1
|
Chen Y, Ma H, Liang J, Sun C, Wang D, Chen K, Zhao J, Ji S, Ma C, Ye X, Cao J, Wang Y, Sun C. Hepatoprotective potential of four fruit extracts rich in different structural flavonoids against alcohol-induced liver injury via gut microbiota-liver axis. Food Chem 2024; 460:140460. [PMID: 39068798 DOI: 10.1016/j.foodchem.2024.140460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024]
Abstract
Alcoholic liver injury (ALI) accounts for a major share of the global burden of non-viral liver disease. In the absence of specialized medications, research on using fruit flavonoids as a treatment is gaining momentum. This study investigated the hepatoprotective effects of four fruits rich in structurally diverse flavonoids: ougan (Citrus reticulata cv. Suavissima, OG), mulberry (Morus alba L., MB), apple (Malus × domestica Borkh., AP), and turnjujube (Hovenia dulcis Thunnb., TJ). A total of one flavanone glycoside, three polymethoxyflavones, two anthocyanins, one flavonol glycoside, and one dihydroflavonol were identified through UPLC analysis. In an acute ethanol-induced ALI mouse model, C57BL/6J mice were supplemented with 200 mg/kg·BW/day of different fruit extracts for three weeks. Our results showed that the four extracts exhibited promising benefits in improving lipid metabolism disorders, iron overload, and oxidative stress. RT-PCR and Western blot tests suggested that the potential mechanism may partially be attributed to the activation of the NRF2-mediated antioxidant response and the inhibition of ferroptosis pathways. Furthermore, fruit extracts administration demonstrated a specific regulatory role in intestinal microecology, with increases in beneficial bacteria such as Dubosiella, Lactobacillus, and Bifidobacterium. Spearman correlation analysis revealed strong links between intestinal flora, lipid metabolism, and iron homeostasis, implying that the fruit extracts mitigated ALI via the gut microbiota-liver axis. In vitro experiments reaffirmed the activity against ethanol-induced oxidative damage and highlighted the positive effects of flavonoid components. These findings endorse the prospective application of OG, MB, AP, and TJ as dietary supplements or novel treatments for ALI.
Collapse
Affiliation(s)
- Yunyi Chen
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Hanbing Ma
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Jiaojiao Liang
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Cui Sun
- Hainan Institute of Zhejiang University, Sanya, Hainan, People's Republic of China
| | - Dengliang Wang
- Institute of Fruit Tree Research, Quzhou Academy of Agriculture and Forestry Science, Quzhou, China
| | - Kang Chen
- Liandu Agriculture and Rural Bureau, Lishui, China
| | - Jinmiao Zhao
- Liandu Agriculture and Rural Bureau, Lishui, China
| | - Shiyu Ji
- Lishui Agriculture and Rural Bureau, Zhejiang, China
| | - Chao Ma
- Zhejiang NongZhen Food Co., Ltd., Hangzhou, China
| | - Xianming Ye
- Zhejiang JiaNong Fruit & Vegetable Co., Ltd., Quzhou, China
| | - Jinping Cao
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Chongde Sun
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Liu J, An Y, Yang N, Xu Y, Wang G. Longitudinal associations of dietary fiber and its source with 48-week weight loss maintenance, cardiometabolic risk factors and glycemic status under metformin or acarbose treatment: a secondary analysis of the March randomized trial. Nutr Diabetes 2024; 14:81. [PMID: 39358341 PMCID: PMC11447090 DOI: 10.1038/s41387-024-00340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS To examine longitudinal and dose-d ependent associations between dietary fiber intake and various clinical outcomes over 48 weeks of pharmacological treatment in T2DM patients. METHODS In this secondary analysis, we used data from the MARCH trial, which was designed to compare the efficacy of acarbose or metformin monotherapy as the initial therapy in Chinese patients newly diagnosed with T2DM. Dietary data were obtained using a 24-h dietary recall method to evaluate the intakes of dietary fiber from different sources as well as the carbohydrate-to-fiber ratio. RESULTS A total of 551 newly-diagnosed patients with T2DM complete dietary records (286 in the acarbose group and 265 in the metformin group) were included. Higher intake of total fiber and whole grain fiber was positively associated with better β-cell function, insulin sensitivity and postprandial glycemic control under acarbose treatment. Higher intake of legume fiber was associated with better glycemic control under both acarbose and metformin treatment but with better weight loss only under metformin treatment. A high-carbohydrate-low-fiber diet was associated with worse glycemic control and lower HDL-C under acarbose treatment but with higher insulin sensitivity and better weight loss under metformin treatment. CONCLUSIONS The notable effects of various dietary fibers when combined with different oral glucose-lowering medications should be considered to maximize therapeutic benefit.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ning Yang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Pam P, Goudarzi MA, Ghotboddin Mohammadi S, Asbaghi O, Aghakhani L, Clark CCT, Hashempur MH, Haghighat N. The effects of kiwifruit consumption on anthropometric and cardiometabolic indices in adults: A systematic review and meta-analysis. Food Sci Nutr 2024; 12:7017-7032. [PMID: 39479621 PMCID: PMC11521643 DOI: 10.1002/fsn3.4385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 11/02/2024] Open
Abstract
The current systematic review and meta-analysis was conducted to evaluate the effects of kiwifruit intake on anthropometric indices and key cardiometabolic parameters. Related articles were found by searching PubMed, ISI Web of Science, and Scopus to detect relevant Randomized Clinical Trials (RCTs) and novel systematic reviews relating to kiwi consumption in adults, up to August 2023. The weighted mean difference (WMD) and 95% confidence intervals (CIs) were calculated using a random-effects model. Heterogeneity, sensitivity analysis, and publication bias were assessed and reported using standard methods. Six RCTs were included in the meta-analysis. Analyzing overall effect sizes demonstrated a significant reduction in low-density lipoprotein cholesterol (LDL) levels (WMD: -9.30 mg/dL; 95% CI: -17.56 to -1.04, p = .027), whereas no significant alterations of triglycerides (TG) (WMD: -12.91 mg/dL; 95% CI: -28.17 to 2.34, p = .097), total cholesterol (TC) (WMD: -7.66 mg/dL; 95% CI: -17.85 to 2.52, p = .141), high-density lipoprotein cholesterol (HDL) (WMD: 2.87 mg/dL; 95% CI: -0.36 to 6.11, p = .141), fasting blood glucose (FBG) (WMD: 1.06 mg/dL; 95% CI: -1.43 to 3.56, p = .404), C-reactive protein (CRP) (WMD: 0.15 mg/dL; 95% CI: -0.40, 0.70, p = .0598), body weight (BW) (WMD: 0.85 kg; 95% CI: -1.34 to 3.04, p = .448), body mass index (BMI) (WMD: 0.04 kg/m2; 95% CI: -0.75 to 0.83, p = .920), and waist circumference (WC) (WMD: 0.18 cm; 95% CI: -1.81 to 2.19, p = .855) were found. Our findings suggest that consuming kiwifruit does not have a significant impact on anthropometric indices and cardiometabolic factors, except for LDL-C levels.
Collapse
Affiliation(s)
- Pedram Pam
- Student Research Committee, Department of Clinical NutritionTabriz University of Medical SciencesTabrizIran
- Department of Clinical NutritionTabriz University of Medical SciencesTabrizIran
| | | | - Shirin Ghotboddin Mohammadi
- Department of Clinical Nutrition, School of Nutrition and Food SciencesIsfahan University of Medical SciencesIsfahanIran
| | - Omid Asbaghi
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
- Student Research CommitteeShahid Beheshti University of Medical SciencesTehranIran
| | - Ladan Aghakhani
- Laparoscopy Research CenterShiraz University of Medical SciencesShirazIran
| | | | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of MedicineShiraz University of Medical SciencesShirazIran
| | - Neda Haghighat
- Laparoscopy Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
4
|
Neufeld PM, Nettersheim RA, Matschke V, Vorgerd M, Stahlke S, Theiss C. Unraveling the gut-brain axis: the impact of steroid hormones and nutrition on Parkinson's disease. Neural Regen Res 2024; 19:2219-2228. [PMID: 38488556 PMCID: PMC11034592 DOI: 10.4103/1673-5374.391304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 04/24/2024] Open
Abstract
This comprehensive review explores the intricate relationship between nutrition, the gut microbiome, steroid hormones, and Parkinson's disease within the context of the gut-brain axis. The gut-brain axis plays a pivotal role in neurodegenerative diseases like Parkinson's disease, encompassing diverse components such as the gut microbiota, immune system, metabolism, and neural pathways. The gut microbiome, profoundly influenced by dietary factors, emerges as a key player. Nutrition during the first 1000 days of life shapes the gut microbiota composition, influencing immune responses and impacting both child development and adult health. High-fat, high-sugar diets can disrupt this delicate balance, contributing to inflammation and immune dysfunction. Exploring nutritional strategies, the Mediterranean diet's anti-inflammatory and antioxidant properties show promise in reducing Parkinson's disease risk. Microbiome-targeted dietary approaches and the ketogenic diet hold the potential in improving brain disorders. Beyond nutrition, emerging research uncovers potential interactions between steroid hormones, nutrition, and Parkinson's disease. Progesterone, with its anti-inflammatory properties and presence in the nervous system, offers a novel option for Parkinson's disease therapy. Its ability to enhance neuroprotection within the enteric nervous system presents exciting prospects. The review addresses the hypothesis that α-synuclein aggregates originate from the gut and may enter the brain via the vagus nerve. Gastrointestinal symptoms preceding motor symptoms support this hypothesis. Dysfunctional gut-brain signaling during gut dysbiosis contributes to inflammation and neurotransmitter imbalances, emphasizing the potential of microbiota-based interventions. In summary, this review uncovers the complex web of interactions between nutrition, the gut microbiome, steroid hormones, and Parkinson's disease within the gut-brain axis framework. Understanding these connections not only offers novel therapeutic insights but also illuminates the origins of neurodegenerative diseases such as Parkinson's disease.
Collapse
Affiliation(s)
- Paula Maria Neufeld
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Ralf A. Nettersheim
- Department of Visceral Surgery, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
5
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
6
|
Guraka A, Duff R, Waldron J, Tripathi G, Kermanizadeh A. Co-Culture of Gut Bacteria and Metabolite Extraction Using Fast Vacuum Filtration and Centrifugation. Methods Protoc 2024; 7:74. [PMID: 39311375 PMCID: PMC11417889 DOI: 10.3390/mps7050074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024] Open
Abstract
This protocol describes a robust method for the extraction of intra and extracellular metabolites of gut bacterial mono and co-cultures. In recent years, the co-culture techniques employed in the field of microbiology have demonstrated significant importance in regard to understanding cell-cell interactions, cross-feeding, and the metabolic interactions between different bacteria, fungi, and microbial consortia which enable the mimicking of complex co-habitant conditions. This protocol highlights a robust reproducible physiologically relevant culture and extraction protocol for the co-culture of gut bacterium. The novel extraction steps are conducted without using quenching and cell disruption through bead-cell methods, freeze-thaw cycles, and sonication, which tend to affect the physical and biochemical properties of intracellular metabolites and secretome. The extraction procedure of inoculated bacterial co-cultures and monocultures use fast vacuum filtration and centrifugation. The extraction methodology is fast, effective, and robust, requiring 4 h to complete.
Collapse
Affiliation(s)
- Asha Guraka
- College of Science and Engineering, University of Derby, Derby DE22 1GB, UK
| | - Richard Duff
- College of Science and Engineering, University of Derby, Derby DE22 1GB, UK
| | - Joe Waldron
- College of Science and Engineering, University of Derby, Derby DE22 1GB, UK
| | - Gyanendra Tripathi
- School of Science and Technology, Nottingham Trent University, Nottingham NG1 4BU, UK;
| | - Ali Kermanizadeh
- College of Science and Engineering, University of Derby, Derby DE22 1GB, UK
| |
Collapse
|
7
|
Hinojosa-Nogueira D, Subiri-Verdugo A, Díaz-Perdigones CM, Rodríguez-Muñoz A, Vilches-Pérez A, Mela V, Tinahones FJ, Moreno-Indias I. Precision or Personalized Nutrition: A Bibliometric Analysis. Nutrients 2024; 16:2922. [PMID: 39275239 PMCID: PMC11397555 DOI: 10.3390/nu16172922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Food systems face the challenge of maintaining adequate nutrition for all populations. Inter-individual responses to the same diet have made precision or personalized nutrition (PN) an emerging and relevant topic. The aim of this study is to analyze the evolution of the PN field, identifying the principal actors and topics, and providing a comprehensive overview. Therefore, a bibliometric analysis of the scientific research available through the Web of Science (WOS) database was performed, revealing 2148 relevant papers up to June 2024. VOSviewer and the WOS platform were employed for the processing and analysis, and included an evaluation of diverse data such as country, author or most frequent keywords, among others. The analysis revealed a period of exponential growth from 2015 to 2023, with the USA, Spain, and England as the top contributors. The field of "Nutrition and Dietetics" is particularly significant, comprising nearly 33% of the total publications. The most highly cited institutions are the universities of Tufts, College Dublin, and Navarra. The relationship between nutrition, genetics, and omics sciences, along with dietary intervention studies, has been a defining factor in the evolution of PN. In conclusion, PN represents a promising field of research with significant potential for further advancement and growth.
Collapse
Affiliation(s)
- Daniel Hinojosa-Nogueira
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Alba Subiri-Verdugo
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Cristina Mª Díaz-Perdigones
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Alba Rodríguez-Muñoz
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Alberto Vilches-Pérez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Virginia Mela
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J Tinahones
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Isabel Moreno-Indias
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Vliex LMM, Penders J, Nauta A, Zoetendal EG, Blaak EE. The individual response to antibiotics and diet - insights into gut microbial resilience and host metabolism. Nat Rev Endocrinol 2024; 20:387-398. [PMID: 38486011 DOI: 10.1038/s41574-024-00966-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 06/16/2024]
Abstract
Antibiotic use disrupts microbial composition and activity in humans, but whether this disruption in turn affects host metabolic health is unclear. Cohort studies show associations between antibiotic use and an increased risk of developing obesity and type 2 diabetes mellitus. Here, we review available clinical trials and show the disruptive effect of antibiotic use on the gut microbiome in humans, as well as its impact on bile acid metabolism and microbial metabolites such as short-chain fatty acids. Placebo-controlled human studies do not show a consistent effect of antibiotic use on body weight and insulin sensitivity at a population level, but rather an individual-specific or subgroup-specific response. This response to antibiotic use is affected by the resistance and resilience of the gut microbiome, factors that determine the extent of disruption and the speed of recovery afterwards. Nutritional strategies to improve the composition and functionality of the gut microbiome, as well as its recovery after antibiotic use (for instance, with prebiotics), require a personalized approach to increase their efficacy. Improved insights into key factors that influence the individual-specific response to antibiotics and dietary intervention may lead to better efficacy in reversing or preventing antibiotic-induced microbial dysbiosis as well as strategies for preventing cardiometabolic diseases.
Collapse
Affiliation(s)
- Lars M M Vliex
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
9
|
Hong Y, Shen H, Chen X, Li G. Gender differences in the association between dietary protein intake and constipation: findings from NHANES. Front Nutr 2024; 11:1393596. [PMID: 38962434 PMCID: PMC11220262 DOI: 10.3389/fnut.2024.1393596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024] Open
Abstract
Purpose Dietary factors play a crucial role in the development and management of chronic constipation, yet the relationship between dietary protein intake and constipation remains underexplored. This study aims to investigate the association between dietary protein intake and the prevalence of constipation among American adults, with a focus on potential gender differences, using large-scale national data. Materials and methods Data from 14,048 participants aged 20 and above (7,072 men and 6,976 women) from the National Health and Nutrition Examination Survey (NHANES) 2005-2010 were analyzed. The Bristol Stool Form Scale's types 1 (separate hard lumps, resembling nuts) and 2 (sausage-shaped, but lumpy) were used to define constipation. A 24-h dietary recall technique was used to measure dietary protein intake. After controlling for covariates, the association between protein consumption and constipation risk was examined using multivariable logistic regression, smooth curve fitting, and testing for gender interaction effects. We then further determined the threshold effect between dietary protein intake and constipation risk. Results Constipation was present in 7.49% of people overall, with a higher proportion among women (10.19%) than among males (4.82%). In men, higher protein intake was significantly associated with a lower rate of constipation. However, in women, higher protein intake correlated with an increased risk of constipation, and the interaction between gender was significant (P for interaction = 0.0298). These results were corroborated by smooth curve fits, which also demonstrated a dose-response effect. Further threshold effect analysis showed that the turning points of dietary protein intake differed between male and female participants (119.42 gm/day for men; 40.79 gm/day for women). Conclusion The association between dietary protein intake and constipation was different in different genders with threshold effect. For men, moderately increasing protein intake could be beneficial, while for women, exceeding a certain level may increase the risk of constipation. These insights are crucial for guiding dietary protein recommendations for different genders and have significant clinical implications.
Collapse
Affiliation(s)
- Yongping Hong
- Department of Anorectal Surgery, The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Hongchen Shen
- The Second Department of Medicine, Renji College of Wenzhou Medical University, Wenzhou, China
| | - Xingxing Chen
- Department of Clinical Research, The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Guofeng Li
- Department of Anorectal Surgery, The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| |
Collapse
|
10
|
Lou X, Li P, Luo X, Lei Z, Liu X, Liu Y, Gao L, Xu W, Liu X. Dietary patterns interfere with gut microbiota to combat obesity. Front Nutr 2024; 11:1387394. [PMID: 38953044 PMCID: PMC11215203 DOI: 10.3389/fnut.2024.1387394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/07/2024] [Indexed: 07/03/2024] Open
Abstract
Obesity and obesity-related metabolic disorders are global epidemics that occur when there is chronic energy intake exceeding energy expenditure. Growing evidence suggests that healthy dietary patterns not only decrease the risk of obesity but also influence the composition and function of the gut microbiota. Numerous studies manifest that the development of obesity is associated with gut microbiota. One promising supplementation strategy is modulating gut microbiota composition by dietary patterns to combat obesity. In this review, we discuss the changes of gut microbiota in obesity and obesity-related metabolic disorders, with a particular emphasis on the impact of dietary components on gut microbiota and how common food patterns can intervene in gut microbiota to prevent obesity. While there is promise in intervening with the gut microbiota to combat obesity through the regulation of dietary patterns, numerous key questions remain unanswered. In this review, we critically review the associations between dietary patterns, gut microbes, and obesity, aiming to contribute to the further development and application of dietary patterns against obesity in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaomeng Liu
- Nutrition and Food Hygiene Laboratory, School of Public Health, Xinxiang Medical College, Xinxiang, China
| |
Collapse
|
11
|
Yadegar A, Bar-Yoseph H, Monaghan TM, Pakpour S, Severino A, Kuijper EJ, Smits WK, Terveer EM, Neupane S, Nabavi-Rad A, Sadeghi J, Cammarota G, Ianiro G, Nap-Hill E, Leung D, Wong K, Kao D. Fecal microbiota transplantation: current challenges and future landscapes. Clin Microbiol Rev 2024; 37:e0006022. [PMID: 38717124 PMCID: PMC11325845 DOI: 10.1128/cmr.00060-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
SUMMARYGiven the importance of gut microbial homeostasis in maintaining health, there has been considerable interest in developing innovative therapeutic strategies for restoring gut microbiota. One such approach, fecal microbiota transplantation (FMT), is the main "whole gut microbiome replacement" strategy and has been integrated into clinical practice guidelines for treating recurrent Clostridioides difficile infection (rCDI). Furthermore, the potential application of FMT in other indications such as inflammatory bowel disease (IBD), metabolic syndrome, and solid tumor malignancies is an area of intense interest and active research. However, the complex and variable nature of FMT makes it challenging to address its precise functionality and to assess clinical efficacy and safety in different disease contexts. In this review, we outline clinical applications, efficacy, durability, and safety of FMT and provide a comprehensive assessment of its procedural and administration aspects. The clinical applications of FMT in children and cancer immunotherapy are also described. We focus on data from human studies in IBD in contrast with rCDI to delineate the putative mechanisms of this treatment in IBD as a model, including colonization resistance and functional restoration through bacterial engraftment, modulating effects of virome/phageome, gut metabolome and host interactions, and immunoregulatory actions of FMT. Furthermore, we comprehensively review omics technologies, metagenomic approaches, and bioinformatics pipelines to characterize complex microbial communities and discuss their limitations. FMT regulatory challenges, ethical considerations, and pharmacomicrobiomics are also highlighted to shed light on future development of tailored microbiome-based therapeutics.
Collapse
Affiliation(s)
- Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tanya Marie Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sepideh Pakpour
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Andrea Severino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ed J Kuijper
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Wiep Klaas Smits
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Sukanya Neupane
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Sadeghi
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Estello Nap-Hill
- Department of Medicine, Division of Gastroenterology, St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dickson Leung
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Wong
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Jin DX, Jia CY, Yang B, Wu YH, Chen L, Liu R, Wu MG, Yu H, Ge QF. The ameliorative mechanism of Lactiplantibacillus plantarum NJAU-01 against D-galactose induced oxidative stress: a hepatic proteomics and gut microbiota analysis. Food Funct 2024; 15:6174-6188. [PMID: 38770619 DOI: 10.1039/d4fo00406j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Probiotic intervention is an effective strategy to alleviate oxidative stress-related diseases. Our previous studies found that Lactiplantibacillus plantarum NJAU-01 (NJAU-01) exhibited antioxidant effects in a D-galactose (D-gal)-induced aging mouse model. However, the underlying mechanism remains to be unveiled. This study was aimed to investigate the ameliorative effect and mechanism of NJAU-01 against oxidative stress induced by D-gal. The results showed that NJAU-01 could reverse the tendency of a slow body weight gain induced by D-gal. NJAU-01 relieved hepatic oxidative stress via increasing the hepatic total antioxidant capacity and antioxidant enzyme activities including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT). Moreover, the malondialdehyde (MDA) level was reversed after NJAU-01 supplementation. The proteomic results showed that there were 201 differentially expressed proteins (DEPs) between NJAU-01 and D-gal groups. NJAU-01 regulated the expressions of glutathione S-transferase Mu 5 (Gstm5), glutathione S-transferase P2 (Gstp2) and NADH dehydrogenase 1α subcomplex subunit 7 (Ndufa7) related to oxidative stress, and autophagy protein 5 (Atg5) and plasma alpha-L-fucosidase (Fuca2) involved in autophagy, etc. 16S rDNA sequencing results showed that NJAU-01 supplementation could regulate the gut microbiota dysbiosis induced by D-gal via increasing the relative abundances of the phylum Firmicutes and the genus Lactobacillus and reducing the relative abundances of the phylum Bacteroidetes and the genera Lachnospiraceae_NK4A136_group as well as Prevotellaceae_UCG-001, etc.. Spearman correlation analysis results showed that the altered gut microbiota composition had a significant correlation with antioxidant enzyme activities and the DEPs related to oxidative stress. Overall, NJAU-01 alleviated hepatic oxidative stress induced by D-gal via manipulating the gut microbiota composition and hepatic protein expression profile.
Collapse
Affiliation(s)
- Du-Xin Jin
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Chao-Yang Jia
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Bo Yang
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Yue-Hao Wu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Lei Chen
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Rui Liu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Man-Gang Wu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Hai Yu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| | - Qing-Feng Ge
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, P. R. China.
| |
Collapse
|
13
|
Adhikary K, Sarkar R, Maity S, Banerjee I, Chatterjee P, Bhattacharya K, Ahuja D, Sinha NK, Maiti R. The underlying causes, treatment options of gut microbiota and food habits in type 2 diabetes mellitus: a narrative review. J Basic Clin Physiol Pharmacol 2024; 35:153-168. [PMID: 38748886 DOI: 10.1515/jbcpp-2024-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/01/2024] [Indexed: 07/05/2024]
Abstract
Type 2 diabetes mellitus is a long-lasting endocrine disorder characterized by persistent hyperglycaemia, which is often triggered by an entire or relative inadequacy of insulin production or insulin resistance. As a result of resistance to insulin (IR) and an overall lack of insulin in the body, type 2 diabetes mellitus (T2DM) is a metabolic illness that is characterized by hyperglycaemia. Notably, the occurrence of vascular complications of diabetes and the advancement of IR in T2DM are accompanied by dysbiosis of the gut microbiota. Due to the difficulties in managing the disease and the dangers of multiple accompanying complications, diabetes is a chronic, progressive immune-mediated condition that plays a significant clinical and health burden on patients. The frequency and incidence of diabetes among young people have been rising worldwide. The relationship between the gut microbiota composition and the physio-pathological characteristics of T2DM proposes a novel way to monitor the condition and enhance the effectiveness of therapies. Our knowledge of the microbiota of the gut and how it affects health and illness has changed over the last 20 years. Species of the genus Eubacterium, which make up a significant portion of the core animal gut microbiome, are some of the recently discovered 'generation' of possibly helpful bacteria. In this article, we have focused on pathogenesis and therapeutic approaches towards T2DM, with a special reference to gut bacteria from ancient times to the present day.
Collapse
Affiliation(s)
- Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology & Management, Bhubaneswar, Odisha, India
| | - Riya Sarkar
- Department of Medical Laboratory Technology, 231513 Dr. B. C. Roy Academy of Professional Courses , Durgapur, West Bengal, India
| | - Sriparna Maity
- Department of Medical Laboratory Technology, 231513 Dr. B. C. Roy Academy of Professional Courses , Durgapur, West Bengal, India
| | - Ipsita Banerjee
- Department of Nutrition, Paramedical College Durgapur, Durgapur, West Bengal, India
| | - Prity Chatterjee
- Department of Biotechnology, Paramedical College Durgapur, Durgapur, West Bengal, India
| | - Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology & Management, Bhubaneswar, Odisha, India
| | - Deepika Ahuja
- School of Paramedics and Allied Health Sciences, Centurion University of Technology & Management, Bhubaneswar, Odisha, India
| | - Nirmalya Kumar Sinha
- Department of Nutrition and Department of NSS, Raja Narendra Lal Khan Women's College (Autonomous), Midnapore, West Bengal, India
| | - Rajkumar Maiti
- Department of Physiology, 326624 Bankura Christian College , Bankura, West Bengal, India
| |
Collapse
|
14
|
Reis SK, Socca EAR, de Souza BR, Genaro SC, Durán N, Fávaro WJ. Effects of probiotic supplementation on chronic inflammatory process modulation in colorectal carcinogenesis. Tissue Cell 2024; 87:102293. [PMID: 38244400 DOI: 10.1016/j.tice.2023.102293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
The current study investigated the potential effects of probiotic supplementation on colorectal carcinogenesis chemically induced with 1,2-dimethylhydrazine (DMH) and treated with 5-fluorouracil (5FU)-based chemotherapy in mice. Animals were randomly allocated in five different groups: Control: which not receive any treatment throughout the experimental course; Colitis model group (DMH): treated with DMH; DMH+ 5FU: animals received I.P. (intraperitoneal) dose of chemotherapy on a weekly basis; DMH+PROB: animals received daily administrations (via gavage) of probiotics (Lactobacillus: acidophilus and paracasei, Bifidobacterium lactis and bifidum); and DMH+ PROB+ 5FU: animals received the same treatment as the previous groups. After ten-week treatment, mice's large intestine was collected and subjected to colon length, histopathological, periodic acid-schiff (PAS) staining and immunohistochemistry (TLR2, MyD88, NF-κB, IL-6, TLR4, TRIF, IRF-3, IFN-γ, Ki-67, KRAS, p53, IL-10, and TGF-β) analyzes. Variance (ANOVA) and Kruskal-Wallis tests were used for statistical analysis, at significance level p 0.05. Probiotics' supplementation has increased the production of Ki-67 cell-proliferation marker, reduced body weight, and colon shortening, as well as modulated the chronic inflammatory process in colorectal carcinogenesis by inhibiting NF-κB expression and mitigating mucin depletion. Thus, these findings lay a basis for guide future studies focused on probiotics' action mechanisms in tumor microenvironment which might have implications in clinical practice.
Collapse
Affiliation(s)
- Sabrina Karen Reis
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Eduardo Augusto Rabelo Socca
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Bianca Ribeiro de Souza
- British Columbia's Gynecological Cancer Research (OVCARE) Program and Department of Obstetrics and Gynecology, University of British Columbia, Vancouver General Hospital, Vancouver, BC, Canada.
| | | | - Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Nanomedicine Research Unit (Nanomed), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Wagner José Fávaro
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
15
|
Sánchez-Moya T, López-Nicolás R, Peso-Echarri P, González-Bermúdez CA, Frontela-Saseta C. Effect of pine bark extract and its phenolic compounds on selected pathogenic and probiotic bacterial strains. Front Nutr 2024; 11:1381125. [PMID: 38600993 PMCID: PMC11004382 DOI: 10.3389/fnut.2024.1381125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction Inflammatory bowel disease (IBD) comprises a heterogeneous group of chronic diseases as ulcerative colitis (UC) and Crohn's disease (CD). IBD is the result of a dysregulation of intestinal homeostasis with a host's loss of tolerance toward normal enteric microflora. Plant-based extracts as phenolic compounds can play a role by modulating the intestinal inflammation response. Methods The in vitro antimicrobial activity of French maritime pine bark extract (PBE) and its phenolic constituents has been investigated in this study. Furthermore, the ability of PBE and phenolic compounds (caffeic acid, chlorogenic acid, ferulic acid, gallic acid and taxifolin) to modulate the microbiota has been assessed. Results Phenolic compounds and PBE showed a great inhibitory effect on the pathogens growth at the highest concentration assessed (1.25 mg/mL). The growth of E. sakazakii and E. faecalis were affected by the effect of caffeic acid and ferulic acid. Taxifolin showed a very strong activity against Listeria sp. (with a reduction ~98%). Gallic acid revealed antibacterial effect on S. aureus at different concentrations. The inhibitory effect of PBE was highly significant on the growth of E. coli O157:H7. PBE, caffeic acid and chlorogenic acid seem to provide the greatest beneficial effect on the probiotic bacteria. However, the highest concentrations of taxifolin may have impaired the growth of beneficial microbiota. Conclusion Present findings could be of interest for considering PBE and/or its phenolic constituents as protectors against gastrointestinal disturbances which lead to ulcerative colitis and Crohn's disease.
Collapse
Affiliation(s)
| | - Rubén López-Nicolás
- Department of Food Science and Nutrition, Faculty of Veterinary Sciences, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | | | | | | |
Collapse
|
16
|
Cani PD, Van Hul M. Gut microbiota in overweight and obesity: crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol 2024; 21:164-183. [PMID: 38066102 DOI: 10.1038/s41575-023-00867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 03/02/2024]
Abstract
Overweight and obesity are characterized by excessive fat mass accumulation produced when energy intake exceeds energy expenditure. One plausible way to control energy expenditure is to modulate thermogenic pathways in white adipose tissue (WAT) and/or brown adipose tissue (BAT). Among the different environmental factors capable of influencing host metabolism and energy balance, the gut microbiota is now considered a key player. Following pioneering studies showing that mice lacking gut microbes (that is, germ-free mice) or depleted of their gut microbiota (that is, using antibiotics) developed less adipose tissue, numerous studies have investigated the complex interactions existing between gut bacteria, some of their membrane components (that is, lipopolysaccharides), and their metabolites (that is, short-chain fatty acids, endocannabinoids, bile acids, aryl hydrocarbon receptor ligands and tryptophan derivatives) as well as their contribution to the browning and/or beiging of WAT and changes in BAT activity. In this Review, we discuss the general physiology of both WAT and BAT. Subsequently, we introduce how gut bacteria and different microbiota-derived metabolites, their receptors and signalling pathways can regulate the development of adipose tissue and its metabolic capacities. Finally, we describe the key challenges in moving from bench to bedside by presenting specific key examples.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
17
|
Mansour S, Alkhaaldi SMI, Sammanasunathan AF, Ibrahim S, Farhat J, Al-Omari B. Precision Nutrition Unveiled: Gene-Nutrient Interactions, Microbiota Dynamics, and Lifestyle Factors in Obesity Management. Nutrients 2024; 16:581. [PMID: 38474710 DOI: 10.3390/nu16050581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Obesity is a complex metabolic disorder that is associated with several diseases. Recently, precision nutrition (PN) has emerged as a tailored approach to provide individualised dietary recommendations. AIM This review discusses the major intrinsic and extrinsic components considered when applying PN during the management of obesity and common associated chronic conditions. RESULTS The review identified three main PN components: gene-nutrient interactions, intestinal microbiota, and lifestyle factors. Genetic makeup significantly contributes to inter-individual variations in dietary behaviours, with advanced genome sequencing and population genetics aiding in detecting gene variants associated with obesity. Additionally, PN-based host-microbiota evaluation emerges as an advanced therapeutic tool, impacting disease control and prevention. The gut microbiome's composition regulates diverse responses to nutritional recommendations. Several studies highlight PN's effectiveness in improving diet quality and enhancing adherence to physical activity among obese patients. PN is a key strategy for addressing obesity-related risk factors, encompassing dietary patterns, body weight, fat, blood lipids, glucose levels, and insulin resistance. CONCLUSION PN stands out as a feasible tool for effectively managing obesity, considering its ability to integrate genetic and lifestyle factors. The application of PN-based approaches not only improves current obesity conditions but also holds promise for preventing obesity and its associated complications in the long term.
Collapse
Affiliation(s)
- Samy Mansour
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Saif M I Alkhaaldi
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Ashwin F Sammanasunathan
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Saleh Ibrahim
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Joviana Farhat
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Basem Al-Omari
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
18
|
Zhang YW, Song PR, Wang SC, Liu H, Shi ZM, Su JC. Diets intervene osteoporosis via gut-bone axis. Gut Microbes 2024; 16:2295432. [PMID: 38174650 PMCID: PMC10773645 DOI: 10.1080/19490976.2023.2295432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Osteoporosis is a systemic skeletal disease that seriously endangers the health of middle-aged and older adults. Recently, with the continuous deepening of research, an increasing number of studies have revealed gut microbiota as a potential target for osteoporosis, and the research concept of the gut-bone axis has gradually emerged. Additionally, the intake of dietary nutrients and the adoption of dietary patterns may affect the gut microbiota, and alterations in the gut microbiota might also influence the metabolic status of the host, thus adjusting bone metabolism. Based on the gut-bone axis, dietary intake can also participate in the modulation of bone metabolism by altering abundance, diversity, and composition of gut microbiota. Herein, combined with emerging literatures and relevant studies, this review is aimed to summarize the impacts of different dietary components and patterns on osteoporosis by acting on gut microbiota, as well as underlying mechanisms and proper dietary recommendations.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Pei-Ran Song
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Zhong-Min Shi
- Department of Orthopaedics, Sixth People’s Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jia-Can Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
19
|
Mokhtari P, Holzhausen EA, Chalifour BN, Schmidt KA, Babaei M, Machle CJ, Adise S, Alderete TL, Goran MI. Associations between Dietary Sugar and Fiber with Infant Gut Microbiome Colonization at 6 Mo of Age. J Nutr 2024; 154:152-162. [PMID: 37717629 PMCID: PMC10808822 DOI: 10.1016/j.tjnut.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND The taxonomic composition of the gut microbiome undergoes rapid development during the first 2-3 y of life. Poor diet during complementary feeding has been associated with alterations in infant growth and compromised bone, immune system, and neurodevelopment, but how it may affect gut microbial composition is unknown. OBJECTIVES This cross-sectional study aimed to examine the associations between early-life nutrition and the developing infant gut microbiota at 6 mo of age. METHODS Latino mother-infant pairs from the Mother's Milk Study (n = 105) were included. Infant gut microbiota and dietary intake were analyzed at 6 mo of age using 16S ribosomal RNA amplicon sequencing and 24-h dietary recalls, respectively. Poisson generalized linear regression analysis was performed to examine associations between dietary nutrients and microbial community abundance while adjusting for infants' mode of delivery, antibiotics, infant feeding type, time of introduction of solid foods, energy intake, and body weight. A P value of <0.05 was used to determine the statistical significance in the study. RESULTS Infants with higher consumption of total sugar exhibited a lower relative abundance of the genera Bacteroides (β = -0.01; 95% CI: -0.02, -0.00; P = 0.03) and genus Clostridium belonging to the Lachnospiraceae family (β = -0.02; 95% CI: -0.03, -0.00; P = 0.01). In addition, a higher intake of free sugar (which excludes sugar from milk, dairy, and whole fruit) was associated with several bacteria at the genus level, including Parabacteroides genus (β = 0.03; 95% CI: 0.01, 0.05; P = 0.001). Total insoluble fiber intake was associated with favorable bacteria at the genus level such as Faecalibacterium (β = 0.28; 95% CI: 0.03, 0.52; P = 0.02) and Coprococcus (β = 0.28; 95% CI: 0.02, 0.52; P = 0.03). CONCLUSION These findings demonstrate that early-life dietary intake at 6 mo impacts the developing gut microbiome associated with the presence of both unfavorable gut microbes and dietary fiber-associated commensal microbes.
Collapse
Affiliation(s)
- Pari Mokhtari
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Elizabeth A Holzhausen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Bridget N Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Kelsey A Schmidt
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Mahsa Babaei
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Christopher J Machle
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Shana Adise
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Michael I Goran
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
20
|
Jardon KM, Goossens GH, Most J, Galazzo G, Venema K, Penders J, Blaak EE. Examination of sex-specific interactions between gut microbiota and host metabolism after 12-week combined polyphenol supplementation in individuals with overweight or obesity. Gut Microbes 2024; 16:2392875. [PMID: 39182247 PMCID: PMC11346568 DOI: 10.1080/19490976.2024.2392875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/12/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Polyphenols exert beneficial effects on host metabolism, which may be mediated by the gut microbiota. We investigated sex-specific differences in microbiota composition and interactions with cardiometabolic parameters after polyphenol supplementation in individuals with overweight/obesity. In a double-blind, randomized, placebo-controlled trial, 19 women and 18 men with normal glucose tolerance and body mass index >25 kg/m2 received epigallocatechin-3-gallate and resveratrol (EGCG+RES, 282 + 80 mg/d) or placebo supplements for 12 weeks. Fecal microbiota composition (16S rRNA gene amplicon sequencing, V3-V4 region), in vivo whole-body fat oxidation (indirect calorimetry), and mitochondrial respiration in permeabilized skeletal muscle fibers (SkM-Ox; ex vivo respirometry) were determined pre- and post-intervention. Overall, EGCG+RES supplementation did not affect gut microbiota composition. Akkermansia, Ruminococcaceae UCG-002, Subdoligranulum, and Lachnospiraceae UCG-004 were more abundant, while Veillonella, Tyzzerella 4, Clostridium innocuum group, Ruminococcus gnavus group, Escherichia-Shigella, and an uncultured Ruminococcaceae family genus were less abundant in women compared to men. In women, only baseline Eubacterium ventriosum group abundance correlated with EGCG+RES-induced changes in SkM-Ox. In men, low Dorea, Barnsiella, Anaerotruncus, Ruminococcus, Subdoligranulum, Coprococcus, Eubacterium ventriosum group, Ruminococcaceae UCG-003, and a Ruminococcaceae family genus abundance, and high Blautia abundance at baseline were associated with improvements in SkM-Ox. Changes in whole-body fat oxidation were not associated with gut microbiota features. We conclude that baseline microbiota composition predicts changes in SkM-Ox as a result of EGCG+RES supplementation in men but not in women. Men may be more prone to diet-induced, gut microbiota-related improvements in cardiometabolic health. These sex-differences should be further investigated in future precision-based intervention studies.
Collapse
Affiliation(s)
- Kelly M. Jardon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- TiFN, Wageningen, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jasper Most
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Orthopedics, Zuyderland Medical Center, Sittard-Geleen, The Netherlands
| | - Gianluca Galazzo
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- TiFN, Wageningen, The Netherlands
| |
Collapse
|
21
|
Behrens F, Bartolomaeus H, Wilck N, Holle J. Gut-immune axis and cardiovascular risk in chronic kidney disease. Clin Kidney J 2024; 17:sfad303. [PMID: 38229879 PMCID: PMC10790347 DOI: 10.1093/ckj/sfad303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Indexed: 01/18/2024] Open
Abstract
Patients with chronic kidney disease (CKD) suffer from marked cardiovascular morbidity and mortality, so lowering the cardiovascular risk is paramount to improve quality of life and survival in CKD. Manifold mechanisms are hold accountable for the development of cardiovascular disease (CVD), and recently inflammation arose as novel risk factor significantly contributing to progression of CVD. While the gut microbiome was identified as key regulator of immunity and inflammation in several disease, CKD-related microbiome-immune interaction gains increasing importance. Here, we summarize the latest knowledge on microbiome dysbiosis in CKD, subsequent changes in bacterial and host metabolism and how this drives inflammation and CVD in CKD. Moreover, we outline potential therapeutic targets along the gut-immune-cardiovascular axis that could aid the combat of CVD development and high mortality in CKD.
Collapse
Affiliation(s)
- Felix Behrens
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| |
Collapse
|
22
|
Duncanson K, Williams G, Hoedt EC, Collins CE, Keely S, Talley NJ. Diet-microbiota associations in gastrointestinal research: a systematic review. Gut Microbes 2024; 16:2350785. [PMID: 38725230 PMCID: PMC11093048 DOI: 10.1080/19490976.2024.2350785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Interactions between diet and gastrointestinal microbiota influence health status and outcomes. Evaluating these relationships requires accurate quantification of dietary variables relevant to microbial metabolism, however current dietary assessment methods focus on dietary components relevant to human digestion only. The aim of this study was to synthesize research on foods and nutrients that influence human gut microbiota and thereby identify knowledge gaps to inform dietary assessment advancements toward better understanding of diet-microbiota interactions. Thirty-eight systematic reviews and 106 primary studies reported on human diet-microbiota associations. Dietary factors altering colonic microbiota included dietary patterns, macronutrients, micronutrients, bioactive compounds, and food additives. Reported diet-microbiota associations were dominated by routinely analyzed nutrients, which are absorbed from the small intestine but analyzed for correlation to stool microbiota. Dietary derived microbiota-relevant nutrients are more challenging to quantify and underrepresented in included studies. This evidence synthesis highlights advancements needed, including opportunities for expansion of food composition databases to include microbiota-relevant data, particularly for human intervention studies. These advances in dietary assessment methodology will facilitate translation of microbiota-specific nutrition therapy to practice.
Collapse
Affiliation(s)
- Kerith Duncanson
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - Georgina Williams
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - Emily C. Hoedt
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - Clare E. Collins
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - Simon Keely
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| | - Nicholas J. Talley
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine & Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
23
|
Qazi AS, Rahman UU, Ahmad B, Safdar W, Ahmad S, Mumtaz S. Diet, Gut Microbes, and Cancer. Cancer Treat Res 2024; 191:163-190. [PMID: 39133408 DOI: 10.1007/978-3-031-55622-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Gut microbes are important and may play important role in spreading cancers specially the gastrointestinal malignancies preferably colorectal cancers. Gut microbes and diet can influence the tissues in gastrointestinal tract increasing the risk of cancer spread. Insufficient nutrient intake and imbalance diet can disturb the microbiome of gastrointestinal tract causing metabolism of xenobiotics which is beneficial as well as detrimental. Dietary imbalance may also weaken the immune system which is another reason for spreading and development of cancers. The triage of gut microbiome, host immune system, and dietary patterns may help the initiation of mechanism of carcinogenesis. In addition to its role in carcinogenesis and tumor development, there is still growing evidence as to how intestinal microflora influences the efficacy and toxicity of chemotherapy and immunotherapy by the gut microbiome. It can therefore be used as a biomarker to predict treatment response or poor response and can also be modified to improve cancer treatment.
Collapse
Affiliation(s)
- Asma Saleem Qazi
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan.
| | - Ubaid Ur Rahman
- Department of Microbiology, Quaid e Azam University, Islamabad, Pakistan
| | - Bilal Ahmad
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Waseem Safdar
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Saeed Ahmad
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Sara Mumtaz
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| |
Collapse
|
24
|
van Vorstenbosch R, van Munster K, Pachen D, Mommers A, Stavropoulos G, van Schooten FJ, Ponsioen C, Smolinska A. The Detection of Primary Sclerosing Cholangitis Using Volatile Metabolites in Fecal Headspace and Exhaled Breath. Metabolites 2023; 14:23. [PMID: 38248826 PMCID: PMC10819709 DOI: 10.3390/metabo14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Up to 5% of inflammatory bowel disease patients may at some point develop primary sclerosing cholangitis (PSC). PSC is a rare liver disease that ultimately results in liver damage, cirrhosis and liver failure. It typically remains subclinical until irreversible damage has been inflicted. Hence, it is crucial to screen IBD patients for PSC, but its early detection is challenging, and the disease's etiology is not well understood. This current study aimed at the early detection of PSC in an IBD population using Volatile Organic Compounds in fecal headspace and exhaled breath. To this aim, fecal material and exhaled breath were collected from 73 patients (n = 16 PSC/IBD; n = 8 PSC; n = 49 IBD), and their volatile profile were analyzed using Gas Chromatography-Mass Spectrometry. Using the most discriminatory features, PSC detection resulted in areas under the ROC curve (AUCs) of 0.83 and 0.84 based on fecal headspace and exhaled breath, respectively. Upon data fusion, the predictive performance increased to AUC 0.92. The observed features in the fecal headspace relate to detrimental microbial dysbiosis and exogenous exposure. Future research should aim for the early detection of PSC in a prospective study design.
Collapse
Affiliation(s)
- Robert van Vorstenbosch
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| | - Kim van Munster
- Department of Gastroenterology and Hepathology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (K.v.M.); (C.P.)
| | - Danielle Pachen
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| | - Alex Mommers
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| | - Georgios Stavropoulos
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| | - Frederik-Jan van Schooten
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| | - Cyriel Ponsioen
- Department of Gastroenterology and Hepathology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (K.v.M.); (C.P.)
| | - Agnieszka Smolinska
- Department of Toxicology, Nutrition and Toxicology Research Institute, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.P.); (A.M.); (F.-J.v.S.)
| |
Collapse
|
25
|
Vernocchi P, Marangelo C, Guerrera S, Del Chierico F, Guarrasi V, Gardini S, Conte F, Paci P, Ianiro G, Gasbarrini A, Vicari S, Putignani L. Gut microbiota functional profiling in autism spectrum disorders: bacterial VOCs and related metabolic pathways acting as disease biomarkers and predictors. Front Microbiol 2023; 14:1287350. [PMID: 38192296 PMCID: PMC10773764 DOI: 10.3389/fmicb.2023.1287350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental disorder. Major interplays between the gastrointestinal (GI) tract and the central nervous system (CNS) seem to be driven by gut microbiota (GM). Herein, we provide a GM functional characterization, based on GM metabolomics, mapping of bacterial biochemical pathways, and anamnestic, clinical, and nutritional patient metadata. Methods Fecal samples collected from children with ASD and neurotypical children were analyzed by gas-chromatography mass spectrometry coupled with solid phase microextraction (GC-MS/SPME) to determine volatile organic compounds (VOCs) associated with the metataxonomic approach by 16S rRNA gene sequencing. Multivariate and univariate statistical analyses assessed differential VOC profiles and relationships with ASD anamnestic and clinical features for biomarker discovery. Multiple web-based and machine learning (ML) models identified metabolic predictors of disease and network analyses correlated GM ecological and metabolic patterns. Results The GM core volatilome for all ASD patients was characterized by a high concentration of 1-pentanol, 1-butanol, phenyl ethyl alcohol; benzeneacetaldehyde, octadecanal, tetradecanal; methyl isobutyl ketone, 2-hexanone, acetone; acetic, propanoic, 3-methyl-butanoic and 2-methyl-propanoic acids; indole and skatole; and o-cymene. Patients were stratified based on age, GI symptoms, and ASD severity symptoms. Disease risk prediction allowed us to associate butanoic acid with subjects older than 5 years, indole with the absence of GI symptoms and low disease severity, propanoic acid with the ASD risk group, and p-cymene with ASD symptoms, all based on the predictive CBCL-EXT scale. The HistGradientBoostingClassifier model classified ASD patients vs. CTRLs by an accuracy of 89%, based on methyl isobutyl ketone, benzeneacetaldehyde, phenyl ethyl alcohol, ethanol, butanoic acid, octadecane, acetic acid, skatole, and tetradecanal features. LogisticRegression models corroborated methyl isobutyl ketone, benzeneacetaldehyde, phenyl ethyl alcohol, skatole, and acetic acid as ASD predictors. Conclusion Our results will aid the development of advanced clinical decision support systems (CDSSs), assisted by ML models, for advanced ASD-personalized medicine, based on omics data integrated into electronic health/medical records. Furthermore, new ASD screening strategies based on GM-related predictors could be used to improve ASD risk assessment by uncovering novel ASD onset and risk predictors.
Collapse
Affiliation(s)
- Pamela Vernocchi
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Chiara Marangelo
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Silvia Guerrera
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Federica Del Chierico
- Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | | | - Federica Conte
- Institute for Systems Analysis and Computer Science “Antonio Ruberti”, National Research Council, Rome, Italy
| | - Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Gianluca Ianiro
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Life Sciences and Public Health Department, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
26
|
Wang A, Guan B, Zhang H, Xu H. Danger-associated metabolites trigger metaflammation: A crowbar in cardiometabolic diseases. Pharmacol Res 2023; 198:106983. [PMID: 37931790 DOI: 10.1016/j.phrs.2023.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Cardiometabolic diseases (CMDs) are characterized by a series of metabolic disorders and chronic low-grade inflammation. CMDs contribute to a high burden of mortality and morbidity worldwide. Host-microbial metabolic regulation that triggers metaflammation is an emerging field of study that promotes a new perspective for perceiving cardiovascular risks. The term metaflammation denotes the entire cascade of immune responses activated by a new class of metabolites known as "danger-associated metabolites" (DAMs). It is being proposed by the present review for the first time. We summarize current studies covering bench to bedside aspects of DAMs to better understand CMDs in the context of DAMs. We have focused on the involvement of DAMs in the pathophysiological development of CMDs, including the disruption of immune homeostasis and chronic inflammation-triggered damage leading to CMD-related adverse events, as well as emerging therapeutic approaches for targeting DAM metabolism in CMDs.
Collapse
Affiliation(s)
- Anlu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China.
| |
Collapse
|
27
|
Zhang Y, Fang XM. The pan-liver network theory: From traditional chinese medicine to western medicine. CHINESE J PHYSIOL 2023; 66:401-436. [PMID: 38149555 DOI: 10.4103/cjop.cjop-d-22-00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
In traditional Chinese medicine (TCM), the liver is the "general organ" that is responsible for governing/maintaining the free flow of qi over the entire body and storing blood. According to the classic five elements theory, zang-xiang theory, yin-yang theory, meridians and collaterals theory, and the five-viscera correlation theory, the liver has essential relationships with many extrahepatic organs or tissues, such as the mother-child relationships between the liver and the heart, and the yin-yang and exterior-interior relationships between the liver and the gallbladder. The influences of the liver to the extrahepatic organs or tissues have been well-established when treating the extrahepatic diseases from the perspective of modulating the liver by using the ancient classic prescriptions of TCM and the acupuncture and moxibustion. In modern medicine, as the largest solid organ in the human body, the liver has the typical functions of filtration and storage of blood; metabolism of carbohydrates, fats, proteins, hormones, and foreign chemicals; formation of bile; storage of vitamins and iron; and formation of coagulation factors. The liver also has essential endocrine function, and acts as an immunological organ due to containing the resident immune cells. In the perspective of modern human anatomy, physiology, and pathophysiology, the liver has the organ interactions with the extrahepatic organs or tissues, for example, the gut, pancreas, adipose, skeletal muscle, heart, lung, kidney, brain, spleen, eyes, skin, bone, and sexual organs, through the circulation (including hemodynamics, redox signals, hepatokines, metabolites, and the translocation of microbiota or its products, such as endotoxins), the neural signals, or other forms of pathogenic factors, under normal or diseases status. The organ interactions centered on the liver not only influence the homeostasis of these indicated organs or tissues, but also contribute to the pathogenesis of cardiometabolic diseases (including obesity, type 2 diabetes mellitus, metabolic [dysfunction]-associated fatty liver diseases, and cardio-cerebrovascular diseases), pulmonary diseases, hyperuricemia and gout, chronic kidney disease, and male and female sexual dysfunction. Therefore, based on TCM and modern medicine, the liver has the bidirectional interaction with the extrahepatic organ or tissue, and this established bidirectional interaction system may further interact with another one or more extrahepatic organs/tissues, thus depicting a complex "pan-hepatic network" model. The pan-hepatic network acts as one of the essential mechanisms of homeostasis and the pathogenesis of diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Physiology; Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong; Issue 12th of Guangxi Apprenticeship Education of Traditional Chinese Medicine (Shi-Cheng Class of Guangxi University of Chinese Medicine), College of Continuing Education, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xian-Ming Fang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine (Guangxi Hospital of Integrated Chinese Medicine and Western Medicine, Ruikang Clinical Faculty of Guangxi University of Chinese Medicine), Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
28
|
West ML, Hart S, Loughman A, Jacka FN, Staudacher HM, Abbaspour A, Phillipou A, Ruusunen A, Rocks T. Challenges and priorities for researching the gut microbiota in individuals living with anorexia nervosa. Int J Eat Disord 2023; 56:2001-2011. [PMID: 37548294 DOI: 10.1002/eat.24033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVE The gut microbiota is implicated in several symptoms and biological pathways relevant to anorexia nervosa (AN). Investigations into the role of the gut microbiota in AN are growing, with a specific interest in the changes that occur in response to treatment. Findings suggest that microbial species may be associated with some of the symptoms common in AN, such as depression and gastrointestinal disturbances (GID). Therefore, researchers believe the gut microbiota may have therapeutic relevance. Whilst research in this field is rapidly expanding, the unique considerations relevant to conducting gut microbiota research in individuals with AN must be addressed. METHOD We provide an overview of the published literature investigating the relationship between the gut microbiota and symptoms and behaviors present in AN, discuss important challenges in gut microbiota research, and offer recommendations for addressing these. We conclude by summarizing research design priorities for the field to move forward. RESULTS Several ways exist to reduce participant burden and accommodate challenges when researching the gut microbiota in individuals with AN. DISCUSSION Recommendations from this article are foreseen to encourage scientific rigor and thoughtful protocol planning for microbiota research in AN, including ways to reduce participant burden. Employing such methods will contribute to a better understanding of the role of the gut microbiota in AN pathophysiology and treatment. PUBLIC SIGNIFICANCE The field of gut microbiota research is rapidly expanding, including the role of the gut microbiota in anorexia nervosa. Thoughtful planning of future research will ensure appropriate data collection for meaningful interpretation while providing a positive experience for the participant. We present current challenges, recommendations for research design and priorities to facilitate the advancement of research in this field.
Collapse
Affiliation(s)
- Madeline L West
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Susan Hart
- Eating and Nutrition Research Group, School of Medicine, Western Sydney University, Cambelltown, Australia
- Nutrition Services, St Vincent's Health Network, Darlinghurst, Australia
- Translational Health Research Institute, Eating Disorders and Body Image, School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
| | - Amy Loughman
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Felice N Jacka
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Black Dog Institute, Randwick, New South Wales, Australia
- James Cook University, Townsville, Queensland, Australia
| | - Heidi M Staudacher
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Afrouz Abbaspour
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutetet, Solna, Stockholm, Sweden
| | - Andrea Phillipou
- Orygen, Melbourne, Victoria, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Psychological Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
- Department of Mental Health, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Mental Health, Austin Health, Melbourne, Victoria, Australia
| | - Anu Ruusunen
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Psychiatry, Kuopio University Hospital, Kuopio, Finland
| | - Tetyana Rocks
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
- Translational Health Research Institute, Eating Disorders and Body Image, School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
| |
Collapse
|
29
|
Yu X, Ma F, Dai H, Liu J, Hashem NM, Sun P. Effects of Different Galacto-Oligosaccharide Supplementation on Growth Performance, Immune Function, Serum Nutrients, and Appetite-Related Hormones in Holstein Calves. Animals (Basel) 2023; 13:3366. [PMID: 37958121 PMCID: PMC10649109 DOI: 10.3390/ani13213366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Our previous study showed that early supplementation with 10 g/(d·head) of galacto-oligosaccharides (GOS) in newborn Holstein dairy calves reduced the incidence of diarrhea and improved growth performance and mineral absorption. Since the dose of 10 g/(d·head) was the lowest by dose screening in our previous study, the present study was designed to investigate whether a lower amount of GOS has similar effects on growth performance, immune function, serum nutrients in newborn Holstein heifer calves, and to further investigate its effect on appetite-related hormones. Twenty-eight healthy newborn (1 day of age) Holstein heifers with similar average body weight (41.18 ± 1.90 kg) were randomly divided into four groups (n = 7): the control group (CON group), which received heated raw milk, and three experimental groups, which received heated raw milk supplemented with 2.5 (GOS2.5 group), 5 (GOS5 group), and 10 g/(d·head) (GOS10 group) GOS. All heifer calves were fed the same starter for 28 d. Supplementation with GOS linearly increased the final body weight, average daily gain, and feed efficiency in heifer calves (p < 0.01). Compared with the control group, the average daily gain and feed efficiency of heifer calves were significantly higher in the GOS5 and GOS10 groups than in the control group (p < 0.05). Furthermore, supplementation with GOS quadratically enhanced the starter and total average daily feed intake of the heifers (p < 0.01), especially in the GOS2.5 and GOS5 groups, (p < 0.05 vs. CON). The serum concentration of immunoglobulin A was linearly increased by GOS supplementation (p < 0.05), and the levels in the GOS5 and GOS10 groups were significantly higher than those in the CON group. Meanwhile, GOS linearly decreased serum interleukin-1β and interleukin-6 concentrations (p < 0.05). The serum concentration of triglycerides was also linearly decreased (p < 0.05), whereas total protein and blood urea nitrogen were linearly increased (p < 0.05). Supplementation with GOS linearly decreased the serum concentration of leptin (p < 0.05) but increased cholecystokinin and glucagon-like peptide-1 (p < 0.05). Increasing doses of GOS linearly improved serum calcium and copper concentrations (p < 0.01) and quadratically enhanced the concentration of magnesium, which peaked in the GOS5 group (p < 0.05). In conclusion, GOS supplementation reduced the incidence of diarrhea and improved the growth performance and immune function of Holstein heifer calves.
Collapse
Affiliation(s)
- Xin Yu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Haonan Dai
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junhao Liu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nesrein M. Hashem
- Department of Animal and Fish Production, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt;
| | - Peng Sun
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
30
|
Ni Y, Wang X, Wu Q, Yao Y, Xu Y, Li Y, Feng Q, Zhou M, Gou X. Qushi Huayu decoction ameliorates non-alcoholic fatty liver disease in rats by modulating gut microbiota and serum lipids. Front Endocrinol (Lausanne) 2023; 14:1272214. [PMID: 37900123 PMCID: PMC10600383 DOI: 10.3389/fendo.2023.1272214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease. As a clinical empirical prescription of traditional Chinese medicine, Qushi Huayu decoction (QHD) has attracted considerable attention for its advantages in multi-target treatment of NAFLD. However, the intervention mechanism of QHD on abnormal lipid levels and gut microbiota in NAFLD has not been reported. Methods Therefore, we verified the therapeutic effect of QHD on high-fat diet (HFD)-induced NAFLD in rats by physiological parameters and histopathological examination. In addition, studies on gut microbiota and serum lipidomics based on 16S rRNA sequencing and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) were conducted to elucidate the therapeutic mechanism of NAFLD in QHD. Results The changes in gut microbiota in NAFLD rats are mainly reflected in their diversity and composition, while QHD treated rats restored these changes. The genera Blautia, Lactobacillus, Allobaculum, Lachnoclostridium and Bacteroides were predominant in the NAFLD group, whereas, Turicibacter, Blautia, Sporosarcina, Romboutsia, Clostridium_sensu_stricto_1, Allobaculum, and Psychrobacter were predominant in the NAFLD+QHD group. Lipid subclasses, including diacylglycerol (DG), triglycerides (TG), phosphatidylethanolamine (PE), phosphatidylcholine (PC), phosphatidic acid (PA), phosphatidylserine (PS), lysophosphatidylinositol (LPI), and phosphatidylglycerol (PG), were significantly different between the NAFLD and the control groups, while QHD treatment significantly altered the levels of DG, TG, PA, lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), and platelet activating factor (PAF). Finally, Spearman's correlation analysis showed that NAFLD related differential lipid molecules were mainly associated with the genera of Bacteroides, Blautia, Lachnoclostridium, Clostridium_sensu_stricto_1, and Turicibacter, which were also significantly correlated with the biological parameters of NAFLD. Discussion Taken together, QHD may exert beneficial effects by regulating the gut microbiota and thus intervening in serum lipids.
Collapse
Affiliation(s)
- Yiming Ni
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Wang
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Wu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Yichen Yao
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Xu
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Qin Feng
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Li L, Yan S, Liu S, Wang P, Li W, Yi Y, Qin S. In-depth insight into correlations between gut microbiota and dietary fiber elucidates a dietary causal relationship with host health. Food Res Int 2023; 172:113133. [PMID: 37689844 DOI: 10.1016/j.foodres.2023.113133] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 09/11/2023]
Abstract
Dietary fiber exerts a wide range of biological benefits on host health, which not only provides a powerful source of nutrition for gut microbiota but also supplies key microbial metabolites that directly affect host health. This review mainly focuses on the decomposition and metabolism of dietary fiber and the essential genera Bacteroides and Bifidobacterium in dietary fiber fermentation. Dietary fiber plays an essential role in host health by impacting outcomes related to obesity, enteritis, immune health, cancer and neurodegenerative diseases. Additionally, the gut microbiota-independent pathway of dietary fiber affecting host health is also discussed. Personalized dietary fiber intake combined with microbiome, genetics, epigenetics, lifestyle and other factors has been highlighted for development in the future. A higher level of evidence is needed to demonstrate which microbial phenotype benefits from which kind of dietary fiber. In-depth insights into the correlation between gut microbiota and dietary fiber provide strong theoretical support for the precise application of dietary fiber, which elucidates a dietary causal relationship with host health.
Collapse
Affiliation(s)
- Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Shuling Yan
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuangjiang Liu
- Shandong University, Qingdao 266237, China; Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Ping Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Wenjun Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Yuetao Yi
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|
32
|
Yang S, Yang H, Zhang Y. Yao-Shan of traditional Chinese medicine: an old story for metabolic health. Front Pharmacol 2023; 14:1194026. [PMID: 37663255 PMCID: PMC10468577 DOI: 10.3389/fphar.2023.1194026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 07/21/2023] [Indexed: 09/05/2023] Open
Abstract
Type 2 diabetes mellitus, nonalcoholic fatty liver disease (NAFLD), cardio-cerebrovascular diseases (CCVDs), hyperuricemia and gout, and metabolic-related sexual dysfunction are metabolic diseases that affect human health in modern society. Scientists have made great efforts to investigate metabolic diseases using cell models in vitro or animal models in the past. However, the findings from cells or animals are difficult to translate into clinical applications due to factors such as the in vitro and in vivo differences; the differences in anatomy, physiology, and genetics between humans and animals; and the differences in microbiome-host interaction. The Chinese have extensively used the medicated diet of traditional Chinese medicine (TCM) (also named as Yao-Shan of TCM, Chinese Yao-Shan et al.) to maintain or improve cardiometabolic health for more than 2,200 years. These ancient classic diets of TCM are essential summaries of long-term life and clinical practices. Over the past 5 years, our group has made every effort to collect and sort out the classic Yao-Shan of TCM from the ancient TCM literature since Spring and Autumn and Warring States Period, especially these are involved in the prevention and treatment of metabolic diseases, such as diabetes, NAFLD, CCVDs, hyperuricemia and gout, and sexual dysfunction. Here, we summarized and discussed the classic Yao-Shan of TCM for metabolic diseases according to the time recorded in the ancient literature, and revised the Latin names of the raw materials in these Yao-Shan of TCM. Moreover, the modern medicine evidences of some Yao-Shan of TCM on metabolic diseases have also been summarized and emphasized in here. However, the exact composition (in terms of ratios), preparation process, and dosage of many Yao-Shan are not standardized, and their main active ingredients are vague. Uncovering the mystery of Yao-Shan of TCM through modern biological and chemical strategies will help us open a door, which is ancient but now looks new, to modulate metabolic homeostasis and diseases.
Collapse
Affiliation(s)
- Shuangling Yang
- School of Health Sciences, Guangzhou Xinhua University, Guangzhou, Guangdong, China
| | - Hongzhi Yang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yaxing Zhang
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Ben-Yacov O, Godneva A, Rein M, Shilo S, Lotan-Pompan M, Weinberger A, Segal E. Gut microbiome modulates the effects of a personalised postprandial-targeting (PPT) diet on cardiometabolic markers: a diet intervention in pre-diabetes. Gut 2023; 72:1486-1496. [PMID: 37137684 PMCID: PMC10359530 DOI: 10.1136/gutjnl-2022-329201] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/17/2023] [Indexed: 05/05/2023]
Abstract
OBJECTIVE To explore the interplay between dietary modifications, microbiome composition and host metabolic responses in a dietary intervention setting of a personalised postprandial-targeting (PPT) diet versus a Mediterranean (MED) diet in pre-diabetes. DESIGN In a 6-month dietary intervention, adults with pre-diabetes were randomly assigned to follow an MED or PPT diet (based on a machine-learning algorithm for predicting postprandial glucose responses). Data collected at baseline and 6 months from 200 participants who completed the intervention included: dietary data from self-recorded logging using a smartphone application, gut microbiome data from shotgun metagenomics sequencing of faecal samples, and clinical data from continuous glucose monitoring, blood biomarkers and anthropometrics. RESULTS PPT diet induced more prominent changes to the gut microbiome composition, compared with MED diet, consistent with overall greater dietary modifications observed. Particularly, microbiome alpha-diversity increased significantly in PPT (p=0.007) but not in MED arm (p=0.18). Post hoc analysis of changes in multiple dietary features, including food-categories, nutrients and PPT-adherence score across the cohort, demonstrated significant associations between specific dietary changes and species-level changes in microbiome composition. Furthermore, using causal mediation analysis we detect nine microbial species that partially mediate the association between specific dietary changes and clinical outcomes, including three species (from Bacteroidales, Lachnospiraceae, Oscillospirales orders) that mediate the association between PPT-adherence score and clinical outcomes of hemoglobin A1c (HbA1c), high-density lipoprotein cholesterol (HDL-C) and triglycerides. Finally, using machine-learning models trained on dietary changes and baseline clinical data, we predict personalised metabolic responses to dietary modifications and assess features importance for clinical improvement in cardiometabolic markers of blood lipids, glycaemic control and body weight. CONCLUSIONS Our findings support the role of gut microbiome in modulating the effects of dietary modifications on cardiometabolic outcomes, and advance the concept of precision nutrition strategies for reducing comorbidities in pre-diabetes. TRIAL REGISTRATION NUMBER NCT03222791.
Collapse
Affiliation(s)
- Orly Ben-Yacov
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Rein
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- School of Public Health, University of Haifa, Haifa, Israel
| | - Smadar Shilo
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Maya Lotan-Pompan
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
34
|
Fan P, Shen Q, Du O, Chen Y, Tang W, Ma J, Ma R, Zhang T, Luo ZC, Liu Z, Ouyang F. Urinary antibiotics concentrations, their related affecting factors and infant growth in the first 6 months of life: A prospective cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115196. [PMID: 37506555 DOI: 10.1016/j.ecoenv.2023.115196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023]
Abstract
Antibiotic exposure even in low-dose could have potential adverse health effects, especially during early life. There is a lack of data on antibiotic burdens in early infancy. We aim to assess antibiotic exposure in infants from birth to 6 months of age, their related affecting factors and the association between antibiotic exposure and infancy growth. Urine samples were collected at ages of 3 days, 42 days, 3 months and 6 months from 197 term-born Chinese infants. A total of 33 representative antibiotics were measured by high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Urinary antibiotics were detectable in 69.4%, 63.2%, 75.0% and 84.3% of infants at ages of 3 days, 42 days, 3 and 6 months, respectively. The dominant antibiotic categories detected were: Preferred as Veterinary Antibiotics (PVAs), Human Antibiotics (HAs), and Veterinary Antibiotics (VAs). The detectable rates were 30.6%, 45.8%, 58.9%, and 81.4% for PVAs, 34.1%, 20.8%, 28.6%, and 45.1% for HAs, and 36.5%, 12.5%, 6.3%, and 5.9% for VAs, at age 3 days, 42 days, 3 and 6 months, respectively. Urinary concentrations of HAs and preferred as human antibiotics (PHAs) in newborns at age 3 days were not associated with maternal intrapartum antibiotic prophylaxis. Similarly, no associations were observed between urinary antibiotics concentration and antibiotics use in infants at age 42 days or 6 months. The numbers and concentrations of urine detectable antibiotics were similar between infants with exclusive breastfeeding and infants fed with formula or mixed-feeding at all ages of 42 days, 3 and 6 months. At age of 42 days, infants in the low tertile of total antibiotics concentration or with one antibiotic detected had higher weight-for-length Z score and greater head circumference, compared to infants with no antibiotics detected. No associations were found between urinary antibiotics and any of the infant anthropometric measures at age 6 months. In conclusion, urinary antibiotics were detectable in most infants during the first 6 months of life, and PVAs, HAs and VAs were the most commonly detected antibiotics. This suggested the possibility of a foods-originated antibiotics exposure in children. No strong nor consistent associations were found between urinary antibiotic concentration and infant growth at the first six months of life. Still, attention is needed on the adverse health effect of early life exposure to antibiotics in future studies.
Collapse
Affiliation(s)
- Pianpian Fan
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianwen Shen
- Department of Neonatology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ouyang Du
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Electrical and Systems Engineering, McKelvey School of Engineering, Washington University in St. Louis, USA
| | - Yuanzhi Chen
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weifeng Tang
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinqian Ma
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Ma
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Zhang
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong-Cheng Luo
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Obstetrics and Gynecology, Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Zhiwei Liu
- Department of Neonatology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fengxiu Ouyang
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
He F, Jin X, Wang C, Hu J, Su S, Zhao L, Geng T, Zhao Y, Pan L, Bao N, Sun H. Lactobacillus rhamnosus GG ATCC53103 and Lactobacillus plantarum JL01 improved nitrogen metabolism in weaned piglets by regulating the intestinal flora structure and portal vein metabolites. Front Microbiol 2023; 14:1200594. [PMID: 37455717 PMCID: PMC10338925 DOI: 10.3389/fmicb.2023.1200594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
At present, most studies have shown that probiotics have a positive regulatory effect on the nutritional metabolism of the body, but the mechanism is still unclear. Here, 48 piglets were divided into four groups. The control group was not fed probiotics, the Lac group was fed L. Rhamnosus GG ATCC53103, the Rha group was fed L. Plantarum JL01, and the mix group was fed two types of probiotics. Nitrogen metabolism and mRNA levels of mTOR and S6K in skeletal muscle were observed in each group. Then, metagenome and non-targeted metabonomics were used to observe the changes of intestinal microorganisms and plasma metabolites in portal channels after probiotics feeding. Finally, we combined the results of omics analysis to reveal the mechanism of probiotics on nitrogen metabolism in weaned piglets. The results showed that L. Rhmnosus GG ATCC53103 and L. Plantarum JL01 increased nitrogen apparent digestibility, nitrogen deposition rate, and nitrogen utilization rate of weaned piglets (P < 0.05); the relative expression of mTOR and SK6 mRNA in skeletal muscle increased significantly (P < 0.05). When L. rhamnosus GG ATCC53103 and L. plantarum JL01 were combined, we found that Clostridium and Prevotella significantly increased in the jejunum (P < 0.05). The relative abundance of Lactobacillus, Ruminococcus, Streptococcus, and Prevotella in the ileum increased significantly (P < 0.05). Compared with the control group, L-Tryptophan, 3-Phosphonyloxypyruvate, cis-Aconitate, and Carbamoyl phosphate were significantly increased in the mixed group portal vein. The result of the combinatorial analysis showed that the significantly increased microorganisms could encode the enzyme genes for the synthesis of L-Tryptophan, 3-Phosphonooxypyruvate, cis-Aconitate, and Carbamoyl phosphate. In summary, our results demonstrated that L. Rhamnosus GG ATCC53103 and L. Plantarum JL01 could stimulate the expression of skeletal muscle protein synthesis genes of weaned piglets by modulating the structure of the gut microbiota and its metabolites, thereby improving nitrogen metabolism in weaned piglets.
Collapse
Affiliation(s)
- Feng He
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xueying Jin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Shuai Su
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Lei Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Tingting Geng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Yuan Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Li Pan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Nan Bao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| |
Collapse
|
36
|
Wang Y, Jian C. Novel plant-based meat alternatives: Implications and opportunities for consumer nutrition and health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023; 106:241-274. [PMID: 37722774 DOI: 10.1016/bs.afnr.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Against the backdrop of the global protein transition needed to remain within planetary boundaries, there is an influx of plant-based meat alternatives that seek to approximate the texture, flavor and/or nutrient profiles of conventional animal meat. These novel plant-based meat alternatives, enabled by advances in food technology, can be fundamentally different from the whole-plant foods from which they are derived. One of the reasons is the necessity to use food additives on various occasions, since consumers' acceptance of plant-based meat products primarily depends on the organoleptic properties. Consequently, a high degree of heterogeneity in formulation and nutritional profiles exists both within and between product categories of plant-based meat alternatives with unknown effects on several aspects of human health. This is further complicated by the differences in digestibility and bioavailability between proteins from animal and plant sources, which have a profound impact on colonic fermentation, nutritional adequacy and potential health effects. On the other hand, emerging strategies provide opportunities to develop affordable, delicious and nutritious plant-based meat alternatives that align with consumer interests.
Collapse
Affiliation(s)
- Yaqin Wang
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland.
| | - Ching Jian
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland; Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
37
|
Sami AS, Frazer LC, Miller CM, Singh DK, Clodfelter LG, Orgel KA, Good M. The role of human milk nutrients in preventing necrotizing enterocolitis. Front Pediatr 2023; 11:1188050. [PMID: 37334221 PMCID: PMC10272619 DOI: 10.3389/fped.2023.1188050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.
Collapse
Affiliation(s)
- Ahmad S. Sami
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lynda G. Clodfelter
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
38
|
Wang Y, Salonen A, Jian C. Can prebiotics help tackle the childhood obesity epidemic? Front Endocrinol (Lausanne) 2023; 14:1178155. [PMID: 37305030 PMCID: PMC10253620 DOI: 10.3389/fendo.2023.1178155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Globally, excess weight during childhood and adolescence has become a public health crisis with limited treatment options. Emerging evidence suggesting the involvement of gut microbial dysbiosis in obesity instills hope that targeting the gut microbiota could help prevent or treat obesity. In pre-clinical models and adults, prebiotic consumption has been shown to reduce adiposity partially via restoring symbiosis. However, there is a dearth of clinical research into its potential metabolic benefits in the pediatric population. Here, we provide a succinct overview of the common characteristics of the gut microbiota in childhood obesity and mechanisms of action of prebiotics conferring metabolic benefits. We then summarize available clinical trials in children with overweight or obesity investigating the effects of prebiotics on weight management. This review highlights several controversial aspects in the microbiota-dependent mechanisms by which prebiotics are thought to affect host metabolism that warrant future investigation in order to design efficacious interventions for pediatric obesity.
Collapse
Affiliation(s)
- Yaqin Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan, China
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ching Jian
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Törős G, El-Ramady H, Prokisch J, Velasco F, Llanaj X, Nguyen DHH, Peles F. Modulation of the Gut Microbiota with Prebiotics and Antimicrobial Agents from Pleurotus ostreatus Mushroom. Foods 2023; 12:foods12102010. [PMID: 37238827 DOI: 10.3390/foods12102010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Pleurotus ostreatus (Jacq. ex Fr.) P. Kumm mushroom contains bioactive compounds with both antimicrobial and prebiotic properties, which are distributed in the mushroom mycelium, fruiting body, and spent substrate. The mushroom is rich in nondigestible carbohydrates like chitin and glucan, which act as prebiotics and support the growth and activity of beneficial gut bacteria, thereby maintaining a healthy balance of gut microbiota and reducing the risk of antibiotic resistance. The bioactive compounds in P. ostreatus mushrooms, including polysaccharides (glucans, chitin) and secondary metabolites (phenolic compounds, terpenoids, and lectins), exhibit antibacterial, antiviral, and antifungal activities. When mushrooms are consumed, these compounds can help preventing the growth and spread of harmful bacteria in the gut, reducing the risk of infections and the development of antibiotic resistance. Nonetheless, further research is necessary to determine the efficacy of P. ostreatus against different pathogens and to fully comprehend its prebiotic and antimicrobial properties. Overall, consuming a diet rich in mushroom-based foods can have a positive impact on human digestion health. A mushroom-based diet can support a healthy gut microbiome and reduce the need for antibiotics.
Collapse
Affiliation(s)
- Gréta Törős
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Animal Husbandry, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Hassan El-Ramady
- Soil and Water Department, Faculty of Agriculture, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - József Prokisch
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Fernando Velasco
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Xhensila Llanaj
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Food Science, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Duyen H H Nguyen
- Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Food Science, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Tay Nguyen Institute for Scientific Research, Vietnam Academy of Science and Technology, Dalat 70072, Vietnam
| | - Ferenc Peles
- Institute of Food Science, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| |
Collapse
|
40
|
Small Intestinal Bacterial Overgrowth and Non-Alcoholic Fatty Liver Disease: What Do We Know in 2023? Nutrients 2023; 15:nu15061323. [PMID: 36986052 PMCID: PMC10052062 DOI: 10.3390/nu15061323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease associated with the pathological accumulation of lipids inside hepatocytes. Untreated NAFL can progress to non-alcoholic hepatitis (NASH), followed by fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The common denominator of the above-mentioned metabolic disorders seems to be insulin resistance, which occurs in NAFLD patients. Obesity is the greatest risk factor for lipid accumulation inside hepatocytes, but a part of the NAFLD patient population has a normal body weight according to the BMI index. Obese people with or without NAFLD have a higher incidence of small intestinal bacterial overgrowth (SIBO), and those suffering from NAFLD show increased intestinal permeability, including a more frequent presence of bacterial overgrowth in the small intestine (SIBO). The health consequences of SIBO are primarily malabsorption disorders (vitamin B12, iron, choline, fats, carbohydrates and proteins) and bile salt deconjugation. Undetected and untreated SIBO may lead to nutrient and/or energy malnutrition, thus directly impairing liver function (e.g., folic acid and choline deficiency). However, whether SIBO contributes to liver dysfunction, decreased intestinal barrier integrity, increased inflammation, endotoxemia and bacterial translocation is not yet clear. In this review, we focus on gut–liver axis and discuss critical points, novel insights and the role of nutrition, lifestyle, pre- and probiotics, medication and supplements in the therapy and prevention of both SIBO and NAFLD.
Collapse
|
41
|
Bastings JJAJ, Venema K, Blaak EE, Adam TC. Influence of the gut microbiota on satiety signaling. Trends Endocrinol Metab 2023; 34:243-255. [PMID: 36870872 DOI: 10.1016/j.tem.2023.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 03/06/2023]
Abstract
Recent studies show a link between the gut microbiota and the regulation of satiety and energy intake, processes that contribute to the development and pathophysiology of metabolic diseases. However, this link is predominantly established in animal and in vitro studies, whereas human intervention studies are scarce. In this review we focus on recent evidence linking satiety and the gut microbiome, with specific emphasis on gut microbial short-chain fatty acids (SCFAs). Based on a systematic search we provide an overview of human studies linking the intake of prebiotics with gut microbial alterations and satiety signaling. Our outcomes highlight the importance of in-depth examination of the gut microbiota in relation to satiety and provide insights into recent and future studies in this field.
Collapse
Affiliation(s)
- Jacco J A J Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Koen Venema
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands; Centre for Healthy Eating and Food Innovation, Maastricht University, Campus Venlo, Venlo, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Tanja C Adam
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
42
|
Valles-Colomer M, Menni C, Berry SE, Valdes AM, Spector TD, Segata N. Cardiometabolic health, diet and the gut microbiome: a meta-omics perspective. Nat Med 2023; 29:551-561. [PMID: 36932240 PMCID: PMC11258867 DOI: 10.1038/s41591-023-02260-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/16/2023] [Indexed: 03/19/2023]
Abstract
Cardiometabolic diseases have become a leading cause of morbidity and mortality globally. They have been tightly linked to microbiome taxonomic and functional composition, with diet possibly mediating some of the associations described. Both the microbiome and diet are modifiable, which opens the way for novel therapeutic strategies. High-throughput omics techniques applied on microbiome samples (meta-omics) hold the unprecedented potential to shed light on the intricate links between diet, the microbiome, the metabolome and cardiometabolic health, with a top-down approach. However, effective integration of complementary meta-omic techniques is an open challenge and their application on large cohorts is still limited. Here we review meta-omics techniques and discuss their potential in this context, highlighting recent large-scale efforts and the novel insights they provided. Finally, we look to the next decade of meta-omics research and discuss various translational and clinical pathways to improving cardiometabolic health.
Collapse
Affiliation(s)
- Mireia Valles-Colomer
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Cristina Menni
- Department of Twin Research, King's College London, London, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Ana M Valdes
- School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham National Institute for Health Research Biomedical Research Centre, Nottingham, UK
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
- European Institute of Oncology, Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy.
| |
Collapse
|
43
|
Wang Y, Jian C, Salonen A, Dong M, Yang Z. Designing healthier bread through the lens of the gut microbiota. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
44
|
Wang W, Liu Y, Li Y, Luo B, Lin Z, Chen K, Liu Y. Dietary patterns and cardiometabolic health: Clinical evidence and mechanism. MedComm (Beijing) 2023; 4:e212. [PMID: 36776765 PMCID: PMC9899878 DOI: 10.1002/mco2.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 02/08/2023] Open
Abstract
For centuries, the search for nutritional interventions to underpin cardiovascular treatment and prevention guidelines has contributed to the rapid development of the field of dietary patterns and cardiometabolic disease (CMD). Numerous studies have demonstrated that healthy dietary patterns with emphasis on food-based recommendations are the gold standard for extending lifespan and reducing the risks of CMD and mortality. Healthy dietary patterns include various permutations of energy restriction, macronutrients, and food intake patterns such as calorie restriction, intermittent fasting, Mediterranean diet, plant-based diets, etc. Early implementation of healthy dietary patterns in patients with CMD is encouraged, but an understanding of the mechanisms by which these patterns trigger cardiometabolic benefits remains incomplete. Hence, this review examined several dietary patterns that may improve cardiometabolic health, including restrictive dietary patterns, regional dietary patterns, and diets based on controlled macronutrients and food groups, summarizing cutting-edge evidence and potential mechanisms for CMD prevention and treatment. Particularly, considering individual differences in responses to dietary composition and nutritional changes in organ tissue diversity, we highlighted the critical role of individual gut microbiota in the crosstalk between diet and CMD and recommend a more precise and dynamic nutritional strategy for CMD by developing dietary patterns based on individual gut microbiota profiles.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yanfei Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yiwen Li
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Binyu Luo
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Zhixiu Lin
- Faculty of MedicineThe Chinese University of Hong KongHong Kong
| | - Keji Chen
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yue Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
45
|
MetaboVariation: Exploring Individual Variation in Metabolite Levels. Metabolites 2023; 13:metabo13020164. [PMID: 36837783 PMCID: PMC9965648 DOI: 10.3390/metabo13020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
To date, most metabolomics biomarker research has focused on identifying disease biomarkers. However, there is a need for biomarkers of early metabolic dysfunction to identify individuals who would benefit from lifestyle interventions. Concomitantly, there is a need to develop strategies to analyse metabolomics data at an individual level. We propose "MetaboVariation", a method that models repeated measurements on individuals to explore fluctuations in metabolite levels at an individual level. MetaboVariation employs a Bayesian generalised linear model to flag individuals with intra-individual variations in their metabolite levels across multiple measurements. MetaboVariation models repeated metabolite levels as a function of explanatory variables while accounting for intra-individual variation. The posterior predictive distribution of metabolite levels at the individual level is available, and is used to flag individuals with observed metabolite levels outside the 95% highest posterior density prediction interval at a given time point. MetaboVariation was applied to a dataset containing metabolite levels for 20 metabolites, measured once every four months, in 164 individuals. A total of 28% of individuals with intra-individual variations in three or more metabolites were flagged. An R package for MetaboVariation was developed with an embedded R Shiny web application. To summarize, MetaboVariation has made considerable progress in developing strategies for analysing metabolomics data at the individual level, thus paving the way toward personalised healthcare.
Collapse
|
46
|
Tuska RM, Helm SM, Graf CF, James C, Kong G, Stiemsma LT, Green DB, Helm SE. Surfeit folic acid, protein, and exercise modify oncogenic inflammatory biomarkers and fecal microbiota. Front Nutr 2023; 9:1060212. [PMID: 36742002 PMCID: PMC9894611 DOI: 10.3389/fnut.2022.1060212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
Intestinal microbiota, diet, and physical activity are inextricably linked to inflammation occurring in the presence of tumor progression and declining neurocognition. This study aimed to explore how fecal microbiota, inflammatory biomarkers, and neurocognitive behavior are influenced by voluntary exercise and surplus dietary protein and folic acid which are common health choices. Dietary treatments provided over 8 weeks to C57BL/CJ male mice (N = 76) were: Folic Acid (FA) Protein (P) Control (FPC, 17.9% P; 2 mgFA/kg); Folic Acid Deficient (FAD); Folic Acid Supplemented (FAS; 8 mgFA/kg); Low Protein Diet (LPD, 6% P); and High Protein Diet (HPD, 48% P). FAS mice had decreased plasma HCys (p < 0.05), therefore confirming consumption of FA. Objectives included examining influence of exercise using Voluntary Wheel Running (VWR) upon fecal microbiota, inflammatory biomarkers C - reactive protein (CRP), Vascular Endothelial Growth Factor (VEGF), Interleukin-6 (IL-6), nuclear factor kappa ß subunit (NF-κßp65), Caspase-3 (CASP3), Tumor Necrosis Factor-alpha (TNF-α), and neurocognitive behavior. CRP remained stable, while a significant exercise and dietary effect was notable with decreased VEGF (p < 0.05) and increased CASP3 (p < 0.05) for exercised HPD mice. Consumption of FAS did significantly increase (p < 0.05) muscle TNF-α and the ability to build a nest (p < 0.05) was significantly decreased for both FAD and LPD exercised mice. Rearing behavior was significantly increased (p < 0.05) in mice fed HPD. An emerging pattern with increased dietary protein intake revealed more distance explored in Open Field Testing. At week 1, both weighted and unweighted UniFrac principal coordinates analysis yielded significant clustering (permanova, p ≤ 0.05) associated with the specific diets. Consumption of a HPD diet resulted in the most distinct fecal microbiota composition. At the phylum level-comparing week 1 to week 8-we report a general increase in the Firmicutes/Bacteroidetes ratio, characterized by an outgrowth of Firmicutes by week 8 in all groups except the HPD. MaAsLin2 analysis corroborates this finding and emphasizes an apparent inversion of the microbiome composition at week 8 after HPD. Explicit modification of oncogenic inflammatory biomarkers and fecal microbiome post high FA and protein intake along with voluntary exercise contributed to current underlying evidence that this diet and exercise relationship has broader effects on human health and disease-perhaps importantly as a practical modulation of cancer progression and declining neurocognition.
Collapse
|
47
|
Zeb F, Osaili T, Obaid RS, Naja F, Radwan H, Cheikh Ismail L, Hasan H, Hashim M, Alam I, Sehar B, Faris ME. Gut Microbiota and Time-Restricted Feeding/Eating: A Targeted Biomarker and Approach in Precision Nutrition. Nutrients 2023; 15:259. [PMID: 36678130 PMCID: PMC9863108 DOI: 10.3390/nu15020259] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Each individual has a unique gut microbiota; therefore, the genes in our microbiome outnumber the genes in our genome by about 150 to 1. Perturbation in host nutritional status influences gut microbiome composition and vice versa. The gut microbiome can help in producing vitamins, hormones, and other active metabolites that support the immune system; harvest energy from food; aid in digestion; protect against pathogens; improve gut transit and function; send signals to the brain and other organs; oscillate the circadian rhythm; and coordinate with the host metabolism through multiple cellular pathways. Gut microbiota can be influenced by host genetics, medications, diet, and lifestyle factors from preterm to aging. Aligning with precision nutrition, identifying a personalized microbiome mandates the provision of the right nutrients at the right time to the right patient. Thus, before prescribing a personalized treatment, it is crucial to monitor and count the gut flora as a focused biomarker. Many nutritional approaches that have been developed help in maintaining and restoring an optimal microbiome such as specific diet therapy, nutrition interventions, and customized eating patterns. One of these approaches is time-restricted feeding/eating (TRF/E), a type of intermittent fasting (IF) in which a subject abstains from food intake for a specific time window. Such a dietary modification might alter and restore the gut microbiome for proper alignment of cellular and molecular pathways throughout the lifespan. In this review, we have highlighted that the gut microbiota would be a targeted biomarker and TRF/E would be a targeted approach for restoring the gut-microbiome-associated molecular pathways such as hormonal signaling, the circadian system, metabolic regulators, neural responses, and immune-inflammatory pathways. Consequently, modulation of the gut microbiota through TRF/E could contribute to proper utilization and availability of the nutrients and in this way confer protection against diseases for harnessing personalized nutrition approaches to improve human health.
Collapse
Affiliation(s)
- Falak Zeb
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Tareq Osaili
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Reyad Shakir Obaid
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Farah Naja
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hadia Radwan
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Leila Cheikh Ismail
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hayder Hasan
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mona Hashim
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Iftikhar Alam
- Department of Human Nutrition and Dietetics, Bacha Khan University Charsadda, Peshawar 24540, KP, Pakistan
| | - Bismillah Sehar
- Department of Health and Social Sciences, University of Bedfordshire, Luton LU1 3JU, UK
| | - MoezAllslam Ezzat Faris
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
48
|
Trouwborst I, Gijbels A, Jardon KM, Siebelink E, Hul GB, Wanders L, Erdos B, Péter S, Singh-Povel CM, de Vogel-van den Bosch J, Adriaens ME, Arts ICW, Thijssen DHJ, Feskens EJM, Goossens GH, Afman LA, Blaak EE. Cardiometabolic health improvements upon dietary intervention are driven by tissue-specific insulin resistance phenotype: A precision nutrition trial. Cell Metab 2023; 35:71-83.e5. [PMID: 36599304 DOI: 10.1016/j.cmet.2022.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/12/2022] [Accepted: 11/13/2022] [Indexed: 01/05/2023]
Abstract
Precision nutrition based on metabolic phenotype may increase the effectiveness of interventions. In this proof-of-concept study, we investigated the effect of modulating dietary macronutrient composition according to muscle insulin-resistant (MIR) or liver insulin-resistant (LIR) phenotypes on cardiometabolic health. Women and men with MIR or LIR (n = 242, body mass index [BMI] 25-40 kg/m2, 40-75 years) were randomized to phenotype diet (PhenoDiet) group A or B and followed a 12-week high-monounsaturated fatty acid (HMUFA) diet or low-fat, high-protein, and high-fiber diet (LFHP) (PhenoDiet group A, MIR/HMUFA and LIR/LFHP; PhenoDiet group B, MIR/LFHP and LIR/HMUFA). PhenoDiet group B showed no significant improvements in the primary outcome disposition index, but greater improvements in insulin sensitivity, glucose homeostasis, serum triacylglycerol, and C-reactive protein compared with PhenoDiet group A were observed. We demonstrate that modulating macronutrient composition within the dietary guidelines based on tissue-specific insulin resistance (IR) phenotype enhances cardiometabolic health improvements. Clinicaltrials.gov registration: NCT03708419, CCMO registration NL63768.068.17.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Anouk Gijbels
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Kelly M Jardon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gabby B Hul
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Lisa Wanders
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Balázs Erdos
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | | | | | | | - Michiel E Adriaens
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Dick H J Thijssen
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lydia A Afman
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands.
| |
Collapse
|
49
|
Suriano F, Vieira-Silva S, Falony G, de Wouters d'Oplinter A, Paone P, Delzenne NM, Everard A, Raes J, Van Hul M, Cani PD. Fat and not sugar as the determining factor for gut microbiota changes, obesity, and related metabolic disorders in mice. Am J Physiol Endocrinol Metab 2023; 324:E85-E96. [PMID: 36516223 DOI: 10.1152/ajpendo.00141.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diet-induced obesity contributes to the development of type 2 diabetes, insulin resistance, metabolic inflammation, oxidative and endoplasmic reticulum (ER) stress. Overall, obesity is associated with deviations in the composition and functionality of the gut microbiota. There are many divergent findings regarding the link between the excessive intake of certain dietary components (i.e., fat and sugar) and obesity development. We therefore investigated the effect of specific diets, with a different content of sugar and fat, in promoting obesity and related comorbidities as well as their impact on microbial load and gut microbiota composition/diversity. C57BL/6J mice were fed either a low-sugar, low-fat control diet (CT), a high-sugar diet (HS), a high-fat, high-sugar diet (HF/HS), or a high-fat diet (HF) for 8 wk. The impact of the different diets on obesity, glucose metabolism, inflammation, and oxidative and ER stress was determined. Diet-induced changes in the gut microbiota composition and density were also analyzed. HF diet-fed mice showed the highest body weight and fat mass gains and displayed the most impaired glucose and insulin profiles. HS, HF/HS, and HF diets differently affected hepatic cholesterol content and mRNA expression of several markers associated with immune cells, inflammation, oxidative and ER stress in several organs/tissues. In addition, HF diet feeding resulted in a decreased microbial load at the end of the experiment. When analyzing the gut microbiota composition, we found that HS, HF/HS, and HF diets induced specific changes in the abundance of certain bacterial taxa. This was not associated with a specific change in systemic inflammatory markers, but HS mice exhibited higher FGF21 plasma levels compared with HF diet-fed mice. Taken together, our results highlight that dietary intake of different macronutrients distinctively impacts the development of an obese/diabetic state and the regulation of metabolic inflammation in specific organs. We propose that these differences are not only obesity-driven but that changes in the gut microbiota composition may play a key role in this context.NEW & NOTEWORTHY To our knowledge, this study is the first to demonstrate that dietary macronutrients (i.e., sugar and fat) have an impact on fecal bacterial cell counting and quantitative microbiome profiling in mice. Yet, we demonstrate that dietary fat is the determining factor to promote obesity and diabetes progression, and local inflammation in different body sites. These observations can help to disentangle the conundrum of the detrimental effects of fat and sugar in our dietary habits.
Collapse
Affiliation(s)
- Francesco Suriano
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Sara Vieira-Silva
- Department of Microbiology and Immunology, Rega Institute for Medical Research, VIB Center for Microbiology, University of Leuven, Leuven, Belgium
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Gwen Falony
- Department of Microbiology and Immunology, Rega Institute for Medical Research, VIB Center for Microbiology, University of Leuven, Leuven, Belgium
| | - Alice de Wouters d'Oplinter
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Paola Paone
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, VIB Center for Microbiology, University of Leuven, Leuven, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
50
|
Systematic Review: Contribution of the Gut Microbiome to the Volatile Metabolic Fingerprint of Colorectal Neoplasia. Metabolites 2022; 13:metabo13010055. [PMID: 36676980 PMCID: PMC9865897 DOI: 10.3390/metabo13010055] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) has been associated with changes in volatile metabolic profiles in several human biological matrices. This enables its non-invasive detection, but the origin of these volatile organic compounds (VOCs) and their relation to the gut microbiome are not yet fully understood. This systematic review provides an overview of the current understanding of this topic. A systematic search using PubMed, Embase, Medline, Cochrane Library, and the Web of Science according to PRISMA guidelines resulted in seventy-one included studies. In addition, a systematic search was conducted that identified five systematic reviews from which CRC-associated gut microbiota data were extracted. The included studies analyzed VOCs in feces, urine, breath, blood, tissue, and saliva. Eight studies performed microbiota analysis in addition to VOC analysis. The most frequently reported dysregulations over all matrices included short-chain fatty acids, amino acids, proteolytic fermentation products, and products related to the tricarboxylic acid cycle and Warburg metabolism. Many of these dysregulations could be related to the shifts in CRC-associated microbiota, and thus the gut microbiota presumably contributes to the metabolic fingerprint of VOC in CRC. Future research involving VOCs analysis should include simultaneous gut microbiota analysis.
Collapse
|