomacropeptide (GMP) represents a good alternative protein source in Phenylketonuria (PKU). In a mouse model, it has been suggested to exert a prebiotic role on beneficial gut bacteria. In this study, we performed the 16S rRNA sequencing to evaluate the effect of 6 months of GMP supplementation on the gut microbiota of nine PKU patients, comparing their bacterial composition and clinical parameters before and after the intervention. GMP seems to be safe from both the microbiological and the clinical point of view. Indeed, we did not observe dramatic changes in the gut microbiota but a specific prebiotic effect on the butyrate-producer Agathobacter spp. and, to a lesser extent, of Subdoligranulum. Clinically, GMP intake did not show a significant impact on both metabolic control, as phenylalanine values were kept below the age target and nutritional parameters. On the other hand, an amelioration of calcium phosphate homeostasis was observed, with an increase in plasmatic vitamin D and a decrease in alkaline phosphatase. Our results suggest GMP as a safe alternative in the PKU diet and its possible prebiotic role on specific taxa without causing dramatic changes in the commensal microbiota.
Collapse
Affiliation(s)
- Chiara Montanari
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (C.M.); (A.C.); (G.Z.); (E.V.)
| | - Camilla Ceccarani
- Institute of Biomedical Technologies, National Research Council, 20090 Segrate, Italy;
| | - Antonio Corsello
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (C.M.); (A.C.); (G.Z.); (E.V.)
| | - Juri Zuvadelli
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo, San Paolo Hospital, University of Milan, 20142 Milan, Italy; (J.Z.); (G.B.)
| | - Emerenziana Ottaviano
- Department of Health Science, University of Milan, 20142 Milan, Italy; (E.O.); (M.D.C.)
| | - Michele Dei Cas
- Department of Health Science, University of Milan, 20142 Milan, Italy; (E.O.); (M.D.C.)
| | - Giuseppe Banderali
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo, San Paolo Hospital, University of Milan, 20142 Milan, Italy; (J.Z.); (G.B.)
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (C.M.); (A.C.); (G.Z.); (E.V.)
| | - Elisa Borghi
- Department of Health Science, University of Milan, 20142 Milan, Italy; (E.O.); (M.D.C.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (C.M.); (A.C.); (G.Z.); (E.V.)
- Department of Health Science, University of Milan, 20142 Milan, Italy; (E.O.); (M.D.C.)
| |
Collapse
|
113
|
Debédat J, Le Roy T, Voland L, Belda E, Alili R, Adriouch S, Bel Lassen P, Kasahara K, Hutchison E, Genser L, Torres L, Gamblin C, Rouault C, Zucker JD, Kapel N, Poitou C, Marcelin G, Rey FE, Aron-Wisnewsky J, Clément K. The human gut microbiota contributes to type-2 diabetes non-resolution 5-years after Roux-en-Y gastric bypass. Gut Microbes 2022; 14:2050635. [PMID: 35435140 PMCID: PMC9037437 DOI: 10.1080/19490976.2022.2050635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is efficient at inducing drastic albeit variable weight loss and type-2 diabetes (T2D) improvements in patients with severe obesity and T2D. We hypothesized a causal implication of the gut microbiota (GM) in these metabolic benefits, as RYGB is known to deeply impact its composition. In a cohort of 100 patients with baseline T2D who underwent RYGB and were followed for 5-years, we used a hierarchical clustering approach to stratify subjects based on the severity of their T2D (Severe vs Mild) throughout the follow-up. We identified via nanopore-based GM sequencing that the more severe cases of unresolved T2D were associated with a major increase of the class Bacteroidia, including 12 species comprising Phocaeicola dorei, Bacteroides fragilis, and Bacteroides caecimuris. A key observation is that patients who underwent major metabolic improvements do not harbor this enrichment in Bacteroidia, as those who presented mild cases of T2D at all times. In a separate group of 36 patients with similar baseline clinical characteristics and preoperative GM sequencing, we showed that this increase in Bacteroidia was already present at baseline in the most severe cases of T2D. To explore the causal relationship linking this enrichment in Bacteroidia and metabolic alterations, we selected 13 patients across T2D severity clusters at 5-years and performed fecal matter transplants in mice. Our results show that 14 weeks after the transplantations, mice colonized with the GM of Severe donors have impaired glucose tolerance and insulin sensitivity as compared to Mild-recipients, all in the absence of any difference in body weight and composition. GM sequencing of the recipient animals revealed that the hallmark T2D-severity associated bacterial features were transferred and were associated with the animals' metabolic alterations. Therefore, our results further establish the GM as a key contributor to long-term glucose metabolism improvements (or lack thereof) after RYGB.
Collapse
Affiliation(s)
- Jean Debédat
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Tiphaine Le Roy
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Lise Voland
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | | | - Rohia Alili
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Solia Adriouch
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Pierre Bel Lassen
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Kazuyuki Kasahara
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Evan Hutchison
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Laurent Genser
- Visceral Surgery Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Licia Torres
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Camille Gamblin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Christine Rouault
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Jean-Daniel Zucker
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Universités, Institut de Recherche pour le Développement (IRD), France
| | - Nathalie Kapel
- Functional Coprology Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Christine Poitou
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Geneviève Marcelin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Judith Aron-Wisnewsky
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,CONTACT Judith Aron-Wisnewsky Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Nutrition and obesities; systemic approaches (NutriOmics)75013, Paris, France
| | - Karine Clément
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,Karine Clément Nutrition and obesities; systemic approaches (NutriOmics) Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, ParisFranceNutrition Department
| |
Collapse
|
114
|
Tang Y, Zhang X, Wang Y, Guo Y, Zhu P, Li G, Zhang J, Ma Q, Zhao L. Dietary ellagic acid ameliorated Clostridium perfringens-induced subclinical necrotic enteritis in broilers via regulating inflammation and cecal microbiota. J Anim Sci Biotechnol 2022; 13:47. [PMID: 35436978 PMCID: PMC9016943 DOI: 10.1186/s40104-022-00694-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Background Subclinical necrotic enteritis (SNE), a common intestinal disease of broiler caused by Clostridium perfringens, could reduce production performance of broilers by chronic intestinal damage and poor absorption of nutrients. Ellagic acid (EA) has been reported to present antioxidant and anti-inflammatory properties on human and animals in many aspects. This study was conducted to evaluate the effect and mechanism of EA in relieving SNE in broilers induced by C. perfringens. Results C. perfringens challenge decreased body weight (BW), average daily gain (ADG), jejunal villi height/crypt depth (V/C) ratio, the activity of catalase (CAT) and the mRNA expression of zonula occludens 1 (ZO-1) in jejunal mucosa of broilers. While feed conversion ratios (FCR), jejunal crypt depth (CD), the activities of myeloperoxidase (MPO) and diamine oxidase (DAO), as well as the concentrations of interleukin 6 (IL-6), C-reactive protein (CRP) and procalcitonin (PCT) in serum, the activities of inducible nitric oxide synthase (iNOS) and lysozyme (LZM), the concentration of malondialdehyde (MDA), and the mRNA expressions of claudin-2, TNF-α, IL-1β, TLR-4, TLR-2, NF-κB, JAK3, STAT6 and iNOS in jejunal mucosa of broilers were increased by C. perfringens challenge. Dietary EA supplement relieved these adverse effects, and heightened jejunal villi height (VH), the concentration of D-xylose in plasma, activity of superoxide dismutase (SOD), and the mRNA expression of occludin in jejunal mucosa of broilers. The alpha diversity of cecal microbiota indicated that dietary EA supplement increased observed species and Shannon index. C. perfringens challenge increased the relative abundance of Firmicutes and decreased the relative abundance of Desulfobacterota in cecal microbiota. EA increased the relative abundance of Firmicutes in cecal microbiota. LEfSe analysis showed that C. perfringens challenge triggered the imbalance of cecal microbiota in broilers, dietary EA supplementation led to a small beneficial effect on microbiota, while the simultaneous effect of them seemed to stimulate the immune function of broilers by improving the microbiota balance. Conclusions Dietary EA ameliorated C. perfringens-induced SNE in broilers via regulating jejunal inflammation signaling pathways TLR/NF-κB and JAK3/STAT6, relieving jejunal oxidative stress and balancing cecal microbiota to inhibit intestinal barrier damage, prevent systemic inflammatory response and improve nutrient absorption capacity, finally protect and enhance growth performance of broilers. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-022-00694-3.
Collapse
Affiliation(s)
- Yu Tang
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Xinyue Zhang
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Yanan Wang
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Yongpeng Guo
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Peiqi Zhu
- Jiangsu Lihua animal husbandry Co., Ltd. No. 500, Hexi Village, Luxi Village Committee, Niutang Town, Wujin District, Changzhou City, Jiangsu Province, 213168, People's Republic of China
| | - Guiguan Li
- COFCO feed Co., Ltd, 4th Floor, No. 6, Nandaan Hutong, Xicheng District, Beijing, 100193, People's Republic of China
| | - Jianyun Zhang
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. West Road Yuanming yuan, Beijing, 100193, People's Republic of China.
| |
Collapse
|
115
|
Garzarella EU, Navajas-Porras B, Pérez-Burillo S, Ullah H, Esposito C, Santarcangelo C, Hinojosa-Nogueira D, Pastoriza S, Zaccaria V, Xiao J, Rufián-Henares JÁ, Daglia M. Evaluating the effects of a standardized polyphenol mixture extracted from poplar-type propolis on healthy and diseased human gut microbiota. Biomed Pharmacother 2022; 148:112759. [PMID: 35248845 DOI: 10.1016/j.biopha.2022.112759] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION A large body of evidence suggests that propolis exerts antioxidant, anti-inflammatory, and antimicrobial activities, mostly ascribed to its polyphenol content. Growing evidence suggests that propolis could modulate gut microbiota exerting a positive impact on several pathological conditions. The aim of this study was to determine the in vitro impact of a poplar-type propolis extract with a standardized polyphenol content, on the composition and functionality of gut microbiota obtained from fecal material of five different donors (healthy adults, and healthy, obese, celiac, and food allergic children). METHODS The standardized polyphenol mixture was submitted to a simulated in vitro digestion-fermentation process, designed to mimic natural digestion in the human oral, gastric, and intestinal chambers. The antioxidant profile of propolis before and after the digestion-fermentation process was determined. 16 S rRNA amplicon next-generation sequencing (NGS) was used to test the effects on the gut microbiota of propolis extract. The profile of the short-chain fatty acids (SCFA) produced by the microbiota was also investigated through a chromatographic method coupled with UV detection. RESULTS In vitro digestion and fermentation induced a decrease in the antioxidant profile of propolis (i.e., decrease of total polyphenol content, antiradical and reducing activities). Propolis fermentation exhibited a modulatory effect on gut microbiota composition and functionality of healthy and diseased subjects increasing the concentration of SCFA. CONCLUSIONS Overall, these data suggest that propolis might contribute to gut health and could be a candidate for further studies in view of its use as a prebiotic ingredient.
Collapse
Affiliation(s)
- Emanuele Ugo Garzarella
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Naples 80131,Italy
| | - Beatriz Navajas-Porras
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Sergio Pérez-Burillo
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Hammad Ullah
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Naples 80131,Italy
| | - Cristina Esposito
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Naples 80131,Italy
| | - Cristina Santarcangelo
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Naples 80131,Italy
| | - Daniel Hinojosa-Nogueira
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Silvia Pastoriza
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | | | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Science, University of Vigo, Vigo, Spain; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain; Instituto de Investigación Biosanitaria (ibs.GRANADA), Universidad de Granada, Granada 18140, Spain.
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, Naples 80131,Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
116
|
Stark KG, Falkowski NR, Brown CA, McDonald RA, Huffnagle GB. Contribution of the Microbiome, Environment, and Genetics to Mucosal Type 2 Immunity and Anaphylaxis in a Murine Food Allergy Model. FRONTIERS IN ALLERGY 2022; 3:851993. [PMID: 35769569 PMCID: PMC9234882 DOI: 10.3389/falgy.2022.851993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
There is heterogeneity inherent in the immune responses of individual mice in murine models of food allergy, including anaphylaxis, similar to the clinical heterogeneity observed in humans with food allergies to a defined food. One major driver of this heterogeneity may be differences in the microbiome between sensitized individuals. Our laboratory and others have reported that disruption of the microbiome (dysbiosis) by broad spectrum antibiotics and/or yeast colonization can alter systemic immunity and favor the development of mucosal Type 2 immunity to aeroallergens. Our objective was to use a well-characterized murine model (Balb/c mice) of food allergies (chicken egg ovalbumin, OVA) and determine if antibiotic-mediated dysbiosis (including C. albicans colonization) could enhance the manifestation of food allergies. Furthermore, we sought to identify elements of the microbiome and host response that were associated with this heterogeneity in the anaphylactic reaction between individual food allergen-sensitized mice. In our dataset, the intensity of the anaphylactic reactions was most strongly associated with a disrupted microbiome that included colonization by C. albicans, loss of a specific Lachnoclostridium species (tentatively, Lachnoclostridium YL32), development of a highly polarized Type 2 response in the intestinal mucosa and underlying tissue, and activation of mucosal mast cells. Serum levels of allergen-specific IgE were not predictive of the response and a complete absence of a microbiome did not fully recapitulate the response. Conventionalization of germ-free mice resulted in Akkermansia muciniphila outgrowth and a higher degree of heterogeneity in the allergic response. C57BL/6 mice remained resistant even under the same dysbiosis-inducing antibiotic regimens, while changes in the microbiome markedly altered the reactivity of Balb/c mice to OVA, as noted above. Strikingly, we also observed that genetically identical mice from different rooms in our vivarium develop different levels of a Type 2 response, as well as anaphylactic reactions. The intestinal microbiome in these mice also differed between rooms. Thus, our data recapitulate the heterogeneity in anaphylactic reactions, ranging from severe to none, seen in patients that have circulating levels of food allergen-reactive IgE and support the concept that alterations in the microbiome can be one factor underlying this heterogeneity.
Collapse
Affiliation(s)
- Kelsey G. Stark
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Nicole R. Falkowski
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Christopher A. Brown
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Institute for Research on Innovation and Science (IRIS), Institute for Social Research (ISR), University of Michigan, Ann Arbor, MI, United States
| | - Roderick A. McDonald
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Gary B. Huffnagle
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Gary B. Huffnagle
| |
Collapse
|
117
|
Abot A, Brochot A, Pomié N, Wemelle E, Druart C, Régnier M, Delzenne NM, de Vos WM, Knauf C, Cani PD. Camu-Camu Reduces Obesity and Improves Diabetic Profiles of Obese and Diabetic Mice: A Dose-Ranging Study. Metabolites 2022; 12:metabo12040301. [PMID: 35448490 PMCID: PMC9025096 DOI: 10.3390/metabo12040301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Overweight, obesity, and their comorbidities are currently considered a major public health concern. Today considerable efforts are still needed to develop efficient strategies able to attenuate the burden of these diseases. Nutritional interventions, some with plant extracts, present promising health benefits. In this study, we evaluated the action of Camu-Camu (Myrciaria dubia), an Amazonian fruit rich in polyphenols and vitamin C, on the prevention of obesity and associated disorders in mice and the abundance of Akkermansia muciniphila in both cecum and feces. Methods: We investigated the dose-response effects of Camu-Camu extract (CCE) in the context of high-fat-diet (HFD)-induced obesity. After 5 weeks of supplementation, we demonstrated that the two doses of CCE differently improved glucose and lipid homeostasis. The lowest CCE dose (62.5 mg/kg) preferentially decreased non-HDL cholesterol and free fatty acids (FFA) and increased the abundance of A. muciniphila without affecting liver metabolism, while only the highest dose of CCE (200 mg/kg) prevented excessive body weight gain, fat mass gain, and hepatic steatosis. Both doses decreased fasting hyperglycemia induced by HFD. In conclusion, the use of plant extracts, and particularly CCE, may represent an additional option in the support of weight management strategies and glucose homeostasis alteration by mechanisms likely independent from the modulation of A. muciniphila abundance.
Collapse
Affiliation(s)
- Anne Abot
- Enterosys SAS, 31670 Labège, France; (A.A.); (N.P.)
| | - Amandine Brochot
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | | | - Eve Wemelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
| | - Céline Druart
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | - Marion Régnier
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Nathalie M. Delzenne
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- Correspondence: (C.K.); (P.D.C.)
| | - Patrice D. Cani
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
- Correspondence: (C.K.); (P.D.C.)
| |
Collapse
|
118
|
Moughaizel M, Dagher E, Jablaoui A, Thorin C, Rhimi M, Desfontis JC, Mallem Y. Long-term high-fructose high-fat diet feeding elicits insulin resistance, exacerbates dyslipidemia and induces gut microbiota dysbiosis in WHHL rabbits. PLoS One 2022; 17:e0264215. [PMID: 35196347 PMCID: PMC8865649 DOI: 10.1371/journal.pone.0264215] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/04/2022] [Indexed: 01/12/2023] Open
Abstract
The metabolic syndrome (MetS) has become a global public health burden due to its link to cardiovascular disease and diabetes mellitus. The present study was designed to characterize the metabolic and cardiovascular disturbances, as well as changes in gut microbiota associated with high-fructose high-fat diet (HFFD)-induced MetS in Watanabe heritable hyperlipidemic (WHHL) rabbits. Twenty-one Watanabe rabbits were assigned to a control (n = 9) and HFFD (n = 12) groups, receiving a chow diet and a HFFD, respectively. During a 12-weeks protocol, morphological parameters were monitored; plasma fasting levels of lipids, glucose and insulin were measured and a glucose tolerance test (GTT) was performed. HOMA-IR was calculated. Cardiac function and vascular reactivity were evaluated using the Langendorff isolated heart and isolated carotid arteries methods, respectively. 16S rRNA sequencing of stool samples was used to determine gut microbial composition and abundance. HFFD-fed Watanabe rabbits exhibited increased fasting insulin (p < 0.03, 12th week vs. Baseline), HOMA-IR (p < 0.03 vs. Control), area under the curve of the GTT (p < 0.02 vs. Control), triglycerides (p < 0.05, 12th week vs. Baseline), TC (p < 0.01 vs. Control), LDL-C (p < 0.001 vs. Control). The HFFD group also displayed a significant decrease in intestinal microbial richness, evenness and diversity (FDR < 0.001, FDR < 0.0001, FDR < 0.01, respectively vs. Control group) and an increase in its Firmicutes/Bacteroidetes ratio (R = 3.39 in control vs. R = 28.24 in the HFFD group) indicating a shift in intestinal microbial composition and diversity. Our results suggest that HFFD induces insulin resistance and gut microbiota dysbiosis and accentuates dyslipidemia; and that, when subjected to HFFD, Watanabe rabbits might become a potential diet-induced MetS animal models with two main features, dyslipidemia and insulin resistance.
Collapse
Affiliation(s)
- Michelle Moughaizel
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
- * E-mail: (MM); (YM)
| | - Elie Dagher
- Laboniris, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Amin Jablaoui
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Chantal Thorin
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Moez Rhimi
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jean-Claude Desfontis
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Yassine Mallem
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering, Nantes, France
- * E-mail: (MM); (YM)
| |
Collapse
|
119
|
Le Roy T, Moens de Hase E, Van Hul M, Paquot A, Pelicaen R, Régnier M, Depommier C, Druart C, Everard A, Maiter D, Delzenne NM, Bindels LB, de Barsy M, Loumaye A, Hermans MP, Thissen JP, Vieira-Silva S, Falony G, Raes J, Muccioli GG, Cani PD. Dysosmobacter welbionis is a newly isolated human commensal bacterium preventing diet-induced obesity and metabolic disorders in mice. Gut 2022; 71:534-543. [PMID: 34108237 PMCID: PMC8862106 DOI: 10.1136/gutjnl-2020-323778] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/20/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To investigate the abundance and the prevalence of Dysosmobacter welbionis J115T, a novel butyrate-producing bacterium isolated from the human gut both in the general population and in subjects with metabolic syndrome. To study the impact of this bacterium on host metabolism using diet-induced obese and diabetic mice. DESIGN We analysed the presence and abundance of the bacterium in 11 984 subjects using four human cohorts (ie, Human Microbiome Project, American Gut Project, Flemish Gut Flora Project and Microbes4U). Then, we tested the effects of daily oral gavages with live D. welbionis J115T on metabolism and several hallmarks of obesity, diabetes, inflammation and lipid metabolism in obese/diabetic mice. RESULTS This newly identified bacterium was detected in 62.7%-69.8% of the healthy population. Strikingly, in obese humans with a metabolic syndrome, the abundance of Dysosmobacter genus correlates negatively with body mass index, fasting glucose and glycated haemoglobin. In mice, supplementation with live D. welbionis J115T, but not with the pasteurised bacteria, partially counteracted diet-induced obesity development, fat mass gain, insulin resistance and white adipose tissue hypertrophy and inflammation. In addition, live D. welbionis J115T administration protected the mice from brown adipose tissue inflammation in association with increased mitochondria number and non-shivering thermogenesis. These effects occurred with minor impact on the mouse intestinal microbiota composition. CONCLUSIONS These results suggest that D. welbionis J115T directly and beneficially influences host metabolism and is a strong candidate for the development of next-generation beneficial bacteria targeting obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Tiphaine Le Roy
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Emilie Moens de Hase
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Adrien Paquot
- Louvain Drug Research Institute (LDRI), Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Rudy Pelicaen
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marion Régnier
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Clara Depommier
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Céline Druart
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Dominique Maiter
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marie de Barsy
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Audrey Loumaye
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Michel P Hermans
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Institut de Recherches Expérimentales et Cliniques (IREC), Pôle EDIN, UCLouvain, Université catholique de Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium,Center for Microbiology, VIB, Leuven, Belgium
| | - Giulio G Muccioli
- Louvain Drug Research Institute (LDRI), Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
120
|
Liu X, Lu S, Shao Y, Zhang D, Tu J, Chen J. Disorders of gut microbiota in children with Tetralogy of Fallot. Transl Pediatr 2022; 11:385-395. [PMID: 35378966 PMCID: PMC8976677 DOI: 10.21037/tp-22-33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/25/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Gut microbiota plays an important role in cardiovascular health and disease, including congenital heart disease (CHD). Tetralogy of Fallot (TOF) is the most common form of cyanotic CHD characterized by systemic chronic hypoxia and sustained pressure overload of the right ventricle. It is well-known that hypoxia and pressure overload can affect gut microbiota. However, the effects of TOF on the gut microbiota remain little understood. This study explored the profile of the gut microbiota in children with unrepaired TOF. METHODS A total of 12 pediatric patients diagnosed with TOF and 9 healthy age- and gender-matched children were enrolled in this study. Fecal samples were collected from every participant and subjected to 16S rDNA gene sequencing. The raw sequencing data were processed using the Quantitative Insights Into Microbial Ecology pipeline. RESULTS A comparison of the gut microbiota revealed that pediatric patients with TOF had developed dysbiosis as reflected by the altered taxonomic composition and impaired functional profile. A total of 14 indicative bacterial genera were identified as differential biomarkers capable of distinguishing between healthy children and TOF patients. Furthermore, functional annotations revealed that the gut microbiota in TOF patients was characterized by increased levels of inflammatory, oxidative, and immune responses, and decreased activities of adaptation, synthesis, and metabolism. CONCLUSIONS Pediatric patients with unrepaired TOF have intestinal dysbacteriosis that is characterized by altered taxonomic composition and impaired functional profile. These findings suggested that the interplay between gut microbiota and the host may be dysregulated in patients with TOF.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Shaoyou Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yijia Shao
- Department of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Duo Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Jiazichao Tu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| |
Collapse
|
121
|
Houtman TA, Eckermann HA, Smidt H, de Weerth C. Gut microbiota and BMI throughout childhood: the role of firmicutes, bacteroidetes, and short-chain fatty acid producers. Sci Rep 2022; 12:3140. [PMID: 35210542 PMCID: PMC8873392 DOI: 10.1038/s41598-022-07176-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Childhood obesity is a risk factor for numerous health conditions. A critical factor in the etiology of obesity appears to be the gut microbiota, which is the microbial community that resides in the human gut. The ratio of the phyla Firmicutes and Bacteroidetes (F/B) and gut bacterial genera that produce short-chain fatty acids (SCFA) have been suggested to contribute to obesity. The current study investigated (1) whether differences in F/B ratio can be observed in infancy and childhood in relation to zBMI in healthy children, and (2) whether an innovative proxy measure adds evidence to a relationship between SCFA producers and the etiology of obesity. Stool samples were collected at five time points, and zBMI was assessed at eight time points throughout the first 12 years of life. Our confirmatory analyses with Bayesian multilevel models showed no relationship between the F/B ratio and zBMI. Also, a proxy measure constructed from known SCFA producers was unrelated to zBMI throughout the first 12 years of life. Exploratory analyses using multilevel and random forest models suggest that the relative abundances of Firmicutes and Bacteroidetes were independently negatively associated with zBMI from infancy through childhood, and the SCFA producing genera Subdoligranulum and Alistipes were negatively related to future BMI in childhood.
Collapse
Affiliation(s)
- Timothy A Houtman
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Henrik A Eckermann
- Donders Institute for Brain, Cognition and Behavior, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Carolina de Weerth
- Donders Institute for Brain, Cognition and Behavior, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
122
|
Gu X, Sim JX, Lee WL, Cui L, Chan YF, Chang ED, Teh YE, Zhang AN, Armas F, Chandra F, Chen H, Zhao S, Lee Z, Thompson JR, Ooi EE, Low JG, Alm EJ, Kalimuddin S. Gut Ruminococcaceae levels at baseline correlate with risk of antibiotic-associated diarrhea. iScience 2022; 25:103644. [PMID: 35005566 PMCID: PMC8718891 DOI: 10.1016/j.isci.2021.103644] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) affects a significant proportion of patients receiving antibiotics. We sought to understand if differences in the gut microbiome would influence the development of AAD. We administered a 3-day course of amoxicillin-clavulanate to 30 healthy adult volunteers, and analyzed their stool microbiome, using 16S rRNA gene sequencing, at baseline and up to 4 weeks post antibiotic administration. Lower levels of gut Ruminococcaceae were significantly and consistently observed from baseline until day 7 in participants who developed AAD. Overall, participants who developed AAD experienced a greater decrease in microbial diversity. The probability of AAD could be predicted based on qPCR-derived levels of Faecalibacterium prausnitzii at baseline. Our findings suggest that a lack of gut Ruminococcaceae influences development of AAD. Quantification of F. prausnitzii in stool prior to antibiotic administration may help identify patients at risk of AAD, and aid clinicians in devising individualized treatment regimens to minimize such adverse effects.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Jean X.Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Liang Cui
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yvonne F.Z. Chan
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Ega Danu Chang
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - An-Ni Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Zhanyi Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Janelle R. Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Asian School of the Environment, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, Singapore 117549, Singapore
| | - Jenny G. Low
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
| | - Eric J. Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Building E25-321, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
123
|
Vavreckova M, Galanova N, Kostovcik M, Krystynik O, Ivanovova E, Roubalova R, Jiraskova Zakostelska Z, Friedecky D, Friedecka J, Haluzik M, Karasek D, Kostovcikova K. Specific gut bacterial and fungal microbiota pattern in the first half of pregnancy is linked to the development of gestational diabetes mellitus in the cohort including obese women. Front Endocrinol (Lausanne) 2022; 13:970825. [PMID: 36133313 PMCID: PMC9484836 DOI: 10.3389/fendo.2022.970825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
AIMS Gestation is linked to changes in gut microbiota composition and function. Since gestational diabetes mellitus (GDM) can develop at any time of the pregnancy, we stratified the women into four groups according to the time and test used for the diagnosis. We focused on the gut microbiota pattern in early pregnancy to detect changes which could be linked to later GDM development. METHODS We collected stool samples from 104 pregnant women including obese individuals (first trimester body mass index median was 26.73). We divided the women into four groups according to routine screening of fasting plasma glucose (FPG) levels and oral glucose tolerance test (oGTT) in the first and third trimesters, respectively. We processed the stool samples for bacterial 16S rRNA and fungal ITS1 genes sequencing by Illumina MiSeq approach and correlated the gut microbiota composition with plasma short-chain fatty acid levels (SCFA). RESULTS We found that gut bacterial microbiota in the first trimester significantly differs among groups with different GDM onset based on unweighted UniFrac distances (p=0.003). Normoglycemic women had gut microbiota associated with higher abundance of family Prevotellaceae, and order Fusobacteriales, and genus Sutterella. Women diagnosed later during pregnancy either by FGP levels or by oGTT had higher abundances of genera Enterococcus, or Erysipelotrichaceae UCG-003, respectively. We observed significant enrichment of fungal genus Mucor in healthy pregnant women whereas Candida was more abundant in the group of pregnant women with impaired oGTT. Using correlation analysis, we found that Holdemanella negatively correlated with Blautia and Candida abundances and that Escherichia/Shigella abundance positively correlated and Subdoligranulum negatively correlated with plasma lipid levels. Coprococcus, Akkermansia, Methanobrevibacter, Phascolarctobacterium and Alistipes positively correlated with acetate, valerate, 2-hydroxybutyrate and 2-methylbutyrate levels, respectively, in women with GDM. CONCLUSIONS We conclude that there are significant differences in the gut microbiota composition between pregnant women with and without GDM already at the early stage of pregnancy in our cohort that included also overweight and obese individuals. Specific microbial pattern associated with GDM development during early pregnancy and its correlation to plasma lipid or SCFA levels could help to identify women in higher risk of GDM development.
Collapse
Affiliation(s)
- Marketa Vavreckova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Natalie Galanova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Kostovcik
- Laboratory of Fungal Genetics and Metabolism, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Ondrej Krystynik
- Third Department of Internal Medicine – Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc, Olomouc, Czechia
| | - Eliska Ivanovova
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc, and Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Jiraskova Zakostelska
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - David Friedecky
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc, and Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Jaroslava Friedecka
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital Olomouc, and Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - David Karasek
- Third Department of Internal Medicine – Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc, Olomouc, Czechia
| | - Klara Kostovcikova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
- *Correspondence: Klara Kostovcikova,
| |
Collapse
|
124
|
Nabavi-Rad A, Sadeghi A, Asadzadeh Aghdaei H, Yadegar A, Smith SM, Zali MR. The double-edged sword of probiotic supplementation on gut microbiota structure in Helicobacter pylori management. Gut Microbes 2022; 14:2108655. [PMID: 35951774 PMCID: PMC9373750 DOI: 10.1080/19490976.2022.2108655] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/13/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
As Helicobacter pylori management has become more challenging and less efficient over the last decade, the interest in innovative interventions is growing by the day. Probiotic co-supplementation to antibiotic therapies is reported in several studies, presenting a moderate reduction in drug-related side effects and a promotion in positive treatment outcomes. However, the significance of gut microbiota involvement in the competence of probiotic co-supplementation is emphasized by a few researchers, indicating the alteration in the host gastrointestinal microbiota following probiotic and drug uptake. Due to the lack of long-term follow-up studies to determine the efficiency of probiotic intervention in H. pylori eradication, and the delicate interaction of the gut microbiota with the host wellness, this review aims to discuss the gut microbiota alteration by probiotic co-supplementation in H. pylori management to predict the comprehensive effectiveness of probiotic oral administration.Abbreviations: acyl-CoA- acyl-coenzyme A; AMP- antimicrobial peptide; AMPK- AMP-activated protein kinase; AP-1- activator protein 1; BA- bile acid; BAR- bile acid receptor; BCAA- branched-chain amino acid; C2- acetate; C3- propionate; C4- butyrate; C5- valeric acid; CagA- Cytotoxin-associated gene A; cAMP- cyclic adenosine monophosphate; CD- Crohn's disease; CDI- C. difficile infection; COX-2- cyclooxygenase-2; DC- dendritic cell; EMT- epithelial-mesenchymal transition; FMO- flavin monooxygenases; FXR- farnesoid X receptor; GPBAR1- G-protein-coupled bile acid receptor 1; GPR4- G protein-coupled receptor 4; H2O2- hydrogen peroxide; HCC- hepatocellular carcinoma; HSC- hepatic stellate cell; IBD- inflammatory bowel disease; IBS- irritable bowel syndrome; IFN-γ- interferon-gamma; IgA immunoglobulin A; IL- interleukin; iNOS- induced nitric oxide synthase; JAK1- janus kinase 1; JAM-A- junctional adhesion molecule A; LAB- lactic acid bacteria; LPS- lipopolysaccharide; MALT- mucosa-associated lymphoid tissue; MAMP- microbe-associated molecular pattern; MCP-1- monocyte chemoattractant protein-1; MDR- multiple drug resistance; mTOR- mammalian target of rapamycin; MUC- mucin; NAFLD- nonalcoholic fatty liver disease; NF-κB- nuclear factor kappa B; NK- natural killer; NLRP3- NLR family pyrin domain containing 3; NOC- N-nitroso compounds; NOD- nucleotide-binding oligomerization domain; PICRUSt- phylogenetic investigation of communities by reconstruction of unobserved states; PRR- pattern recognition receptor; RA- retinoic acid; RNS- reactive nitrogen species; ROS- reactive oxygen species; rRNA- ribosomal RNA; SCFA- short-chain fatty acids; SDR- single drug resistance; SIgA- secretory immunoglobulin A; STAT3- signal transducer and activator of transcription 3; T1D- type 1 diabetes; T2D- type 2 diabetes; Th17- T helper 17; TLR- toll-like receptor; TMAO- trimethylamine N-oxide; TML- trimethyllysine; TNF-α- tumor necrosis factor-alpha; Tr1- type 1 regulatory T cell; Treg- regulatory T cell; UC- ulcerative colitis; VacA- Vacuolating toxin A.
Collapse
Affiliation(s)
- Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sinéad Marian Smith
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
125
|
Doll JPK, Vázquez-Castellanos JF, Schaub AC, Schweinfurth N, Kettelhack C, Schneider E, Yamanbaeva G, Mählmann L, Brand S, Beglinger C, Borgwardt S, Raes J, Schmidt A, Lang UE. Fecal Microbiota Transplantation (FMT) as an Adjunctive Therapy for Depression-Case Report. Front Psychiatry 2022; 13:815422. [PMID: 35250668 PMCID: PMC8891755 DOI: 10.3389/fpsyt.2022.815422] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 12/20/2022] Open
Abstract
Depression is a debilitating disorder, and at least one third of patients do not respond to therapy. Associations between gut microbiota and depression have been observed in recent years, opening novel treatment avenues. Here, we present the first two patients with major depressive disorder ever treated with fecal microbiota transplantation as add-on therapy. Both improved their depressive symptoms 4 weeks after the transplantation. Effects lasted up to 8 weeks in one patient. Gastrointestinal symptoms, constipation in particular, were reflected in microbiome changes and improved in one patient. This report suggests further FMT studies in depression could be worth pursuing and adds to awareness as well as safety assurance, both crucial in determining the potential of FMT in depression treatment.
Collapse
Affiliation(s)
- Jessica P K Doll
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Jorge F Vázquez-Castellanos
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Leuven, Belgium
| | | | - Nina Schweinfurth
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Cedric Kettelhack
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Else Schneider
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | | | - Laura Mählmann
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Serge Brand
- Center for Affective, Stress- and Sleep Disorders (ZASS), Psychiatric Clinics (UPK), University of Basel, Basel, Switzerland.,Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Division of Sport Science and Psychosocial Health, Department of Sport, Exercise, and Health, University of Basel, Basel, Switzerland.,Department of Psychiatry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Stefan Borgwardt
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland.,Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Leuven, Belgium
| | - André Schmidt
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Undine E Lang
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| |
Collapse
|
126
|
Santarossa S, Sitarik AR, Johnson CC, Li J, Lynch SV, Ownby DR, Ramirez A, Yong GLM, Cassidy-Bushrow AE. Associations of physical activity with gut microbiota in pre-adolescent children. Phys Act Nutr 2021; 25:24-37. [PMID: 35152621 PMCID: PMC8843867 DOI: 10.20463/pan.2021.0023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To determine whether physical activity (PA), primarily the recommended 60 minutes of moderate-to-vigorous PA, is associated with gut bacterial microbiota in 10-year-old children. METHODS The Block Physical Activity Screener, which provides minutes/day PA variables, was used to determine whether the child met the PA recommendations. 16S rRNA sequencing was performed on stool samples from the children to profile the composition of their gut bacterial microbiota. Differences in alpha diversity metrics (richness, Pielou's evenness, and Faith's phylogenetic diversity) by PA were determined using linear regression, whereas beta diversity (unweighted and weighted UniFrac) relationships were assessed using PERMANOVA. Taxon relative abundance differentials were determined using DESeq2. RESULTS The analytic sample included 321 children with both PA and 16S rRNA sequencing data (mean age [SD] =10.2 [0.8] years; 54.2% male; 62.9% African American), where 189 (58.9%) met the PA recommendations. After adjusting for covariates, meeting the PA recommendations as well as minutes/day PA variables were not significantly associated with gut richness, evenness, or diversity (p ≥ 0.19). However, meeting the PA recommendations (weighted UniFrac R2 = 0.014, p = 0.001) was significantly associated with distinct gut bacterial composition. These compositional differences were partly characterized by increased abundance of Megamonas and Anaerovorax as well as specific Christensenellaceae_R-7_group taxa in children with higher PA. CONCLUSION Children who met the recommendations of PA had altered gut microbiota compositions. Whether this translates to a reduced risk of obesity or associated metabolic diseases is still unclear.
Collapse
Affiliation(s)
- Sara Santarossa
- Department of Public Health Sciences, Henry Ford Health System, Michigan, USA
| | | | | | - Jia Li
- Department of Public Health Sciences, Henry Ford Health System, Michigan, USA
| | - Susan V. Lynch
- Department of Medicine, University of California, California, USA
| | - Dennis R. Ownby
- Department of Pediatrics, Georgia Regents University, Georgia, USA
| | - Alex Ramirez
- Department of Public Health Sciences, Henry Ford Health System, Michigan, USA
- Wayne State University School of Medicine Detroit, Michigan, USA
| | | | | |
Collapse
|
127
|
Díaz-Perdigones CM, Muñoz-Garach A, Álvarez-Bermúdez MD, Moreno-Indias I, Tinahones FJ. Gut microbiota of patients with type 2 diabetes and gastrointestinal intolerance to metformin differs in composition and functionality from tolerant patients. Biomed Pharmacother 2021; 145:112448. [PMID: 34844104 DOI: 10.1016/j.biopha.2021.112448] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Metformin modifies the gut microbiome in type 2 diabetes and gastrointestinal tolerance to metformin could be mediated by the gut microbiome. METHODS We enrolled 35 patients with type 2 diabetes not receiving treatment with metformin due to suspected gastrointestinal intolerance. Metformin was reintroduced at 425 mg, increasing 425 mg every two weeks until reaching 1700 mg per day. According to the occurrence of metformin-related gastrointestinal symptoms, patients were classified into three groups: early intolerance, non-tolerant, and tolerant. Gut microbiota was profiled with 16 S rRNA. This sequencing aimed to determine the differences in the baseline gut microbiota in all groups and prospectively in the tolerant and non-tolerant groups. RESULTS The classification resulted in 15 early intolerant, 10 tolerant, and 10 non-tolerant subjects. Early tolerance was characterized by a higher abundance of Subdoligranulum; while Veillonella and Serratia were higher in the non-tolerant group. The tolerant group showed enrichment of Megamonas, Megamonas rupellensis, and Phascolarctobacterium spp; Ruminococcus gnavus was lower in the longitudinal analysis. At the end point Prevotellaceae, Prevotella stercorea, Megamonas funiformis, Bacteroides xylanisolvens, and Blautia producta had a higher relative abundance in the tolerant group compared to the non-tolerant group. Subdoligranulum, Ruminococcus torques_1, Phascolarctobacterium faecium, and Eubacterium were higher in the non-tolerant group. The PICRUSt analysis showed a lower activity of the amino acid biosynthesis pathways and a higher sugar degradation pathway in the intolerant groups. CONCLUSIONS Gut microbiota of subjects with gastrointestinal intolerance depicted taxonomic and functional differences compared to tolerant patients, and this changed differently after metformin administration.
Collapse
Affiliation(s)
- Cristina Ma Díaz-Perdigones
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain
| | - Araceli Muñoz-Garach
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain
| | - María Dolores Álvarez-Bermúdez
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain
| | - Isabel Moreno-Indias
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain.
| | - Francisco J Tinahones
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain.
| |
Collapse
|
128
|
Galié S, Papandreou C, Arcelin P, Garcia D, Palau-Galindo A, Gutiérrez-Tordera L, Folch À, Bulló M. Examining the Interaction of the Gut Microbiome with Host Metabolism and Cardiometabolic Health in Metabolic Syndrome. Nutrients 2021; 13:nu13124318. [PMID: 34959869 PMCID: PMC8706982 DOI: 10.3390/nu13124318] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/20/2021] [Accepted: 11/28/2021] [Indexed: 12/05/2022] Open
Abstract
(1) Background: The microbiota-host cross-talk has been previously investigated, while its role in health is not yet clear. This study aimed to unravel the network of microbial-host interactions and correlate it with cardiometabolic risk factors. (2) Methods: A total of 47 adults with overweight/obesity and metabolic syndrome from the METADIET study were included in this cross-sectional analysis. Microbiota composition (151 genera) was assessed by 16S rRNA sequencing, fecal (m = 203) and plasma (m = 373) metabolites were profiled. An unsupervised sparse generalized canonical correlation analysis was used to construct a network of microbiota-metabolite interactions. A multi-omics score was derived for each cluster of the network and associated with cardiometabolic risk factors. (3) Results: Five multi-omics clusters were identified. Thirty-one fecal metabolites formed these clusters and were correlated with plasma sphingomyelins, lysophospholipids and medium to long-chain acylcarnitines. Seven genera from Ruminococcaceae and a member from the Desulfovibrionaceae family were correlated with fecal and plasma metabolites. Positive correlations were found between the multi-omics scores from two clusters with cholesterol and triglycerides levels. (4) Conclusions: We identified a correlated network between specific microbial genera and fecal/plasma metabolites in an adult population with metabolic syndrome, suggesting an interplay between gut microbiota and host lipid metabolism on cardiometabolic health.
Collapse
Affiliation(s)
- Serena Galié
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University RoviraiVirgili (URV), 43201 Reus, Spain; (S.G.); (A.P.-G.); (L.G.-T.); (À.F.)
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
| | - Christopher Papandreou
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (C.P.); (M.B.); Tel.: +34-977-759388 (M.B.)
| | - Pierre Arcelin
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
- Atención Basica de Salut (ABS) Reus V. Centre d’Assistència Primària Marià Fortuny, SAGESSA, 43204 Reus, Spain
| | - David Garcia
- ABS Alt Camp Oest, Centre d’Atenció Primària, 43460 Alcover, Spain;
| | - Antoni Palau-Galindo
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University RoviraiVirgili (URV), 43201 Reus, Spain; (S.G.); (A.P.-G.); (L.G.-T.); (À.F.)
- Atención Basica de Salut (ABS) Reus V. Centre d’Assistència Primària Marià Fortuny, SAGESSA, 43204 Reus, Spain
| | - Laia Gutiérrez-Tordera
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University RoviraiVirgili (URV), 43201 Reus, Spain; (S.G.); (A.P.-G.); (L.G.-T.); (À.F.)
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
| | - Àlex Folch
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University RoviraiVirgili (URV), 43201 Reus, Spain; (S.G.); (A.P.-G.); (L.G.-T.); (À.F.)
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
| | - Mònica Bulló
- Department of Biochemistry and Biotechnology, Faculty of Medicine and Health Sciences, University RoviraiVirgili (URV), 43201 Reus, Spain; (S.G.); (A.P.-G.); (L.G.-T.); (À.F.)
- Institute of Health Pere Virgili—IISPV, University Hospital Sant Joan, 43202 Reus, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (C.P.); (M.B.); Tel.: +34-977-759388 (M.B.)
| |
Collapse
|
129
|
Alberdi A, Martin Bideguren G, Aizpurua O. Diversity and compositional changes in the gut microbiota of wild and captive vertebrates: a meta-analysis. Sci Rep 2021; 11:22660. [PMID: 34811423 PMCID: PMC8608908 DOI: 10.1038/s41598-021-02015-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
The gut microbiota is recognised as an essential asset for the normal functioning of animal biology. When wild animals are moved into captivity, the modified environmental pressures are expected to rewire the gut microbiota, yet whether this transition follows similar patterns across vertebrates is still unresolved due to the absence of systematic multi-species analyses. We performed a meta-analysis of gut microbiota profiles of 322 captive and 322 wild specimens from 24 vertebrate species. Our analyses yielded no overall pattern of diversity and compositional variation between wild and captive vertebrates, but a heterogeneous landscape of responses, which differed depending on the components of diversity considered. Captive populations showed enrichment patterns of human-associated microorganisms, and the minimal host phylogenetic signal suggests that changes between wild and captive populations are mainly driven by case-specific captivity conditions. Finally, we show that microbiota differences between wild and captive populations can impact evolutionary and ecological inferences that rely on hierarchical clustering-based comparative analyses of gut microbial communities across species.
Collapse
Affiliation(s)
- Antton Alberdi
- Center for Evolutionary Hologenomics, GLOBE Institute, University of Copenhagen, 1353, Copenhagen, Denmark.
| | - Garazi Martin Bideguren
- Center for Evolutionary Hologenomics, GLOBE Institute, University of Copenhagen, 1353, Copenhagen, Denmark
| | - Ostaizka Aizpurua
- Center for Evolutionary Hologenomics, GLOBE Institute, University of Copenhagen, 1353, Copenhagen, Denmark.
| |
Collapse
|
130
|
Hasani A, Ebrahimzadeh S, Hemmati F, Khabbaz A, Hasani A, Gholizadeh P. The role of Akkermansia muciniphila in obesity, diabetes and atherosclerosis. J Med Microbiol 2021; 70. [PMID: 34623232 DOI: 10.1099/jmm.0.001435] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alteration in the composition of the gut microbiota can lead to a number of chronic clinical diseases. Akkermansia muciniphila is an anaerobic bacteria constituting 3-5% of the gut microbial community in healthy adults. This bacterium is responsible for degenerating mucin in the gut; its scarcity leads to diverse clinical disorders. In this review, we focus on the role of A. muciniphila in diabetes, obesity and atherosclerosis, as well as the use of this bacterium as a next-generation probiotic. In regard to obesity and diabetes, human and animal trials have shown that A. muciniphila controls the essential regulatory system of glucose and energy metabolism. However, the underlying mechanisms by which A. muciniphila alleviates the complications of obesity, diabetes and atherosclerosis are unclear. At the same time, its abundance suggests improved metabolic disorders, such as metabolic endotoxemia, adiposity insulin resistance and glucose tolerance. The role of A. muciniphila is implicated in declining aortic lesions and atherosclerosis. Well-characterized virulence factors, antigens and cell wall extracts of A. muciniphila may act as effector molecules in these diseases. These molecules may provide novel mechanisms and strategies by which this bacterium could be used as a probiotic for the treatment of obesity, diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Alka Hasani
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Ebrahimzadeh
- Department of Food Science and Technology, Faculty of Agriculture, Urmia University, Urmia, Iran
| | - Fatemeh Hemmati
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aytak Khabbaz
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pourya Gholizadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
131
|
Niccolai E, Di Pilato V, Nannini G, Baldi S, Russo E, Zucchi E, Martinelli I, Menicatti M, Bartolucci G, Mandrioli J, Amedei A. The Gut Microbiota-Immunity Axis in ALS: A Role in Deciphering Disease Heterogeneity? Biomedicines 2021; 9:753. [PMID: 34209688 PMCID: PMC8301418 DOI: 10.3390/biomedicines9070753] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disorder with an unknown etiology and no effective treatment, and is characterized by large phenotypic heterogeneity, including variable sites, ages of symptom onset and rates of disease progression. Increasing data support the role of the microbiota-immunity axis in the pathogenesis of neurodegenerative diseases. In the present study, we compared the inflammatory and microbiota profile of ALS patients with different clinical characteristics, with healthy family caregivers. Measuring a panel of 30 inflammatory cytokines in serum and fecal samples, we observed a distinct cytokine profile both at the systemic and intestinal level in patients compared to controls and even in patients with different clinical phenotypes and progression rates. The 16S targeted metagenome analysis revealed slight differences in patients compared to controls as well as in patients with slow progression, marked by the reduction of butyrate-producing bacteria and a decrease of the Firmicutes/Bacteroidetes ratio in ALS. Finally, the short chain fatty acid analysis did not show a different distribution among the groups. If confirmed in a larger number of patients, the inflammatory cytokine profile and the microbial composition could be appropriate biomarker candidates for deciphering ALS heterogeneity.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy; (E.N.); (G.N.); (S.B.); (E.R.)
| | - Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy;
| | - Giulia Nannini
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy; (E.N.); (G.N.); (S.B.); (E.R.)
| | - Simone Baldi
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy; (E.N.); (G.N.); (S.B.); (E.R.)
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy; (E.N.); (G.N.); (S.B.); (E.R.)
| | - Elisabetta Zucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Ilaria Martinelli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria di Modena, 41125 Modena, Italy;
| | - Marta Menicatti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, 50139 Florence, Italy; (M.M.); (G.B.)
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, 50139 Florence, Italy; (M.M.); (G.B.)
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria di Modena, 41125 Modena, Italy;
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy; (E.N.); (G.N.); (S.B.); (E.R.)
| |
Collapse
|
132
|
Habitual Dietary Intake Affects the Altered Pattern of Gut Microbiome by Acarbose in Patients with Type 2 Diabetes. Nutrients 2021; 13:nu13062107. [PMID: 34205413 PMCID: PMC8235473 DOI: 10.3390/nu13062107] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
The aim of this research was to reveal the characteristics of gut microbiome altered by acarbose intervention in Japanese patients with type 2 diabetes (T2D) and its possible association with habitual dietary intake. Eighteen patients with T2D were administered acarbose for four weeks. The abundances of two major phyla, namely Actinobacteria and Bacteroidetes, were reciprocally changed accompanied by the acarbose intervention. There were also significant changes in the abundances of ten genera, including the greater abundance of Bifidobacterium, Eubacterium, and Lactobacillus and the lower abundance of Bacteroides in the group after the intervention than that before the intervention. Hierarchical clustering of habitual dietary intake was performed based on the pattern of changes in the gut microbiota and were classified into distinct three clusters. Cluster I consisted of sucrose, cluster II mainly included fat intake, and cluster III mainly included carbohydrate intake. Moreover, the amount of change in Faecalibacterium was positively correlated with the intake of rice, but negatively correlated with the intake of bread. The intake of potato was negatively correlated with the amount of change in Akkermansia and Subdoligranulum. Acarbose altered the composition of gut microbiome in Japanese patients with T2D, which might be linked to the habitual dietary intake.
Collapse
|
133
|
Gut Microbiota and Host Metabolism: From Proof of Concept to Therapeutic Intervention. Microorganisms 2021; 9:microorganisms9061302. [PMID: 34203876 PMCID: PMC8232674 DOI: 10.3390/microorganisms9061302] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/12/2021] [Accepted: 06/13/2021] [Indexed: 12/16/2022] Open
Abstract
The field of the gut microbiota is still a relatively young science area, yet many studies have already highlighted the translational potential of microbiome research in the context of human health and disease. However, like in many new fields, discoveries are occurring at a fast pace and have provided new hope for the development of novel clinical applications in many different medical conditions, not in the least in metabolic disorders. This rapid progress has left the field vulnerable to premature claims, misconceptions and criticism, both from within and outside the sector. Tackling these issues requires a broad collaborative effort within the research field and is only possible by acknowledging the difficulties and challenges that are faced and that are currently hindering clinical implementation. These issues include: the primarily descriptive nature of evidence, methodological concerns, disagreements in analysis techniques, lack of causality, and a rather limited molecular-based understanding of underlying mechanisms. In this review, we discuss various studies and models that helped identifying the microbiota as an attractive tool or target for developing various translational applications. We also discuss some of the limitations and try to clarify some common misconceptions that are still prevalent in the field.
Collapse
|
134
|
Affiliation(s)
- Patrice D Cani
- UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium.
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| |
Collapse
|