1
|
Martinez P, Sabatier JM. Malignant Tumors in Vagal-innervated Organs: Exploring Its Homeostatic Role. Cancer Lett 2025:217539. [PMID: 39954934 DOI: 10.1016/j.canlet.2025.217539] [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: 12/01/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Cancer remains a significant global health challenge, with its progression shaped by complex and multifactorial mechanisms. Recent research suggests that the vagus nerve could play a critical role in mediating communication between the tumor microenvironment and the central nervous system (CNS). This review highlights the diversity of vagal afferent receptors, which could position the vagus nerve as a unique pathway for transmitting immune, metabolic, mechanical, and chemical signals from tumors to the CNS. Such signaling could influence systemic disease progression and tumor-related responses. Additionally, the vagus nerve's interactions with the microbiome and the renin-angiotensin system (RAS)-both implicated in cancer biology-further underscore its potential central role in modulating tumor-related processes. Contradictions in the literature, particularly concerning vagal fibers, illustrate the complexity of its involvement in tumor progression, with both tumor-promoting and tumor-suppressive effects reported depending on cancer type and context. These contradictions often overlook certain experimental biases, such as the failure to distinguish between vagal afferent and efferent fibers during vagotomies or the localized parasympathetic effects that cannot always be extrapolated to the systemic level. By focusing on the homeostatic role of the vagus nerve, understanding these mechanisms could open the door to new perspectives in cancer research related to the vagus nerve and lead to potential therapeutic innovations.
Collapse
Affiliation(s)
| | - Jean-Marc Sabatier
- Institut de NeuroPhysiopathologie (INP), CNRS UMR 7051, 27 Bd Jean Moulin, 13005, Marseille
| |
Collapse
|
2
|
Wei W, Wang S, Yuan Z, Ren Y, Wu J, Gao X, Wang R, Li J. Plant-based diets and the risk of lung cancer: a large prospective cohort study. Eur J Nutr 2025; 64:73. [PMID: 39891747 PMCID: PMC11787154 DOI: 10.1007/s00394-024-03570-0] [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: 07/07/2024] [Accepted: 12/12/2024] [Indexed: 02/03/2025]
Abstract
PURPOSE Plant-based diets are increasingly recognized for cancer prevention, yet their specific impact on lung cancer (LC) risk remains insufficiently examined. This study aims to assess the relationship between plant-based diets adherence and the incidence of LC. METHODS Data from the Prostate, Lung, Colorectal, and Ovarian cancer screening trial were analyzed. The plant-based diet index (PDI) was developed to assess adherence to plant-based diets. Multivariable Cox regression model was performed to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Restricted cubic spline (RCS) model was performed to examine risk across the PDI spectrum. Prespecified subgroup analyses identified potential modifiers, and sensitivity analyses tested the association's robustness. RESULTS Of the 98,459 participants included, 1,642 developed LC over an average follow-up of 8.83 years. Higher PDI scores were associated with a lower LC risk (HR quartile 4 vs. 1: 0.75, 95% CI: 0.65-0.87, P trend < 0.001), evident in both non-small cell lung cancer (HR quartile 4 vs. 1: 0.76, 95% CI: 0.65-0.88, P trend < 0.001) and small cell lung cancer (HR quartile 4 vs. 1: 0.73, 95% CI: 0.49-1.09, P trend = 0.046). RCS analyses further confirmed these relationships. The association was stronger among participants with lower BMI, smokers, those without a history of emphysema or diabetes, those without a family history of LC, and those with lower physical activity (all P trend < 0.001). Sensitivity analyses consistently supported these findings. CONCLUSION Our findings reveal an inverse correlation between PDI and LC risk, supporting the potential benefits of plant-based diets in LC prevention. CLINICAL TRIAL REGISTRY NUMBER ClinicalTrials.gov ID: NCT00339495 (URL: https://www. CLINICALTRIALS gov/study/NCT00339495 ).
Collapse
Affiliation(s)
- Wei Wei
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Shuyuan Wang
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhen Yuan
- School of Medicine, Nankai University, Tianjin, China
| | - Yifan Ren
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, China
| | - Jiaxing Wu
- School of Medicine, Nankai University, Tianjin, China
| | - Xiaohui Gao
- Department of Oncology, The Nuclear Industry 416 Hospital, Chengdu, China
| | - Rong Wang
- Department of Radiation Oncology, Zhongshan City People's Hospital, No. 2, Sunwen East Road, Zhongshan, Guangdong, 528400, China.
| | - Jianxiong Li
- Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
3
|
Huang H, Zhao L, Kong X, Zhu J, Lu J. Vinegar powder exerts immunomodulatory effects through alleviating immune system damage and protecting intestinal integrity and microbiota homeostasis. FOOD BIOSCI 2025; 63:105687. [DOI: 10.1016/j.fbio.2024.105687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Ericson U, Hellstrand S, Larsson A, Miari M, Sayols-Baixeras S, Dekkers KF, Bergström G, Malinovschi A, Engström G, Ärnlöv J, Fall T, Orho-Melander M. A Swedish dietary guideline index, gut microbial α-diversity and prevalence of metabolic syndrome - observations in the Swedish CArdioPulmonary bioImage Study (SCAPIS). Food Nutr Res 2024; 68:10547. [PMID: 39691688 PMCID: PMC11650442 DOI: 10.29219/fnr.v68.10547] [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: 01/25/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 12/19/2024] Open
Abstract
Background Metabolic syndrome (MetS) is characterized by coexisting risk factors for type 2 diabetes and cardiovascular disease. Diet is of importance in their aetiology, and gut microbiota (GM) may constitute a link between diet and metabolic health. Understanding the interplay between diet and GM could contribute novel insights for future dietary guidelines, and aid in preventive actions to motivate adherence to dietary guidelines. Objective We intended to create a Swedish dietary guideline index (SweDGI) measuring adherence to 12 Swedish dietary guidelines and examine whether SweDGI and its components are associated with GM α-diversity (Shannon index) and prevalent MetS, and if the association between the Shannon index and MetS differs depending on SweDGI. Design SweDGI was based on food-frequency data assessed 2014-2018 in 10,396 diabetes-free participants from the Malmö and Uppsala-sites of the Swedish CArdioPulmonary bioImage Study (SCAPIS) (50-64 y, 53% women). We estimated the Shannon index from shotgun metagenomic sequencing-data to assess microbial richness and evenness. We used a general linear model to examine cross-sectional SweDGI-Shannon associations and logistic regression for associations with MetS. Results Most guidelines were followed by less than half of the participants. Men showed poorer adherence. Higher SweDGI was linked to higher Shannon index (P-trend across five SweDGI-groups = 1.7 × 10-12). Most guidelines contributed to this observation. Higher SweDGI and Shannon index were associated with lower MetS-prevalence, where the lowest prevalence was observed among those with both high SweDGI and high Shannon index (odds ratio:0.43; 95% confidence interval:0.35, 0.52). Both the Shannon index and SweDGI were associated with MetS, independently of the level of the other factor (P-interaction = 0.82). Conclusions We created a new index to comprehensively reflect adherence to the Swedish dietary guidelines in sub-cohorts within the large multicentre SCAPIS study. Better adherence was associated with a richer and more even GM and lower prevalence of MetS. The inverse association between the Shannon index and MetS was consistent at different levels of adherence to dietary guidelines.
Collapse
Affiliation(s)
- Ulrika Ericson
- Department of Clinical Sciences in Malmö, Diabetes and Cardiovascular Disease, Lund University, Malmö, Sweden
| | - Sophie Hellstrand
- Department of Clinical Sciences in Malmö, Diabetes and Cardiovascular Disease, Lund University, Malmö, Sweden
| | - Anna Larsson
- Department of Clinical Sciences in Malmö, Diabetes and Cardiovascular Disease, Lund University, Malmö, Sweden
| | - Mariam Miari
- Department of Clinical Sciences in Malmö, Diabetes and Cardiovascular Disease, Lund University, Malmö, Sweden
| | - Sergi Sayols-Baixeras
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala University, Uppsala, Sweden
- CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Cardiovascular Research-Epidemiology, Lund University, Malmö, Sweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences in Malmö, Diabetes and Cardiovascular Disease, Lund University, Malmö, Sweden
| |
Collapse
|
5
|
Shen L, Zhao H, Xi Y, Wang Z, Deng K, Gou W, Zhang K, Hu W, Tang J, Xu F, Jiang Z, Fu Y, Zhu Y, Zhou D, Chen YM, Zheng JS. Mapping the gut microbial structural variations in healthy aging within the Chinese population. Cell Rep 2024; 43:114968. [PMID: 39520681 DOI: 10.1016/j.celrep.2024.114968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Mapping gut microbial structural variants (SVs) during human aging may provide fundamental knowledge and mechanistic understanding of the gut microbiome's relationship with healthy aging. We characterize gut microbial SVs from 3,230 Chinese participants, identifying key SVs associated with aging, healthy aging, and age-related chronic diseases. Our findings reveal a pattern of copy number loss in aging-related SVs, with 35 core SVs consistently detected. Additionally, eight SVs distinguish healthy from unhealthy aging, regardless of age. Notably, a 3-kbp deletion SV of Bifidobacterium pseudocatenulatum, encoding plant polysaccharide degradation, is regulated by plant-based diet and contributes to healthy aging through bile acid metabolism. Our analysis also connects SVs to age-related diseases, such as chronic kidney disease, via genes in the methionine-homocysteine pathway. This study deepens our understanding of the gut microbiome's role in aging and could inform future efforts to enhance lifespan and healthspan.
Collapse
Affiliation(s)
- Luqi Shen
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Hui Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yue Xi
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhaoping Wang
- Department of Epidemiology & Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kui Deng
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China
| | - Wanglong Gou
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Ke Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China
| | - Wei Hu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jun Tang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China
| | - Fengzhe Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Zengliang Jiang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China
| | - Yuanqing Fu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Ju-Sheng Zheng
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
| |
Collapse
|
6
|
Ross FC, Patangia D, Grimaud G, Lavelle A, Dempsey EM, Ross RP, Stanton C. The interplay between diet and the gut microbiome: implications for health and disease. Nat Rev Microbiol 2024; 22:671-686. [PMID: 39009882 DOI: 10.1038/s41579-024-01068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/17/2024]
Abstract
Diet has a pivotal role in shaping the composition, function and diversity of the gut microbiome, with various diets having a profound impact on the stability, functionality and diversity of the microbial community within our gut. Understanding the profound impact of varied diets on the microbiome is crucial, as it will enable us not only to make well-informed dietary decisions for better metabolic and intestinal health, but also to prevent and slow the onset of specific diet-related diseases that stem from suboptimal diets. In this Review, we explore how geographical location affects the gut microbiome and how different diets shape its composition and function. We examine the mechanisms by which whole dietary regimes, such as the Mediterranean diet, high-fibre diet, plant-based diet, high-protein diet, ketogenic diet and Western diet, influence the gut microbiome. Furthermore, we underscore the need for exhaustive studies to better understand the causal relationship between diet, host and microorganisms for the development of precision nutrition and microbiome-based therapies.
Collapse
Affiliation(s)
- Fiona C Ross
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Dhrati Patangia
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Cork, Ireland
| | - Ghjuvan Grimaud
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Cork, Ireland
| | - Aonghus Lavelle
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Centre, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| |
Collapse
|
7
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
8
|
Jiang Y, Li Y. Nutrition Intervention and Microbiome Modulation in the Management of Breast Cancer. Nutrients 2024; 16:2644. [PMID: 39203781 PMCID: PMC11356826 DOI: 10.3390/nu16162644] [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: 07/18/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Breast cancer (BC) is one of the most common cancers worldwide and a leading cause of cancer-related deaths among women. The escalating incidence of BC underscores the necessity of multi-level treatment. BC is a complex and heterogeneous disease involving many genetic, lifestyle, and environmental factors. Growing evidence suggests that nutrition intervention is an evolving effective prevention and treatment strategy for BC. In addition, the human microbiota, particularly the gut microbiota, is now widely recognized as a significant player contributing to health or disease status. It is also associated with the risk and development of BC. This review will focus on nutrition intervention in BC, including dietary patterns, bioactive compounds, and nutrients that affect BC prevention and therapeutic responses in both animal and human studies. Additionally, this paper examines the impacts of these nutrition interventions on modulating the composition and functionality of the gut microbiome, highlighting the microbiome-mediated mechanisms in BC. The combination treatment of nutrition factors and microbes is also discussed. Insights from this review paper emphasize the necessity of comprehensive BC management that focuses on the nutrition-microbiome axis.
Collapse
Affiliation(s)
| | - Yuanyuan Li
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
9
|
Rauber F, Laura da Costa Louzada M, Chang K, Huybrechts I, Gunter MJ, Monteiro CA, Vamos EP, Levy RB. Implications of food ultra-processing on cardiovascular risk considering plant origin foods: an analysis of the UK Biobank cohort. THE LANCET REGIONAL HEALTH. EUROPE 2024; 43:100948. [PMID: 39210945 PMCID: PMC11360147 DOI: 10.1016/j.lanepe.2024.100948] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 09/04/2024]
Abstract
Background Comprehensive research evidence is lacking on the role of ultra-processed foods (UPF) in the relationship between the consumption of plant-sourced foods and their impact on cardiovascular disease (CVD) outcomes. This study aims to assess CVD risk associated with the dietary contribution of food groups that consider both plant or animal origin and food processing categories, within a large cohort of British adults. Methods Data from the UK Biobank participants (40-69 y) who completed at least two 24-h dietary recalls between 2009 and 2012 (n = 126,842; median follow-up: 9 y), with subsequent data linkage to hospital and mortality records, were used. Food groups were classified as either plant-sourced or non-plant/animal-sourced foods. These groups were further divided into non-UPF and UPF, and expressed as a percentage of total energy intake. Findings Every 10 percentage points increase in plant-sourced non-UPF consumption was associated with a 7% lower risk of CVD (95% CI 0.91-0.95) and a 13% lower risk of CVD mortality (0.80-0.94). Conversely, plant-sourced UPF consumption was associated with a 5% increased risk (1.03-1.07) and a 12% higher mortality (1.05-1.20). The contribution of all UPF was linked to higher CVD risk and mortality, and no evidence for an association between contribution of all plant-sourced foods and CVD incidence and mortality was observed. Interpretation The dietary contribution of plant-sourced non-UPF inversely linked to CVD risk, while plant-sourced UPF contribution showed a positive association. Recognizing the role of food processing is crucial for favourable CVD outcomes, even in plant-sourced diets. Funding World Cancer Research Fund.
Collapse
Affiliation(s)
- Fernanda Rauber
- Department of Preventive Medicine, School of Medicine, University of São Paulo, São Paulo, 01246 903, Brazil
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, 01246-904, Brazil
| | - Maria Laura da Costa Louzada
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, 01246-904, Brazil
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, 01246-904, Brazil
| | - Kiara Chang
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, London, W6 8RP, United Kingdom
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, 69372, France
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, 69372, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, United Kingdom
| | - Carlos Augusto Monteiro
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, 01246-904, Brazil
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, 01246-904, Brazil
| | - Eszter P. Vamos
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, London, W6 8RP, United Kingdom
| | - Renata Bertazzi Levy
- Department of Preventive Medicine, School of Medicine, University of São Paulo, São Paulo, 01246 903, Brazil
- Center for Epidemiological Research in Nutrition and Health, University of São Paulo, São Paulo, 01246-904, Brazil
| |
Collapse
|
10
|
Li Z, Xing J, Ma X, Zhang W, Wang C, Wang Y, Qi X, Liu Y, Jian D, Cheng X, Zhu Y, Shi C, Guo Y, Zhao H, Jiang W, Tang H. An orally administered bacterial membrane protein nanodrug ameliorates doxorubicin cardiotoxicity through alleviating impaired intestinal barrier. Bioact Mater 2024; 37:517-532. [PMID: 38698916 PMCID: PMC11063951 DOI: 10.1016/j.bioactmat.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
The cardiotoxicity caused by Dox chemotherapy represents a significant limitation to its clinical application and is a major cause of late death in patients undergoing chemotherapy. Currently, there are no effective treatments available. Our analysis of 295 clinical samples from 132 chemotherapy patients and 163 individuals undergoing physical examination revealed a strong positive correlation between intestinal barrier injury and the development of cardiotoxicity in chemotherapy patients. We developed a novel orally available and intestinal targeting protein nanodrug by assembling membrane protein Amuc_1100 (obtained from intestinal bacteria Akkermansia muciniphila), fluorinated polyetherimide, and hyaluronic acid. The protein nanodrug demonstrated favorable stability against hydrolysis compared with free Amuc_1100. The in vivo results demonstrated that the protein nanodrug can alleviate Dox-induced cardiac toxicity by improving gut microbiota, increasing the proportion of short-chain fatty acid-producing bacteria from the Lachnospiraceae family, and further enhancing the levels of butyrate and pentanoic acids, ultimately regulating the homeostasis repair of lymphocytes in the spleen and heart. Therefore, we believe that the integrity of the intestinal barrier plays an important role in the development of chemotherapy-induced cardiotoxicity. Protective interventions targeting the intestinal barrier may hold promise as a general clinical treatment regimen for reducing Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Wanjun Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Chuan Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xinkun Qi
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yanhui Liu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaolei Cheng
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yanjie Zhu
- Department of Pathology, Central Hospital of Kaifeng City, Kaifeng, Henan, 475000, China
| | - Chao Shi
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Yongjun Guo
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| |
Collapse
|
11
|
Lee S, Tejesvi MV, Hurskainen E, Aasmets O, Plaza-Díaz J, Franks S, Morin-Papunen L, Tapanainen JS, Ruuska TS, Altmäe S, Org E, Salumets A, Arffman RK, Piltonen TT. Gut bacteriome and mood disorders in women with PCOS. Hum Reprod 2024; 39:1291-1302. [PMID: 38614956 PMCID: PMC11145006 DOI: 10.1093/humrep/deae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/19/2024] [Indexed: 04/15/2024] Open
Abstract
STUDY QUESTION How does the gut bacteriome differ based on mood disorders (MDs) in women with polycystic ovary syndrome (PCOS), and how can the gut bacteriome contribute to the associations between these two conditions? SUMMARY ANSWER Women with PCOS who also have MDs exhibited a distinct gut bacteriome with reduced alpha diversity and a significantly lower abundance of Butyricicoccus compared to women with PCOS but without MDs. WHAT IS KNOWN ALREADY Women with PCOS have a 4- to 5-fold higher risk of having MDs compared to women without PCOS. The gut bacteriome has been suggested to influence the pathophysiology of both PCOS and MDs. STUDY DESIGN, SIZE, DURATION This population-based cohort study was derived from the Northern Finland Birth Cohort 1966 (NFBC1966), which includes all women born in Northern Finland in 1966. Women with PCOS who donated a stool sample at age 46 years (n = 102) and two BMI-matched controls for each case (n = 205), who also responded properly to the MD criteria scales, were included. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 102 women with PCOS and 205 age- and BMI-matched women without PCOS were included. Based on the validated MD criteria, the subjects were categorized into MD or no-MD groups, resulting in the following subgroups: PCOS no-MD (n = 84), PCOS MD (n = 18), control no-MD (n = 180), and control MD (n = 25). Clinical characteristics were assessed at age 31 years and age 46 years, and stool samples were collected from the women at age 46 years, followed by the gut bacteriome analysis using 16 s rRNA sequencing. Alpha diversity was assessed using observed features and Shannon's index, with a focus on genera, and beta diversity was characterized using principal components analysis (PCA) with Bray-Curtis Dissimilarity at the genus level. Associations between the gut bacteriome and PCOS-related clinical features were explored by Spearman's correlation coefficient. A P-value for multiple testing was adjusted with the Benjamini-Hochberg false discovery rate (FDR) method. MAIN RESULTS AND THE ROLE OF CHANCE We observed changes in the gut bacteriome associated with MDs, irrespective of whether the women also had PCOS. Similarly, PCOS MD cases showed a lower alpha diversity (Observed feature, PCOS no-MD, median 272; PCOS MD, median 208, FDR = 0.01; Shannon, PCOS no-MD, median 5.95; PCOS MD, median 5.57, FDR = 0.01) but also a lower abundance of Butyricicoccus (log-fold changeAnalysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC)=-0.90, FDRANCOM-BC=0.04) compared to PCOS no-MD cases. In contrast, in the controls, the gut bacteriome did not differ based on MDs. Furthermore, in the PCOS group, Sutterella showed positive correlations with PCOS-related clinical parameters linked to obesity (BMI, r2=0.31, FDR = 0.01; waist circumference, r2=0.29, FDR = 0.02), glucose metabolism (fasting glucose, r2=0.46, FDR < 0.001; fasting insulin, r2=0.24, FDR = 0.05), and gut barrier integrity (zonulin, r2=0.25, FDR = 0.03). LIMITATIONS, REASONS FOR CAUTION Although this was the first study to assess the link between the gut bacteriome and MDs in PCOS and included the largest PCOS dataset for the gut microbiome analysis, the number of subjects stratified by the presence of MDs was limited when contrasted with previous studies that focused on MDs in a non-selected population. WIDER IMPLICATIONS OF THE FINDINGS The main finding is that gut bacteriome is associated with MDs irrespective of the PCOS status, but PCOS may also modulate further the connection between the gut bacteriome and MDs. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the European Union's Horizon 2020 Research and Innovation Programme under the Marie Sklodowska-Curie Grant Agreement (MATER, No. 813707), the Academy of Finland (project grants 315921, 321763, 336449), the Sigrid Jusélius Foundation, Novo Nordisk Foundation (NNF21OC0070372), grant numbers PID2021-12728OB-100 (Endo-Map) and CNS2022-135999 (ROSY) funded by MCIN/AEI/10.13039/501100011033 and ERFD A Way of Making Europe. The study was also supported by EU QLG1-CT-2000-01643 (EUROBLCS) (E51560), NorFA (731, 20056, 30167), USA/NIH 2000 G DF682 (50945), the Estonian Research Council (PRG1076, PRG1414), EMBO Installation (3573), and Horizon 2020 Innovation Grant (ERIN, No. EU952516). The funders did not participate in any process of the study. We have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- S Lee
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - M V Tejesvi
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Ecology and Genetics, University of Oulu, Oulu, Finland
| | - E Hurskainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - O Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - J Plaza-Díaz
- Faculty of Pharmacy, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - S Franks
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - L Morin-Papunen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - J S Tapanainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR—Cantonal Hospital of and University of Fribourg, Fribourg, Switzerland
| | - T S Ruuska
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - S Altmäe
- Faculty of Pharmacy, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - E Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - A Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| | - R K Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - T T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
12
|
Schorr KA, Agayn V, de Groot LCPGM, Slagboom PE, Beekman M. A plant-based diet index to study the relation between diet and disease risk among adults: a narrative review. J Nutr Health Aging 2024; 28:100272. [PMID: 38815475 DOI: 10.1016/j.jnha.2024.100272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024]
Abstract
Plant-based diets (PBD) may offer various health benefits and contribute to a sustainable way of life, but, if not planned correctly, may also confer risks, e.g., by focusing on plant foods with low nutrient density, such as foods primarily consisting of refined carbohydrates. A plant-based diet index (PDI) differentiating between a healthful, unhealthful, and overall PBD, offers a promising approach to standardize and compare studies and integrate results. In this review we (1) summarize current evidence on the PDI and disease risk of relevance to public health, (2) discuss the methodology of the PDI and how it can be sensibly applied in further studies and (3) indicate areas with a lack of knowledge, such as vulnerable populations. In summary, our amalgamation shows, that adherence to a healthier plant-based diet is associated with an 8-68% lower risk for metabolic risk factors, diabetes, and cardiovascular disease, while adherence to an unhealthier plant-based diet is associated with a 10-63% higher risk. Although differences in calculation methods and underlying diet patterns between populations should be accounted for, the PDI can be a useful tool to assess adherence to different plant-based diet patterns and their association with health outcomes in cohort studies across cultures.
Collapse
Affiliation(s)
- Kerstin A Schorr
- Innoso BV, Den Haag, The Neterhlands; Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | - Marian Beekman
- Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Rostgaard-Hansen AL, Esberg A, Dicksved J, Hansen T, Pelve E, Brunius C, Halkjær J, Tjønneland A, Johansson I, Landberg R. Temporal gut microbiota variability and association with dietary patterns: From the one-year observational Diet, Cancer, and Health - Next Generations MAX study. Am J Clin Nutr 2024; 119:1015-1026. [PMID: 38301827 DOI: 10.1016/j.ajcnut.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Knowledge about the variability of gut microbiota within an individual over time is important to allow meaningful investigations of the gut microbiota in relation to diet and health outcomes in observational studies. Plant-based dietary patterns have been associated with a lower risk of morbidity and mortality and may alter gut microbiota in a favorable direction. OBJECTIVES To assess the gut microbiota variability during one year and investigate the association between adherence to diet indexes and the gut microbiota in a Danish population. METHODS Four hundred forty-four participants were included in the Diet, Cancer, and Health - Next Generations MAX study (DCH-NG MAX). Stool samples collected up to three times during a year were analyzed by 16S ribosomal ribonucleic acid gene sequencing. Diet was obtained by 24-hour dietary recalls. Intraclass correlation coefficient (ICC) was calculated to assess temporal microbial variability based on 214 individuals. Diet indexes (Nordic, Mediterranean, and plant-based diets) and food groups thereof were associated with gut microbiota using linear regression analyses. RESULTS We found that 91 out of 234 genera had an ICC >0.5. We identified three subgroups dominated by Bacteroides, Prevotella 9, and Ruminococcaceae and adherence to diet indexes differed between subgroups. Higher adherence to diet indexes was associated with the relative abundance of 22 genera. Across diet indexes, higher intakes of fruit, vegetables, whole grains/cereals, and nuts were most frequently associated with these genera. CONCLUSIONS In the DCH-NG MAX study, 39% of the genera had an ICC >0.5 over one year, suggesting that these genera could be studied with health outcomes in prospective analyses with acceptable precision. Adherence to the Nordic, Mediterranean, and plant-based diets differed between bacterial subgroups and was associated with a higher abundance of genera with fiber-degrading properties. Fruits, vegetables, whole grains/cereals, and nuts were frequently associated with these genera.
Collapse
Affiliation(s)
- Agnetha L Rostgaard-Hansen
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden; Department of Diet, Cancer, and Health, Danish Cancer Institute, Copenhagen, Denmark.
| | - Anders Esberg
- Department of Odontology, Umeå University, Umeå, Sweden
| | - Johan Dicksved
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Pelve
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Carl Brunius
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Jytte Halkjær
- Department of Diet, Cancer, and Health, Danish Cancer Institute, Copenhagen, Denmark
| | - Anne Tjønneland
- Department of Diet, Cancer, and Health, Danish Cancer Institute, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
14
|
Kashyap Y, Wang ZJ. Gut microbiota dysbiosis alters chronic pain behaviors in a humanized transgenic mouse model of sickle cell disease. Pain 2024; 165:423-439. [PMID: 37733476 PMCID: PMC10843763 DOI: 10.1097/j.pain.0000000000003034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Pain is the most common symptom experienced by patients with sickle cell disease (SCD) throughout their lives and is the main cause of hospitalization. Despite the progress that has been made towards understanding the disease pathophysiology, major gaps remain in the knowledge of SCD pain, the transition to chronic pain, and effective pain management. Recent evidence has demonstrated a vital role of gut microbiota in pathophysiological features of SCD. However, the role of gut microbiota in SCD pain is yet to be explored. We sought to evaluate the compositional differences in the gut microbiota of transgenic mice with SCD and nonsickle control mice and investigate the role of gut microbiota in SCD pain by using antibiotic-mediated gut microbiota depletion and fecal material transplantation (FMT). The antibiotic-mediated gut microbiota depletion did not affect evoked pain but significantly attenuated ongoing spontaneous pain in mice with SCD. Fecal material transplantation from mice with SCD to wild-type mice resulted in tactile allodynia (0.95 ± 0.17 g vs 0.08 ± 0.02 g, von Frey test, P < 0.001), heat hyperalgesia (15.10 ± 0.79 seconds vs 8.68 ± 1.17 seconds, radiant heat, P < 0.01), cold allodynia (2.75 ± 0.26 seconds vs 1.68 ± 0.08 seconds, dry ice test, P < 0.01), and anxiety-like behaviors (Elevated Plus Maze Test, Open Field Test). On the contrary, reshaping gut microbiota of mice with SCD with FMT from WT mice resulted in reduced tactile allodynia (0.05 ± 0.01 g vs 0.25 ± 0.03 g, P < 0.001), heat hyperalgesia (5.89 ± 0.67 seconds vs 12.25 ± 0.76 seconds, P < 0.001), and anxiety-like behaviors. These findings provide insights into the relationship between gut microbiota dysbiosis and pain in SCD, highlighting the importance of gut microbial communities that may serve as potential targets for novel pain interventions.
Collapse
Affiliation(s)
- Yavnika Kashyap
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
| | - Zaijie Jim Wang
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
- Department of Neurology & Rehabilitation, and Sickle Cell Center, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, United States
| |
Collapse
|
15
|
Wang D, Feng Y, Yang M, Sun H, Zhang Q, Wang R, Tong S, Su R, Jin Y, Wang Y, Lu Z, Han L, Sun Y. Variations in the oral microbiome and metabolome of methamphetamine users. mSystems 2024; 9:e0099123. [PMID: 38112416 PMCID: PMC10804968 DOI: 10.1128/msystems.00991-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023] Open
Abstract
Drug addiction can seriously damage human physical and mental health, while detoxification is a long and difficult process. Although studies have reported changes in the oral microbiome of methamphetamine (METH) users, the role that the microbiome plays in the process of drug addiction is still unknown. This study aims to explore the function of the microbiome based on analysis of the variations in the oral microbiome and metabolome of METH users. We performed the 16S rRNA sequencing analysis based on the oral saliva samples collected from 278 METH users and 105 healthy controls (CTL). In addition, the untargeted metabolomic profiling was conducted based on 220 samples. Compared to the CTL group, alpha diversity was reduced in the group of METH users and the relative abundances of Peptostreptococcus and Gemella were significantly increased, while the relative abundances of Campylobacter and Aggregatibacter were significantly decreased. Variations were also detected in oral metabolic pathways, including enhanced tryptophan metabolism, lysine biosynthesis, purine metabolism, and steroid biosynthesis. Conversely, the metabolic pathways of porphyrin metabolism, glutathione metabolism, and pentose phosphate were significantly reduced. It was speculated that four key microbial taxa, i.e., Peptostreptococcus, Gemella, Campylobacter, and Aggregatibacter, could be involved in the toxicity and addiction mechanisms of METH by affecting the above metabolic pathways. It was found that with the increase of drug use years, the content of tryptamine associated with neuropsychiatric disorders was gradually increased. Our study provides novel insights into exploring the toxic damage and addiction mechanisms underlying the METH addiction.IMPORTANCEIt was found that with the increase of drug use years, the content of tryptamine associated with neuropsychiatric disorders gradually increased. The prediction models based on oral microbiome and metabolome could effectively predict the methamphetamine (METH) smoking. Our study provides novel insights into the exploration of the molecular mechanisms regulating the toxic damage and addiction of METH as well as new ideas for early prevention and treatment strategies of METH addiction.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yu Feng
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Min Yang
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haihui Sun
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingchen Zhang
- Department of Orthopedics, Central hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rongrong Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Shuqing Tong
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rui Su
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lihui Han
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yundong Sun
- Department of Microbiology, Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
Gul F, Herrema H, Davids M, Keating C, Nasir A, Ijaz UZ, Javed S. Gut microbial ecology and exposome of a healthy Pakistani cohort. Gut Pathog 2024; 16:5. [PMID: 38254227 PMCID: PMC10801943 DOI: 10.1186/s13099-024-00596-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Pakistan is a multi-ethnic society where there is a disparity between dietary habits, genetic composition, and environmental exposures. The microbial ecology of healthy Pakistani gut in the context of anthropometric, sociodemographic, and dietary patterns holds interest by virtue of it being one of the most populous countries, and also being a Lower Middle Income Country (LMIC). METHODS 16S rRNA profiling of healthy gut microbiome of normo-weight healthy Pakistani individuals from different regions of residence is performed with additional meta-data collected through filled questionnaires. The current health status is then linked to dietary patterns through [Formula: see text] test of independence and Generalized Linear Latent Variable Model (GLLVM) where distribution of individual microbes is regressed against all recorded sources of variability. To identify the core microbiome signature, a dynamic approach is used that considers into account species occupancy as well as consistency across assumed grouping of samples including organization by gender and province of residence. Fitting neutral modeling then revealed core microbiome that is selected by the environment. RESULTS A strong determinant of disparity is by province of residence. It is also established that the male microbiome is better adapted to the local niche than the female microbiome, and that there is microbial taxonomic and functional diversity in different ethnicities, dietary patterns and lifestyle habits. Some microbial genera, such as, Megamonas, Porphyromonas, Haemophilus, Klebsiella and Finegoldia showed significant associations with consumption of pickle, fresh fruits, rice, and cheese. Our analyses suggest current health status being associated with the diet, sleeping patterns, employment status, and the medical history. CONCLUSIONS This study provides a snapshot of the healthy core Pakistani gut microbiome by focusing on the most populous provinces and ethnic groups residing in predominantly urban areas. The study serves a reference dataset for exploring variations in disease status and designing personalized dietary and lifestyle interventions to promote gut health, particularly in LMICs settings.
Collapse
Affiliation(s)
- Farzana Gul
- Department of Biosciences, COMSATS University Islamabad, Islamabad, 45550, Pakistan
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Mark Davids
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Ciara Keating
- School of Biodiversity, One Health & Veterinary Medicine, Graham Kerr Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Arshan Nasir
- Department of Biosciences, COMSATS University Islamabad, Islamabad, 45550, Pakistan
- Moderna, Inc., Cambridge, MA, USA
| | - Umer Zeeshan Ijaz
- Water & Environment Research Group, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK.
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 7BE, UK.
- National University of Ireland, Galway, University Road, Galway, H91 TK33, Ireland.
| | - Sundus Javed
- Department of Biosciences, COMSATS University Islamabad, Islamabad, 45550, Pakistan.
| |
Collapse
|
17
|
Dimina L, Tremblay-Franco M, Deveaux A, Tardivel C, Fouillet H, Polakof S, Martin JC, Mariotti F. Plasma Metabolome Analysis Suggests That L-Arginine Supplementation Affects Microbial Activity Resulting in a Decrease in Trimethylamine N-oxide-A Randomized Controlled Trial in Healthy Overweight Adults with Cardiometabolic Risk Factors. Curr Dev Nutr 2023; 7:102038. [PMID: 38162999 PMCID: PMC10754708 DOI: 10.1016/j.cdnut.2023.102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Background The effects of supplementation with L-arginine (L-arg), the precursor of nitric oxide (NO), on vascular and cardiometabolic health have largely been explored. Whether other mechanisms of the action of L-arg exist remains unknown, as arginine metabolism is complicated. Objective We aimed to characterize the effect of low dose L-arg supplementation on overall human metabolism both in a fasting state and in response to an allostatic stress. Methods In a randomized, double-blind, crossover study, 32 healthy overweight adults (mean age 45 y) with cardiometabolic risk (fasting plasma triglycerides >150 mg/dL; waist circumference >94 cm [male] or >80 cm [female]) were treated with 1.5 g sustained-release L-arg 3 times/d (4.5 g/d) or placebo for 4 wk. On the last day of treatment, volunteers consumed a high-fat meal challenge (900 kcal, 80% as fat, 13% as carbohydrate, and 7% as protein). Plasma was collected at fasting, 2, 4, and 6 h after the challenge, and the metabolome was analyzed by high-resolution liquid chromatography-mass spectrometry. Metabolic profiles were analyzed using linear mixed models-principal component analysis. Results The challenge meal explained most of the changes in the metabolome. The overall effect of L-arg supplementation significantly explained 0.5% of the total variance, irrespective of the response to the challenge meal (P < 0.05). Among the metabolites that explain most of the L-arg effect, we found many amino acids, including branched-chain amino acids, that were decreased by L-arg supplementation. L-arg also decreased trimethylamine N-oxide (TMAO). Other changes suggest that L-arg increased methyl demand. Conclusions Analysis of the effect of 4 wk of L-arg supplementation on the metabolome reveals important effects on methyl balance and gut microbiota activity, such as a decrease in TMAO. Further studies are needed to investigate those mechanisms and the implications of these changes for long-term health.This trial was registered at clinicaltrials.gov as NCT02354794.
Collapse
Affiliation(s)
- Laurianne Dimina
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Marie Tremblay-Franco
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, Toulouse, France
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, Toulouse, France
| | - Ambre Deveaux
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | | | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
| | | | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| |
Collapse
|
18
|
Gou W, Miao Z, Deng K, Zheng JS. Nutri-microbiome epidemiology, an emerging field to disentangle the interplay between nutrition and microbiome for human health. Protein Cell 2023; 14:787-806. [PMID: 37099800 PMCID: PMC10636640 DOI: 10.1093/procel/pwad023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/02/2023] [Indexed: 04/28/2023] Open
Abstract
Diet and nutrition have a substantial impact on the human microbiome, and interact with the microbiome, especially gut microbiome, to modulate various diseases and health status. Microbiome research has also guided the nutrition field to a more integrative direction, becoming an essential component of the rising area of precision nutrition. In this review, we provide a broad insight into the interplay among diet, nutrition, microbiome, and microbial metabolites for their roles in the human health. Among the microbiome epidemiological studies regarding the associations of diet and nutrition with microbiome and its derived metabolites, we summarize those most reliable findings and highlight evidence for the relationships between diet and disease-associated microbiome and its functional readout. Then, the latest advances of the microbiome-based precision nutrition research and multidisciplinary integration are described. Finally, we discuss several outstanding challenges and opportunities in the field of nutri-microbiome epidemiology.
Collapse
Affiliation(s)
- Wanglong Gou
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Research Center for Industries of the Future, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310030, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Zelei Miao
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Research Center for Industries of the Future, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310030, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Kui Deng
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Research Center for Industries of the Future, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310030, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Ju-Sheng Zheng
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Research Center for Industries of the Future, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310030, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
19
|
Lanuza F, Meroño T, Zamora-Ros R, Bondonno NP, Rostgaard-Hansen AL, Sánchez-Pla A, Miro B, Carmona-Pontaque F, Riccardi G, Tjønneland A, Landberg R, Halkjær J, Andres-Lacueva C. Plasma metabolomic profiles of plant-based dietary indices reveal potential pathways for metabolic syndrome associations. Atherosclerosis 2023; 382:117285. [PMID: 37778133 DOI: 10.1016/j.atherosclerosis.2023.117285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND AND AIMS Plant-based dietary patterns have been associated with improved health outcomes. This study aims to describe the metabolomic fingerprints of plant-based diet indices (PDI) and examine their association with metabolic syndrome (MetS) and its components in a Danish population. METHODS The MAX study comprised 676 participants (55% women, aged 18-67 y) from Copenhagen. Sociodemographic and dietary data were collected using questionnaires and three 24-h dietary recalls over one year (at baseline, and at 6 and 12 months). Mean dietary intakes were computed, as well as overall PDI, healthful (hPDI) and unhealthful (uPDI) scores, according to food groups for each plant-based index. Clinical variables were also collected at the same time points in a health examination that included complete blood tests. MetS was defined according to the International Diabetes Federation criteria. Plasma metabolites were measured using a targeted metabolomics approach. Metabolites associated with PDI were selected using random forest models and their relationships with PDIs and MetS were analyzed using generalized linear mixed models. RESULTS The mean prevalence of MetS was 10.8%. High, compared to low, hPDI and uPDI scores were associated with a lower and higher odd of MetS, respectively [odds ratio (95%CI); hPDI: 0.56 (0.43-0.74); uPDI: 1.61 (1.26-2.05)]. Out of 411 quantified plasma metabolites, machine-learning metabolomics fingerprinting revealed 13 metabolites, including food and food-related microbial metabolites, like hypaphorine, indolepropionic acid and lignan-derived enterolactones. These metabolites were associated with all PDIs and were inversely correlated with MetS components (p < 0.05). Furthermore, they had an explainable contribution of 12% and 14% for the association between hPDI or uPDI, respectively, and MetS only among participants with overweight/obesity. CONCLUSIONS Metabolites associated with PDIs were inversely associated with MetS and its components, and may partially explain the effects of plant-based diets on cardiometabolic risk factors.
Collapse
Affiliation(s)
- Fabian Lanuza
- Biomarkers and Nutrimetabolomics Laboratory, Department de Nutrició, Ciències de L'Alimentació i Gastronomia, Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Facultat de Farmàcia i Ciències de L'Alimentació, Universitat de Barcelona (UB), 08028, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Tomas Meroño
- Biomarkers and Nutrimetabolomics Laboratory, Department de Nutrició, Ciències de L'Alimentació i Gastronomia, Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Facultat de Farmàcia i Ciències de L'Alimentació, Universitat de Barcelona (UB), 08028, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, 28029, Spain.
| | - Raul Zamora-Ros
- Biomarkers and Nutrimetabolomics Laboratory, Department de Nutrició, Ciències de L'Alimentació i Gastronomia, Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Facultat de Farmàcia i Ciències de L'Alimentació, Universitat de Barcelona (UB), 08028, Barcelona, Spain; Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
| | - Nicola P Bondonno
- Danish Cancer Society Research Center, Strandboulevarden 49, DK 2100, Copenhagen, Denmark; Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | | | - Alex Sánchez-Pla
- Statistics and Bioinformatics Research Group, Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Berta Miro
- Biomarkers and Nutrimetabolomics Laboratory, Department de Nutrició, Ciències de L'Alimentació i Gastronomia, Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Facultat de Farmàcia i Ciències de L'Alimentació, Universitat de Barcelona (UB), 08028, Barcelona, Spain; Statistics and Bioinformatics Research Group, Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Francesc Carmona-Pontaque
- Statistics and Bioinformatics Research Group, Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Strandboulevarden 49, DK 2100, Copenhagen, Denmark
| | - Rikard Landberg
- Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Jytte Halkjær
- Danish Cancer Society Research Center, Strandboulevarden 49, DK 2100, Copenhagen, Denmark
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department de Nutrició, Ciències de L'Alimentació i Gastronomia, Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), Facultat de Farmàcia i Ciències de L'Alimentació, Universitat de Barcelona (UB), 08028, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, 28029, Spain
| |
Collapse
|
20
|
Aljuraiban GS, Aljazairy EA, Alsahli AS, Sabico S, Al-Musharaf S. Plant-based dietary index in relation to gut microbiota in Arab women. Medicine (Baltimore) 2023; 102:e35262. [PMID: 37747018 PMCID: PMC10519475 DOI: 10.1097/md.0000000000035262] [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: 01/03/2023] [Revised: 07/31/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Plant-based foods may influence gut microbiota profiles and contribute to overall human health. However, not all plant-based diets are nutritionally equivalent. We aimed to assess the association between a plant-based dietary index (PDI), specifically unhealthy PDI and healthy PDI (hPDI), and gut microbial composition and diversity in young women in Saudi Arabia. This observational study included 92 healthy women aged 18 to 25 years. Dietary and anthropometric data were collected. Fecal samples were analyzed using a novel whole-genome shotgun sequencing technique. Alpha and beta diversities measured the richness and composition of the gastrointestinal system. Relationships were examined with Pearson correlation, linear regression, and Wilcoxon Rank-Sum tests. Participants with higher PDI had higher levels of Bacteroides_u_s than those with lower PDI. hPDI was positively correlated with Bifidobacterium pseudocatenulatum, Bifidobacterium longum, Oscillibacter, and Lactobacillus acidophilus and inversely correlated with Clostridioides difficile (P < .05). Unhealthy plant-based dietary index was inversely correlated with B pseudocatenulatum, B longum, and L acidophilus and positively correlated with C difficile (P < .05) and other species of interest. In conclusion, hPDI scores were significantly associated with microbiota species linked with favorable health outcomes, independent of body mass index and gut microbial richness and composition in Arab women. Future studies should investigate the modulating effect of plant-based diets on the species identified in the current study.
Collapse
Affiliation(s)
- Ghadeer S. Aljuraiban
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Esra’a A. Aljazairy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz S. Alsahli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Shaun Sabico
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sara Al-Musharaf
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
21
|
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: 0.5] [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
|
22
|
Zambrano AK, Cadena-Ullauri S, Guevara-Ramírez P, Frias-Toral E, Ruiz-Pozo VA, Paz-Cruz E, Tamayo-Trujillo R, Chapela S, Montalván M, Sarno G, Guerra CV, Simancas-Racines D. The Impact of a Very-Low-Calorie Ketogenic Diet in the Gut Microbiota Composition in Obesity. Nutrients 2023; 15:2728. [PMID: 37375632 DOI: 10.3390/nu15122728] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The very-low-calorie KD (VLCKD) is characterized by a caloric intake of under 800 kcal/day divided into less than 50 g/day of carbohydrate (13%) and 1 to 1.5 g of protein/kg of body weight (44%) and 43% of fat. This low carbohydrate intake changes the energy source from glucose to ketone bodies. Moreover, clinical trials have consistently shown a beneficial effect of VLCKD in several diseases, such as heart failure, schizophrenia, multiple sclerosis, Parkinson's, and obesity, among others. The gut microbiota has been associated with the metabolic conditions of a person and is regulated by diet interactions; furthermore, it has been shown that the microbiota has a role in body weight homeostasis by regulating metabolism, appetite, and energy. Currently, there is increasing evidence of an association between gut microbiota dysbiosis and the pathophysiology of obesity. In addition, the molecular pathways, the role of metabolites, and how microbiota modulation could be beneficial remain unclear, and more research is needed. The objective of the present article is to contribute with an overview of the impact that VLCKD has on the intestinal microbiota composition of individuals with obesity through a literature review describing the latest research regarding the topic and highlighting which bacteria phyla are associated with obesity and VLCKD.
Collapse
Affiliation(s)
- Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Evelyn Frias-Toral
- School of Medicine, Universidad Católica Santiago de Guayaquil, Guayaquil 090615, Ecuador
| | - Viviana A Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador
| | - Sebastián Chapela
- Departamento de Bioquímica, Facultad de Ciencias Médicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1121ABE, Argentina
- Hospital Británico de Buenos Aires, Equipo de Soporte Nutricional, Ciudad Autónoma de Buenos Aires C1280AEB, Argentina
| | - Martha Montalván
- School of Medicine, Universidad Espíritu Santo, Samborondón 091952, Ecuador
| | - Gerardo Sarno
- "San Giovanni di Dio e Ruggi D'Aragona" University Hospital, Scuola Medica Salernitana, 84131 Salerno, Italy
| | - Claudia V Guerra
- Centro de Investigación de Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito 170527, Ecuador
| | - Daniel Simancas-Racines
- Centro de Investigación de Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito 170527, Ecuador
| |
Collapse
|
23
|
Thompson AS, Tresserra-Rimbau A, Karavasiloglou N, Jennings A, Cantwell M, Hill C, Perez-Cornago A, Bondonno NP, Murphy N, Rohrmann S, Cassidy A, Kühn T. Association of Healthful Plant-based Diet Adherence With Risk of Mortality and Major Chronic Diseases Among Adults in the UK. JAMA Netw Open 2023; 6:e234714. [PMID: 36976560 PMCID: PMC10051114 DOI: 10.1001/jamanetworkopen.2023.4714] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/08/2023] [Indexed: 03/29/2023] Open
Abstract
Importance Plant-based diets have gained popularity for both environmental and health reasons, but a comprehensive assessment of their quality in relation to risk of mortality and major chronic diseases is lacking. Objective To examine whether healthful vs unhealthful plant-based dietary patterns are associated with mortality and major chronic diseases among UK adults. Design, Setting, and Participants This prospective cohort study used data from adults in the UK Biobank, a large-scale population-based study. Participants were recruited between 2006 and 2010 and followed up using record linkage data until 2021; follow-up for different outcomes ranged between 10.6 and 12.2 years. Data analysis was conducted from November 2021 to October 2022. Exposures Adherence to a healthful vs unhealthful plant-based diet index (hPDI vs uPDI) derived from 24-hour dietary assessments. Main Outcomes and Measures The main outcomes were hazard ratios (HRs) and 95% CIs of mortality (overall and cause specific), cardiovascular disease (CVD [total, myocardial infarction, ischemic stroke, and hemorrhagic stroke]), cancer (total, breast, prostate, and colorectal), and fracture (total, vertebrae, and hip) across quartiles of hPDI and uPDI adherence. Results This study included 126 394 UK Biobank participants. They had a mean (SD) age of 56.1 (7.8) years; 70 618 (55.9%) were women. The majority of participants (115 371 [91.3%]) were White. Greater adherence to the hPDI was associated with lower risks of total mortality, cancer, and CVD, with HRs (95% CIs) of 0.84 (0.78-0.91), 0.93 (0.88-0.99), and 0.92 (0.86-0.99), respectively, for participants in the highest hPDI quartile compared with the lowest. The hPDI was also associated with lower risks of myocardial infarction and ischemic stroke, with HRs (95% CIs) of 0.86 (0.78-0.95) and 0.84 (0.71-0.99), respectively. By contrast, higher uPDI scores were associated with higher risks of mortality, CVD, and cancer. The associations observed did not show heterogeneity across strata of sex, smoking status, body mass index, or socioeconomic status or with polygenic risk scores (specifically with regard to CVD end points). Conclusions and Relevance The findings of this cohort study of middle-aged UK adults suggest that a diet characterized by high-quality plant-based foods and lower intakes of animal products may be beneficial for health, irrespective of established chronic disease risk factors and genetic predisposition.
Collapse
Affiliation(s)
- Alysha S. Thompson
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
| | - Anna Tresserra-Rimbau
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
- Department of Nutrition, Food Science, and Gastronomy, School of Pharmacy and Food Sciences, Institute for Research on Nutrition and Food Safety, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición, Institute of Health Carlos III, Madrid, Spain
| | - Nena Karavasiloglou
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Amy Jennings
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
| | - Marie Cantwell
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
| | - Claire Hill
- Centre for Public Health, Queen’s University, Belfast, United Kingdom
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Nicola P. Bondonno
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
- Danish Cancer Society Research Centre, Copenhagen, Denmark
- Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Sabine Rohrmann
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Aedín Cassidy
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
| | - Tilman Kühn
- Institute for Global Food Security, School of Biological Sciences, Queen’s University, Belfast, United Kingdom
- Heidelberg Institute of Global Health, Faculty of Medicine and University Hospital, Heidelberg, Germany
| |
Collapse
|
24
|
Zhong GC, Li Z, You AJ, Zhu Q, Wang CR, Yang PF. Plant-based diets and the risk of pancreatic cancer: a large prospective multicenter study. Am J Clin Nutr 2023; 117:235-242. [PMID: 36863825 DOI: 10.1016/j.ajcnut.2022.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Plant-based diets have been recommended for improving health outcomes, including cancer. However, previous studies on plant-based diets and the risk of pancreatic cancer are scarce and fail to consider plant food quality. OBJECTIVES We sought to examine the potential associations of 3 plant-based diet indices (PDIs) with the risk of pancreatic cancer in a US population. METHODS A population-based cohort of 101,748 US adults was identified from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. The overall PDI, healthful PDI (hPDI), and unhealthful PDI (uPDI) were constructed to qualify adherence to overall, healthy, and less healthy plant-based diets, respectively, with higher scores indicating better adherence. Multivariable Cox regression was used to compute hazard ratios (HRs) for pancreatic cancer incidence. Subgroup analysis was conducted to identify the potential effect modifiers. RESULTS Over a mean follow-up of 8.86 years, 421 pancreatic cancer cases occurred. Participants in the highest compared with the lowest quartiles of overall PDI had a lower risk of pancreatic cancer [HRquartile 4 versus 1: 0.74; 95% confidence interval (CI): 0.57, 0.96; Ptrend = 0.023]. A stronger inverse association was observed for hPDI (HRquartile 4 versus 1: 0.56; 95% CI: 0.42, 0.75; Ptrend < 0.001). Conversely, uPDI was positively associated with the risk of pancreatic cancer (HRquartile 4 versus 1: 1.38; 95% CI: 1.02, 1.85; Ptrend = 0.012). Subgroup analyses revealed a stronger positive association for uPDI in participants with BMI <25 (HRquartile 4 versus 1: 3.22; 95% CI: 1.56, 6.65) than in those with BMI ≥25 (HRquartile 4 versus 1: 1.08; 95% CI: 0.78, 1.51) (Pinteraction = 0.001). CONCLUSIONS In this US population, adherence to a healthy plant-based diet confers a lower risk of pancreatic cancer, whereas adherence to a less healthy plant-based diet confers a higher risk. These findings highlight the importance of considering plant food quality in preventing pancreatic cancer.
Collapse
Affiliation(s)
- Guo-Chao Zhong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zhi Li
- Department of Breast and Thyroid Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ai-Jing You
- The Second Student Office, the Second College of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Qian Zhu
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Chun-Rui Wang
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng-Fei Yang
- Department of Nephrology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
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: 16] [Impact Index Per Article: 8.0] [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
|
26
|
Variations in oral responsiveness associate with specific signatures in the gut microbiota and modulate dietary habits. Food Qual Prefer 2022. [DOI: 10.1016/j.foodqual.2022.104790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Ma PJ, Wang MM, Wang Y. Gut microbiota: A new insight into lung diseases. Biomed Pharmacother 2022; 155:113810. [DOI: 10.1016/j.biopha.2022.113810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022] Open
|
28
|
Song Y, Chen K, Lv L, Xiang Y, Du X, Zhang X, Zhao G, Xiao Y. Uncovering the biogeography of the microbial commmunity and its association with nutrient metabolism in the intestinal tract using a pig model. Front Nutr 2022; 9:1003763. [PMID: 36238459 PMCID: PMC9552906 DOI: 10.3389/fnut.2022.1003763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiota is a complex ecosystem that is essential for the metabolism, immunity and health of the host. The gut microbiota also plays a critical role in nutrient absorption and metabolism, and nutrients can influence the growth and composition of the gut microbiota. To gain a better understanding of the relationship between the gut microbial composition and nutrient metabolism, we used a pig model by collecting the contents of the different intestinal locations from six pigs to investigate microbial composition in different intestinal locations based on 16S rRNA gene sequencing and the concentrations of short-chain fatty acids (SCFAs), amino acids, fat, and crude ash in different intestinal locations using gas chromatography and chemical analysis. The results showed that the richness and diversity of intestinal microbial communities gradually increased from the small intestine to the large intestine. The relative abundance of Proteobacteria was higher in the jejunum and ileum, whereas the proportion of Firmicutes was higher in the cecum and colon. The concentrations of SCFAs were higher in the cecum and colon (P < 0.05). The concentrations of amino acids were higher in the small intestine than in the large intestine, while the amino acid content was significantly higher in the ascending colon than in the transverse colon and descending colon. The correlation analysis revealed that Ruminococcaceae UCG-005, Coriobacteriaceae_uncultured, [Eubacterium] hallii group, Mogibacterium and Lachnospiraceae AC2044 group had a higher positive correlation with SCFAs, crude ash and fat but had a negative correlation with amino acids in different gut locations of pigs. These findings may serve as fundamental data for using nutrient metabolism to regulate human and animal gut microbes and health and provide guidance for exploring host-microbe bidirectional interaction mechanisms and driving pathways.
Collapse
Affiliation(s)
- Yuanyuan Song
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, China
| | - Kai Chen
- Quality and Safety of Animal Products Group, Zhejiang Center of Animal Disease Control, Hangzhou, China
| | - Lu Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yun Xiang
- Institute of Animal Husbandry and Veterinary Medicine, Jinhua Academy of Agricultural Sciences, Jinhua, China
| | - Xizhong Du
- Institute of Animal Husbandry and Veterinary Medicine, Jinhua Academy of Agricultural Sciences, Jinhua, China
| | - Xiaojun Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jinhua Academy of Agricultural Sciences, Jinhua, China
| | - Guangmin Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- *Correspondence: Yingping Xiao
| |
Collapse
|