1
|
Delzenne NM, Bindels LB, Neyrinck AM, Walter J. The gut microbiome and dietary fibres: implications in obesity, cardiometabolic diseases and cancer. Nat Rev Microbiol 2025; 23:225-238. [PMID: 39390291 DOI: 10.1038/s41579-024-01108-z] [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: 09/04/2024] [Indexed: 10/12/2024]
Abstract
Dietary fibres constitute a heterogeneous class of nutrients that are key in the prevention of various chronic diseases. Most dietary fibres are fermented by the gut microbiome and may, thereby, modulate the gut microbial ecology and metabolism, impacting human health. Dietary fibres may influence the occurrence of specific bacterial taxa, with this effect varying between individuals. The effect of dietary fibres on microbial diversity is a matter of debate. Most intervention studies with dietary fibres in the context of obesity and related metabolic disorders reveal the need for an accurate assessment of the microbiome to better understand the variable response to dietary fibres. Epidemiological studies confirm that a high dietary fibre intake is strongly associated with a reduced occurrence of many types of cancer. However, there is a need to determine the impact of intervention with specific dietary fibres on cancer risk, therapy efficacy and toxicity, as well as in cancer cachexia. In this Review, we summarize the mechanisms by which the gut microbiome can mediate the physiological benefits of dietary fibres in the contexts of obesity, cardiometabolic diseases and cancer, their incidence being clearly linked to low dietary fibre intake.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
3
|
Costa A, Lucarini E. Treating chronic stress and chronic pain by manipulating gut microbiota with diet: can we kill two birds with one stone? Nutr Neurosci 2025; 28:221-244. [PMID: 38889540 DOI: 10.1080/1028415x.2024.2365021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Background: Chronic stress and chronic pain are closely linked by the capacity to exacerbate each other, sharing common roots in the brain and in the gut. The strict intersection between these two neurological diseases makes important to have a therapeutic strategy aimed at preventing both to maintain mental health in patients. Diet is an modifiable lifestyle factor associated with gut-brain axis diseases and there is growing interest in its use as adjuvant to main therapies. Several evidence attest the impact of specific diets or nutrients on chronic stress-related disorders and pain with a good degree of certainty. A daily adequate intake of foods containing micronutrients such as amino acids, minerals and vitamins, as well as the reduction in the consumption of processed food products can have a positive impact on microbiota and gut health. Many nutrients are endowed of prebiotic, anti-inflammatory, immunomodulatory and neuroprotective potential which make them useful tools helping the management of chronic stress and pain in patients. Dietary regimes, as intermittent fasting or caloric restriction, are promising, although further studies are needed to optimize protocols according to patient's medical history, age and sex. Moreover, by supporting gut microbiota health with diet is possible to attenuate comorbidities such as obesity, gastrointestinal dysfunction and mood disorders, thus reducing healthcare costs related to chronic stress or pain.Objective: This review summarize the most recent evidence on the microbiota-mediated beneficial effects of macro- and micronutrients, dietary-related factors, specific nutritional regimens and dietary intervention on these pathological conditions.
Collapse
Affiliation(s)
- Alessia Costa
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
4
|
Klingbeil EA, Schade R, Lee SH, Kirkland R, de La Serre CB. Manipulation of feeding patterns in high fat diet fed rats improves microbiota composition dynamics, inflammation and gut-brain signaling. Physiol Behav 2024; 285:114643. [PMID: 39059597 DOI: 10.1016/j.physbeh.2024.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Chronic consumption of high fat (HF) diets has been shown to increase meal size and meal frequency in rodents, resulting in overeating. Reducing meal frequency and establishing periods of fasting, independently of caloric intake, may improve obesity-associated metabolic disorders. Additionally, diet-driven changes in microbiota composition have been shown to play a critical role in the development and maintenance of metabolic disorders. In this study, we used a pair-feeding paradigm to reduce meal frequency and snacking episodes while maintaining overall intake and body weight in HF fed rats. We hypothesized that manipulation of feeding patterns would improve microbiota composition and metabolic outcomes. Male Wistar rats were placed in three groups consuming either a HF, low fat diet (LF, matched for sugar), or pair-fed HF diet for 7 weeks (n = 11-12/group). Pair-fed animals received the same amount of food consumed by the HF fed group once daily before dark onset (HF-PF). Rats underwent oral glucose tolerance and gut peptide cholecystokinin sensitivity tests. Bacterial DNA was extracted from the feces collected during both dark and light cycles and sequenced via Illumina MiSeq sequencing of the 16S V4 region. Our pair-feeding paradigm reduced meal numbers, especially small meals in the inactive phase, without changing total caloric intake. This shift in feeding patterns reduced relative abundances of obesity-associated bacteria and maintained circadian fluctuations in microbial abundances. These changes were associated with improved gastrointestinal (GI) function, reduced inflammation, and improved glucose tolerance and gut to brain signaling. We concluded from these data that targeting snacking may help improve metabolic outcomes, independently of energy content of the diet and hyperphagia.
Collapse
Affiliation(s)
- E A Klingbeil
- Department of Nutritional Sciences, The University of Texas at Austin, United States
| | - R Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, United States
| | - S H Lee
- Department of Food Sciences, Sun Moon University, South Korea
| | - R Kirkland
- Office of Research, University of Georgia, United States
| | - C B de La Serre
- Department of Nutritional Sciences, University of Georgia, United States; Department of Biomedical Sciences, Colorado State University, United States.
| |
Collapse
|
5
|
Blais A, Denis I, Andriamihaja M, Gratio V, Champeil-Potokar G, Laouirem S, Chassac A, Couvelard A, Paradis V, Voisin T, Davila AM, Couvineau A. Orexins mitigate obesity-associated dysfunctions in mice. Obesity (Silver Spring) 2024; 32:1897-1909. [PMID: 39315414 DOI: 10.1002/oby.24120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Obesity is a chronic disease that affects more than 400 million adults with severe comorbidities. The search for new treatments to reduce its negative consequences is necessary. Orexins are hypothalamic neuropeptides involved in various physiological processes related to obesity. The aim of this study was to investigate the consequences of chronic orexin-A treatment in mouse models. METHODS Female wild-type C57BL/6 mice that were obesity-prone or obesity-resistant and mice that were deficient for orexin receptors were fed with a high-fat diet. Glucose tolerance, indirect calorimetry, expression of brain neuropeptides and receptors, microglial activation, and microbiota were determined to evaluate the role of orexins on metabolic flexibility. RESULTS Orexin-A reduces weight gain in obesity-prone mice. This reduction is associated with a decrease in body fat, food intake, steatosis, and insulin resistance, as well as alterations of intestinal microbiota composition. A decreased expression of orexin receptors and neuropeptides involved in food intake was also observed in the hypothalamus. CONCLUSIONS Our data support the notion that orexin receptor signaling is involved in different aspects of energy metabolism and can mitigate several dysfunctions associated with obesity, suggesting that orexin receptors can represent new targets for obesity treatment.
Collapse
Affiliation(s)
- Anne Blais
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, Palaiseau, France
| | - Isabelle Denis
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, Palaiseau, France
| | | | - Valérie Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | | | - Samira Laouirem
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | - Anais Chassac
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | - Anne Couvelard
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | - Valérie Paradis
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | - Thierry Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| | | | - Alain Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Paris, France
| |
Collapse
|
6
|
Luecke SM, Aryee G, Holman DB, Schmidt KN, King LE, Crouse MS, Ward AK, Dahlen CR, Caton JS, Amat S. Effects of dietary restriction and one-carbon metabolite supplementation during the first 63 days of gestation on the maternal gut, vaginal, and blood microbiota in cattle. Anim Microbiome 2024; 6:48. [PMID: 39210404 PMCID: PMC11360793 DOI: 10.1186/s42523-024-00335-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Maternal diet quality and quantity have significant impacts on both maternal and fetal health and development. The composition and function of the maternal gut microbiome is also significantly influenced by diet; however, little is known about the impact of gestational nutrient restriction on the bovine maternal microbiome during early gestation, which is a critical stage for maternal microbiome-mediated fetal programming to take place. The objective of the present study was to evaluate the impacts of diet restriction and one-carbon metabolite (OCM) supplementation during early gestation on maternal ruminal, vaginal, and blood microbiota in cattle. Thirty-three beef heifers (approx. 14 months old) were used in a 2 × 2 factorial experiment with main factors of target gain (control [CON]; targeted 0.45 kg/d gain vs restricted [RES]; targeted - 0.23 kg/d gain), and OCM supplementation (+ OCM vs - OCM; n = 8/treatment; except n = 9 for RES-OCM). Heifers were individually fed, starting treatment at breeding (d 0) and concluding at d 63 of gestation. Ruminal fluid and vaginal swabs were collected on d - 2, d 35, and d 63 (at necropsy) and whole blood was collected on d 63 (necropsy). Bacterial microbiota was assessed using 16S rRNA gene (V3-V4) sequencing. RESULTS Overall ruminal microbiota structure was affected by gain, OCM, time, and their interactions. The RES heifers had greater microbial richness (observed ASVs) but neither Shannon nor Inverse Simpson diversity was significantly influenced by gain or OCM supplementation; however, on d 63, 34 bacterial genera showed differential abundance in the ruminal fluid, with 25 genera enriched in RES heifers as compared to CON heifers. In addition, the overall interaction network structure of the ruminal microbiota changed due to diet restriction. The vaginal microbiota community structure was influenced by gain and time. Overall microbial richness and diversity of the vaginal microbiota steadily increased as pregnancy progressed. The vaginal ecological network structure was distinctive between RES and CON heifers with genera-genera interactions being intensified in RES heifers. A relatively diverse bacterial community was detected in blood samples, and the composition of the blood microbiota differed from that of ruminal and vaginal microbiota. CONCLUSION Restricted dietary intake during early gestation induced significant alterations in the ruminal microbiota which also extended to the vaginal microbiota. The composition of these two microbial communities was largely unaffected by OCM supplementation. Blood associated microbiota was largely distinctive from the ruminal and vaginal microbiota.
Collapse
Affiliation(s)
- Sarah M Luecke
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, USA
| | - Godson Aryee
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, USA
| | - Devin B Holman
- Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, AB, Canada
| | - Kaycie N Schmidt
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, USA
| | - Layla E King
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA
| | - Matthew S Crouse
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA
| | - Alison K Ward
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA
| | - Carl R Dahlen
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA
| | - Joel S Caton
- Department of Animal Sciences, and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA
| | - Samat Amat
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
7
|
Ribas-Latre A, Fernández-Veledo S, Vendrell J. Time-restricted eating, the clock ticking behind the scenes. Front Pharmacol 2024; 15:1428601. [PMID: 39175542 PMCID: PMC11338815 DOI: 10.3389/fphar.2024.1428601] [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: 05/06/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Maintaining metabolic balance relies on accumulating nutrients during feeding periods and their subsequent release during fasting. In obesity and metabolic disorders, strategies aimed at reducing food intake while simulating fasting have garnered significant attention for weight loss. Caloric restriction (CR) diets and intermittent fasting (IF) interventions have emerged as effective approaches to improving cardiometabolic health. Although the comparative metabolic benefits of CR versus IF remain inconclusive, this review focuses on various forms of IF, particularly time-restricted eating (TRE). Methods This study employs a narrative review methodology, systematically collecting, synthesizing, and interpreting the existing literature on TRE and its metabolic effects. A comprehensive and unbiased search of relevant databases was conducted to identify pertinent studies, including pre-clinical animal studies and clinical trials in humans. Keywords such as "Obesity," "Intermittent Fasting," "Time-restricted eating," "Chronotype," and "Circadian rhythms" guided the search. The selected studies were critically appraised based on predefined inclusion and exclusion criteria, allowing for a thorough exploration and synthesis of current knowledge. Results This article synthesizes pre-clinical and clinical studies on TRE and its metabolic effects, providing a comprehensive overview of the current knowledge and identifying gaps for future research. It explores the metabolic outcomes of recent clinical trials employing different TRE protocols in individuals with overweight, obesity, or type II diabetes, emphasizing the significance of individual chronotype, which is often overlooked in practice. In contrast to human studies, animal models underscore the role of the circadian clock in mitigating metabolic disturbances induced by obesity through time-restricted feeding (TRF) interventions. Consequently, we examine pre-clinical evidence supporting the interplay between the circadian clock and TRF interventions. Additionally, we provide insights into the role of the microbiota, which TRE can modulate and its influence on circadian rhythms.
Collapse
Affiliation(s)
- Aleix Ribas-Latre
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joan Vendrell
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| |
Collapse
|
8
|
Corriero A, Giglio M, Inchingolo F, Moschetta A, Varrassi G, Puntillo F. Gut Microbiota Modulation and Its Implications on Neuropathic Pain: A Comprehensive Literature Review. Pain Ther 2024; 13:33-51. [PMID: 38087070 PMCID: PMC10796891 DOI: 10.1007/s40122-023-00565-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/13/2023] [Indexed: 01/19/2024] Open
Abstract
Neuropathic pain (NP) is a chronic pain disorder arising from somatosensory nervous system impairment. Extensive evidence supports the notion that the gut microbiota (GM) is crucial in maintaining human health by performing vital tasks. At the same time, its disruption has been linked to the emergence and advancement of an expanding range of disorders, including NP, in which GM could play a role in its pathophysiology. The crosstalk between the nervous system and GM happens through immune mediators, metabolites, and nervous structures and involves both central and peripheral nervous systems. This literature review aims to thoroughly investigate the function of modulating GM in the treatment of NP. It will achieve this by integrating existing knowledge, identifying underlying mechanisms, and evaluating the possible clinical consequences of exploiting the gut-brain axis. We will cover the main therapeutic applications of the described GM-modulators, such as probiotics, faecal microbiota transplantation, dietary supplements and emotional support, to the main kinds of NP in which any evidence, even if only pre-clinical, has been unravelled in recent years. The explored NP areas include chemotherapy-induced peripheral neuropathy, diabetic neuropathy, trauma-induced neuropathic pain, trigeminal neuralgia, postherpetic neuralgia and low back pain.
Collapse
Affiliation(s)
- Alberto Corriero
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy.
| | - Mariateresa Giglio
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine - Dental Medicine Section, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124, Bari, Italy
| | | | - Filomena Puntillo
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
9
|
Minaya DM, Kim JS, Kirkland R, Allen J, Cullinan S, Maclang N, de Lartigue G, de La Serre C. Transfer of microbiota from lean donors in combination with prebiotics prevents excessive weight gain and improves gut-brain vagal signaling in obese rats. Gut Microbes 2024; 16:2421581. [PMID: 39485288 PMCID: PMC11540078 DOI: 10.1080/19490976.2024.2421581] [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: 06/20/2024] [Revised: 10/03/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024] Open
Abstract
Gastrointestinal (GI) microbiota plays an active role in regulating the host's immune system and metabolism, as well as certain pathophysiological processes. Diet is the main factor modulating GI microbiota composition and studies have shown that high fat (HF) diets induce detrimental changes (dysbiosis) in the GI bacterial makeup. HF diet induced dysbiosis has been associated with structural and functional changes in gut-brain vagally mediated signaling system, associated with overeating and obesity. Although HF-driven changes in microbiota composition are sufficient to alter vagal signaling, it is unknown if improving microbiota composition after diet-induced obesity has been established can ameliorate gut-brain signaling and metabolic outcomes. In this study, we evaluated the effect of lean gut microbiota transfer in obese, vagally compromised, rats on gut-brain communication, food intake, and body weight. Male rats were maintained on regular chow or 45% HF diet for nine weeks followed by three weeks of microbiota depletion using antibiotics. The animals were then divided into four groups (n = 10 each): LF - control fed regular chow, LF-LF - chow fed animals that received microbiota from chow fed donors, HF-LF - HF fed animals that received microbiota from chow fed donors, and HF-HF - HF fed animals that received microbiota from HF fed donors. HF-LF animals received inulin as a prebiotic to aid the establishment of the lean microbiome. We found that transferring a LF microbiota to HF fed animals (HF-LF) reduced caloric intake during the light phase when compared with HF-HF rats and prevented additional excessive weight gain. HF-LF animals displayed an increase in postprandial activation of both primary sensory neurons innervating the GI tract and brainstem secondary neurons. We concluded from these data that improving microbiota composition in obese rats is sufficient to ameliorate gut-brain communication and restore normal feeding patterns which was associated with a reduction in weight gain.
Collapse
Affiliation(s)
- Dulce M. Minaya
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Jiyoung S Kim
- Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca Kirkland
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Jillian Allen
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Sitara Cullinan
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Neil Maclang
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | | | - Claire de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
10
|
Jiang J, Liu Y, Wang A, Zhuo Z, Shi H, Zhang X, Li W, Sun M, Jiang S, Wang Y, Zou X, Zhang Y, Jia Z, Xu J. Development and validation of a nutrition-related genetic-clinical-radiological nomogram associated with behavioral and psychological symptoms in Alzheimer's disease. Chin Med J (Engl) 2023:00029330-990000000-00878. [PMID: 38031345 PMCID: PMC11407811 DOI: 10.1097/cm9.0000000000002914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Few evidence is available in the early prediction models of behavioral and psychological symptoms of dementia (BPSD) in Alzheimer's disease (AD). This study aimed to develop and validate a novel genetic-clinical-radiological nomogram for evaluating BPSD in patients with AD and explore its underlying nutritional mechanism. METHODS This retrospective study included 165 patients with AD from the Chinese Imaging, Biomarkers, and Lifestyle (CIBL) cohort between June 1, 2021, and March 31, 2022. Data on demoimagedatas, neuropsychological assessments, single-nucleotide polymorphisms of AD risk genes, and regional brain volumes were collected. A multivariate logistic regression model identified BPSD-associated factors, for subsequently constructing a diagnostic nomogram. This nomogram was internally validated through 1000-bootstrap resampling and externally validated using a time-series split based on the CIBL cohort data between June 1, 2022, and February 1, 2023. Area under receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA) were used to assess the discrimination, calibration, and clinical applicability of the nomogram. RESULTS Factors independently associated with BPSD were: CETP rs1800775 (odds ratio [OR] = 4.137, 95% confidence interval [CI]: 1.276-13.415, P = 0.018), decreased Mini Nutritional Assessment score (OR = 0.187, 95% CI: 0.086-0.405, P <0.001), increased caregiver burden inventory score (OR = 8.993, 95% CI: 3.830-21.119, P <0.001), and decreased brain stem volume (OR = 0.006, 95% CI: 0.001-0.191, P = 0.004). These variables were incorporated into the nomogram. The area under the ROC curve was 0.925 (95% CI: 0.884-0.967, P <0.001) in the internal validation and 0.791 (95% CI: 0.686-0.895, P <0.001) in the external validation. The calibration plots showed favorable consistency between the prediction of nomogram and actual observations, and the DCA showed that the model was clinically useful in both validations. CONCLUSION A novel nomogram was established and validated based on lipid metabolism-related genes, nutritional status, and brain stem volumes, which may allow patients with AD to benefit from early triage and more intensive monitoring of BPSD. REGISTRATION Chictr.org.cn, ChiCTR2100049131.
Collapse
Affiliation(s)
- Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Yaou Liu
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Zhizheng Zhuo
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing 100081, China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Shirui Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Yanli Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
11
|
Ahmad J, Khan I, Zengin G, Mahomoodally MF. The gut microbiome in the fight against obesity: The potential of dietary factors. FASEB J 2023; 37:e23258. [PMID: 37843880 DOI: 10.1096/fj.202300864rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Obesity as a global public health burden has experienced a drastic growing trend recently. The management of obesity is challenging because of its complex etiology, and various factors are involved in its development, such as genetic and environmental factors. Different approaches are available to treat and/or manage obesity, including diet, physical activity, lifestyle changes, medications, and surgery. However, some of these approaches have inherent limitations and are closely associated with adverse effects. Therefore, probing into a novel/safe approach to treat and/or manage obesity is of fundamental importance. One such approach gaining renewed interest is the potential role of gut microbiota in obesity and its effectiveness in treating this condition. However, there is a dearth of comprehensive compilation of data on the potential role of the gut microbiome in obesity, particularly regarding dietary factors as a therapeutic approach. Therefore, this review aims to provide an updated overview of the role of gut microbiota in obesity, further highlighting the importance of dietary factors, particularly diet, prebiotics, and probiotics, as potential complementary and/or alternative therapeutic options. Moreover, the association of gut microbiota with obese or lean individuals has also been discussed.
Collapse
Affiliation(s)
- Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Imran Khan
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Mohamad Fawzi Mahomoodally
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
12
|
Ismael S, Rodrigues C, Santos GM, Castela I, Barreiros-Mota I, Almeida MJ, Calhau C, Faria A, Araújo JR. IPA and its precursors differently modulate the proliferation, differentiation, and integrity of intestinal epithelial cells. Nutr Res Pract 2023; 17:616-630. [PMID: 37529264 PMCID: PMC10375328 DOI: 10.4162/nrp.2023.17.4.616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES Indole-3-propionic acid (IPA) is a tryptophan-derived microbial metabolite that has been associated with protective effects against inflammatory and metabolic diseases. However, there is a lack of knowledge regarding the effects of IPA under physiological conditions and at the intestinal level. MATERIALS/METHODS Human intestinal epithelial Caco-2 cells were treated for 2, 24, and/or 72 h with IPA or its precursors - indole, tryptophan, and propionate - at 1, 10, 100, 250, or 500 μM to assess cell viability, integrity, differentiation, and proliferation. RESULTS IPA induced cell proliferation and this effect was associated with a higher expression of extracellular signal-regulated kinase 2 (ERK2) and a lower expression of c-Jun. Although indole and propionate also induced cell proliferation, this involved ERK2 and c-Jun independent mechanisms. On the other hand, both tryptophan and propionate increased cell integrity and reduced the expression of claudin-1, whereas propionate decreased cell differentiation. CONCLUSIONS In conclusion, these findings suggested that IPA and its precursors distinctly contribute to the proliferation, differentiation, and barrier function properties of human intestinal epithelial cells. Moreover, the pro-proliferative effect of IPA in intestinal epithelial cells was not explained by its precursors and is rather related to its whole chemical structure. Maintaining IPA at physiological levels, e.g., through IPA-producing commensal bacteria, may be important to preserve the integrity of the intestinal barrier and play an integral role in maintaining metabolic homeostasis.
Collapse
Affiliation(s)
- Shámila Ismael
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CINTESIS, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CHRC, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Catarina Rodrigues
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CHRC, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Gilberto Maia Santos
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Inês Castela
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CINTESIS, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CHRC, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Inês Barreiros-Mota
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CHRC, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Maria João Almeida
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Conceição Calhau
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CINTESIS, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- Unidade Universitária Lifestyle Medicine José de Mello Saúde by NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Ana Faria
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CHRC, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - João Ricardo Araújo
- Nutrição e Metabolismo, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- CINTESIS, NOVA Medical School - Faculdade de Ciências Médicas (NMS - FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| |
Collapse
|
13
|
Nguyen NM, Cho J, Lee C. Gut Microbiota and Alzheimer's Disease: How to Study and Apply Their Relationship. Int J Mol Sci 2023; 24:ijms24044047. [PMID: 36835459 PMCID: PMC9958597 DOI: 10.3390/ijms24044047] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Gut microbiota (GM), the microorganisms in the gastrointestinal tract, contribute to the regulation of brain homeostasis through bidirectional communication between the gut and the brain. GM disturbance has been discovered to be related to various neurological disorders, including Alzheimer's disease (AD). Recently, the microbiota-gut-brain axis (MGBA) has emerged as an enticing subject not only to understand AD pathology but also to provide novel therapeutic strategies for AD. In this review, the general concept of the MGBA and its impacts on the development and progression of AD are described. Then, diverse experimental approaches for studying the roles of GM in AD pathogenesis are presented. Finally, the MGBA-based therapeutic strategies for AD are discussed. This review provides concise guidance for those who wish to obtain a conceptual and methodological understanding of the GM and AD relationship with an emphasis on its practical application.
Collapse
|
14
|
Gillespie KM, Kemps E, White MJ, Bartlett SE. The Impact of Free Sugar on Human Health-A Narrative Review. Nutrients 2023; 15:889. [PMID: 36839247 PMCID: PMC9966020 DOI: 10.3390/nu15040889] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The importance of nutrition in human health has been understood for over a century. However, debate is ongoing regarding the role of added and free sugars in physiological and neurological health. In this narrative review, we have addressed several key issues around this debate and the major health conditions previously associated with sugar. We aim to determine the current evidence regarding the role of free sugars in human health, specifically obesity, diabetes, cardiovascular diseases, cognition, and mood. We also present some predominant theories on mechanisms of action. The findings suggest a negative effect of excessive added sugar consumption on human health and wellbeing. Specific class and source of carbohydrate appears to greatly influence the impact of these macronutrients on health. Further research into individual effects of carbohydrate forms in diverse populations is needed to understand the complex relationship between sugar and health.
Collapse
Affiliation(s)
- Kerri M. Gillespie
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Eva Kemps
- College of Education, Psychology and Social Work, Flinders University, Bedford Park, SA 5042, Australia
| | - Melanie J. White
- School of Psychology and Counselling, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Selena E. Bartlett
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
15
|
Schade R, Song L, Cordner ZA, Ding H, Peterson DA, Moran TH, Tamashiro KL, Serre CBDL. Rat offspring's microbiota composition is predominantly shaped by the postnatal maternal diet rather than prenatal diet. Physiol Behav 2023; 258:113987. [PMID: 36198343 PMCID: PMC10088501 DOI: 10.1016/j.physbeh.2022.113987] [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: 06/23/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 01/26/2023]
Abstract
This study assessed the impact of maternal diet during pregnancy versus lactation on offspring gut microbiota. Sprague-Dawley dams were fed high fat (HF) or Chow diets during pregnancy, and their male offspring were raised by a different dam consuming the same or opposite diet (Chow-Chow, Chow-HF, HF-Chow, and HF-HF). Microbiota analysis showed that maternal lactation diet, rather than pregnancy diet, determined offspring microbiota profiles at weaning. Increased abundances of Turicibacter, Staphylococcus , and Ruminococcus were characteristic of chow lactation groups. Lactococcus , Streptococcus , and Parabacteroides were characteristic of HF lactation groups and positively correlated with offspring body weight.
Collapse
Affiliation(s)
- Ruth Schade
- Department of Nutritional Sciences, University of Georgia, Athens, GA, U.S.A; Current address: Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, U.S.A
| | - Lin Song
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zachary A Cordner
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Hua Ding
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Daniel A Peterson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Timothy H Moran
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Kellie L Tamashiro
- Departments of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A.
| | | |
Collapse
|
16
|
Corriero A, Gadaleta RM, Puntillo F, Inchingolo F, Moschetta A, Brienza N. The central role of the gut in intensive care. Crit Care 2022; 26:379. [PMID: 36476497 PMCID: PMC9730662 DOI: 10.1186/s13054-022-04259-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Critically ill patients undergo early impairment of their gut microbiota (GM) due to routine antibiotic therapies and other environmental factors leading to intestinal dysbiosis. The GM establishes connections with the rest of the human body along several axes representing critical inter-organ crosstalks that, once disrupted, play a major role in the pathophysiology of numerous diseases and their complications. Key players in this communication are GM metabolites such as short-chain fatty acids and bile acids, neurotransmitters, hormones, interleukins, and toxins. Intensivists juggle at the crossroad of multiple connections between the intestine and the rest of the body. Harnessing the GM in ICU could improve the management of several challenges, such as infections, traumatic brain injury, heart failure, kidney injury, and liver dysfunction. The study of molecular pathways affected by the GM in different clinical conditions is still at an early stage, and evidence in critically ill patients is lacking. This review aims to describe dysbiosis in critical illness and provide intensivists with a perspective on the potential as adjuvant strategies (e.g., nutrition, probiotics, prebiotics and synbiotics supplementation, adsorbent charcoal, beta-lactamase, and fecal microbiota transplantation) to modulate the GM in ICU patients and attempt to restore eubiosis.
Collapse
Affiliation(s)
- Alberto Corriero
- Department of Interdisciplinary Medicine - ICU Section, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Filomena Puntillo
- Department of Interdisciplinary Medicine - ICU Section, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Francesco Inchingolo
- Dental Medicine Section, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Nicola Brienza
- Department of Interdisciplinary Medicine - ICU Section, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| |
Collapse
|
17
|
Amalia R, Pramono A, Afifah DN, Noer ER, Muniroh M, Kumoro AC. Mangrove fruit ( Bruguiera gymnorhiza) increases circulating GLP-1 and PYY, modulates lipid profiles, and reduces systemic inflammation by improving SCFA levels in obese wistar rats. Heliyon 2022; 8:e10887. [PMID: 36254287 PMCID: PMC9568852 DOI: 10.1016/j.heliyon.2022.e10887] [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: 04/21/2022] [Revised: 07/17/2022] [Accepted: 09/28/2022] [Indexed: 11/26/2022] Open
Abstract
Bruguiera gymnorhiza (BG) has potential as a functional food because of its dietary fibre content and bioactive components such as flavonoids and phenolic compounds. However, it is not studied in the context of diet-related disease prevention. In the present study, we aimed to investigate the effects of Bruguiera gymnorhiza fruit flour (BGF) on satiety hormone, lipid profile, systemic inflammation, body weight, and caecum SCFA levels in diet-induced obese rats. A total of 28 obese male Wistar rats were divided into four groups. Group 1 (K1) was given a standard chow, group 2 (K2) standard chow + orlistat, group 3 (P1) standard chow + BGF 2 g/200 g BW/day, and group 4 (P2) standard chow + BGF 4 g/200 g BW/day for 28 days. The levels of GLP-1, PYY, total cholesterol (TC), triglyceride (TG), HDL, IL-6, TNF-α, and body weight were measured before and after the intervention; meanwhile, the caecum SCFA levels were assessed only after the intervention. In this study, BGF intervention increased the dose-dependent plasma GLP-1 and PYY levels (P < 0.000). In addition, BGF intervention also decreased lipid profiles (TC & TG) (P < 0.000, respectively) and systemic inflammation in a dose-dependent manner. Finally, acetate, propionate, and total SCFA concentrations were higher in the BGF intervention group (P2) compared to the other groups (p < 0.05). The SCFA levels were associated with satiety hormones, lipids, and systemic inflammation (P < 0.05). The BGF intervention improved satiety hormone, lipid profile, systemic inflammation, and SCFA levels.
Collapse
Affiliation(s)
- Rinta Amalia
- Department of Nutrition, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Adriyan Pramono
- Department of Nutrition, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia,Center of Nutrition Research, Universitas Diponegoro, Semarang, Indonesia,Corresponding author.
| | - Diana Nur Afifah
- Department of Nutrition, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia,Center of Nutrition Research, Universitas Diponegoro, Semarang, Indonesia
| | - Etika Ratna Noer
- Department of Nutrition, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia,Center of Nutrition Research, Universitas Diponegoro, Semarang, Indonesia
| | - Muflihatul Muniroh
- Department of Physiology, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Andri Cahyo Kumoro
- Department of Chemical Engineering, Faculty of Engineering, Semarang, Indonesia
| |
Collapse
|
18
|
Montenegro Junior RM, Ponte CMM, Castelo MHCG, de Oliveira Silveira AC, Fernandes VO, D'Alva CB, Oliveira LFV, Hristov AD, Bandeira SP, da Cruz Paiva GE, Levi JE. Reduced gut microbiota diversity in patients with congenital generalized lipodystrophy. Diabetol Metab Syndr 2022; 14:136. [PMID: 36153588 PMCID: PMC9508722 DOI: 10.1186/s13098-022-00908-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Previous studies suggest intestinal dysbiosis is associated with metabolic diseases. However, the causal relationship between them is not fully elucidated. Gut microbiota evaluation of patients with congenital generalized lipodystrophy (CGL), a disease characterized by the absence of subcutaneous adipose tissue, insulin resistance, and diabetes since the first years of life, could provide insights into these relationships. METHODS A cross-sectional study was conducted with patients with CGL (n = 17) and healthy individuals (n = 17). The gut microbiome study was performed by sequencing the 16S rRNA gene through High-Throughput Sequencing (BiomeHub Biotechnologies, Brazil). RESULTS The median age was 20.0 years old, and 64.7% were female. There was no difference between groups in pubertal stage, BMI, ethnicity, origin (rural or urban), delivery, breastfeeding, caloric intake, macronutrient, or fiber consumption. Lipodystrophic patients presented a lower alpha diversity (Richness index: 54.0 versus 67.5; p = 0.008). No differences were observed in the diversity parameters when analyzing the presence of diabetes, its complications, or the CGL subtype. CONCLUSION In this study, we demonstrate for the first time a reduced gut microbiota diversity in individuals with CGL. Dysbiosis was present despite dietary treatment and was also observed in young patients. Our findings allow us to speculate that the loss of intestinal microbiota diversity may be due to metabolic abnormalities present since the first years of life in CGL. Longitudinal studies are needed to confirm these findings, clarifying the possible causal link between dysbiosis and insulin resistance in humans.
Collapse
Affiliation(s)
| | - Clarisse Mourão Melo Ponte
- University Hospitals, Federal University of Ceará/Ebserh, Fortaleza, CE, Brazil.
- DASA, São Paulo, SP, Brazil.
- Christus University Center, CE, Fortaleza, Brazil.
| | - Maria Helane Costa Gurgel Castelo
- University Hospitals, Federal University of Ceará/Ebserh, Fortaleza, CE, Brazil
- DASA, São Paulo, SP, Brazil
- Christus University Center, CE, Fortaleza, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Squizani S, Jantsch J, Rodrigues FDS, Braga MF, Eller S, de Oliveira TF, Silveira AK, Moreira JCF, Giovenardi M, Porawski M, Guedes RP. Zinc Supplementation Partially Decreases the Harmful Effects of a Cafeteria Diet in Rats but Does Not Prevent Intestinal Dysbiosis. Nutrients 2022; 14:3921. [PMID: 36235574 PMCID: PMC9571896 DOI: 10.3390/nu14193921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/21/2022] Open
Abstract
Zinc (Zn) plays an important role in metabolic homeostasis and may modulate neurological impairment related to obesity. The present study aimed to evaluate the effect of Zn supplementation on the intestinal microbiota, fatty acid profile, and neurofunctional parameters in obese male Wistar rats. Rats were fed a cafeteria diet (CAF), composed of ultra-processed and highly caloric and palatable foods, for 20 weeks to induce obesity. From week 16, Zn supplementation was started (10 mg/kg/day). At the end of the experiment, we evaluated the colon morphology, composition of gut microbiota, intestinal fatty acids, integrity of the intestinal barrier and blood-brain barrier (BBB), and neuroplasticity markers in the cerebral cortex and hippocampus. Obese rats showed dysbiosis, morphological changes, short-chain fatty acid (SCFA) reduction, and increased saturated fatty acids in the colon. BBB may also be compromised in CAF-fed animals, as claudin-5 expression is reduced in the cerebral cortex. In addition, synaptophysin was decreased in the hippocampus, which may affect synaptic function. Our findings showed that Zn could not protect obese animals from intestinal dysbiosis. However, an increase in acetate levels was observed, which suggests a partial beneficial effect of Zn. Thus, Zn supplementation may not be sufficient to protect from obesity-related dysfunctions.
Collapse
Affiliation(s)
- Samia Squizani
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Fernanda da Silva Rodrigues
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Matheus Filipe Braga
- Acadêmico do Curso de Biomedicina, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Sarah Eller
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Tiago Franco de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Alexandre Kleber Silveira
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - Marcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Marilene Porawski
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
- Programa de Pós-Graduação em Medicina: Hepatologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| |
Collapse
|
20
|
Dietary carbohydrate-to-protein ratio influences growth performance, hepatic health and dynamic of gut microbiota in atlantic salmon (Salmo salar). ANIMAL NUTRITION 2022; 10:261-279. [PMID: 35785253 PMCID: PMC9234083 DOI: 10.1016/j.aninu.2022.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 01/11/2022] [Accepted: 04/10/2022] [Indexed: 11/24/2022]
Abstract
Atlantic salmon (Salmo salar) fed a carbohydrate-rich diet exhibit suboptimal growth performance, along with other metabolic disturbances. It is well known that gut microbes play a pivotal role in influencing metabolism of the host, and these microbes can be modified by the diet. The main goal of the present study was to determine the effect of feeding graded levels of digestible carbohydrates to Atlantic salmon on the distal intestine digesta microbiota at 3 sampling times (i.e., weeks 4, 8 and 12), during a 12-week trial. A low carbohydrate-to-high protein diet (LC/HP, 0% wheat starch), a medium carbohydrate-to-medium protein diet (MC/MP, 15% wheat starch) or a high carbohydrate-to-low protein diet (HC/LP, 30% wheat starch) was fed to triplicate fish tanks (27 to 28 fish per tank). We performed an in-depth characterization of the distal intestine digesta microbiota. Further, growth parameters, liver histology and the expression of genes involved in hepatic neolipogenesis in fish were measured. Fish fed a HC/LP diet showed greater hepatosomatic and viscerosomatic indexes (P = 0.026 and P = 0.018, respectively), lower final weight (P = 0.005), weight gain (P = 0.003), feed efficiency (P = 0.033) and growth rate (P = 0.003) compared with fish fed the LC/HP diet. Further, feeding salmon a high digestible carbohydrate diet caused greater lipid vacuolization, steatosis index (P = 0.007) and expression of fatty acid synthase (fas) and delta-6 fatty acyl desaturase (d6fad) (P = 0.001 and P = 0.001, respectively) in the liver compared with fish fed the LC/HP diet. Although, the major impact of feeding a carbohydrate-rich diet to Atlantic salmon in beta diversity of distal intestine digesta microbiota was observed at week 4 (HC/LP vs MC/MP and HC/LP vs LC/HP; P = 0.007 and P = 0.008, respectively) and week 8 (HC/LP vs MC/MP; P = 0.04), no differences between experimental groups were detected after 12 weeks of feeding. Finally, at the end of the trial, there was a negative correlation between lactic acid bacteria (LAB) members, including Leuconostoc and Lactobacillus, with hepatic steatosis level, the hepatosomatic and viscerosomatic indexes as well as the expression of fas and d6fad. Weissella showed negative correlation with hepatic steatosis level and the hepatosomatic index. Finally, further research to explore the potential use of LAB as probiotics to improve liver health in carnivorous fish fed fatty liver-induced diet is warranted.
Collapse
|
21
|
Patrakeeva VP, Shtaborov VA. Nutrition and the state of the intestinal microflora in the formation of the metabolic syndrome. OBESITY AND METABOLISM 2022. [DOI: 10.14341/omet12893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The literature review presents the results of modern studies of the relationship between diet and intestinal microbiota in the regulation of metabolic disorders. Metabolic syndrome, which is a symptom complex that combines abdominal obesity, insulin resistance, hyperglycemia, dyslipidemia and arterial hypertension, remains an important problem, being a risk factor for cardiovascular, neurodegenerative, oncological diseases and the development of type 2 diabetes mellitus. Although the pathogenesis of the metabolic syndrome has not yet been fully elucidated, it is known that visceral obesity and its associated complications, such as dyslipidemia and increased levels of pro-inflammatory cytokines, play a central role. The article presents data on the impact of the consumption of certain food products, the inclusion of plant biologically active substances (flavonoids, polyphenols, etc.) in the diet, as well as the use of elimination diets with the exclusion of carbohydrates or fats from the diet, on reducing the risk of cardiovascular accidents, levels of fasting glucose, total cholesterol, LDL, triglycerides, C-reactive protein, leptin, insulin, reduction in body weight and waist circumference, reduction in the level of circulating endotoxins and changes in the activity of immunocompetent cells. Data are presented on the possible influence of the intestinal microbiota in maintaining inflammation and the formation of degenerative changes in the body. The role of changes in the ratio of the levels of pathogenic microflora, bifidobacteria and lactobacilli in the formation of a pathological condition is shown.
Collapse
Affiliation(s)
- V. P. Patrakeeva
- N. Laverov Federal Center for Integrated Arctic Research of the Ural Branch of the Russian Academy of Sciences
| | - V. A. Shtaborov
- N. Laverov Federal Center for Integrated Arctic Research of the Ural Branch of the Russian Academy of Sciences
| |
Collapse
|
22
|
Ben Fradj S, Nédélec E, Salvi J, Fouesnard M, Huillet M, Pallot G, Cansell C, Sanchez C, Philippe C, Gigot V, Lemoine A, Trompier D, Henry T, Petrilli V, Py BF, Guillou H, Loiseau N, Ellero-Simatos S, Nahon JL, Rovère C, Grober J, Boudry G, Douard V, Benani A. Evidence for Constitutive Microbiota-Dependent Short-Term Control of Food Intake in Mice: Is There a Link with Inflammation, Oxidative Stress, Endotoxemia, and GLP-1? Antioxid Redox Signal 2022; 37:349-369. [PMID: 35166124 DOI: 10.1089/ars.2021.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aims: Although prebiotics, probiotics, and fecal transplantation can alter the sensation of hunger and/or feeding behavior, the role of the constitutive gut microbiota in the short-term regulation of food intake during normal physiology is still unclear. Results: An antibiotic-induced microbiota depletion study was designed to compare feeding behavior in conventional and microbiota-depleted mice. Tissues were sampled to characterize the time profile of microbiota-derived signals in mice during consumption of either standard or high-fat food for 1 h. Pharmacological and genetic tools were used to evaluate the contribution of postprandial endotoxemia and inflammatory responses in the short-term regulation of food intake. We observed constitutive microbial and macronutrient-dependent control of food intake at the time scale of a meal; that is, within 1 h of food introduction. Specifically, microbiota depletion increased food intake, and the microbiota-derived anorectic effect became significant during the consumption of high-fat but not standard food. This anorectic effect correlated with a specific postprandial microbial metabolic signature, and did not require postprandial endotoxemia or an NOD-, LRR-, and Pyrin domain-containing protein 3-inflammasome-mediated inflammatory response. Innovation and Conclusion: These findings show that the gut microbiota controls host appetite at the time scale of a meal under normal physiology. Interestingly, a microbiota-derived anorectic effect develops specifically with a high-fat meal, indicating that gut microbiota activity is involved in the satietogenic properties of foods. Antioxid. Redox Signal. 37, 349-369.
Collapse
Affiliation(s)
- Selma Ben Fradj
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Mélanie Fouesnard
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Institut NuMeCan, INRAE (UMR1341), INSERM (UMR1241), Université de Rennes 1, St-Gilles, France
| | - Marine Huillet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Gaëtan Pallot
- Centre de Recherche Lipides, Nutrition, Cancer, INSERM (UMR1231), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Céline Cansell
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Clara Sanchez
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Catherine Philippe
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Vincent Gigot
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Aleth Lemoine
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Inserm (U1111), CNRS (UMR5308), ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Virginie Petrilli
- Centre de Recherche en Cancérologie de Lyon, Inserm (U1052), CNRS (UMR5286), Université de Lyon 1, Lyon, France
| | - Benedicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Inserm (U1111), CNRS (UMR5308), ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Jean-Louis Nahon
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Jacques Grober
- Centre de Recherche Lipides, Nutrition, Cancer, INSERM (UMR1231), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Gaelle Boudry
- Institut NuMeCan, INRAE (UMR1341), INSERM (UMR1241), Université de Rennes 1, St-Gilles, France
| | - Véronique Douard
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
23
|
Neyrinck AM, Rodriguez J, Zhang Z, Nazare JA, Bindels LB, Cani PD, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM. Breath volatile metabolome reveals the impact of dietary fibres on the gut microbiota: Proof of concept in healthy volunteers. EBioMedicine 2022; 80:104051. [PMID: 35561452 PMCID: PMC9108873 DOI: 10.1016/j.ebiom.2022.104051] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 01/06/2023] Open
Abstract
Background Current data suggest that dietary fibre (DF) interaction with the gut microbiota largely contributes to their physiological effects. The bacterial fermentation of DF leads to the production of metabolites, most of them are volatile. This study analyzed the breath volatile metabolites (BVM) profile in healthy individuals (n=15) prior and after a 3-week intervention with chitin-glucan (CG, 4.5 g/day), an insoluble fermentable DF. Methods The present exploratory study presents the original data related to the secondary outcomes, notably the analysis of BVM. BVM were analyzed throughout the test days -in fasting state and after standardized meals - using selected ion flow tube mass spectrometry (SIFT-MS). BVM production was correlated to the gut microbiota composition (Illumina sequencing, primary outcome), analyzed before and after the intervention. Findings The data reveal that the post-prandial state versus fasting state is a key determinant of BVM fingerprint. Correlation analyses with fecal microbiota spotlighted butyrate-producing bacteria, notably Faecalibacterium, as dominant bacteria involved in butyrate and other BVM expiration. CG intervention promotes interindividual variations of fasting BVM, and decreases or delays the expiration of most exhaled BVM in favor of H2 expiration, without any consequence on gastrointestinal tolerance. Interpretation Assessing BVM is a non-invasive methodology allowing to analyze the influence of DF intervention on the gut microbiota. Funding FiberTAG project was initiated from a European Joint Programming Initiative “A Healthy Diet for a Healthy Life” (JPI HDHL) and was supported by the Service Public de Wallonie (SPW-EER, convention 1610365, Belgium).
Collapse
Affiliation(s)
- Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada; College of Food and Biological Engineering, Jimei University, Xiamen, China
| | - Julie-Anne Nazare
- Rhône-Alpes Research Center for Human Nutrition, CarMeN Laboratory, Hospices Civils de Lyon, Université-Lyon, France
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium; WELBIO- Walloon Excellence in Life Sciences and Biotechnology, UCLouvain (Université catholique de Louvain), Brussels, Belgium
| | - Véronique Maquet
- KitoZyme, Parc Industriel des Hauts-Sart, Zone 2, Rue de Milmort 680, Herstal 4040, Belgium
| | - Martine Laville
- Rhône-Alpes Research Center for Human Nutrition, CarMeN Laboratory, Hospices Civils de Lyon, Université-Lyon, France
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, APC Microbiome Ireland, School of Microbiology, University College Cork, Cork, Ireland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium.
| |
Collapse
|
24
|
Liu T, Xu Y, Yi CX, Tong Q, Cai D. The hypothalamus for whole-body physiology: from metabolism to aging. Protein Cell 2022; 13:394-421. [PMID: 33826123 PMCID: PMC9095790 DOI: 10.1007/s13238-021-00834-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 01/05/2023] Open
Abstract
Obesity and aging are two important epidemic factors for metabolic syndrome and many other health issues, which contribute to devastating diseases such as cardiovascular diseases, stroke and cancers. The brain plays a central role in controlling metabolic physiology in that it integrates information from other metabolic organs, sends regulatory projections and orchestrates the whole-body function. Emerging studies suggest that brain dysfunction in sensing various internal cues or processing external cues may have profound effects on metabolic and other physiological functions. This review highlights brain dysfunction linked to genetic mutations, sex, brain inflammation, microbiota, stress as causes for whole-body pathophysiology, arguing brain dysfunction as a root cause for the epidemic of aging and obesity-related disorders. We also speculate key issues that need to be addressed on how to reveal relevant brain dysfunction that underlines the development of these disorders and diseases in order to develop new treatment strategies against these health problems.
Collapse
Affiliation(s)
- Tiemin Liu
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Institute of Metabolism and Integrative Biology, Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yong Xu
- grid.39382.330000 0001 2160 926XChildren’s Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Chun-Xia Yi
- grid.7177.60000000084992262Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Qingchun Tong
- grid.453726.10000 0004 5906 7293Brown Foundation Institute of Molecular Medicine, Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Graduate Program in Neuroscience of MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030 USA
| | - Dongsheng Cai
- grid.251993.50000000121791997Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461 USA
| |
Collapse
|
25
|
Varesi A, Pierella E, Romeo M, Piccini GB, Alfano C, Bjørklund G, Oppong A, Ricevuti G, Esposito C, Chirumbolo S, Pascale A. The Potential Role of Gut Microbiota in Alzheimer’s Disease: from Diagnosis to Treatment. Nutrients 2022; 14:nu14030668. [PMID: 35277027 PMCID: PMC8840394 DOI: 10.3390/nu14030668] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/04/2022] Open
Abstract
Gut microbiota is emerging as a key regulator of many disease conditions and its dysregulation is implicated in the pathogenesis of several gastrointestinal and extraintestinal disorders. More recently, gut microbiome alterations have been linked to neurodegeneration through the increasingly defined gut microbiota brain axis, opening the possibility for new microbiota-based therapeutic options. Although several studies have been conducted to unravel the possible relationship between Alzheimer’s Disease (AD) pathogenesis and progression, the diagnostic and therapeutic potential of approaches aiming at restoring gut microbiota eubiosis remain to be fully addressed. In this narrative review, we briefly summarize the role of gut microbiota homeostasis in brain health and disease, and we present evidence for its dysregulation in AD patients. Based on these observations, we then discuss how dysbiosis might be exploited as a new diagnostic tool in early and advanced disease stages, and we examine the potential of prebiotics, probiotics, fecal microbiota transplantation, and diets as complementary therapeutic interventions on disease pathogenesis and progression, thus offering new insights into the diagnosis and treatment of this devastating and progressive disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy;
- Almo Collegio Borromeo, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (E.P.); (A.O.)
| | - Marcello Romeo
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy;
| | | | - Claudia Alfano
- Department of Emergency Medicine and Surgery, IRCCS Fondazione Policlinico San Matteo, 27100 Pavia, Italy;
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), 8610 Mo i Rana, Norway;
| | - Abigail Oppong
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (E.P.); (A.O.)
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37121 Verona, Italy;
| | - Alessia Pascale
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
26
|
Singh G, Haileselassie Y, Briscoe L, Bai L, Patel A, Sanjines E, Hendler S, Singh PK, Garud NR, Limketkai BN, Habtezion A. The effect of gastric acid suppression on probiotic colonization in a double blinded randomized clinical trial. Clin Nutr ESPEN 2022; 47:70-77. [DOI: 10.1016/j.clnesp.2021.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022]
|
27
|
Rodrigues VST, Moura EG, Peixoto TC, Soares P, Lopes BP, Bertasso IM, Silva BS, Cabral S, Kluck GEG, Atella GC, Trindade PL, Daleprane JB, Oliveira E, Lisboa PC. The model of litter size reduction induces long-term disruption of the gut-brain axis: An explanation for the hyperphagia of Wistar rats of both sexes. Physiol Rep 2022; 10:e15191. [PMID: 35146951 PMCID: PMC8831958 DOI: 10.14814/phy2.15191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/10/2021] [Accepted: 01/04/2022] [Indexed: 04/26/2023] Open
Abstract
The gut microbiota affects the host's metabolic phenotype, impacting health and disease. The gut-brain axis unites the intestine with the centers of hunger and satiety, affecting the eating behavior. Deregulation of this axis can lead to obesity onset. Litter size reduction is a well-studied model for infant obesity because it causes overnutrition and programs for obesity. We hypothesize that animals raised in small litters (SL) have altered circuitry between the intestine and brain, causing hyperphagia. We investigated vagus nerve activity, the expression of c-Fos, brain-derived neurotrophic factor (BDNF), gastrointestinal (GI) hormone receptors, and content of bacterial phyla and short-chain fatty acids (SCFAs) in the feces of adult male and female Wistar rats overfed during lactation. On the 3rd day after birth, litter size was reduced to 3 pups/litter (SL males or SL females) until weaning. Controls had normal litter size (10 pups/litter: 5 males and 5 females). The rats were killed at 5 months of age. The male and female offspring were analyzed separately. The SL group of both sexes showed higher food consumption and body adiposity than the respective controls. SL animals presented dysbiosis (increased Firmicutes, decreased Bacteroidetes) and had increased vagus nerve activity. Only the SL males had decreased hypothalamic GLP-1 receptor expression, while only the SL females had lower acetate and propionate in the feces and higher CCK receptor expression in the hypothalamus. Thus, overfeeding during lactation differentially changes the gut-brain axis, contributing to hyperphagia of the offspring of both sexes.
Collapse
Affiliation(s)
- Vanessa S. T. Rodrigues
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Egberto G. Moura
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Thamara C. Peixoto
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia N. Soares
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Bruna P. Lopes
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Iala M. Bertasso
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Beatriz S. Silva
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - S. S. Cabral
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - G. E. G. Kluck
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - G. C. Atella
- Laboratory of Lipids and Lipoprotein BiochemistryBiochemistry InstituteFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - P. L. Trindade
- Laboratory for studies of Interactions between Nutrition and GeneticsNutrition InstituteRio de Janeiro State UniversityRio de JaneiroBrazil
| | - J. B. Daleprane
- Laboratory for studies of Interactions between Nutrition and GeneticsNutrition InstituteRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Elaine Oliveira
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia Cristina Lisboa
- Laboratory of Endocrine PhysiologyBiology InstituteState University of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
28
|
Rodrigues VST, Moura EG, Peixoto TC, Soares PN, Lopes BP, Oliveira E, Manhães AC, Atella GC, Kluck GEG, Cabral SS, Trindade PL, Daleprane JB, Lisboa PC. Changes in gut-brain axis parameters in adult rats of both sexes with different feeding pattern that were early nicotine-exposed. Food Chem Toxicol 2021; 158:112656. [PMID: 34740714 DOI: 10.1016/j.fct.2021.112656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 01/09/2023]
Abstract
Nicotine is an endocrine disruptor and imprinting factor during breastfeeding that can cause food intake imbalance in the adulthood. As nicotine affects the intestinal microbiota, altering the composition of the bacterial communities and short-chain fatty acids (SCFAs) synthesis in a sex-dependent manner, we hypothesized that nicotine could program the gut-brain axis, consequently modifying the eating pattern of adult male and female rats in a model of maternal nicotine exposure (MNE) during breastfeeding. Lactating Wistar rat dams received minipumps that release 6 mg/kg/day of nicotine (MNE group) or saline for 14 days. The progeny received standard diet from weaning until euthanasia (26 weeks of age). We measured: in vivo electrical activity of the vagus nerve; c-Fos expression in the nucleus tractus solitarius, gastrointestinal peptides receptors, intestinal brain-derived neurotrophic factor (BDNF), SCFAs and microbiota. MNE females showed hyperphagia despite normal adiposity, while MNE males had unchanged food intake, despite obesity. Adult MNE offspring showed decreased Bacteroidetes and increased Firmicutes, Actinobacteria and Proteobacteria. MNE females had lower fecal acetate while MNE males showed higher vagus nerve activity. In summary nicotine exposure through the milk induces long-term intestinal dysbiosis, which may affect eating patterns of adult offspring in a sex-dependent manner.
Collapse
Affiliation(s)
- V S T Rodrigues
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E G Moura
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - T C Peixoto
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - P N Soares
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - B P Lopes
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E Oliveira
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - A C Manhães
- Neurophysiology Laboratory, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - G C Atella
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - G E G Kluck
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - S S Cabral
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - P L Trindade
- Laboratory for Studies of Interactions Between Nutrition and Genetics, Nutrition Institute, Rio de Janeiro State University, RJ, Brazil
| | - J B Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, Nutrition Institute, Rio de Janeiro State University, RJ, Brazil
| | - P C Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
29
|
Varesi A, Deumer US, Ananth S, Ricevuti G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J Clin Med 2021; 10:jcm10215077. [PMID: 34768601 PMCID: PMC8584653 DOI: 10.3390/jcm10215077] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
The well-known symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) are chronic pain, cognitive dysfunction, post-exertional malaise and severe fatigue. Another class of symptoms commonly reported in the context of ME/CFS are gastrointestinal (GI) problems. These may occur due to comorbidities such as Crohn's disease or irritable bowel syndrome (IBS), or as a symptom of ME/CFS itself due to an interruption of the complex interplay between the gut microbiota (GM) and the host GI tract. An altered composition and overall decrease in diversity of GM has been observed in ME/CFS cases compared to controls. In this review, we reflect on genetics, infections, and other influences that may factor into the alterations seen in the GM of ME/CFS individuals, we discuss consequences arising from these changes, and we contemplate the therapeutic potential of treating the gut to alleviate ME/CFS symptoms holistically.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Undine-Sophie Deumer
- Department of Biological Sciences, Faculty of Natural Sciences and Mathematics, University of Cologne, 50674 Cologne, Germany;
| | - Sanjana Ananth
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Giovanni Ricevuti
- Department of Drug Sciences, School of Pharmacy, University of Pavia, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| |
Collapse
|
30
|
Więckowska-Gacek A, Mietelska-Porowska A, Wydrych M, Wojda U. Western diet as a trigger of Alzheimer's disease: From metabolic syndrome and systemic inflammation to neuroinflammation and neurodegeneration. Ageing Res Rev 2021; 70:101397. [PMID: 34214643 DOI: 10.1016/j.arr.2021.101397] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
An excess of saturated fatty acids and simple sugars in the diet is a known environmental risk factor of Alzheimer's disease (AD) but the holistic view of the interacting processes through which such diet may contribute to AD pathogenesis is missing. We addressed this need through extensive analysis of published studies investigating the effects of western diet (WD) on AD development in humans and laboratory animals. We reviewed WD-induced systemic alterations comprising metabolic changes, induction of obesity and adipose tissue inflammation, gut microbiota dysbiosis and acceleration of systemic low-grade inflammation. Next we provide an overview of the evidence demonstrating that WD-associated systemic alterations drive impairment of the blood-brain barrier (BBB) and development of neuroinflammation paralleled by accumulation of toxic amyloid. Later these changes are followed by dysfunction of synaptic transmission, neurodegeneration and finally memory and cognitive impairment. We conclude that WD can trigger AD by acceleration of inflammaging, and that BBB impairment induced by metabolic and systemic inflammation play the central role in this process. Moreover, the concurrence of neuroinflammation and Aβ dyshomeostasis, which by reciprocal interactions drive the vicious cycle of neurodegeneration, contradicts Aβ as the primary trigger of AD. Given that in 2019 the World Health Organization recommended focusing on modifiable risk factors in AD prevention, this overview of the sequential, complex pathomechanisms initiated by WD, which can lead from peripheral disturbances to neurodegeneration, can support future prevention strategies.
Collapse
|
31
|
Romero Marcia AD, Yao T, Chen MH, Oles RE, Lindemann SR. Fine Carbohydrate Structure of Dietary Resistant Glucans Governs the Structure and Function of Human Gut Microbiota. Nutrients 2021; 13:nu13092924. [PMID: 34578800 PMCID: PMC8467459 DOI: 10.3390/nu13092924] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/01/2023] Open
Abstract
Increased dietary fiber consumption has been shown to increase human gut microbial diversity, but the mechanisms driving this effect remain unclear. One possible explanation is that microbes are able to divide metabolic labor in consumption of complex carbohydrates, which are composed of diverse glycosidic linkages that require specific cognate enzymes for degradation. However, as naturally derived fibers vary in both sugar composition and linkage structure, it is challenging to separate out the impact of each of these variables. We hypothesized that fine differences in carbohydrate linkage structure would govern microbial community structure and function independently of variation in glycosyl residue composition. To test this hypothesis, we fermented commercially available soluble resistant glucans, which are uniformly composed of glucose linked in different structural arrangements, in vitro with fecal inocula from each of three individuals. We measured metabolic outputs (pH, gas, and short-chain fatty acid production) and community structure via 16S rRNA amplicon sequencing. We determined that community metabolic outputs from identical glucans were highly individual, emerging from divergent initial microbiome structures. However, specific operational taxonomic units (OTUs) responded similarly in growth responses across individuals’ microbiota, though in context-dependent ways; these data suggested that certain taxa were more efficient in competing for some structures than others. Together, these data support the hypothesis that variation in linkage structure, independent of sugar composition, governs compositional and functional responses of microbiota.
Collapse
Affiliation(s)
- Arianna D. Romero Marcia
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (A.D.R.M.); (T.Y.); (M.-H.C.); (R.E.O.)
| | - Tianming Yao
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (A.D.R.M.); (T.Y.); (M.-H.C.); (R.E.O.)
| | - Ming-Hsu Chen
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (A.D.R.M.); (T.Y.); (M.-H.C.); (R.E.O.)
| | - Renee E. Oles
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (A.D.R.M.); (T.Y.); (M.-H.C.); (R.E.O.)
| | - Stephen R. Lindemann
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (A.D.R.M.); (T.Y.); (M.-H.C.); (R.E.O.)
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-765-494-9207
| |
Collapse
|
32
|
Wang Z, Ma K, Fujino M, Kusano K, Yi SQ, Iwai A, Li XK. The effects of oral administration of Aureobasidium pullulans-cultured fluid containing β-glucan on concanavalin A injected mice. Heliyon 2021; 7:e07277. [PMID: 34195409 PMCID: PMC8233140 DOI: 10.1016/j.heliyon.2021.e07277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/17/2021] [Accepted: 06/07/2021] [Indexed: 01/07/2023] Open
Abstract
A black yeast, Aureobasidium pullulans, extracellularly produces β-(1,3), (1,6)-D-glucan (β-glucan) under certain conditions. The β-glucan is known to be an immunomodulatory agent, and β-glucan enriched A. pullulans cultured fluid (AP-CF) is used in supplements to maintain human health. Concanavalin A (ConA) is a lectin, and when injected it is known to cause T cell mediated autoimmune hepatitis in mice. The present study investigated the effects of oral administration of AP-CF on ConA injection in mice. The results demonstrated that increases in serum alanine transaminase (ALT) levels after ConA injection were significantly suppressed in an AP-CF administered group of mice. To understand the mechanism of the ALT lowering effects of AP-CF, we used Foxp3 (forkhead box P3) knock-in mice which express the green fluorescent protein (GFP) in Foxp3 induced cells, and the effects of AP-CF on the regulatory T cell (Treg) populations were investigated. The results show that the basal level of Foxp3+ Treg populations in peripheral blood lymphocytes, liver infiltrating lymphocytes, and splenocytes was decreased after 7 days of administration of AP-CF. These findings suggest that oral administration of AP-CF suppresses the basal level of inflammation, and that it may be postulated to be involved in the ALT lowering effects of AP-CF.
Collapse
Affiliation(s)
- Zhidan Wang
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Functional Morphology, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Kuai Ma
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | - Shuang-Qin Yi
- Laboratory of Functional Morphology, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Atsushi Iwai
- Aureo Co., Ltd., Chiba, Japan.,Aureo Science Co., Ltd., Hokkaido, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Functional Morphology, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
33
|
Beckmann L, Künstner A, Freschi ML, Huber G, Stölting I, Ibrahim SM, Hirose M, Freitag M, Langan EA, Matschl U, Galuska CE, Fuchs B, Knobloch JK, Busch H, Raasch W. Telmisartan induces a specific gut microbiota signature which may mediate its antiobesity effect. Pharmacol Res 2021; 170:105724. [PMID: 34116209 DOI: 10.1016/j.phrs.2021.105724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/17/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
Telmisartan prevents diet-induced obesity (DIO) in rodents. Given that the precise underlying mechanism is not known, we examined whether a gut-related mechanism might be involved. Sprague-Dawley rats received cafeteria diet (CD) for 3 months to develop DIO and were administered either telmisartan (8 mg/kgbw) or vehicle. In addition, pair-fed (PF) rats received CD adjusted to TEL and control rats (CON) only received chow. Stool samples were analysed by 16 S rRNA gene amplicon sequencing. CD-fed rats became obese while TEL, PF and CON rats remained lean. Alpha diversity analyses indicated that bacterial diversity was similar before the study but changed over time. Beta diversity revealed a time-, CD- and telmisartan-dependent effect. The Firmicutes/Bacteroidetes ratio and the abundance of Blautia, Allobaculum and Parasutterella were higher in DIO and PF than in CON, but not in TEL. Enterotype (ET)-like clustering analyses, Kleinberg's hub network scoring and random forest analyses also indicated that telmisartan induced a specific signature of gut microbiota. In response to stool transfer from telmisartan-pre-treated donor to high-fat fed acceptor mice, body weight gain was slightly attenuated. We attribute the anti-obesity action of telmisartan treatment to diet-independent alterations in gut microbiota as the microbiota from telmisartan-treated, CD-fed rats clearly differed from those of DIO and PF rats. ET-like clustering network, random forest classification and the higher stability in bacterial co-occurrence network analyses indicate that there is more than one indicator species for telmisartan's specific signature, which is further strengthened by the fact that we cannot identify a single indicator species.
Collapse
Affiliation(s)
- Laura Beckmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck, Germany; Institute for Cardiogenetic, University of Lübeck, Germany
| | - Marco L Freschi
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Gianna Huber
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany; CBBM (Center of Brain, Behavior and Metabolism), Germany
| | - Ines Stölting
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Saleh M Ibrahim
- Institute of Experimental Dermatology, University of Lübeck, Germany
| | - Misa Hirose
- Institute of Experimental Dermatology, University of Lübeck, Germany
| | - Miriam Freitag
- Departement of Dermatology, University of Lübeck, Germany
| | - Ewan A Langan
- Departement of Dermatology, University of Lübeck, Germany; Dermatological Sciences, University of Manchester, UK
| | - Urte Matschl
- Department Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Christina E Galuska
- Leibniz Institute for Farm Animal Biology (FBN) Core Facility Metabolomics, Germany
| | - Beate Fuchs
- Leibniz Institute for Farm Animal Biology (FBN) Core Facility Metabolomics, Germany
| | - Johannes K Knobloch
- Clinic of Infectiology and Microbiology, University Clinic Schleswig-Holstein, Campus Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany; Insitute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck, Germany; Institute for Cardiogenetic, University of Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany; CBBM (Center of Brain, Behavior and Metabolism), Germany.
| |
Collapse
|
34
|
Pairing Binge Drinking and a High-Fat Diet in Adolescence Modulates the Inflammatory Effects of Subsequent Alcohol Consumption in Mice. Int J Mol Sci 2021; 22:ijms22105279. [PMID: 34067897 PMCID: PMC8157004 DOI: 10.3390/ijms22105279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/15/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol binge drinking (BD) and poor nutritional habits are two frequent behaviors among many adolescents that alter gut microbiota in a pro-inflammatory direction. Dysbiotic changes in the gut microbiome are observed after alcohol and high-fat diet (HFD) consumption, even before obesity onset. In this study, we investigate the neuroinflammatory response of adolescent BD when combined with a continuous or intermittent HFD and its effects on adult ethanol consumption by using a self-administration (SA) paradigm in mice. The inflammatory biomarkers IL-6 and CX3CL1 were measured in the striatum 24 h after BD, 3 weeks later and after the ethanol (EtOH) SA. Adolescent BD increased alcohol consumption in the oral SA and caused a greater motivation to seek the substance. Likewise, mice with intermittent access to HFD exhibited higher EtOH consumption, while the opposite effect was found in mice with continuous HFD access. Biochemical analyses showed that after BD and three weeks later, striatal levels of IL-6 and CX3CL1 were increased. In addition, in saline-treated mice, CX3CL1 was increased after continuous access to HFD. After oral SA procedure, striatal IL-6 was increased only in animals exposed to BD and HFD. In addition, striatal CX3CL1 levels were increased in all BD- and HFD-exposed groups. Overall, our findings show that adolescent BD and intermittent HFD increase adult alcohol intake and point to neuroinflammation as an important mechanism modulating this interaction.
Collapse
|
35
|
Oluwagbemigun K, O'Donovan AN, Berding K, Lyons K, Alexy U, Schmid M, Clarke G, Stanton C, Cryan J, Nöthlings U. Long-term dietary intake from infancy to late adolescence is associated with gut microbiota composition in young adulthood. Am J Clin Nutr 2021; 113:647-656. [PMID: 33471048 PMCID: PMC7948843 DOI: 10.1093/ajcn/nqaa340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gut microbiota composition as influenced by long-term diet may be associated with the risk of adult chronic diseases. Thus, establishing the relation of long-term diet, particularly starting from early life, with adult microbiota composition would be an important research advance. OBJECTIVE We aimed to investigate the association of long-term intake of energy, carbohydrate, fiber, protein, and fat from infancy to late adolescence with microbiota composition in adulthood. METHODS Within the prospective DOrtmund Nutritional and Anthropometric Longitudinally Designed (DONALD) Study, we sampled stool 1 or 2 times within 1 y from 128 adults (median age: 29 y). Microbiota composition was profiled by 16S ribosomal RNA sequencing. Annual dietary records from age 1 to 18 y were retrieved. We estimated trajectories of energy, energy-adjusted carbohydrate, fiber, protein, and fat intake with multilevel models, producing predicted intake at age 1 y and rates of change in intake. A multivariate, zero-inflated, logistic-normal model was used to model the association between intake trajectories and the composition of 158 genera in single-sampled individuals. Associations found in this model were confirmed in double-sampled individuals using a zero-inflated Beta regression model. RESULTS Adjusting for covariates and temporal differences in microbiota composition, long-term carbohydrate intake was associated with 3 genera. Specifically, carbohydrate intake at age 1 y was negatively associated with Phascolarctobacterium [coefficient = -4.31; false discovery rate (FDR)-adjusted P = 0.006] and positively associated with Dialister (coefficient = 3.06; FDR-adjusted P = 0.003), and the rate of change in carbohydrate intake was positively associated with Desulfovibrio (coefficient = 13.16; FDR-adjusted P = 0.00039). Energy and other macronutrients were not associated with any genus. CONCLUSIONS This work links long-term carbohydrate intake to microbiota composition. Considering the associations of high carbohydrate intake and microbiota composition with some diseases, these findings could inform the development of gut microbiota-targeted dietary recommendations for disease prevention.
Collapse
Affiliation(s)
- Kolade Oluwagbemigun
- Unit of Nutritional Epidemiology, Department of Nutrition and Food Sciences, University of Bonn, Bonn, Germany
| | - Aoife N O'Donovan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Kirsten Berding
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Katriona Lyons
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Ireland
| | - Ute Alexy
- Unit of Nutritional Epidemiology, Department of Nutrition and Food Sciences, University of Bonn, Bonn, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Ireland
| | - John Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ute Nöthlings
- Unit of Nutritional Epidemiology, Department of Nutrition and Food Sciences, University of Bonn, Bonn, Germany
| |
Collapse
|
36
|
|
37
|
Hsu CK, Su SC, Chang LC, Shao SC, Yang KJ, Chen CY, Chen YT, Wu IW. Effects of Low Protein Diet on Modulating Gut Microbiota in Patients with Chronic Kidney Disease: A Systematic Review and Meta-analysis of International Studies. Int J Med Sci 2021; 18:3839-3850. [PMID: 34790060 PMCID: PMC8579282 DOI: 10.7150/ijms.66451] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Although associations between low protein diet (LPD) and changes of gut microbiota have been reported; however, systematic discernment of the effects of LPD on diet-microbiome-host interaction in patients with chronic kidney disease (CKD) is lacking. Methods: We searched PUBMED and EMBASE for articles published on changes of gut microbiota associated with implementation of LPD in CKD patients until July 2021. Independent researchers extracted data and assessed risks of bias. We conducted meta-analyses of combine p-value, mean differences and random effects for gut microbiota and related metabolites. Study heterogeneity was measured by Tau2 and I2 statistic. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Results: Five articles met inclusion criteria. The meta-analyses of gut microbiota exhibited enrichments of Lactobacillaceae (meta-p= 0.010), Bacteroidaceae (meta-p= 0.048) and Streptococcus anginosus (meta-p< 0.001), but revealed depletion of Bacteroides eggerthii (p=0.017) and Roseburia faecis (meta-p=0.019) in LPD patients compared to patients undergoing normal protein diet. The serum IS levels (mean difference: 0.68 ug/mL, 95% CI: -8.38-9.68, p= 0.89) and pCS levels (mean difference: -3.85 ug/mL, 95% CI: -15.49-7.78, p < 0.52) did not change between groups. We did not find significant differences on renal function associated with change of microbiota between groups (eGFR, mean difference: -7.21 mL/min/1.73 m2, 95% CI: -33.2-18.79, p= 0.59; blood urea nitrogen, mean difference: -6.8 mg/dL, 95% CI: -46.42-32.82, p= 0.74). Other clinical (sodium, potassium, phosphate, albumin, fasting sugar, uric acid, total cholesterol, triglycerides, C-reactive protein and hemoglobin) and anthropometric estimates (body mass index, systolic blood pressure and diastolic blood pressure) did not differ between the two groups. Conclusions: This systematic review and meta-analysis suggested that the effects of LPD on the microbiota were observed predominantly at the families and species levels but minimal on microbial diversity or richness. In the absence of global compositional microbiota shifts, the species-level changes appear insufficient to alter metabolic or clinical outputs.
Collapse
Affiliation(s)
- Cheng-Kai Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Lun-Ching Chang
- Department of Mathematical Sciences, Florida Atlantic University, Florida, US
| | - Shih-Chieh Shao
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Kai-Jie Yang
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chun-Yu Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yih-Ting Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - I-Wen Wu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
38
|
Senizza A, Rocchetti G, Mosele JI, Patrone V, Callegari ML, Morelli L, Lucini L. Lignans and Gut Microbiota: An Interplay Revealing Potential Health Implications. Molecules 2020; 25:E5709. [PMID: 33287261 PMCID: PMC7731202 DOI: 10.3390/molecules25235709] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Plant polyphenols are a broad group of bioactive compounds characterized by different chemical and structural properties, low bioavailability, and several in vitro biological activities. Among these compounds, lignans (a non-flavonoid polyphenolic class found in plant foods for human nutrition) have been recently studied as potential modulators of the gut-brain axis. In particular, gut bacterial metabolism is able to convert dietary lignans into therapeutically relevant polyphenols (i.e., enterolignans), such as enterolactone and enterodiol. Enterolignans are characterized by various biologic activities, including tissue-specific estrogen receptor activation, together with anti-inflammatory and apoptotic effects. However, variation in enterolignans production by the gut microbiota is strictly related to both bioaccessibility and bioavailability of lignans through the entire gastrointestinal tract. Therefore, in this review, we summarized the most important dietary source of lignans, exploring the interesting interplay between gut metabolites, gut microbiota, and the so-called gut-brain axis.
Collapse
Affiliation(s)
- Alice Senizza
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Gabriele Rocchetti
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Juana I. Mosele
- Cátedra de Fisicoquímica, Departamento de Química Analítica y Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina;
| | - Vania Patrone
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Maria Luisa Callegari
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Lorenzo Morelli
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Luigi Lucini
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| |
Collapse
|
39
|
Gómez-Gallego C, García-Mantrana I, Martínez-Costa C, Salminen S, Isolauri E, Collado MC. The Microbiota and Malnutrition: Impact of Nutritional Status During Early Life. Annu Rev Nutr 2020; 39:267-290. [PMID: 31433738 DOI: 10.1146/annurev-nutr-082117-051716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
According to the developmental origins of health and disease hypothesis, our health is determined by events experienced in utero and during early infancy. Indeed, both our prenatal and postnatal nutrition conditions have an impact on the initial architecture and activity of our microbiota. Recent evidence has underlined the importance of the composition of the early gut microbiota in relation to malnutrition, whether it be undernutrition or overnutrition, that is, in terms of both stunted and overweight development. It remains unclear how early microbial contact is linked to the risk of disease, as well as whether alterations in the microbiome underlie the pathogenesis of malnutrition or are merely the end result of it, which indicates that thequestion of causality must urgently be answered. This review provides information on the complex interaction between the microbiota and nutrition during the first 1,000 days of life, taking into account the impact of both undernutrition and overnutrition on the microbiota and on infants' health outcomes in the short- and long-term.
Collapse
Affiliation(s)
- Carlos Gómez-Gallego
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, FI-70210 Kuopio, Finland; .,Functional Foods Forum, University of Turku, FI-20520 Turku, Finland;
| | - Izaskun García-Mantrana
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, 46980 Valencia, Spain; ,
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, 46010 Valencia, Spain.,Pediatric Gastroenterology and Nutrition Section, Hospital Clinico Universitario Valencia, INCLIVA,46010 Valencia, Spain;
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, FI-20520 Turku, Finland;
| | - Erika Isolauri
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, FI-20500 Turku, Finland; .,Department of Clinical Sciences, Faculty of Medicine, University of Turku, FI-20014 Turku, Finland
| | - M Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), National Research Council, 46980 Valencia, Spain; , .,Functional Foods Forum, University of Turku, FI-20520 Turku, Finland;
| |
Collapse
|
40
|
Delzenne NM, Rodriguez J, Olivares M, Neyrinck AM. Microbiome response to diet: focus on obesity and related diseases. Rev Endocr Metab Disord 2020; 21:369-380. [PMID: 32691288 DOI: 10.1007/s11154-020-09572-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Numerous studies in humans and animal models describe disturbances of the gut microbial ecosystem associated with adiposity and hallmarks of the metabolic syndrome, including hepatic and cardiovascular diseases. The manipulation of the microbiome, which is largely influenced by the diet, appears as an innovative therapeutic tool to prevent or control obesity and related diseases. This review describes the impact of nutrients on the gut microbiota composition and/or function and when available, the consequences on host physiology. A special emphasis is made on the contribution of bacterial-derived metabolites in the regulation of key gut functions that may explain their systemic effect.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
41
|
Kim JS, Kirkland RA, Lee SH, Cawthon CR, Rzepka KW, Minaya DM, de Lartigue G, Czaja K, de La Serre CB. Gut microbiota composition modulates inflammation and structure of the vagal afferent pathway. Physiol Behav 2020; 225:113082. [PMID: 32682966 DOI: 10.1016/j.physbeh.2020.113082] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
Vagal afferent neurons (VAN), located in the nodose ganglion (NG) innervate the gut and terminate in the nucleus of solitary tract (NTS) in the brainstem. They are the primary sensory neurons integrating gut-derived signals to regulate meal size. Chronic high-fat diet (HFD) consumption impairs vagally mediated satiety, resulting in overfeeding. There is evidence that HFD consumption leads to alterations in both vagal nerve function and structural integrity. HFD also leads to marked gut microbiota dysbiosis; in rodent models, dysbiosis is sufficient to induce weight gain. In this study, we investigated the effect of microbiota dysbiosis on gut-brain vagal innervation independently of diet. To do so, we recolonized microbiota-depleted rats with gastrointestinal (GI) contents isolated from donor animals fed either a HFD (45 or 60% fat) or a low fat diet (LFD, 13% fat). We used two different depletion models while maintaining the animals on LFD: 1) conventionally raised Fischer and Wistar rats that underwent a depletion paradigm using an antibiotic cocktail and 2) germ free (GF) raised Fischer rats. Following recolonization, receiver animals were designated as ConvLF and ConvHF. Fecal samples were collected throughout these studies and analyzed via 16S Illumina sequencing. In both models, bacteria that were identified as characteristic of HFD were successfully transferred to recipient animals. Three weeks post-colonization, ConvHF rats showed significant increases in ionized calcium-binding adapter molecule-1 (Iba1) positive immune cells in the NG compared to ConvLF animals. Additionally, using isolectin B4 (IB4) staining to identify c-fibers, we found that, compared to ConvLF animals, ConvHF rats displayed decreased innervation at the level of the medial NTS; c-fibers at this level are believed to be primarily of vagal origin. This alteration in vagal structure was associated with a loss in satiety induced by the gut peptide cholecystokinin (CCK). Increased presence of immunocompetent Iba1+ cells along the gut-brain axis and alterations in NTS innervation were still evident in ConvHF rats compared to ConvLF animals 12 weeks post-colonization and were associated with increases in food intake and body weight (BW). We conclude from these data that microbiota dysbiosis can alter gut-brain vagal innervation, potentially via recruitment and/or activation of immune cells.
Collapse
Affiliation(s)
- J S Kim
- Dept. of Foods and Nutrition, USA
| | | | - S H Lee
- Dept. of Foods and Nutrition, USA
| | | | - K W Rzepka
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | - D M Minaya
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | - G de Lartigue
- Dept. of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - K Czaja
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
42
|
Li R, Li Y, Li C, Zheng D, Chen P. Gut Microbiota and Endocrine Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1238:143-164. [DOI: 10.1007/978-981-15-2385-4_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Klingbeil EA, Cawthon C, Kirkland R, de La Serre CB. Potato-Resistant Starch Supplementation Improves Microbiota Dysbiosis, Inflammation, and Gut-Brain Signaling in High Fat-Fed Rats. Nutrients 2019; 11:E2710. [PMID: 31717368 PMCID: PMC6893629 DOI: 10.3390/nu11112710] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023] Open
Abstract
(1) High-fat (HF) diet leads to gut microbiota dysbiosis which is associated with systemic inflammation. Bacterial-driven inflammation is sufficient to alter vagally mediated satiety and induce hyperphagia. Promoting bacterial fermentation improves gastrointestinal (GI) epithelial barrier function and reduces inflammation. Resistant starch escape digestion and can be fermented by bacteria in the distal gut. Therefore, we hypothesized that potato RS supplementation in HF-fed rats would lead to compositional changes in microbiota composition associated with improved inflammatory status and vagal signaling. (2) Male Wistar rats (n = 8/group) were fed a low-fat chow (LF, 13% fat), HF (45% fat), or an isocaloric HF supplemented with 12% potato RS (HFRS) diet. (3) The HFRS-fed rats consumed significantly less energy than HF animals throughout the experiment. Systemic inflammation and glucose homeostasis were improved in the HFRS compared to HF rats. Cholecystokinin-induced satiety was abolished in HF-fed rats and restored in HFRS rats. HF feeding led to a significant decrease in positive c fiber staining in the brainstem which was averted by RS supplementation. (4) The RS supplementation prevented dysbiosis and systemic inflammation. Additionally, microbiota manipulation via dietary potato RS prevented HF-diet-induced reorganization of vagal afferent fibers, loss in CCK-induced satiety, and hyperphagia.
Collapse
Affiliation(s)
| | | | | | - Claire B. de La Serre
- Department of Foods and Nutrition, University of Georgia, Athens, GA 30602, USA (C.C.); (R.K.)
| |
Collapse
|
44
|
de Sousa AR, de Castro Moreira ME, Grancieri M, Toledo RCL, de Oliveira Araújo F, Mantovani HC, Queiroz VAV, Martino HSD. Extruded sorghum (Sorghum bicolor L.) improves gut microbiota, reduces inflammation, and oxidative stress in obese rats fed a high-fat diet. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
45
|
Metabolite-Sensing G Protein-Coupled Receptors Connect the Diet-Microbiota-Metabolites Axis to Inflammatory Bowel Disease. Cells 2019; 8:cells8050450. [PMID: 31091682 PMCID: PMC6562883 DOI: 10.3390/cells8050450] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence has indicated that diet and metabolites, including bacteria- and host-derived metabolites, orchestrate host pathophysiology by regulating metabolism, immune system and inflammation. Indeed, autoimmune diseases such as inflammatory bowel disease (IBD) are associated with the modulation of host response to diets. One crucial mechanism by which the microbiota affects the host is signaling through G protein-coupled receptors (GPCRs) termed metabolite-sensing GPCRs. In the gut, both immune and nonimmune cells express GPCRs and their activation generally provide anti-inflammatory signals through regulation of both the immune system functions and the epithelial integrity. Members of GPCR family serve as a link between microbiota, immune system and intestinal epithelium by which all these components crucially participate to maintain the gut homeostasis. Conversely, impaired GPCR signaling is associated with IBD and other diseases, including hepatic steatosis, diabetes, cardiovascular disease, and asthma. In this review, we first outline the signaling, function, expression and the physiological role of several groups of metabolite-sensing GPCRs. We then discuss recent findings on their role in the regulation of the inflammation, their existing endogenous and synthetic ligands and innovative approaches to therapeutically target inflammatory bowel disease.
Collapse
|