1
|
Hossain MS, Wazed MA, Asha S, Hossen MA, Fime SNM, Teeya ST, Jenny LY, Dash D, Shimul IM. Flavor and Well-Being: A Comprehensive Review of Food Choices, Nutrition, and Health Interactions. Food Sci Nutr 2025; 13:e70276. [PMID: 40384991 PMCID: PMC12082435 DOI: 10.1002/fsn3.70276] [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: 10/30/2024] [Revised: 04/22/2025] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
Human beings are naturally drawn to food flavors and pleasant aromas, which not only guide food choices but also contribute to health by promoting the intake of nutritious foods, aiding digestion, and enhancing emotional well-being. This review explores the complex relationship between flavor, nutrition, and health, highlighting that flavor perception can be affected by genetic susceptibility, age, culture, gender, and early life experiences. They influence emotional and physiological responses through brain mechanisms, directly affecting food selection and health outcomes. The use of natural flavors enhances the taste of food and encourages healthier food choices. In contrast, the widespread use of artificial flavors, while often boosting food sales, often leads to the overconsumption of less nutritious products, thereby increasing potential health risks. There is a growing trend among health-conscious consumers that shows a preference for natural and organic flavors, despite challenges such as low bioavailability and limited evidence of their effectiveness. However, advancements in food processing technologies such as microencapsulation and novel extraction methods offer promising tools to improve flavor stability and sensory acceptance, making healthier products more appealing and widely acceptable. In addition, the use of flavor in a strategic manner is most relevant in food reformulation, dietary interventions, and nutrition education, where it can influence consumers to make more health-conscious and sustainable food choices. Subsequent research needs to focus on human trials to optimize flavor delivery techniques and dosages, along with the role of genetic traits and environmental influences on customized flavor perception. Governments across the world need to impose stricter regulations on synthetic additives to ensure safety and safeguard consumer health.
Collapse
Affiliation(s)
- Md Sakhawot Hossain
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Md Abdul Wazed
- School of Nutrition and Public Health, College of Health Oregon State University Corvallis Oregon USA
| | - Sharmin Asha
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Md Alomgir Hossen
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
- College of Food Science Sichuan Agricultural University Yaan China
| | - Sk Nur Muhammad Fime
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Shamiha Tabassum Teeya
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Lubna Yeasmin Jenny
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Diptho Dash
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| | - Islam Md Shimul
- Department of Nutrition and Food Technology Jashore University of Science and Technology Jashore Bangladesh
| |
Collapse
|
2
|
Ribeiro G, Schellekens H, Cuesta-Marti C, Maneschy I, Ismael S, Cuevas-Sierra A, Martínez JA, Silvestre MP, Marques C, Moreira-Rosário A, Faria A, Moreno LA, Calhau C. A menu for microbes: unraveling appetite regulation and weight dynamics through the microbiota-brain connection across the lifespan. Am J Physiol Gastrointest Liver Physiol 2025; 328:G206-G228. [PMID: 39811913 DOI: 10.1152/ajpgi.00227.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Appetite, as the internal drive for food intake, is often dysregulated in a broad spectrum of conditions associated with over- and under-nutrition across the lifespan. Appetite regulation is a complex, integrative process comprising psychological and behavioral events, peripheral and metabolic inputs, and central neurotransmitter and metabolic interactions. The microbiota-gut-brain axis has emerged as a critical mediator of multiple physiological processes, including energy metabolism, brain function, and behavior. Therefore, the role of the microbiota-gut-brain axis in appetite and obesity is receiving increased attention. Omics approaches such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics in appetite and weight regulation offer new opportunities for featuring obesity phenotypes. Furthermore, gut-microbiota-targeted approaches such as pre-, pro-, post-, and synbiotic, personalized nutrition, and fecal microbiota transplantation are novel avenues for precision treatments. The aim of this narrative review is 1) to provide an overview of the role of the microbiota-gut-brain axis in appetite regulation across the lifespan and 2) to discuss the potential of omics and gut microbiota-targeted approaches to deepen understanding of appetite regulation and obesity.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Harriët Schellekens
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Cristina Cuesta-Marti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ivie Maneschy
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Shámila Ismael
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Amanda Cuevas-Sierra
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - J Alfredo Martínez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - Marta P Silvestre
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cláudia Marques
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - André Moreira-Rosário
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Faria
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Luis A Moreno
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Conceição Calhau
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
3
|
Vartanian M, Endres KJ, Lee YT, Friedrich S, Meemken MT, Schamarek I, Rohde-Zimmermann K, Schürfeld R, Eisenberg L, Hilbert A, Beyer F, Stumvoll M, Sacher J, Villringer A, Christensen JF, Witte AV. Investigating the impact of microbiome-changing interventions on food decision-making: MIFOOD study protocol. BMC Nutr 2025; 11:8. [PMID: 39806493 PMCID: PMC11727427 DOI: 10.1186/s40795-024-00971-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Obesity is a multifactorial disease reaching pandemic proportions with increasing healthcare costs, advocating the development of better prevention and treatment strategies. Previous research indicates that the gut microbiome plays an important role in metabolic, hormonal, and neuronal cross-talk underlying eating behavior. We therefore aim to examine the effects of prebiotic and neurocognitive behavioral interventions on food decision-making and to assay the underlying mechanisms in a Randomized Controlled Trial (RCT). METHOD This study uses a parallel arm RCT design with a 26-week intervention period. We plan to enroll 90 participants (male/diverse/female) living with overweight or obesity, defined as either a Waist-to-Hip Ratio (WHR) ≥ 0.9 (male)/0.85 (diverse, female) or a Body Mass Index (BMI) ≥ 25 kg/m2. Key inclusion criteria are 18-60 years of age and exclusion criteria are type 2 diabetes, psychiatric disease, and Magnetic Resonance Imaging (MRI) contraindications. The interventions comprise either a daily supplementary intake of 30 g soluble fiber (inulin), or weekly neurocognitive behavioral group sessions, compared to placebo (equicaloric maltodextrin). At baseline and follow-up, food decision-making is assessed utilizing task-based MRI. Secondary outcome measures include structural MRI, eating habits, lifestyle factors, personality traits, and mood. Further, we obtain fecal and blood samples to investigate gut microbiome composition and related metabolites. DISCUSSION This study relies on expanding research suggesting that dietary prebiotics could improve gut microbiome composition, leading to beneficial effects on gut-brain signaling and higher-order cognitive functions. In parallel, neurocognitive behavioral interventions have been proposed to improve unhealthy eating habits and metabolic status. However, causal evidence on how these "bottom-up" and "top-down" processes affect food decision-making and neuronal correlates in humans is still scarce. In addition, microbiome, and gut-brain-axis-related mediating mechanisms remain unclear. The present study proposes a comprehensive approach to assess the effects of these gut-brain-related processes influencing food decision-making in overweight and obesity. TRIAL REGISTRATION ClinicalTrials.gov NCT05353504. Retrospectively registered on 29 April 2022.
Collapse
Affiliation(s)
- Meghedi Vartanian
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Konrad Jakob Endres
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Yee Teng Lee
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Silke Friedrich
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Marie-Theres Meemken
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Imke Schamarek
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University of Leipzig and the University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Rohde-Zimmermann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University of Leipzig and the University Hospital Leipzig, Leipzig, Germany
| | - Robin Schürfeld
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Lina Eisenberg
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Anja Hilbert
- Integrated Research and Treatment Center AdiposityDiseases, Behavioral Medicine Research Unit, Department of Psychosomatic Medicine and Psychotherapy, University of Leipzig Medical Center, Leipzig, Germany
| | - Frauke Beyer
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine III, Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University of Leipzig and the University Hospital Leipzig, Leipzig, Germany
| | - Julia Sacher
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Center for Mental Health, Helios Park Clinic, Leipzig, Germany
| | - Arno Villringer
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Julia F Christensen
- Department of Cognitive Neuropsychology, Max Planck Institute for Empirical Aesthetics, Frankfurt/M, Germany
| | - A Veronica Witte
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany.
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
4
|
Choe JY, Jones HP. Methods for Modeling Early Life Stress in Rodents. Methods Mol Biol 2025; 2868:205-219. [PMID: 39546232 DOI: 10.1007/978-1-0716-4200-9_11] [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: 11/17/2024]
Abstract
Animal models of early life stress/adversity (ELS) have provided a foundation from which our understanding of the psychoneuroimmunology of childhood trauma has expanded over recent decades. Rodent models are a cornerstone of the ELS literature with many studies utilizing paradigms based on early life separation/deprivation protocols and manipulating the cage environment. However, no animal model is perfect. In particular, the lack of standardization across ELS models has led to inconsistent results and raised questions regarding the translational value of common preclinical models. In this chapter, we present an overview of the history of ELS rodent models and discuss considerations relevant to the ongoing efforts to both improve existing models and generate novel paradigms to meet the evolving needs of molecular- and mechanism-based ELS research.
Collapse
Affiliation(s)
- Jamie Y Choe
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Harlan P Jones
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA.
- Institute for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
5
|
Huwart SJP, Fayt C, Gangarossa G, Luquet S, Cani PD, Everard A. TLR4-dependent neuroinflammation mediates LPS-driven food-reward alterations during high-fat exposure. J Neuroinflammation 2024; 21:305. [PMID: 39580436 PMCID: PMC11585241 DOI: 10.1186/s12974-024-03297-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Obesity has become a global pandemic, marked by significant shifts in both the homeostatic and hedonic/reward aspects of food consumption. While the precise causes are still under investigation, recent studies have identified the role of gut microbes in dysregulating the reward system within the context of obesity. Unravelling these gut-brain connections is crucial for developing effective interventions against eating and metabolic disorders, particularly in the context of obesity. This study explores the causal role of LPS, as a key relay of microbiota component-induced neuroinflammation in the dysregulation of the reward system following exposure to high-fat diet (HFD). METHODS Through a series of behavioural paradigms related to food-reward events and the use of pharmacological agents targeting the dopamine circuit, we investigated the mechanisms associated with the development of reward dysregulation during HFD-feeding in male mice. A Toll-like receptor 4 (TLR4) full knockout model and intraventricular lipopolysaccharide (LPS) diffusion at low doses, which mimics the obesity-associated neuroinflammatory phenotype, were used to investigate the causal roles of gut microbiota-derived components in neuroinflammation and reward dysregulation. RESULTS Our study revealed that short term exposure to HFD (24 h) tended to affect food-seeking behaviour, and this effect became significant after 1 week of HFD. Moreover, we found that deletion of TLR4 induced a partial protection against HFD-induced neuroinflammation and reward dysregulation. Finally, chronic brain diffusion of LPS recapitulated, at least in part, HFD-induced molecular and behavioural dysfunctions within the reward system. CONCLUSIONS These findings highlight a link between the neuroinflammatory processes triggered by the gut microbiota components LPS and the dysregulation of the reward system during HFD-induced obesity through the TLR4 pathway, thus paving the way for future therapeutic approaches.
Collapse
Affiliation(s)
- Sabrina J P Huwart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
| | - Clémence Fayt
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, F-75013, France
- Institut Universitaire de France (IUF), Paris, France
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, F-75013, France
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium.
| |
Collapse
|
6
|
Van Hul M, Neyrinck AM, Everard A, Abot A, Bindels LB, Delzenne NM, Knauf C, Cani PD. Role of the intestinal microbiota in contributing to weight disorders and associated comorbidities. Clin Microbiol Rev 2024; 37:e0004523. [PMID: 38940505 PMCID: PMC11391702 DOI: 10.1128/cmr.00045-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
SUMMARYThe gut microbiota is a major factor contributing to the regulation of energy homeostasis and has been linked to both excessive body weight and accumulation of fat mass (i.e., overweight, obesity) or body weight loss, weakness, muscle atrophy, and fat depletion (i.e., cachexia). These syndromes are characterized by multiple metabolic dysfunctions including abnormal regulation of food reward and intake, energy storage, and low-grade inflammation. Given the increasing worldwide prevalence of obesity, cachexia, and associated metabolic disorders, novel therapeutic strategies are needed. Among the different mechanisms explaining how the gut microbiota is capable of influencing host metabolism and energy balance, numerous studies have investigated the complex interactions existing between nutrition, gut microbes, and their metabolites. In this review, we discuss how gut microbes and different microbiota-derived metabolites regulate host metabolism. We describe the role of the gut barrier function in the onset of inflammation in this context. We explore the importance of the gut-to-brain axis in the regulation of energy homeostasis and glucose metabolism but also the key role played by the liver. Finally, we present specific key examples of how using targeted approaches such as prebiotics and probiotics might affect specific metabolites, their signaling pathways, and their interactions with the host and reflect on the challenges to move from bench to bedside.
Collapse
Affiliation(s)
- Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
| | - Audrey M Neyrinck
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Amandine Everard
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | | | - Laure B Bindels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Nathalie M Delzenne
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Toulouse, France
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium
| |
Collapse
|
7
|
Ferrario CR, Münzberg-Gruening H, Rinaman L, Betley JN, Borgland SL, Dus M, Fadool DA, Medler KF, Morton GJ, Sandoval DA, de La Serre CB, Stanley SA, Townsend KL, Watts AG, Maruvada P, Cummings D, Cooke BM. Obesity- and diet-induced plasticity in systems that control eating and energy balance. Obesity (Silver Spring) 2024; 32:1425-1440. [PMID: 39010249 PMCID: PMC11269035 DOI: 10.1002/oby.24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024]
Abstract
In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.
Collapse
Grants
- P30 DK048520 NIDDK NIH HHS
- NSF1949989 National Science Foundation
- T32 DC000044 NIDCD NIH HHS
- R01 DK089056 NIDDK NIH HHS
- R01 DK124801 NIDDK NIH HHS
- R01 DK100685 NIDDK NIH HHS
- R01 DK130875 NIDDK NIH HHS
- R01 DK133464 NIDDK NIH HHS
- R01 DK125890 NIDDK NIH HHS
- Z99 DK999999 Intramural NIH HHS
- R01 DK124461 NIDDK NIH HHS
- K26 DK138368 NIDDK NIH HHS
- R01 DK121995 NIDDK NIH HHS
- R01 DK121531 NIDDK NIH HHS
- P30 DK089503 NIDDK NIH HHS
- P01 DK119130 NIDDK NIH HHS
- R01 DK118910 NIDDK NIH HHS
- R01 AT011683 NCCIH NIH HHS
- Reported research was supported by DK130246, DK092587, AT011683, MH059911, DK100685, DK119130, DK124801, DK133399, AG079877, DK133464, T32DC000044, F31DC016817, NSF1949989, DK089056, DK124238, DK138368, DK121995, DK125890, DK118910, DK121531, DK124461, DK130875; Canada Research Chair: 950-232211, CIHRFDN148473, CIHRPJT185886; USDA Predoctoral Fellowship; Endowment from the Robinson Family and Tallahassee Memorial Hospital; Department of Defense W81XWH-20-1-0345 and HT9425-23-1-0244; American Diabetes Association #1-17-ACE-31; W.M. Keck Foundation Award; National Science Foundation CAREER 1941822
- R01 DK133399 NIDDK NIH HHS
- HT9425-23-1-0244 Department of Defense
- R01 DK092587 NIDDK NIH HHS
- W81XWH-20-1-0345 Department of Defense
- 1941822 National Science Foundation
- R01 MH059911 NIMH NIH HHS
- F31 DC016817 NIDCD NIH HHS
- R01 AG079877 NIA NIH HHS
- R01 DK130246 NIDDK NIH HHS
- P30 DK017047 NIDDK NIH HHS
- R01 DK124238 NIDDK NIH HHS
Collapse
Affiliation(s)
- Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Heike Münzberg-Gruening
- Laboratory of Central Leptin Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Debra A Fadool
- Department of Biological Science, Program in Neuroscience, Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Kathryn F Medler
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute at South Lake Union, Seattle, Washington, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alan G Watts
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diana Cummings
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Bradley M Cooke
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Wu Y, Su Q. Harnessing the Gut Microbiome: To What Extent Can Pre-/Probiotics Alleviate Immune Activation in Autism Spectrum Disorder? Nutrients 2024; 16:2382. [PMID: 39125263 PMCID: PMC11314583 DOI: 10.3390/nu16152382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Children diagnosed with autism spectrum disorder (ASD) are at an increased risk of experiencing gastrointestinal (GI) discomfort, which has been linked to dysfunctions in the microbiome-gut-brain axis. The bidirectional communication between gut and brain plays a crucial role in the overall health of individuals, and alterations in the gut microbiome can contribute to immune activation and gut-brain dysfunction in ASD. Despite the limited and controversial results of pre-/probiotic applications in ASD, this review comprehensively maps the association between ASD clinical symptoms and specific bacterial taxa and evaluates the efficacy of pre-/probiotics in modulating microbiota composition, reducing inflammatory biomarkers, alleviating difficulties in GI distress, sleep problems, core and other ASD-associated symptoms, as well as relieving parental concerns, separately, in individuals with ASD. Beyond simply targeting core ASD symptoms, this review highlights the potential of pre-/probiotic supplementations as a strategy to modulate gut homeostasis and immune response, and to delineate the potential mechanisms by which its direct or mediating effects can alleviate gut-brain dysfunction and poor nutritional status in ASD management. Further well-designed randomized controlled trials are needed to strengthen the existing evidence and establish optimal protocols for the use of pre-/probiotics in the context of ASD.
Collapse
Affiliation(s)
- Yuqi Wu
- Microbiota I-Center (MagIC), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Gibbons C, Beaulieu K, Almiron-Roig E, Navas-Carretero S, Martínez JA, O'Hara B, O'Connor D, Nazare JA, Le Bail A, Rannou C, Hardman C, Wilton M, Kjølbæk L, Scott C, Moshoyiannis H, Raben A, Harrold JA, Halford JCG, Finlayson G. Acute and two-week effects of neotame, stevia rebaudioside M and sucrose-sweetened biscuits on postprandial appetite and endocrine response in adults with overweight/obesity-a randomised crossover trial from the SWEET consortium. EBioMedicine 2024; 102:105005. [PMID: 38553262 PMCID: PMC11026940 DOI: 10.1016/j.ebiom.2024.105005] [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: 10/02/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Sweeteners and sweetness enhancers (S&SE) are used to replace energy yielding sugars and maintain sweet taste in a wide range of products, but controversy exists about their effects on appetite and endocrine responses in reduced or no added sugar solid foods. The aim of the current study was to evaluate the acute (1 day) and repeated (two-week daily) ingestive effects of 2 S&SE vs. sucrose formulations of biscuit with fruit filling on appetite and endocrine responses in adults with overweight and obesity. METHODS In a randomised crossover trial, 53 healthy adults (33 female, 20 male) with overweight/obesity in England and France consumed biscuits with fruit filling containing 1) sucrose, or reformulated with either 2) Stevia Rebaudioside M (StRebM) or 3) Neotame daily during three, two-week intervention periods with a two-week washout. The primary outcome was composite appetite score defined as [desire to eat + hunger + (100 - fullness) + prospective consumption]/4. FINDINGS Each formulation elicited a similar reduction in appetite sensations (3-h postprandial net iAUC). Postprandial insulin (2-h iAUC) was lower after Neotame (95% CI (0.093, 0.166); p < 0.001; d = -0.71) and StRebM (95% CI (0.133, 0.205); p < 0.001; d = -1.01) compared to sucrose, and glucose was lower after StRebM (95% CI (0.023, 0.171); p < 0.05; d = -0.39) but not after Neotame (95% CI (-0.007, 0.145); p = 0.074; d = -0.25) compared to sucrose. There were no differences between S&SE or sucrose formulations on ghrelin, glucagon-like peptide 1 or pancreatic polypeptide iAUCs. No clinically meaningful differences between acute vs. two-weeks of daily consumption were found. INTERPRETATION In conclusion, biscuits reformulated to replace sugar using StRebM or Neotame showed no differences in appetite or endocrine responses, acutely or after a two-week exposure, but can reduce postprandial insulin and glucose response in adults with overweight or obesity. FUNDING The present study was funded by the Horizon 2020 program: Sweeteners and sweetness enhancers: Impact on health, obesity, safety and sustainability (acronym: SWEET, grant no: 774293).
Collapse
Affiliation(s)
- Catherine Gibbons
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK.
| | - Kristine Beaulieu
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK
| | - Eva Almiron-Roig
- University of Navarra, Faculty of Pharmacy and Nutrition, Dept. of Food Science and Physiology, Center for Nutrition Research, Pamplona, Spain; Navarra Institute for Health Research (IdiSNa), Pamplona, Spain
| | - Santiago Navas-Carretero
- University of Navarra, Faculty of Pharmacy and Nutrition, Dept. of Food Science and Physiology, Center for Nutrition Research, Pamplona, Spain; Navarra Institute for Health Research (IdiSNa), Pamplona, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, Madrid, Spain
| | - J Alfredo Martínez
- University of Navarra, Faculty of Pharmacy and Nutrition, Dept. of Food Science and Physiology, Center for Nutrition Research, Pamplona, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, Madrid, Spain
| | - Beverley O'Hara
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK
| | - Dominic O'Connor
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK
| | - Julie-Anne Nazare
- Human Nutrition Research Center Rhône-Alpes, Lyon 1 Claude Bernard University, France
| | | | | | - Charlotte Hardman
- Department of Psychology, Institute of Population Health, University of Liverpool, UK
| | - Moon Wilton
- Department of Psychology, Institute of Population Health, University of Liverpool, UK
| | - Louise Kjølbæk
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Corey Scott
- Core Research and Development, Cargill, Inc, USA
| | | | - Anne Raben
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark; Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Joanne A Harrold
- Department of Psychology, Institute of Population Health, University of Liverpool, UK
| | - Jason C G Halford
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK
| | - Graham Finlayson
- School of Psychology, Faculty of Medicine & Health, University of Leeds, UK
| |
Collapse
|
10
|
Mansour S, Alkhaaldi SMI, Sammanasunathan AF, Ibrahim S, Farhat J, Al-Omari B. Precision Nutrition Unveiled: Gene-Nutrient Interactions, Microbiota Dynamics, and Lifestyle Factors in Obesity Management. Nutrients 2024; 16:581. [PMID: 38474710 PMCID: PMC10935146 DOI: 10.3390/nu16050581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Obesity is a complex metabolic disorder that is associated with several diseases. Recently, precision nutrition (PN) has emerged as a tailored approach to provide individualised dietary recommendations. AIM This review discusses the major intrinsic and extrinsic components considered when applying PN during the management of obesity and common associated chronic conditions. RESULTS The review identified three main PN components: gene-nutrient interactions, intestinal microbiota, and lifestyle factors. Genetic makeup significantly contributes to inter-individual variations in dietary behaviours, with advanced genome sequencing and population genetics aiding in detecting gene variants associated with obesity. Additionally, PN-based host-microbiota evaluation emerges as an advanced therapeutic tool, impacting disease control and prevention. The gut microbiome's composition regulates diverse responses to nutritional recommendations. Several studies highlight PN's effectiveness in improving diet quality and enhancing adherence to physical activity among obese patients. PN is a key strategy for addressing obesity-related risk factors, encompassing dietary patterns, body weight, fat, blood lipids, glucose levels, and insulin resistance. CONCLUSION PN stands out as a feasible tool for effectively managing obesity, considering its ability to integrate genetic and lifestyle factors. The application of PN-based approaches not only improves current obesity conditions but also holds promise for preventing obesity and its associated complications in the long term.
Collapse
Affiliation(s)
- Samy Mansour
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.M.); (A.F.S.)
| | - Saif M. I. Alkhaaldi
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.M.); (A.F.S.)
| | - Ashwin F. Sammanasunathan
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.M.); (A.F.S.)
| | - Saleh Ibrahim
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.M.); (A.F.S.)
- Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Joviana Farhat
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Basem Al-Omari
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
11
|
Salaün C, Courvalet M, Rousseau L, Cailleux K, Breton J, Bôle-Feysot C, Guérin C, Huré M, Goichon A, do Rego JC, Déchelotte P, Ribet D, Achamrah N, Coëffier M. Sex-dependent circadian alterations of both central and peripheral clock genes expression and gut-microbiota composition during activity-based anorexia in mice. Biol Sex Differ 2024; 15:6. [PMID: 38217033 PMCID: PMC10785476 DOI: 10.1186/s13293-023-00576-x] [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: 07/20/2023] [Accepted: 12/16/2023] [Indexed: 01/14/2024] Open
Abstract
RATIONALE Patients with anorexia nervosa (AN) often present sleep disorders and circadian hormonal dysregulation. The role of the microbiota-gut-brain axis in the regulation of feeding behavior has emerged during the last decades but its relationships with the circadian rhythm remains poorly documented. Thus, we aimed to characterize the circadian clock genes expression in peripheral and central tissues in the activity-based anorexia mouse model (ABA), as well as the dynamics of the gut-microbiota composition. METHODS From day 1 to day 17, male and female C57Bl/6 mice were submitted or not to the ABA protocol (ABA and control (CT) groups), which combines a progressive limited access to food and a free access to a running wheel. At day 17, fasted CT and ABA mice were euthanized after either resting (EoR) or activity (EoA) phase (n = 10-12 per group). Circadian clock genes expression was assessed by RT-qPCR on peripheral (liver, colon and ileum) and central (hypothalamic suprachiasmatic nucleus or SCN) tissues. Cecal bacterial taxa abundances were evaluated by qPCR. Data were compared by two-way ANOVA followed by post-tests. RESULTS ABA mice exhibited a lower food intake, a body weight loss and an increase of diurnal physical activity that differ according with the sex. Interestingly, in the SCN, only ABA female mice exhibited altered circadian clock genes expression (Bmal1, Per1, Per2, Cry1, Cry2). In the intestinal tract, modification of clock genes expression was also more marked in females compared to males. For instance, in the ileum, female mice showed alteration of Bmal1, Clock, Per1, Per2, Cry1, Cry2 and Rev-erbα mRNA levels, while only Per2 and Cry1 mRNAs were affected by ABA model in males. By contrast, in the liver, clock genes expression was more markedly affected in males compared to females in response to ABA. Finally, circadian variations of gut-bacteria abundances were observed in both male and female mice and sex-dependent alteration were observed in response to the ABA model. CONCLUSIONS This study shows that alteration of circadian clock genes expression at both peripheral and central levels occurs in response to the ABA model. In addition, our data underline that circadian variations of the gut-microbiota composition are sex-dependent.
Collapse
Affiliation(s)
- Colin Salaün
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Marine Courvalet
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Léna Rousseau
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Kévin Cailleux
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Jonathan Breton
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Charlène Guérin
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Marion Huré
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Alexis Goichon
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Jean-Claude do Rego
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
- Univ Rouen Normandie, Inserm, CNRS, Normandie Univ, HERACLES US 51 UAR 2026, Behavioural Analysis Platform SCAC, 76000, Rouen, France
| | - Pierre Déchelotte
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
- Department of Nutrition, CHU Rouen, 76000, Rouen, France
| | - David Ribet
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Najate Achamrah
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
- Department of Nutrition, CHU Rouen, 76000, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR 1073, Nutrition Inflammation and Microbiota Gut Brain Axis, UFR Santé, 22 Boulevard Gambetta, 76183, Rouen Cedex, France.
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France.
- Department of Nutrition, CHU Rouen, 76000, Rouen, France.
| |
Collapse
|
12
|
Concepción-Zavaleta MJ, Quiroz-Aldave JE, Durand-Vásquez MDC, Gamarra-Osorio ER, Valencia de la Cruz JDC, Barrueto-Callirgos CM, Puelles-León SL, Alvarado-León EDJ, Leiva-Cabrera F, Zavaleta-Gutiérrez FE, Concepción-Urteaga LA, Paz-Ibarra J. A comprehensive review of genetic causes of obesity. World J Pediatr 2024; 20:26-39. [PMID: 37725322 DOI: 10.1007/s12519-023-00757-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Obesity is a multifactorial chronic disease with a high, increasing worldwide prevalence. Genetic causes account for 7% of the cases in children with extreme obesity. DATA SOURCES This narrative review was conducted by searching for papers published in the PubMed/MEDLINE, Embase and SciELO databases and included 161 articles. The search used the following search terms: "obesity", "obesity and genetics", "leptin", "Prader-Willi syndrome", and "melanocortins". The types of studies included were systematic reviews, clinical trials, prospective cohort studies, cross-sectional and prospective studies, narrative reviews, and case reports. RESULTS The leptin-melanocortin pathway is primarily responsible for the regulation of appetite and body weight. However, several important aspects of the pathophysiology of obesity remain unknown. Genetic causes of obesity can be grouped into syndromic, monogenic, and polygenic causes and should be assessed in children with extreme obesity before the age of 5 years, hyperphagia, or a family history of extreme obesity. A microarray study, an analysis of the melanocortin type 4 receptor gene mutations and leptin levels should be performed for this purpose. There are three therapeutic levels: lifestyle modifications, pharmacological treatment, and bariatric surgery. CONCLUSIONS Genetic study technologies are in constant development; however, we are still far from having a personalized approach to genetic causes of obesity. A significant proportion of the affected individuals are associated with genetic causes; however, there are still barriers to its approach, as it continues to be underdiagnosed. Video Abstract (MP4 1041807 KB).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - José Paz-Ibarra
- Department of Medicine, School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| |
Collapse
|
13
|
Galmiche M, Déchelotte P. Rôle de l’axe microbiote-intestin-cerveau dans la dérégulation du comportement alimentaire au cours de l’obésité et de l’hyperphagie boulimique : les mécanismes. NUTR CLIN METAB 2023; 37:2S16-2S25. [DOI: 10.1016/s0985-0562(24)00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Zhuang Z, Zhou P, Wang J, Lu X, Chen Y. The Characteristics, Mechanisms and Therapeutics: Exploring the Role of Gut Microbiota in Obesity. Diabetes Metab Syndr Obes 2023; 16:3691-3705. [PMID: 38028999 PMCID: PMC10674108 DOI: 10.2147/dmso.s432344] [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: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Presently, obesity has emerged as a significant global public health concern due to its escalating prevalence and incidence rates. The gut microbiota, being a crucial environmental factor, has emerged as a key player in the etiology of obesity. Nevertheless, the intricate and specific interactions between obesity and gut microbiota, along with the underlying mechanisms, remain incompletely understood. This review comprehensively summarizes the gut microbiota characteristics in obesity, the mechanisms by which it induces obesity, and explores targeted therapies centered on gut microbiota restoration.
Collapse
Affiliation(s)
- Zequn Zhuang
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Peng Zhou
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Jing Wang
- Jiangnan University Medical Center, Wuxi, People’s Republic of China
| | - Xiaojing Lu
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Yigang Chen
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
- Jiangnan University Medical Center, Wuxi, People’s Republic of China
- Wuxi Clinical College, Nantong University, Wuxi, People’s Republic of China
| |
Collapse
|
15
|
Forte N, Roussel C, Marfella B, Lauritano A, Villano R, De Leonibus E, Salviati E, Khalilzadehsabet T, Giorgini G, Silvestri C, Piscitelli F, Mollica MP, Di Marzo V, Cristino L. Olive oil-derived endocannabinoid-like mediators inhibit palatable food-induced reward and obesity. Commun Biol 2023; 6:959. [PMID: 37735539 PMCID: PMC10514336 DOI: 10.1038/s42003-023-05295-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023] Open
Abstract
N-oleoylglycine (OlGly), a lipid derived from the basic component of olive oil, oleic acid, and N-oleoylalanine (OlAla) are endocannabinoid-like mediators. We report that OlGly and OlAla, by activating the peroxisome proliferator-activated receptor alpha (PPARα), reduce the rewarding properties of a highly palatable food, dopamine neuron firing in the ventral tegmental area, and the obesogenic effect of a high-fat diet rich in lard (HFD-L). An isocaloric olive oil HFD (HFD-O) reduced body weight gain compared to the HFD-L, in a manner reversed by PPARα antagonism, and enhanced brain and intestinal OlGly levels and gut microbial diversity. OlGly or OlAla treatment of HFD-L mice resulted in gut microbiota taxonomic changes partly similar to those induced by HFD-O. We suggest that OlGly and OlAla control body weight by counteracting highly palatable food overconsumption, and possibly rebalancing the gut microbiota, and provide a potential new mechanism of action for the obeso-preventive effects of olive oil-rich diets.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Charlène Roussel
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Anna Lauritano
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Rosaria Villano
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | | | - Tina Khalilzadehsabet
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Giada Giorgini
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Cristoforo Silvestri
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80138, Naples, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy.
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada.
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, 61V0AG, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy.
| |
Collapse
|
16
|
Hankir MK, Kovatcheva-Datchary P, Springer R, Hoffmann A, Vogel J, Seyfried F, Arora T. Gut Microbiota Contribution to Weight-Independent Glycemic Improvements after Gastric Bypass Surgery. Microbiol Spectr 2023; 11:e0510922. [PMID: 37022171 PMCID: PMC10269853 DOI: 10.1128/spectrum.05109-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/02/2023] [Indexed: 04/07/2023] Open
Abstract
Roux-en-Y gastric bypass surgery (RYGB) leads to improved glycemic control in individuals with severe obesity beyond the effects of weight loss alone. Here, We addressed the potential contribution of gut microbiota in mediating this favourable surgical outcome by using an established preclinical model of RYGB. 16S rRNA sequencing revealed that RYGB-treated Zucker fatty rats had altered fecal composition of various bacteria at the phylum and species levels, including lower fecal abundance of an unidentified Erysipelotrichaceae species, compared with both sham-operated (Sham) and body weight-matched to RYGB-treated (BWM) rats. Correlation analysis further revealed that fecal abundance of this unidentified Erysipelotrichaceae species linked with multiple indices of glycemic control uniquely in RYGB-treated rats. Sequence alignment of this Erysipelotrichaceae species identified Longibaculum muris to be the most closely related species, and its fecal abundance positively correlated with oral glucose intolerance in RYGB-treated rats. In fecal microbiota transplant experiments, the improved oral glucose tolerance of RYGB-treated compared with BWM rats could partially be transferred to recipient germfree mice, independently of body weight. Unexpectedly, providing L. muris as a supplement to RYGB recipient mice further improved oral glucose tolerance, while administering L. muris alone to chow-fed or Western style diet-challenged conventionally raised mice had minimal metabolic impact. Taken together, our findings provide evidence that the gut microbiota contributes to weight loss-independent improvements in glycemic control after RYGB and demonstrate how correlation of a specific gut microbiota species with a host metabolic trait does not imply causation. IMPORTANCE Metabolic surgery remains the most effective treatment modality for severe obesity and its comorbidities, including type 2 diabetes. Roux-en-Y gastric bypass (RYGB) is a commonly performed type of metabolic surgery that reconfigures gastrointestinal anatomy and profoundly remodels the gut microbiota. While it is clear that RYGB is superior to dieting when it comes to improving glycemic control, the extent to which the gut microbiota contributes to this effect remains untested. In the present study, we uniquely linked fecal Erysipelotrichaceae species, including Longibaculum muris, with indices of glycemic control after RYGB in genetically obese and glucose-intolerant rats. We further show that the weight loss-independent improvements in glycemic control in RYGB-treated rats can be transmitted via their gut microbiota to germfree mice. Our findings provide rare causal evidence that the gut microbiota contributes to the health benefits of metabolic surgery and have implications for the development of gut microbiota-based treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed K. Hankir
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Petia Kovatcheva-Datchary
- Institute for Molecular Infection Biology, University of Wurzburg, Wurzburg, Germany
- Department of Pediatrics, University Hospital Wurzburg, Wurzburg, Germany
| | - Rebecca Springer
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Annett Hoffmann
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Jörg Vogel
- Institute for Molecular Infection Biology, University of Wurzburg, Wurzburg, Germany
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Wurzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Wang S, Zhang B, Chang X, Zhao H, Zhang H, Zhao T, Qi H. Potential use of seaweed polysaccharides as prebiotics for management of metabolic syndrome: a review. Crit Rev Food Sci Nutr 2023; 64:7707-7727. [PMID: 36971135 DOI: 10.1080/10408398.2023.2191135] [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: 03/29/2023]
Abstract
Seaweed polysaccharides (SPs) obtained from seaweeds are a class of functional prebiotics. SPs can regulate glucose and lipid anomalies, affect appetite, reduce inflammation and oxidative stress, and therefore have great potential for managing metabolic syndrome (MetS). SPs are poorly digested by the human gastrointestinal tract but are available to the gut microbiota to produce metabolites and exert a series of positive effects, which may be the mechanism by which SPs render their anti-MetS effects. This article reviews the potential of SPs as prebiotics in the management of MetS-related metabolic disturbances. The structure of SPs and studies related to the process of their degradation by gut bacteria and their therapeutic effects on MetS are highlighted. In summary, this review provides new perspectives on SPs as prebiotics to prevent and treat MetS.
Collapse
Affiliation(s)
- Shaopeng Wang
- College of Pharmacy, Weifang Medical University, Weifang, Shandong, PR China
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Bo Zhang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Xintao Chang
- Department of Pharmacy, People's Hospital of Zhangqiu District, Jinan, Shandong, PR China
| | - Hailing Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Haojun Zhang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Tingting Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, PR China
| | - Huimin Qi
- College of Pharmacy, Weifang Medical University, Weifang, Shandong, PR China
| |
Collapse
|
18
|
Wang W, Liu Y, Li Y, Luo B, Lin Z, Chen K, Liu Y. Dietary patterns and cardiometabolic health: Clinical evidence and mechanism. MedComm (Beijing) 2023; 4:e212. [PMID: 36776765 PMCID: PMC9899878 DOI: 10.1002/mco2.212] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 02/08/2023] Open
Abstract
For centuries, the search for nutritional interventions to underpin cardiovascular treatment and prevention guidelines has contributed to the rapid development of the field of dietary patterns and cardiometabolic disease (CMD). Numerous studies have demonstrated that healthy dietary patterns with emphasis on food-based recommendations are the gold standard for extending lifespan and reducing the risks of CMD and mortality. Healthy dietary patterns include various permutations of energy restriction, macronutrients, and food intake patterns such as calorie restriction, intermittent fasting, Mediterranean diet, plant-based diets, etc. Early implementation of healthy dietary patterns in patients with CMD is encouraged, but an understanding of the mechanisms by which these patterns trigger cardiometabolic benefits remains incomplete. Hence, this review examined several dietary patterns that may improve cardiometabolic health, including restrictive dietary patterns, regional dietary patterns, and diets based on controlled macronutrients and food groups, summarizing cutting-edge evidence and potential mechanisms for CMD prevention and treatment. Particularly, considering individual differences in responses to dietary composition and nutritional changes in organ tissue diversity, we highlighted the critical role of individual gut microbiota in the crosstalk between diet and CMD and recommend a more precise and dynamic nutritional strategy for CMD by developing dietary patterns based on individual gut microbiota profiles.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yanfei Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yiwen Li
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Binyu Luo
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Zhixiu Lin
- Faculty of MedicineThe Chinese University of Hong KongHong Kong
| | - Keji Chen
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yue Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
19
|
Qian X, Liu A, Liang C, He L, Xu Z, Tang S. Analysis of gut microbiota in patients with acute myocardial infarction by 16S rRNA sequencing. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1340. [PMID: 36660636 PMCID: PMC9843380 DOI: 10.21037/atm-22-5671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
Background An increasing number of studies have shown that gut microbiota are associated with human cardiovascular disease, but the characteristics of intestinal flora in patients with acute myocardial infarction (AMI) are still unclear. In this study, we aimed to investigate the difference of intestinal microflora between patients with AMI and healthy people, and to find the effect of percutaneous coronary intervention (PCI) on intestinal microflora. Methods A total of 60 stool samples and 60 peripheral blood samples were collected from 20 previously diagnosed AMI patients and 20 healthy people serving as controls. Gut microbiota communities were analyzed via 16 ribosomal RNA-sequencing (16S rRNA). Gut microbiota-derived metabolites, trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFA), in the blood were detected using stable isotope dilution high-performance liquid chromatography with on line electrospray ionization tandem mass spectrometry (LC/MS/MS). Results The results showed that a distinct pattern of gut microbiota was observed in AMI patients compared to healthy controls. AMI patients had lower microbiological richness but no significant change in diversity. Bacteroidetes and Verrucomicobia showed an upward trend, whereas Proteobacteria showed a downward trend in AMI patients. During a longitudinal study to compare the changes in bacteria before and after treatment, we found routine cardiac admission therapy 1 week after PCI surgery had no effect on the microbial community structure in patients. There were significantly higher levels of plasma TMAO in AMI patients' microbiota than that in the control group. Contrarily, there was no obvious change in SCFA. Conclusions The gut microbiota of patients with AMI differs from that of normal people, and the metabolic products of microflora are more abundant in the plasma of AMI than control cases. Microflora may act on the cardiovascular system through metabolites, and regulation of the microfloral structure may be used in the future treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xueyi Qian
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Ankang Liu
- Department of Cardiology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | - Chen Liang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Lianjun He
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zhenyu Xu
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Shengxing Tang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|