1
|
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
|
2
|
Climent E, Hevilla F, Padial M, Barril-Cuadrado G, Blanca M, Jiménez-Salcedo T, López-Picasso M, Nogueira-Pérez Á, Olveira G. Psychobiotic Protection of Nutritional Supplements and Probiotics in Patients Undergoing Hemodialysis: A Randomized Trial. Nutrients 2025; 17:652. [PMID: 40004979 PMCID: PMC11858206 DOI: 10.3390/nu17040652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The prevalence of depression and anxiety symptoms is remarkably high in malnourished individuals undergoing hemodialysis. The goal of this project was to evaluate the impact of administering an oral nutritional supplement combined with a probiotic blend on the microbiota, intestinal permeability, and depression symptoms in malnourished hemodialysis patients. METHODS With this aim, a randomized trial was conducted with three parallel groups: a control group with individualized diet, a supplement-placebo (SU-PL) group with oral nutritional supplementation (ONS), and a supplement-probiotic (SU-PR) group with ONS in conjunction with a probiotic blend. Blood and fecal samples were collected at basal time, and at 3 and 6 months. Several blood biomarkers, like zonulin, lipopolysaccharide-binding protein (LBP), lipopolysaccharide (LPS), and brain-derived neurotrophic factor (BDNF), were measured, and the fecal microbiome was sequenced with the Illumina platform. The Hospital Anxiety and Depression Scale (HADS) was used for the estimation of depression (HADS-D) and anxiety (HADS-A) symptoms, along with the standardized mental health index SF12-MH from the general health questionnaire SF-12. RESULTS The results showed that patients who consumed the probiotic blend maintained the LPS levels from their baseline readings and decreased their BDNF levels compared to the SU-PL or control groups. Moreover, a significant decrease in HADS-D scores (less depressive symptoms) and an increase in SF12-MH scores (higher quality of life) were found in that group in comparison to the other groups. The intervention produced an impact on the microbiome population, where the SU-PR group had reduced Akkermansia abundance with respect to the other groups, while their Acidaminococcus abundance decreased and their Barnesiella abundance increased with respect to the SU-PL group. CONCLUSIONS Overall, the results indicate that the probiotic with the nutritional supplement could reduce the intestinal permeability biomarkers and improve depressive symptoms and quality of life in malnourished hemodialysis patients.
Collapse
Affiliation(s)
- Eric Climent
- Departamento de Biotecnología, Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Francisco Hevilla
- Servicio de Endocrinología y Nutrición, Hospital Regional Universitario, 29071 Málaga, Spain; (F.H.); (M.P.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND, 29010 Málaga, Spain
- Departamento de Medicina y Dermatología, Universidad de Málaga, 29071 Málaga, Spain
| | - Marina Padial
- Servicio de Endocrinología y Nutrición, Hospital Regional Universitario, 29071 Málaga, Spain; (F.H.); (M.P.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND, 29010 Málaga, Spain
- Departamento de Medicina y Dermatología, Universidad de Málaga, 29071 Málaga, Spain
| | | | - María Blanca
- Servicio de Endocrinología y Nutrición, Hospital Rey Juan Carlos, 28933 Madrid, Spain; (M.B.); (M.L.-P.)
| | | | - Maria López-Picasso
- Servicio de Endocrinología y Nutrición, Hospital Rey Juan Carlos, 28933 Madrid, Spain; (M.B.); (M.L.-P.)
| | - Ángel Nogueira-Pérez
- Servicio de Nefrología, Hospital la Princesa, 28006 Madrid, Spain; (G.B.-C.); (Á.N.-P.)
| | - Gabriel Olveira
- Servicio de Endocrinología y Nutrición, Hospital Regional Universitario, 29071 Málaga, Spain; (F.H.); (M.P.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND, 29010 Málaga, Spain
- Departamento de Medicina y Dermatología, Universidad de Málaga, 29071 Málaga, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 29071 Málaga, Spain
| |
Collapse
|
3
|
Luo Y, Li M, Luo D, Tang B. Gut Microbiota: An Important Participant in Childhood Obesity. Adv Nutr 2025; 16:100362. [PMID: 39733798 PMCID: PMC11786877 DOI: 10.1016/j.advnut.2024.100362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
Increasing prevalence of childhood obesity has emerged as a critical global public health concern. Recent studies have challenged the previous belief that obesity was solely a result of excessive caloric intake. Alterations in early-life gut microbiota can contribute to childhood obesity through their influence on nutrient absorption and metabolism, initiation of inflammatory responses, and regulation of gut-brain communication. The gut microbiota is increasingly acknowledged to play a crucial role in human health, as certain beneficial bacteria have been scientifically proven to possess the capacity to reduce body fat content and enhance intestinal barrier function and their metabolic products to exhibit anti-inflammatory effect. Examples of such microbes include bifidobacteria, Akkermansia muciniphila, and Lactobacillus reuteri. In contrast, an increase in Enterobacteriaceae and propionate-producing bacteria (Prevotellaceae and Veillonellaceae) has been implicated in the induction of low-grade systemic inflammation and disturbances in lipid metabolism, which can predispose individuals to obesity. Studies have demonstrated that modulating the gut microbiota through diet, lifestyle changes, prebiotics, probiotics, or fecal microbiota transplantation may contribute to gut homeostasis and the management of obesity and its associated comorbidities. This review aimed to elucidate the impact of alterations in gut microbiota composition during early life on childhood obesity and explores the mechanisms by which gut microbiota contributes to the pathogenesis of obesity and specifically focused on recent advances in using short-chain fatty acids for regulating gut microbiota and ameliorating obesity. Additionally, it aimed to discuss the therapeutic strategies for childhood obesity from the perspective of gut microbiota, aiming to provide a theoretical foundation for interventions targeting pediatric obesity based on gut microbiota.
Collapse
Affiliation(s)
- Yu Luo
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Maojun Li
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Luo
- Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Binzhi Tang
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
4
|
Gao X, Zhu Z, Bao Y, Li Y, Zhu W, He X, Ge X, Huang W, Wang H, Wei W, Du J, Chen L, Li H, Sheng L. Chrysanthemum morifolium Ramat extract and probiotics combination ameliorates metabolic disorders through regulating gut microbiota and PPARα subcellular localization. Chin Med 2024; 19:76. [PMID: 38831430 PMCID: PMC11149226 DOI: 10.1186/s13020-024-00950-w] [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: 03/12/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Chrysanthemum morifolium Ramat, a traditional Chinese medicine, has the effects on liver clearing, vision improving, and anti-inflammation. C. morifolium and probiotics have been individually studied for their beneficial effects on metabolic diseases. However, the underlying molecular mechanisms were not completely elucidated. This study aims to elucidate the potential molecular mechanisms of C. morifolium and probiotics combination (CP) on alleviating nonalcoholic fatty liver disease (NAFLD) and the dysregulation of glucose metabolism in high-fat diet (HFD)-fed mice. METHODS The therapeutic effect of CP on metabolism was evaluated by liver histology and serum biochemical analysis, as well as glucose tolerance test. The impact of CP on gut microbiota was analyzed by 16S rRNA sequencing and fecal microbiota transplantation. Hepatic transcriptomic analysis was performed with the key genes and proteins validated by RT-qPCR and western blotting. In addition, whole body Pparα knockout (Pparα-/-) mice were used to confirm the CP-mediated pathway. RESULTS CP supplementation ameliorated metabolic disorders by reducing body weight and hepatic steatosis, and improving glucose intolerance and insulin resistance in HFD fed mice. CP intervention mitigated the HFD-induced gut microbiota dysbiosis, which contributed at least in part, to the beneficial effect of improving glucose metabolism. In addition, hepatic transcriptomic analysis showed that CP modulated the expression of genes associated with lipid metabolism. CP downregulated the mRNA level of lipid droplet-binding proteins, such as Cidea and Cidec in the liver, leading to more substrates for fatty acid oxidation (FAO). Meanwhile, the expression of CPT1α, the rate-limiting enzyme of FAO, was significantly increased upon CP treatment. Mechanistically, though CP didn't affect the total PPARα level, it promoted the nuclear localization of PPARα, which contributed to the reduced expression of Cidea and Cidec, and increased expression of CPT1α, leading to activated FAO. Moreover, whole body PPARα deficiency abolished the anti-NAFLD effect of CP, suggesting the importance of PPARα in CP-mediated beneficial effect. CONCLUSION This study revealed the hypoglycemic and hepatoprotective effect of CP by regulating gut microbiota composition and PPARα subcellular localization, highlighting its potential for therapeutic candidate for metabolic disorders.
Collapse
Affiliation(s)
- Xinxin Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhigang Zhu
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, 201203, China
| | - Yiyang Bao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yifan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaofang He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinyu Ge
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hao Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenjing Wei
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, 201203, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, 201203, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Balaguer F, Barrena M, Enrique M, Maicas M, Álvarez B, Tortajada M, Chenoll E, Ramón D, Martorell P. Bifidobacterium animalis subsp. lactis BPL1™ and Its Lipoteichoic Acid Modulate Longevity and Improve Age/Stress-Related Behaviors in Caenorhabditis elegans. Antioxidants (Basel) 2023; 12:2107. [PMID: 38136226 PMCID: PMC10740966 DOI: 10.3390/antiox12122107] [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: 11/10/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Life expectancy has increased globally in recent decades, driving interest in maintaining a healthy life that includes preservation of physical and mental abilities, particularly in elderly people. The gut microbiome becomes increasingly perturbed with aging so the use of probiotics can be a strategy for maintaining a balanced gut microbiome. A previous report showed that Bifidobacterium animalis subsp. lactis BPL1™ induces through its lipoteichoic acid (LTA) fat reduction activities via the insulin/IGF-1 signaling pathway. Here, we have delved into the mechanism of action, eliminating alternative pathways as putative mechanisms. Furthermore, we have identified that BPL1™, its heat treated form (BPL1™ HT) and its LTA prolong longevity in Caenorhabditis elegans (C. elegans) in an insulin/IGF-1-dependent mechanism, and its consumption improves the oxidative stress response, gut permeability and protection against pathogenic infections. Furthermore, positive effects on C. elegans stress-related behaviors and in the Alzheimer's Disease model were found, highlighting the potential of the strain in improving the cognitive functions and proteotoxicity in the nematode. These results indicate the pivotal role of the IGF-1 pathway in the activity of the strain and pave the way for potential applications of BPL1™, BPL1™ HT and its LTA in the field of longevity and age-related markers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Patricia Martorell
- Archer Daniels Midland, Nutrition, Health & Wellness, Biopolis S.L. Parc Científic Universitat de València, C/Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain (M.B.); (M.E.); (M.T.); (E.C.)
| |
Collapse
|
6
|
Ke S, Hartmann J, Ressler KJ, Liu YY, Koenen KC. The emerging role of the gut microbiome in posttraumatic stress disorder. Brain Behav Immun 2023; 114:360-370. [PMID: 37689277 PMCID: PMC10591863 DOI: 10.1016/j.bbi.2023.09.005] [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: 04/15/2023] [Revised: 08/28/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) occurs in some people following exposure to a terrifying or catastrophic event involving actual/threatened death, serious injury, or sexual violence. PTSD is a common and debilitating mental disorder that imposes a significant burden on individuals, their families, health services, and society. Moreover, PTSD is a risk factor for chronic diseases such as coronary heart disease, stroke, diabetes, as well as premature mortality. Furthermore, PTSD is associated with dysregulated immune function. Despite the high prevalence of PTSD, the mechanisms underlying its etiology and manifestations remain poorly understood. Compelling evidence indicates that the human gut microbiome, a complex community of microorganisms living in the gastrointestinal tract, plays a crucial role in the development and function of the host nervous system, complex behaviors, and brain circuits. The gut microbiome may contribute to PTSD by influencing inflammation, stress responses, and neurotransmitter signaling, while bidirectional communication between the gut and brain involves mechanisms such as microbial metabolites, immune system activation, and the vagus nerve. In this literature review, we summarize recent findings on the role of the gut microbiome in PTSD in both human and animal studies. We discuss the methodological limitations of existing studies and suggest future research directions to further understand the role of the gut microbiome in PTSD.
Collapse
Affiliation(s)
- Shanlin Ke
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Center for Artificial Intelligence and Modeling, The Carl R. WoeseInstitute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Loy MH, Usseglio J, Lasalandra D, Gold MA. Probiotic Use in Children and Adolescents with Overweight or Obesity: A Scoping Review. Child Obes 2023; 19:145-159. [PMID: 35723657 DOI: 10.1089/chi.2022.0059] [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] [Indexed: 11/13/2022]
Abstract
Context: Probiotics have been proposed as a prevention or treatment for pediatric overweight and obesity. Objective: Conduct a scoping review on probiotic use in children and adolescents with overweight or obesity and those with weight-related conditions and to identify knowledge gaps and research priorities. Data Sources: Seven databases using keywords and medical subject heading terms for articles reporting probiotic use in children or adolescents with overweight or obesity published from database conception until initiation of the study. Study Selection: Articles reporting primary data on probiotics use in children or adolescents with overweight or obesity. Data Extraction: We utilized the Arksey and O'Malley framework, PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines, followed a predetermined study protocol for level-one abstract and level-two full-text screenings, synthesized information into subject-area domains, and identified research gaps. Limitations: Heterogeneity of probiotic interventions, host factors, and genomics. Results: Database search yielded 1356 unique articles with 19 randomized placebo-controlled studies, 945 participants, duration of interventions from 8 weeks to 9 months. Disease indications included Nonalcoholic Fatty Liver Disease, insulin resistance, hypercholesterolemia, Prader-Willi Syndrome, metabolic syndrome, and obesity. Limited and heterogeneous evidence for probiotic use in children and adolescents with weight-related conditions noted. Heterogeneity among published articles in probiotic strains, doses, design, biomarkers, confirmation, and outcomes observed. Conclusions: Despite complex existing and limited data, studies to date of children and adolescents with overweight and obesity demonstrate potential beneficial treatment effects of probiotics on BMI, adiposity, metabolic parameters, inflammatory markers, fatty liver, transaminase levels, and glucose metabolism. Clinical trials to address heterogeneous results are needed.
Collapse
Affiliation(s)
- Michelle H Loy
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Integrative Health and Well-Being, Weill Cornell Medicine/New York Presbyterian Hospital, New York, NY, USA
| | - John Usseglio
- Health Sciences Library, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Melanie A Gold
- Heilbrunn Department of Population and Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
- Section of Adolescent Medicine, Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Center for Community Health and Education, School-Based Health Centers, NewYork Presbyterian, New York, NY, USA
| |
Collapse
|
8
|
Abstract
A large subset of patients with Angelman syndrome (AS) suffer from concurrent gastrointestinal (GI) issues, including constipation, poor feeding, and reflux. AS is caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. Clinical features of AS, which include developmental delays, intellectual disability, microcephaly, and seizures, are primarily due to the deficient expression or function of the maternally inherited UBE3A allele. The association between neurodevelopmental delay and GI disorders is part of the increasing evidence suggesting a link between the brain and the gut microbiome via the microbiota-gut-brain axis. To investigate the associations between colonization of the gut microbiota in AS, we characterized the fecal microbiome in three animal models of AS involving maternal deletions of Ube3A, including mouse, rat, and pig, using 16S rRNA amplicon sequencing. Overall, we identified changes in bacterial abundance across all three animal models of AS. Specific bacterial groups were significantly increased across all animal models, including Lachnospiraceae Incertae sedis, Desulfovibrios sp., and Odoribacter, which have been correlated with neuropsychiatric disorders. Taken together, these findings suggest that specific changes to the local environment in the gut are driven by a Ube3a maternal deletion, unaffected by varying housing conditions, and are prominent and detectable across multiple small and large animal model species. These findings begin to uncover the underlying mechanistic causes of GI disorders in AS patients and provide future therapeutic options for AS patients. IMPORTANCE Angelman syndrome (AS)-associated gastrointestinal (GI) symptoms significantly impact quality of life in patients. In AS models in mouse, rat, and pig, AS animals showed impaired colonization of the gut microbiota compared to wild-type (healthy) control animals. Common changes in AS microbiomes across all three animal models may play a causal effect for GI symptoms and may help to identify ways to treat these comorbidities in patients in the future.
Collapse
|
9
|
de Cuevillas B, Milagro FI, Tur JA, Gil-Campos M, de Miguel-Etayo P, Martínez JA, Navas-Carretero S. Fecal microbiota relationships with childhood obesity: A scoping comprehensive review. Obes Rev 2022; 23 Suppl 1:e13394. [PMID: 34913242 DOI: 10.1111/obr.13394] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022]
Abstract
Childhood obesity is a costly burden in most regions with relevant and adverse long-term health consequences in adult life. Several studies have associated excessive body weight with a specific profile of gut microbiota. Different factors related to fecal microorganism abundance seem to contribute to childhood obesity, such as gestational weight gain, perinatal diet, antibiotic administration to the mother and/or child, birth delivery, and feeding patterns, among others. This review reports and discusses diverse factors that affect the infant intestinal microbiota with putative or possible implications on the increase of the obesity childhood rates as well as microbiota shifts associated with excessive body weight in children.
Collapse
Affiliation(s)
- Begoña de Cuevillas
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Fermín I Milagro
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| | - Josep A Tur
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS & IDISBA, Palma de Mallorca, Spain
| | - Mercedes Gil-Campos
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Metabolism and Investigation Unit, Reina Sofia University Hospital, Maimónides Institute of Biomedicine Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Pilar de Miguel-Etayo
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza. Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - J Alfredo Martínez
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
- Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food. CSIC-UAM, Madrid, Spain
| | - Santiago Navas-Carretero
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| |
Collapse
|
10
|
Targeting the Gut Microbiome in Prader-Willi Syndrome. J Clin Med 2021; 10:jcm10225328. [PMID: 34830610 PMCID: PMC8625997 DOI: 10.3390/jcm10225328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/24/2022] Open
Abstract
Overwhelming evidence demonstrates an important role of the gut microbiome in the development of a wide range of diseases, including obesity, metabolic disorders, and mental health symptoms. Indeed, interventions targeting the gut microbiome are being actively investigated as a therapeutic strategy to tackle these diseases. Given that obesity and mental health symptoms are both hallmarks of Prader-Willi syndrome, targeting the gut microbiome may be a promising therapeutical strategy. Only a few studies have investigated the gut microbiome in the context of Prader-Willi syndrome and assessed the efficacy of probiotic supplementation as a therapeutic strategy for this disease. Here, we review the knowledge obtained to this date regarding the gut microbiome in individuals with Prader-Willi syndrome. The limited evidence available indicate that probiotic supplementation improves some metabolic and mental health aspects, however further studies are warranted to determine whether targeting the gut microbiome may constitute a safe and efficient strategy to treat individuals with Prader-Willi syndrome.
Collapse
|
11
|
Fernández-Calderón MC, Sánchez-Moro MDH, Rincón EO. In vitro Cholesterol Assimilation by Bifidobacterium animalis subsp. lactis (BPL1) Probiotic Bacteria under Intestinal Conditions. Endocr Metab Immune Disord Drug Targets 2021; 22:433-439. [PMID: 34496737 DOI: 10.2174/1871530321666210908124848] [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] [Received: 02/22/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hypercholesterolemia is one of the principal causes of the development of cardiovascular diseases. Recently, probiotics consumption has been also proposed as a non-pharmacological intervention to control cholesterol concentrations. OBJECTIVE To evaluate in vitro assimilation of cholesterol by Bifidobacterium animalis subsp. lactis (BPL1) under simulated intestinal environment in anaerobic conditions and to review and discuss potential physiological mechanisms in this context. METHODS Bacterial viability and cholesterol assimilation was evaluated in both standard MRS and stimulated intestinal fluid (SIF) medium under anaerobic conditions, and in presence or absence of cholesterol. For assimilation assays, cholesterol concentrations in the different suspensions, containing the probiotic or not, was determined by chromatography coupled to mass spectrometry. RESULTS Results showed that the growth of B. lactis BPL1 under intestinal conditions is favored when cholesterol is present in the culture medium. In addition, a cholesterol assimilation of up to 44.4% under intestinal and anaerobic conditions was observed. CONCLUSION Taking into account the revised literature and the experimental results herein presented, administration of functional foodstuffs together with probiotic bacteria such as B. lactis BPL1 could be a potential effective option to decrease hypercholesterolemia, thus preventing the development of cardiovascular diseases. Nevertheless, further studies on mechanisms of effectiveness in animals and clinical trials are still needed.
Collapse
Affiliation(s)
- María Coronada Fernández-Calderón
- Grupo de Investigación en Microbiología de la UEx. Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz. Spain
| | - María Dolores Hinchado Sánchez-Moro
- Grupo de Investigación en Inmunofisiología. Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz. Spain
| | - Eduardo Ortega Rincón
- Grupo de Investigación en Inmunofisiología. Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz. Spain
| |
Collapse
|
12
|
Kong XJ, Liu K, Zhuang P, Tian R, Liu S, Clairmont C, Lin X, Sherman H, Zhu J, Wang Y, Fong M, Li A, Wang BK, Wang J, Yu Z, Shen C, Cui X, Cao H, Du T, Wan G, Cao X. The Effects of Limosilactobacillus reuteri LR-99 Supplementation on Body Mass Index, Social Communication, Fine Motor Function, and Gut Microbiome Composition in Individuals with Prader-Willi Syndrome: a Randomized Double-Blinded Placebo-Controlled Trial. Probiotics Antimicrob Proteins 2021; 13:1508-1520. [PMID: 34115318 PMCID: PMC8578098 DOI: 10.1007/s12602-021-09800-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2021] [Indexed: 12/16/2022]
Abstract
Prader-Willi syndrome (PWS) is a rare genetic disorder associated with developmental delay, obesity, and neuropsychiatric comorbidities. Limosilactobacillus reuteri (Lactobacillus reuteri, Lact. reuteri) has demonstrated anti-obesity and anti-inflammatory effects in previous studies. In the present study, we aim to evaluate the effects of Lact. reuteri supplementation on body mass index (BMI), social behaviors, and gut microbiota in individuals with PWS. We conducted a 12-week, randomized, double-blind, placebo-controlled trial in 71 individuals with PWS aged 6 to 264 months (64.4 ± 51.0 months). Participants were randomly assigned to either receive daily Lact. reuteri LR-99 probiotic (6 × 1010 colony forming units) or a placebo sachet. Groupwise differences were assessed for BMI, ASQ-3, and GARS-3 at baseline, 6 weeks, and 12 weeks into treatment. Gut microbiome data was analyzed with the QIIME2 software package, and predictive functional profiling was conducted with PICRUSt-2. We found a significant reduction in BMI for the probiotic group at both 6 weeks and 12 weeks relative to the baseline (P < 0.05). Furthermore, we observed a significant improvement in social communication and interaction, fine motor function, and total ASQ-3 score in the probiotics group compared to the placebo group (P < 0.05). Altered gut microbiota was observed in the probiotic group to favor weight loss and improve gut health. The findings suggest a novel therapeutic potential for Lact. reuteri LR-99 probiotic to modulate BMI, social behaviors, and gut microbiota in Prader-Willi syndrome patients, although further investigation is warranted.Trial registration Chinese Clinical Trial Registry: ChiCTR1900022646.
Collapse
Affiliation(s)
- Xue-Jun Kong
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine and Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Kevin Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Zhuang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Ruiyi Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Siyu Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Cullen Clairmont
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | - Hannah Sherman
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | - Yelan Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Michelle Fong
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Alice Li
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Zhehao Yu
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chen Shen
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xianghua Cui
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hanyu Cao
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ting Du
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Guobin Wan
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Xia Cao
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
13
|
|
14
|
Balaguer F, Enrique M, Llopis S, Barrena M, Navarro V, Álvarez B, Chenoll E, Ramón D, Tortajada M, Martorell P. Lipoteichoic acid from Bifidobacterium animalis subsp. lactis BPL1: a novel postbiotic that reduces fat deposition via IGF-1 pathway. Microb Biotechnol 2021; 15:805-816. [PMID: 33620143 PMCID: PMC8913875 DOI: 10.1111/1751-7915.13769] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity and its related metabolic disorders, such as diabetes and cardiovascular disease, are major risk factors for morbidity and mortality in the world population. In this context, supplementation with the probiotic strain Bifidobacterium animalis subsp. lactis BPL1 (CECT8145) has been shown to ameliorate obesity biomarkers. Analyzing the basis of this observation and using the pre-clinical model Caenorhabditis elegans, we have found that lipoteichoic acid (LTA) of BPL1 is responsible for its fat-reducing properties and that this attribute is preserved under hyperglycaemic conditions. This fat-reducing capacity of both BPL1 and LTA-BPL1 is abolished under glucose restriction, as a result of changes in LTA chemical composition. Moreover, we have demonstrated that LTA exerts this function through the IGF-1 pathway, as does BPL1 strain. These results open the possibility of using LTA as a novel postbiotic, whose beneficial properties can be applied therapeutically and/or preventively in metabolic syndrome and diabetes-related disorders.
Collapse
|