1
|
Song CH, Kim N, Choi Y, Kim S, Kim KS, Park MH, Lee SH, Lee DH. Beneficial effect of consuming milk containing only A2 beta-casein on gut microbiota: A single-center, randomized, double-blind, cross-over study. PLoS One 2025; 20:e0323016. [PMID: 40338897 PMCID: PMC12061139 DOI: 10.1371/journal.pone.0323016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/01/2025] [Indexed: 05/10/2025] Open
Abstract
Cow milk contains essential nutrients, with β-casein existing in A1 and A2 forms. Studies suggest that A2 milk (containing only A2 β-casein) may offer gastrointestinal (GI) benefits compared to A1/A2 milk (containing both forms). This study investigated the effects of A2 milk consumption on the gut microbiota of South Korean cohort experiencing GI discomfort after consuming A1/A2 milk. Thirty-five participants with GI discomfort after milk consumption were included. Stool DNA was analyzed using 16S rRNA gene sequencing before and after consuming either A1/A2 or A2 milk. Beta diversity analysis using the generalized UniFrac distance method revealed a significant shift in gut microbiota composition after A2 milk consumption (p = 0.04), but no significant change after consuming A1/A2 milk. Significant differences in gut microbiota composition were found between A1/A2 and A2 milk drinkers after milk consumption (p = 0.031). Alpha diversity indices remained unchanged. Notable increases in beneficial microbes, including Bifidobacterium and Blautia, were observed after A2 milk intake. Linear discriminant analysis Effect Size (LEfSe) analysis identified significant enrichment of Actinobacteria, particularly Bifidobacterium longum and Blautia wexlerae, in the A2 group. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) analysis highlighted enriched transport systems related to energy, peptides, sugars, and raffinose family oligosaccharides in the A2 group. Spearman correlation showed significant associations between Bifidobacterium, Blautia, and enhanced transport systems exclusively in the A2 group. Two weeks of A2 milk consumption led to significant alterations in gut microbiota, promoting beneficial microbes and related functions. A2 milk could be a suitable alternative for subjects who experience milk-intake-related GI discomfort.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seulgi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyung Su Kim
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Min Hee Park
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Sang Hee Lee
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
de la O V, Fernández-Cruz E, Valdés A, Cifuentes A, Walton J, Martínez JA. Exhaustive Search of Dietary Intake Biomarkers as Objective Tools for Personalized Nutrimetabolomics and Precision Nutrition Implementation. Nutr Rev 2025; 83:925-942. [PMID: 39331531 DOI: 10.1093/nutrit/nuae133] [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: 09/29/2024] Open
Abstract
OBJECTIVE To conduct an exhaustive scoping search of existing literature, incorporating diverse bibliographic sources to elucidate the relationships between metabolite biomarkers in human fluids and dietary intake. BACKGROUND The search for biomarkers linked to specific dietary food intake holds immense significance for precision health and nutrition research. Using objective methods to track food consumption through metabolites offers a more accurate way to provide dietary advice and prescriptions on healthy dietary patterns by healthcare professionals. An extensive investigation was conducted on biomarkers associated with the consumption of several food groups and consumption patterns. Evidence is integrated from observational studies, systematic reviews, and meta-analyses to achieve precision nutrition and metabolism personalization. METHODS Tailored search strategies were applied across databases and gray literature, yielding 158 primary research articles that met strict inclusion criteria. The collected data underwent rigorous analysis using STATA and Python tools. Biomarker-food associations were categorized into 5 groups: cereals and grains, dairy products, protein-rich foods, plant-based foods, and a miscellaneous group. Specific cutoff points (≥3 or ≥4 bibliographic appearances) were established to identify reliable biomarkers indicative of dietary consumption. RESULTS Key metabolites in plasma, serum, and urine revealed intake from different food groups. For cereals and grains, 3-(3,5-dihydroxyphenyl) propanoic acid glucuronide and 3,5-dihydroxybenzoic acid were significant. Omega-3 fatty acids and specific amino acids showcased dairy and protein foods consumption. Nuts and seafood were linked to hypaphorine and trimethylamine N-oxide. The miscellaneous group featured compounds like theobromine, 7-methylxanthine, caffeine, quinic acid, paraxanthine, and theophylline associated with coffee intake. CONCLUSIONS Data collected from this research demonstrate potential for incorporating precision nutrition into clinical settings and nutritional advice based on accurate estimation of food intake. By customizing dietary recommendations based on individualized metabolic profiles, this approach could significantly improve personalized food consumption health prescriptions and support integrating multiple nutritional data.This article is part of a Nutrition Reviews special collection on Precision Nutrition.
Collapse
Affiliation(s)
- Victor de la O
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Faculty of Health Sciences, International University of La Rioja, 26006, Logroño, Spain
| | - Edwin Fernández-Cruz
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Faculty of Health Sciences, International University of La Rioja, 26006, Logroño, Spain
| | - Alberto Valdés
- Foodomics Lab, Institute of Food Science Research, Spanish National Research Council, 28049, Madrid, Spain
| | - Alejandro Cifuentes
- Foodomics Lab, Institute of Food Science Research, Spanish National Research Council, 28049, Madrid, Spain
| | - Janette Walton
- Department of Biological Sciences, Munster Technological University, Cork, Republic of Ireland
| | - J Alfredo Martínez
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 28049, Madrid, Spain
- Department of Medicine and Endocrinology, Campus of Soria, University of Valladolid, Valladolid, Spain
| |
Collapse
|
3
|
Ofori‐Kwafo A, Sigdel I, Al Mamun E, Zubcevic J, Tang Y. Gut-on-a-chip platforms: Bridging in vitro and in vivo models for advanced gastrointestinal research. Physiol Rep 2025; 13:e70356. [PMID: 40323242 PMCID: PMC12051376 DOI: 10.14814/phy2.70356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
The gastrointestinal (GI) tract plays a critical role in nutrient absorption, immune responses, and overall health. Traditional models such as two-dimensional cell cultures have provided valuable insights but fail to replicate the dynamic and complex microenvironment of the human gut. Gut-on-a-chip platforms, which incorporate cells located in the gut into microfluidic devices that simulate peristaltic motion and fluid flow, represent a significant advancement in modeling GI physiology and diseases. This review discusses the evolution of gut-on-a-chip technology, from simple cellular mono-cultures models to more sophisticated systems incorporating bi-cultures and tri-cultures that enable studies of drug metabolism, disease modeling, and gut-microbiome interactions. Although challenges remain, including maintaining long-term cell viability and replicating immune responses, these platforms hold great potential for advancing personalized medicine and improving drug discovery efforts targeting gastrointestinal disorders.
Collapse
Affiliation(s)
- Awurama Ofori‐Kwafo
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Indira Sigdel
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Earshed Al Mamun
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Jasenka Zubcevic
- University of South Florida Center for Microbiome ResearchMicrobiomes InstituteTampaFloridaUSA
- Department of Neurosurgery and Brain RepairUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yuan Tang
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| |
Collapse
|
4
|
Rukavina Mikusic NL, Prince PD, Choi MR, Chuffa LGA, Simão VA, Castro C, Manucha W, Quesada I. Microbiota, mitochondria, and epigenetics in health and disease: converging pathways to solve the puzzle. Pflugers Arch 2025; 477:635-655. [PMID: 40111427 DOI: 10.1007/s00424-025-03072-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Dysbiosis, which refers to an imbalance in the composition of the gut microbiome, has been associated with a range of metabolic disorders, including type 2 diabetes, obesity, and metabolic syndrome. Although the exact mechanisms connecting gut dysbiosis to these conditions are not fully understood, various lines of evidence strongly suggest a substantial role for the interaction between the gut microbiome, mitochondria, and epigenetics. Current studies suggest that the gut microbiome has the potential to affect mitochondrial function and biogenesis through the production of metabolites. A well-balanced microbiota plays a pivotal role in supporting normal mitochondrial and cellular functions by providing metabolites that are essential for mitochondrial bioenergetics and signaling pathways. Conversely, in the context of illnesses, an unbalanced microbiota can impact mitochondrial function, leading to increased aerobic glycolysis, reduced oxidative phosphorylation and fatty acid oxidation, alterations in mitochondrial membrane permeability, and heightened resistance to cellular apoptosis. Mitochondrial activity can also influence the composition and function of the gut microbiota. Because of the intricate interplay between nuclear and mitochondrial communication, the nuclear epigenome can regulate mitochondrial function, and conversely, mitochondria can produce metabolic signals that initiate epigenetic changes within the nucleus. Given the epigenetic modifications triggered by metabolic signals from mitochondria in response to stress or damage, targeting an imbalanced microbiota through interventions could offer a promising strategy to alleviate the epigenetic alterations arising from disrupted mitochondrial signaling.
Collapse
Affiliation(s)
- Natalia Lucia Rukavina Mikusic
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Paula Denise Prince
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Químicas, Cátedra de Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina.
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina.
| | - Luiz Gustavo A Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Vinícius Augusto Simão
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Claudia Castro
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
- Laboratorio de Farmacología Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina.
| | - Isabel Quesada
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
5
|
Zhang Y, Ma R, Du X, He X, Zhang Y, Ma N, Liu H, Zhao X. Impact of bacteroides uniformis on fatty liver hemorrhagic syndrome in dawu golden phoenix laying hens: modulation of gut microbiota and arachidonic acid metabolism. Front Microbiol 2025; 16:1560887. [PMID: 40356654 PMCID: PMC12066428 DOI: 10.3389/fmicb.2025.1560887] [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: 01/20/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
This study explored the impact of Bacteroides uniformis (B. uniformis) on fatty liver hemorrhagic syndrome (FLHS) induced by a high-energy and low-protein (HELP) diet in laying hens, mainly focusing on hepatic lipid metabolism, gut microbiota, and arachidonic acid (AA) metabolism. A total of 120 Dawu Golden Phoenix laying hens (210-day-old) were randomly divided into four groups. The control group (CON) was fed a standard diet and received a daily gavage of PBS, while the other groups were fed with a HELP diet to induce FLHS and received a daily gavage of PBS (MOD), 1 × 109 CFU/ml B. uniformis (BUL), and 1 × 1011 CFU/ml B. uniformis (BUH) for 70 days. All hens were administered 1 ml daily by gavage. Each group had 6 replications with 5 hens per replication. The results showed that B. uniformis increased the egg production rate and feed conversion ratio and decreased body weight, liver index, and abdominal fat rate (p < 0.05). B. uniformis treatment reduced liver lipid accumulation by reducing the levels of Triglyceride (TG), Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), alanine transaminases (ALT), and aspartate transaminases (AST) in serum and significantly elevated high-density lipoprotein cholesterol (HDL-C) (p < 0.05). The results indicated that B. uniformis altered the gut microbiota. Specifically, the abundance of Bacteroides was higher, and the relative abundances of Treponema, Helicobacter, and Spirochaetota were lower than those of the MOD group (p < 0.05). Moreover, targeted metabolomic analysis showed that supplementation of B. uniformis significantly elevated 6-keto-PGF1α and AA levels, along with significantly reduced levels of thromboxane B2 (TXB2), leukotriene D4 (LTD4), 8-isoprostaglandin F2α (8-iso-PGF2α), 12S-hydroxyeicosatetraenoic acid (12S-HETE), 15S-hydroxyeicosatetraenoic acid (15S-HETE), 9-S-hydroxy-octadecadienoic acid (9S-HODE), and 13-S-hydroxy-octadecadienoic acid (13S-HODE) (p < 0.05). In conclusion, the oral intake of B. uniformis can improve liver function, gut microbiota, and AA metabolism, thereby helping to ameliorate FLHS in Dawu Golden Phoenix laying hens.
Collapse
Affiliation(s)
- Yu Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Rongfei Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Xicui Du
- Hebei Jinkun Animal Pharmaceutical Co. Ltd., Xinji, China
| | - Xin He
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yan Zhang
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Ning Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Hailong Liu
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| |
Collapse
|
6
|
Fu L, Baranova A, Cao H, Zhang F. Gut microbiome links obesity to type 2 diabetes: insights from Mendelian randomization. BMC Microbiol 2025; 25:253. [PMID: 40289103 PMCID: PMC12034155 DOI: 10.1186/s12866-025-03968-8] [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: 10/27/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Research has established links between the gut microbiome (GM) and both obesity and type 2 diabetes (T2D), which is much discussed, but underexplored. This study employed body mass index (BMI) as the measurement of obesity to delve deeper into the correlations from a genetic perspective. METHODS We performed the Mendelian randomization (MR) analysis to examine the causal effects of GM on T2D and BMI, and vice versa. Genome-wide association study (GWAS) summary datasets were utilized for the analysis, including T2D (N = 933,970), BMI (N = 806,834), and two GM datasets from the international consortium MiBioGen (211 taxa, N = 18,340) and the Dutch Microbiome Project (DMP) (207 taxa, N = 7,738). These datasets mainly cover European populations, with additional cohorts from Asia and other regions. To further explore the potential mediating role of GM in the connections between BMI and T2D, their interaction patterns were summarized into a network. RESULTS MR analysis identified 9 taxa that showed protective properties against T2D. Seven species were within the Firmicutes and Bacteroidales phyla in the DMP, and two were from the MiBioGen (Odds Ratio (OR): 0.94-0.95). Conversely, genetic components contributing to the abundance of 12 taxa were associated with increased risks of T2D (OR: 1.04-1.12). Furthermore, T2D may elevate the abundance of seven taxa (OR: 1.03-1.08) and reduce the abundance of six taxa (OR: 0.93-0.97). In the analysis of the influence of the genetic component of BMI on GM composition, BMI affected 52 bacterial taxa, with 28 decreasing (OR: 0.75-0.92) and 24 increasing (OR: 1.08-1.27). Besides, abundances of 25 taxa were negatively correlated with BMI (OR: 0.95-0.99), while positive correlations were detected for 14 taxa (OR: 1.01-1.05). Notably, we uncovered 11 taxa genetically associated with both BMI and T2D, which formed an interactive network. CONCLUSIONS Our findings provide evidence for the GM-mediated links between obesity and T2D. The identification of relevant GM taxa offers valuable insights into the potential role of the microbiome in these diseases.
Collapse
Affiliation(s)
- Li Fu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
- Research Centre for Medical Genetics, Moscow, 115478, Russia
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
7
|
Dhurandhar Y, Tomar S, Das A, Prajapati JL, Singh AP, Bodake SH, Namdeo KP. Chronic inflammation in obesity and neurodegenerative diseases: exploring the link in disease onset and progression. Mol Biol Rep 2025; 52:424. [PMID: 40274681 DOI: 10.1007/s11033-025-10509-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Obesity, a worldwide health emergency, is defined by excessive fat accumulation and significantly impacts metabolic health. In addition to its recognized association with cardiovascular disease, diabetes, and other metabolic illnesses, recent studies have revealed the connection between obesity and neurodegeneration. The main reason for this link is inflammation caused by the growth of fat tissue, which activates harmful processes that affect how the brain works. Fat tissue, particularly the fat around the organs, produces various substances that cause inflammation, such as cytokines (TNF-α, IL-6), adipokines (leptin, resistin), and free fatty acids. These chemicals cause low-grade, persistent systemic inflammation, which is becoming more widely acknowledged as a major factor in peripheral metabolic dysfunction and pathology of the central nervous system (CNS). Inflammatory signals in the brain cause neuroinflammatory reactions that harm neuronal structures, change neuroplasticity, and disrupt synaptic function. When obesity-related inflammation is present, the brain's resident immune cells, known as microglia, become hyperactivated, which can lead to the production of neurotoxic chemicals, which can cause neuronal death. This neuroinflammation exacerbates the negative effects of obesity on brain health and is linked to cognitive decline, Alzheimer's disease, and other neurodegenerative disorders. Moreover, the blood-brain barrier (BBB) exhibits increased permeability during inflammatory states, facilitating the infiltration of peripheral immune cells and cytokines into the brain, hence exacerbating neurodegeneration. Adipose tissue is a source of chronic inflammatory mediators, which are examined in this review along with the molecular pathways that connect inflammation brought on by obesity to neurodegeneration. Additionally, it addresses various anti-inflammatory treatment approaches, including lifestyle modifications, anti-inflammatory medications, and gut microbiota modulation, to lessen the metabolic and neurological effects of obesity. Recognizing the link between obesity and inflammation opens up new opportunities for early intervention and the development of targeted treatments to prevent or alleviate neurodegenerative disorders.
Collapse
Affiliation(s)
- Yogita Dhurandhar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India
| | - Shubham Tomar
- Pharmacovigilance Programme of India, Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, Ghaziabad, Uttar Pradesh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India
| | - Jeevan Lal Prajapati
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India
| | - As Pee Singh
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India
| | - Surendra H Bodake
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India
| | - Kamta P Namdeo
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, 495009, India.
| |
Collapse
|
8
|
Fang Z, Chang S, Niu P, Wang C, Zhang J. Multidimensional-based exploration of gut microbial and metabolite differences in patients with recurrent stroke. Neuroscience 2025; 572:35-48. [PMID: 39914520 DOI: 10.1016/j.neuroscience.2025.02.004] [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: 07/11/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 03/11/2025]
Abstract
This study aims to explore the differences in gut microbes and their metabolites between patients with original and recurrent stroke, providing insights and justification for the diagnosis and prevention of ischemic stroke progression from the perspective of the gut microbiota-metabolite-brain axis. In this study, fecal samples were collected from patients with Original stroke (Os) and patients with Recurrent stroke (Rs) to assess differences in gut microbiota and to screen for different metabolites that reveal the physiological changes related to the recurrent of ischemic stroke. The results found that there was no significant change in Alpha diversity between the two groups. Beta diversity analysis revealed slight changes in community composition between two groups (Bray-Curtis), although their overall microbial abundance may not have changed (UniFrac). Compared with Os patients, Prevotella, Lachnospiraceae_UCG-010, Holdemanella, and Coprococcus were significantly depleted in the Rs group. Correlation analysis showed that the risk of stroke recurrence was negatively correlated with Lachnospiraceae_UCG-010. In Rs group, metabolites such as carbohydrates and terpene lactones were up-regulated, while those of sesquiterpenoids, triterpenoids, and fatty acids and their couplings were down-regulated. These metabolites are significantly enriched in the pathways of arachidonic acid metabolism, betaine biosynthesis, and linoleic acid metabolism. Compared with the Os, Rs was mainly characterized by minor destruction of anaerobic bacteria and significant depletion of SCFAs-producing bacteria. In addition, the related compounds involved in arachidonic acid metabolism and linoleic acid metabolism pathway may be associated with the progression of ischemic stroke.
Collapse
Affiliation(s)
- Zongwei Fang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Sijie Chang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Chunhua Wang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
9
|
Na ES. Epigenetic Mechanisms of Obesity: Insights from Transgenic Animal Models. Life (Basel) 2025; 15:653. [PMID: 40283207 PMCID: PMC12028693 DOI: 10.3390/life15040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Obesity is a chronic disease with prevalence rates that have risen dramatically over the past four decades. This increase is not due to changes in the human genome but rather to environmental factors that promote maladaptive physiological responses. Emerging evidence suggests that external influences, such as high-fat diets, modify the epigenome-the interface between genes and the environment-leading to persistent alterations in energy homeostasis. This review explores the role of epigenetic mechanisms in obesity, emphasizing insights from transgenic animal models and clinical studies. Additionally, we discuss the evolution of obesity research from homeostatic to allostatic frameworks, highlighting key neuroendocrine regulators of energy balance.
Collapse
Affiliation(s)
- Elisa S Na
- School of Social Work, Psychology, & Philosophy, Texas Woman's University, Denton, TX 76209, USA
| |
Collapse
|
10
|
da Mota JC, Smaira FI, Julio JMG, Carvalho BGD, Carvalho LM, Ribeiro AA, Souza LL, Borba EF, Roschel H, Gualano B, Nicoletti CF. Association between excess body weight and disordered eating attitude among women living with systemic lupus erythematosus. Lupus 2025:9612033251332830. [PMID: 40199379 DOI: 10.1177/09612033251332830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The study aimed to: (i) characterize dietary intake and identify disorders eating attitudes in women with SLE, (ii) evaluate possible differences in both dietary intake and disorders eating attitudes in patients with SLE according to nutritional status, (iii) investigate possible associations between eating disorders attitudes, anthropometric characteristics and food consumption. METHODS This cross-sectional study included 46 premenopausal female patients (18-40 years), with inactive disease, using prednisone <10 mg/day and hydroxychloroquine at a stable dose. Patients were allocated into two groups according to their nutritional status by body mass index (BMI): normal weight (BMI between 18.5 and 24.9 kg/m2) and excess weight (BMI >25 kg/m2). Food consumption was assessed according to the processing level and energy and macronutrient content. The Disordered Eating Attitude Scale (DEAS) was applied. RESULTS Patients with excess weight had a higher DEAS score when compared to those with normal weight (34 ± 8.7 vs 25 ± 5.9, p = .001). A higher percentage of patients with excess weight demonstrated disturbance in their relationship with food and concerns about food and weight gain versus those with normal weight. DEAS score was positively associated with BMI, abdominal circumference, and fat mass percentage and negatively associated with lipid intake. CONCLUSION Disordered eating attitudes differ in SLE patients according to nutritional status, and those with excess weight show higher DEAS scores, which may be related to food and weight gain.
Collapse
Affiliation(s)
- Jhulia Cnl da Mota
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Fabiana I Smaira
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Juliana Maria Gonçalves Julio
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Beatriz Garcia de Carvalho
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Lucas M Carvalho
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Amanda A Ribeiro
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia L Souza
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Eduardo F Borba
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
| | - Hamilton Roschel
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
- Laboratory of Assessment and Conditioning in Rheumatology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruno Gualano
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
- Laboratory of Assessment and Conditioning in Rheumatology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carolina F Nicoletti
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
11
|
Li Z, Ye Y, Lai S, Bao J, Fu A. Puerarin affects adipose lipolysis and ameliorates obesity by gut microbiota. Front Microbiol 2025; 16:1567339. [PMID: 40248428 PMCID: PMC12003275 DOI: 10.3389/fmicb.2025.1567339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Background Obesity has become a widespread metabolic disorder, marked by its escalating global prevalence. Puerarin, extracted from Pueraria lobata, one of the traditional homologies of medicine and food, demonstrates anti-obesity properties. Nonetheless, the mechanisms through which puerarin exerts its anti-obesity effects remain to be elucidated. This study seeks to highlight the potential application of puerarin in obesity management and to investigate the underlying mechanisms involving gut microbiota and lipid metabolism. Method Different doses of puerarin were administered to the high-fat diet mice model, and the structure and composition of gut microbiota were analyzed using 16S rRNA sequencing. Q-PCR evaluated the expression of genes related to lipid metabolism. Result Our findings demonstrate that puerarin treatment significantly reduces body weight and epithelial and beige fat mass. Furthermore, puerarin alters the structure and composition of gut microbiota, which is associated with metabolism. Additionally, puerarin treatment significantly upregulates gene expression, fatty acid transport protein 5 (FATP5) hormone-sensitive lipase (HSL), adipose triglyceride lipase (ATGL) in epithelial and beige adipose. Conclusion Puerarin demonstrates potential in ameliorating obesity by changing the structure and composition of gut microbiota, which enhances the transport of fatty acid and triglycerides hydrolysis within adipose tissue. This study provides a novel perspective on puerarin as a dietary supplement for obesity.
Collapse
Affiliation(s)
- Zhaoyi Li
- Institute of Hepatology and Epidemiology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingyan Ye
- Hangzhou Linan District People's Hospital, Hangzhou, China
| | - Shanglei Lai
- Department of Medical Research Center, Shaoxing People's Hospital, Shaoxing, China
| | - Jianfeng Bao
- Institute of Hepatology and Epidemiology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Hepatology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ai Fu
- Institute of Hepatology and Epidemiology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Hepatology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
12
|
Yuan X, Wang J, Wang W, Song Y, Wu J, Du R. Microbiome alterations in primary Sjögren's syndrome: Regional dysbiosis and microbiome-targeted therapeutic strategies. Clin Immunol 2025; 273:110444. [PMID: 39947272 DOI: 10.1016/j.clim.2025.110444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by diverse clinical manifestations. While xerophthalmia and xerostomia are hallmark symptoms, the disease often involves multiple organ systems, including the kidneys, lungs, nervous system, and gastrointestinal tract, leading to systemic morbidity in severe cases. Despite extensive research, the precise pathogenesis of pSS remains unclear, likely involving infectious, hormonal, and genetic factors. Emerging evidence highlights the microbiome as a key contributor to autoimmune diseases, including pSS. Dysbiosis in the oral, ocular, gut, and genital microbiomes plays a critical role in disease onset, progression, and variability. This review summarizes current findings on microbiome alterations in pSS, emphasizing their role in pathogenesis and clinical features, and explores microbiome-targeted therapies. Understanding the role of the microbiome in pSS pathophysiology could advance disease management and inspire targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xujing Yuan
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jun Wang
- Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiwei Wang
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - You Song
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jiajia Wu
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Rong Du
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
13
|
Aleali MS, Mahapatro A, Maddineni G, Paladiya R, Jeanty H, Mohanty E, Mirchandani M, Jahanshahi A, Devulapally P, Alizadehasl A, Tariq MD, Hosseini Jebelli SF, Aliabadi AY, Hashemi SM, Amini-Salehi E. The impact of gut microbiome modulation on anthropometric indices in metabolic syndrome: an umbrella review. Ann Med Surg (Lond) 2025; 87:2263-2277. [PMID: 40212162 PMCID: PMC11981403 DOI: 10.1097/ms9.0000000000003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/25/2025] [Indexed: 04/13/2025] Open
Abstract
Background Metabolic syndrome (MetS) is a complex disorder characterized by a cluster of metabolic risk factors. Recent research highlights the gut microbiome's role in metabolic regulation, suggesting that modulation through probiotics, prebiotics, and synbiotics may provide a novel approach to managing MetS. This umbrella review aims to integrate insights from existing meta-analyses to explore how changes in gut microbiota influence key body measurement indicators in individuals with MetS. Methods A systematic search of PubMed, Scopus, and Web of Science databases identified meta-analyses that assessed the impact of probiotics, prebiotics, or synbiotics on anthropometric indices in MetS patients. Results The results indicated that microbial therapy leads to a significant reduction in body mass index (BMI) (SMD: -0.22; 95% CI: -0.35 to -0.09; P < 0.01) and waist circumference (WC) (SMD: -0.47; 95% CI: -0.80 to -0.15; P < 0.01). However, microbial therapy did not significantly affect body fat mass (SMD: -0.30; 95% CI: -0.64 to 0.02; P = 0.06), body fat percentage (SMD: -0.29; 95% CI: -0.62 to 0.03; P = 0.07), waist-to-hip ratio (SMD: -0.09; 95% CI: -0.46 to 0.28; P = 0.63), and weight (SMD: -0.06; 95% CI: -0.21 to 0.08; P = 0.37). Conclusions Gut microbial modulation, mainly through probiotics and synbiotics, shows promise in reducing BMI and WC in MetS patients. However, its effects on other anthropometric indices remain uncertain, warranting further high-quality research to fully understand microbial interventions' therapeutic potential.
Collapse
Affiliation(s)
- Maryam Sadat Aleali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Ruchir Paladiya
- University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Herby Jeanty
- The Brooklyn Hospital Center, Brooklyn, New York, USA
| | - Elan Mohanty
- Gautam Maddineni, MD Mary Medical Center Apple Valley, Apple Valley, California, USA
| | | | - Ali Jahanshahi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Pavan Devulapally
- Social Determinants of Health Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Azin Alizadehasl
- Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | | | - Seyyed Mohammad Hashemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
14
|
Ramos-Lopez O. Epigenomic mechanisms of dietary prescriptions for obesity therapy. Epigenomics 2025; 17:423-434. [PMID: 40025880 PMCID: PMC11980491 DOI: 10.1080/17501911.2025.2473309] [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: 06/27/2024] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Dietary modification is a cornerstone and a primary goal for weight loss, whose effects may be related to epigenetic phenomena. In this literature review, a comprehensive search without time restriction was performed in PubMed/Medline, Cochrane, SciELO, and Scopus databases to identify epigenetic signatures related to obesity outcomes upon dietary advice. In this context, experimental studies and clinical trials have identified certain DNA methylation marks, miRNA expression profiles and histone modifications putatively associated with adiposity outcomes after different nutritional interventions. These include traditional dietary patterns, diets with different macronutrient compositions, and supplementation with fatty acids, amino acids and derivatives, methyl donors, vitamins and minerals, probiotics and prebiotics, and bioactive food compounds. Some of these epigenetic signatures have been mapped to genes involved in food intake control, adipogenesis, lipolysis, fatty acid oxidation, body fat deposition, and gut microbiota modulation. However, additional studies are still required to address dosage and follow-up variability, validation of epigenetic marks, genome-wide approaches, and appropriate statistical settings. Although more investigation is required, these insights may contribute to the characterization of epigenetic biomarkers of body weight regulation toward the prescription of tailored dietary strategies targeting the epigenome for a more precise obesity management and control.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana, Baja California, Mexico
| |
Collapse
|
15
|
Li W, Zeng Y, Zhong J, Hu Y, Xiong X, Zhou Y, Fu L. Probiotics Exert Gut Immunomodulatory Effects by Regulating the Expression of Host miRNAs. Probiotics Antimicrob Proteins 2025; 17:557-568. [PMID: 39754704 DOI: 10.1007/s12602-024-10443-9] [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] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
Probiotics exert a diverse range of immunomodulatory effects on the human gut immune system. These mechanisms encompass strengthening the intestinal mucosal barrier, inhibiting pathogen adhesion and colonization, stimulating immune modulation, and fostering the production of beneficial substances. As a result, probiotics hold significant potential in the prevention and treatment of various conditions, including inflammatory bowel disease and colorectal cancer. A pivotal mechanism by which probiotics achieve these effects is through modulating the expression of host miRNAs. miRNAs, non-coding RNA molecules, are vital regulators of fundamental biological processes like cell growth, differentiation, and apoptosis. By interacting with mRNAs, miRNAs can either promote their degradation or repress their translation, thereby regulating gene expression post-transcriptionally and modulating the immune system. This review provides a comprehensive overview of how probiotics modulate gut immune responses by altering miRNA expression levels, both upregulating and downregulating specific miRNAs. It further delves into how this modulation impacts the host's resistance to pathogens and susceptibility to diseases, offering a theoretical foundation and practical insights for the clinical utilization of probiotics in disease prevention and therapy.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Yongwei Zeng
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Jiayu Zhong
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Youyu Hu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yingshun Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Public Center of Experimental Technology of Pathogen Biology Technology Platform, Southwest Medical University, Luzhou, 646000, China.
| | - Li Fu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
16
|
Ponsuksili S, Hadlich F, Li S, Trakooljul N, Reyer H, Oster M, Abitew YA, Sommerfeld V, Rodehutscord M, Wimmers K. DNA methylation dynamics in the small intestine of egg-selected laying hens along egg production stages. Physiol Genomics 2025; 57:125-139. [PMID: 39869094 DOI: 10.1152/physiolgenomics.00063.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
Decades of artificial selection have markedly enhanced egg production efficiency, yet the epigenetic underpinnings, notably DNA methylation dynamics in the gut, remain largely unexplored. Here, we investigate how breeds and developmental stages influence DNA methylation profiles in laying hens, and their potential relationship to laying performance and gut health. We compared two highly selected laying hen strains, Lohmann Brown-Classic (LB) and Lohmann Selected Leghorn-Classic (LSL), which exhibited similar egg production but divergent physiological, metabolic, and immunological characteristics. Our sampling encompassed key developmental stages: the pullet stage (10 and 16 wk old), peak production (24 and 30 wk old), and later stage (60 wk old) (n = 99; 10 per group), allowing us to elucidate the temporal dynamics of epigenetic regulation. Our findings highlight a crucial window of epigenetic modulation during the prelaying period, characterized by stage-specific methylation alterations and the involvement of predicted transcription factor motifs within methylated regions. This observation was consistent with the expression patterns of DNA methyltransferases (DNMTs), including DNMT1, DNMT3A, and DNMT3B. In addition, a higher methylation level was observed in specific loci or regions in the LSL compared with the LB strain. Notably, we uncover strain-specific differences in methylation levels, particularly pronounced in genomic regions associated with intestinal integrity, inflammation, and energy homeostasis. Our research contributes to the multidisciplinary framework of epigenetics and egg-laying performance, offering valuable implications for poultry production and welfare.NEW & NOTEWORTHY Our study reveals key methylation changes in the jejunum mucosa of laying hens across developmental stages and between strains, with implications for gut health, immune function, and egg production. These findings highlight a crucial role of epigenetic regulation in optimizing performance.
Collapse
Affiliation(s)
| | - Frieder Hadlich
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Shuaichen Li
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Nares Trakooljul
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | | | - Vera Sommerfeld
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | | | - Klaus Wimmers
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| |
Collapse
|
17
|
Yılmaz HÖ, Şahin K, Ayvaz H. A comparative study of cognitive function and reaction time in obese and non-obese adults. Neurol Res 2025; 47:201-210. [PMID: 39904741 DOI: 10.1080/01616412.2025.2462739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE Obesity may negatively affect the physical health and cognitive functions of individuals and delay their reaction time to stimuli. However, the association among obesity, cognitive functions, and reaction times is yet to be fully elucidated. The aim of this study was to assess the effect of obesity on cognitive functions and visual and auditory reaction times in adults. METHODS Data of 100 participants (50 obese and 50 normal) were analyzed in the study. Anthropometric parameters and 24-h dietary recall data were recorded. The Montreal Cognitive Assessment (MoCA) was used to evaluate the cognitive functions, Simple Reaction Time Task (SRTT)-Visual and SRTT-Auditory were used to assess visual and auditory reaction times of the participants, respectively. RESULTS The mean MoCA score of the obese was significantly lower than normal (17.46 and 25.22, respectively; p < 0.001). In addition, the mean auditory (p < 0.001) and visual (p < 0.05) reaction times of obese were significantly longer than normal. Similarly, this condition was also observed for the fastest and lowest values of auditory and visual reaction times. Additionally, obesity caused a decrease in the MoCA score (β = -0.762; p < 0.001) and delayed visual (β = 0.423; p < 0.001) and auditory (β = 0.590; p < 0.001) reactions. The negative effect of obesity was maintained after controlling for potential factors (MoCA, β = -0.594; p < 0.001; SRTT-Auditory, β = 0.409; p < 0.01; SRTT-Visual, β = 0.330; p < 0.05). CONCLUSION Obese participants showed worse cognitive, auditory and visual performance. Additional research will be necessary in the future to shed light on the fundamental mechanisms involved.
Collapse
Affiliation(s)
- Hacı Ömer Yılmaz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Bandırma Onyedi Eylul University, Balıkesir, Türkiye
| | - Kezban Şahin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Bandırma Onyedi Eylul University, Balıkesir, Türkiye
| | - Hilal Ayvaz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gümüşhane University, Gümüşhane, Türkiye
| |
Collapse
|
18
|
Lan F, Wang X, Zhou Q, Li X, Jin J, Zhang W, Wen C, Wu G, Li G, Yan Y, Yang N, Sun C. Deciphering the coordinated roles of the host genome, duodenal mucosal genes, and microbiota in regulating complex traits in chickens. MICROBIOME 2025; 13:62. [PMID: 40025569 PMCID: PMC11871680 DOI: 10.1186/s40168-025-02054-5] [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] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/01/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The complex interactions between host genetics and the gut microbiome are well documented. However, the specific impacts of gene expression patterns and microbial composition on each other remain to be further explored. RESULTS Here, we investigated this complex interplay in a sizable population of 705 hens, employing integrative analyses to examine the relationships among the host genome, mucosal gene expression, and gut microbiota. Specific microbial taxa, such as the cecal family Christensenellaceae, which showed a heritability of 0.365, were strongly correlated with host genomic variants. We proposed a novel concept of regulatability ( r b 2 ), which was derived from h2, to quantify the cumulative effects of gene expression on the given phenotypes. The duodenal mucosal transcriptome emerged as a potent influencer of duodenal microbial taxa, with much higher r b 2 values (0.17 ± 0.01, mean ± SE) than h2 values (0.02 ± 0.00). A comparative analysis of chickens and humans revealed similar average microbiability values of genes (0.18 vs. 0.20) and significant differences in average r b 2 values of microbes (0.17 vs. 0.04). Besides, cis ( h cis 2 ) and trans heritability ( h trans 2 ) were estimated to assess the effects of genetic variations inside and outside the cis window of the gene on its expression. Higher h trans 2 values than h cis 2 values and a greater prevalence of trans-regulated genes than cis-regulated genes underscored the significant role of loci outside the cis window in shaping gene expression levels. Furthermore, our exploration of the regulatory effects of duodenal mucosal genes and the microbiota on 18 complex traits enhanced our understanding of the regulatory mechanisms, in which the CHST14 gene and its regulatory relationships with Lactobacillus salivarius jointly facilitated the deposition of abdominal fat by modulating the concentration of bile salt hydrolase, and further triglycerides, total cholesterol, and free fatty acids absorption and metabolism. CONCLUSIONS Our findings highlighted a novel concept of r b 2 to quantify the phenotypic variance attributed to gene expression and emphasize the superior role of intestinal mucosal gene expressions over host genomic variations in elucidating host‒microbe interactions for complex traits. This understanding could assist in devising strategies to modulate host-microbe interactions, ultimately improving economic traits in chickens.
Collapse
Affiliation(s)
- Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiqiong Wang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiaming Jin
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wenxin Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guiqin Wu
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Guangqi Li
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Yiyuan Yan
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
19
|
Yang D, Xu HX, Wang WJ, Yin ZP, Chen JG, Yuan E, Zhang QF. Roselle Extract Ameliorates Abnormal Glucolipid Metabolism and Gut Microbiota in Obese Mice Fed With High-Fat Diet. Mol Nutr Food Res 2025; 69:e202400756. [PMID: 39935166 DOI: 10.1002/mnfr.202400756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
Roselle extract (RE) is rich in anthocyanins and chlorogenic acids. This study investigated the health-promoting effects of RE on lipid metabolism, oxidative stress, glycometabolism, and gut microbiota in obese mice fed a high-fat diet (HFD). The obesity model was induced by feeding mice a HFD, with RE supplementation added to their drinking water at concentrations of 2 and 4 mg/mL for 12 weeks. RE significantly reduced body weight gain and fat accumulation compared to the control group, alleviated hepatic steatosis, and improved insulin sensitivity. Additionally, RE restored antioxidative enzyme activities such as SOD and GSH-PX while reducing MDA levels. Transcriptomic analysis of the liver was performed to evaluate gene expression related to lipid metabolism, particularly in the PPAR signaling pathway. Gut microbiota analysis showed that RE increased beneficial bacteria and reduced the Firmicutes-to-Bacteroidetes ratio, suggesting an improvement in gut dysbiosis caused by the HFD. RE enhanced lipid metabolism, reduced oxidative stress, and improved insulin sensitivity in obese mice, potentially through modulation of the PPAR signaling pathway and gut microbiota, suggesting its potential as a therapeutic candidate for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Dan Yang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Hai-Xia Xu
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Wen-Jun Wang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Zhong-Ping Yin
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Ji-Guang Chen
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - En Yuan
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qing-Feng Zhang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
20
|
Dunyach-Remy C, Pouget C, Pers YM, Gaujoux-Viala C, Demattei C, Salipante F, Grenga L, Armengaud J, Lavigne JP, Jorgensen C. Participation of gut microbiota and bacterial translocation in chronic systemic inflammation in recently diagnosed rheumatoid arthritis patients. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100366. [PMID: 40123592 PMCID: PMC11928969 DOI: 10.1016/j.crmicr.2025.100366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
The objective of this study was to investigate the link between gut microbiota (GM) dysbiosis, gut inflammation, and bacterial translocation (BT) in recently diagnosed rheumatoid arthritis (RA). This case-control, observational study prospectively recruited recently diagnosed (<12 months) RA patients and age-matched healthy controls (HC) from two French hospitals between July 2014 to March 2018. The primary objective was to investigate GM composition in each group using 16S rRNA sequencing and metaproteomics approaches. Three plasmatic BT markers (sCD14, LPS-binding protein, and number of 16S rRNA gene copies) and one intestinal permeability marker (I-FABP) were quantified in blood samples. Twenty-five were included in each group, and 50 stools and blood samples were analyzed. 16S rRNA gene analysis showed an decrease in Coprococcus in RA patients after Body Mass Index and HLA status. Circulating bacterial DNA (number of copies of the 16S rRNA gene) and plasmatic I-FABP were higher in RA patients compared to HCs (p < 0.01), indicating increased BT and intestinal permeability in these patients. Metaproteomics from stool samples highlighted an increased host humoral immune response in RA, with elevated levels of inflammatory proteins (azurocidin, cathepsin G, neutrophil defensing 1). Gut inflammation may contribute to increased intestinal permeability, leading to BT into the systemic circulation and thus chronic inflammation.
Collapse
Affiliation(s)
- Catherine Dunyach-Remy
- Bacterial Virulence and Chronic Infections, INSERM U1047, Univ Montpellier, Department of Microbiology and Hospital Hygiene, CHU Nîmes, Nîmes, France
| | - Cassandra Pouget
- Bacterial Virulence and Chronic Infections, INSERM U1047, Univ Montpellier, Department of Microbiology and Hospital Hygiene, CHU Nîmes, Nîmes, France
| | - Yves-Marie Pers
- Department of Rheumatology, Stem cells, Cellular plasticity, Regenerative medicine and Immunotherapies, IRMB, INSERM UMR1183, University of Montpellier & University Hospital of Montpellier, Montpellier, France
| | - Cécile Gaujoux-Viala
- Desbrest Institute of Epidemiology and Public Health, University of Montpellier, INSERM, Department of Rheumatology, CHU Nîmes, Montpellier, France
| | - Christophe Demattei
- Department of Biostatistics, Epidemiology, Public Health and Innovation in Methodology (BESPIM), CHU Nîmes, Univ Montpellier, Nîmes, France
| | - Florian Salipante
- Department of Biostatistics, Epidemiology, Public Health and Innovation in Methodology (BESPIM), CHU Nîmes, Univ Montpellier, Nîmes, France
| | - Lucia Grenga
- Department of Medicines and Technologies for Health, Atomic Energy and Alternative Energies Commission (CEA), Paris-Saclay University, Bagnols-sur-Cèze, France
| | - Jean Armengaud
- Department of Medicines and Technologies for Health, Atomic Energy and Alternative Energies Commission (CEA), Paris-Saclay University, Bagnols-sur-Cèze, France
| | - Jean-Philippe Lavigne
- Bacterial Virulence and Chronic Infections, INSERM U1047, Univ Montpellier, Department of Microbiology and Hospital Hygiene, CHU Nîmes, Nîmes, France
| | - Christian Jorgensen
- Department of Rheumatology, Stem cells, Cellular plasticity, Regenerative medicine and Immunotherapies, IRMB, INSERM UMR1183, University of Montpellier & University Hospital of Montpellier, Montpellier, France
| |
Collapse
|
21
|
Sharma D, Patel D, Mandal P. In Vitro, In Vivo, and In Silico Investigation of Synbiotic-Mediated Activation of PPAR- α Curtails Nonalcoholic Steatohepatitis (NASH) in Wistar Rats by Inhibiting PNPLA3/SREBP1-c Lead Inflammatory Injury of Hepatic Cells. Mediators Inflamm 2025; 2025:9948679. [PMID: 40017524 PMCID: PMC11865469 DOI: 10.1155/mi/9948679] [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: 02/24/2024] [Revised: 07/31/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is an inflammation of the liver and a menace to human health. To treat NASH various pharmaceutical products have been used, but their prohibitive side effects limit their effectiveness. NASH, a multihit hypothesis involves high-fat diet and signals from the gut to the liver. Lactobacillus plantarum (probiotic) and aged garlic extract (AGE, a prebiotic) are antioxidative and anti-inflammatory and may be a latent combination therapy for NASH. The NASH model was developed using Wistar rats and treatments were administered to understand the mechanism. Initially, in the in vitro models, transepithelial electrical resistance (TEER) 2'-7'-dichlorodihydrofluorescein diacetate (DCFDA), 4-6-diamidino-2-phenylindole (DAPI) labeling and Oil Red O (ORO) conducted on HepG2 and Caco2 cells. Afterwards, in in vivo studies rat liver tissues were examined through confocal microscopy using the ORO staining and hematoxylin and eosin (H/E) stain, malondialdehyde (MDA), and biochemical indices were recorded. The levels of patatin-like phospholipase domain-containing protein 3 (PNPLA3) and sterol regulatory element binding protein-1c (SREBP-1c), peroxisome proliferators activated receptors (PPARs)-α, inflammatory, and apoptotic biomarkers were quantified by qRT-PCR. Synbiotic reduced the hepatic inflammation and apoptosis examined through the levels of PNPLA3, SREBP-1c, IL-6, TGF-β, Bcl-2, and caspase-3 in NASH models. In turn, the gram-negative species and bacterial translocation associated were reduced. Consequently, the Insilco analysis supports the theory that each (eight) bioactive compound of AGE targets PNPLA3 and enhances the PPAR-α activity. Additionally, PPAR-α inhibitors upregulated the PNPLA3 and SREBP-1C expression. As a result, the synbiotic may inhibit NASH progression by affecting PNPLA3/SREBP1-c through PPAR-α.
Collapse
Affiliation(s)
- Dixa Sharma
- Department of Biology, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand 3888421, Gujarat, India
| | - Dhara Patel
- Department of Biology, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand 3888421, Gujarat, India
| | - Palash Mandal
- Department of Biology, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Anand 3888421, Gujarat, India
| |
Collapse
|
22
|
Aliberti SM, Capunzo M. The Power of Environment: A Comprehensive Review of the Exposome's Role in Healthy Aging, Longevity, and Preventive Medicine-Lessons from Blue Zones and Cilento. Nutrients 2025; 17:722. [PMID: 40005049 PMCID: PMC11858149 DOI: 10.3390/nu17040722] [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: 01/21/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Aging and longevity are shaped by the exposome, a dynamic network of environmental, social, and biological factors. Understanding how these exposures interact with biological mechanisms can inform strategies for healthier aging. Background/Objectives: This review explores the exposome as a dynamic system encompassing both protective and risk factors, with a specific focus on how beneficial environmental exposures, microbiome diversity, lifestyle behaviors, and resilience mechanisms contribute to successful aging. By analyzing high-longevity populations, such as the Blue Zones and Cilento, it aims to identify common determinants of successful aging. Methods: A mixed-method study was conducted, combining a systematic review of the English literature (2003-2024) with a comparative analysis of longevity regions. A structured search was performed in PubMed, Scopus, and Google Scholar using keywords such as "longevity", "Blue Zones", "Cilento", "microbiome", "environmental factors", and related terms. Additionally, qualitative and quantitative analysis were applied to assess key protective factors across different aging models. Results: This study identified key factors contributing to successful aging in longevity hotspots, including sustained exposure to biodiverse natural environments, adherence to Mediterranean or plant-based diet rich in polyphenols and probiotics, regular physical activity, strong social networks, and psychological resilience. A novel aspect of this review is the role of the gut microbiome as a mediator between environmental exposures and immune-metabolic health, influencing inflammation modulation and cellular aging. Despite geographic and cultural differences, case studies reveal a shared pattern of protective factors that collectively enhance lifespan and healthspan. Conclusions: The exposome is a critical determinant of aging trajectories, acting through complex interactions between environmental and biological mechanisms. By integrating insights from high-longevity populations, this mixed-method study proposes a comprehensive framework for optimizing microbiome health, enhancing resilience, and promoting protective environmental exposures. These findings provide a translational perspective to guide future interventions in aging research and global health initiatives.
Collapse
Affiliation(s)
- Silvana Mirella Aliberti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Salerno, Italy;
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Salerno, Italy;
- Complex Operational Unit Health Hygiene, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
23
|
Zhang J, Wei J, Lai W, Sun J, Bai Y, Cao H, Guo J, Su Z. Focus on Glucagon-like Peptide-1 Target: Drugs Approved or Designed to Treat Obesity. Int J Mol Sci 2025; 26:1651. [PMID: 40004115 PMCID: PMC11855704 DOI: 10.3390/ijms26041651] [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: 01/07/2025] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity is closely related to metabolic diseases, which brings a heavy burden to the health care system. It is urgent to formulate and implement effective treatment strategies. Glucagon-like peptide-1 (GLP-1) is a protein with seven transmembrane domains connected by type B and G proteins, which is widely distributed and expressed in many organs and tissues. GLP-1 analogues can reduce weight, lower blood pressure, and improve blood lipids. Obesity, diabetes, cardiovascular diseases, and other diseases have caused scientists' research and development boom. Among them, GLP-1R agonist drugs have developed rapidly in weight-loss drugs. In this paper, based on the target of GLP-1, the mechanism of action of GLP-1 in obesity treatment was deeply studied, and the drugs approved and designed for obesity treatment based on GLP-1 target were elaborated in detail. Innovatively put forward and summarized the double and triple GLP-1 targeted drugs in the treatment of obesity with better effects and less toxic and side effects, and this can make full use of multi-target methods to treat other diseases in the future. Finally, it is pointed out that intestinal flora and microorganisms have many benefits in the treatment of obesity, and fecal bacteria transplantation may be a potential treatment for obesity with less harm to the body. This article provides some promising methods to treat obesity, which have strong practical value.
Collapse
Affiliation(s)
- Jiahua Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.Z.); (J.W.); (W.L.); (J.S.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jintao Wei
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.Z.); (J.W.); (W.L.); (J.S.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Weiwen Lai
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.Z.); (J.W.); (W.L.); (J.S.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiawei Sun
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.Z.); (J.W.); (W.L.); (J.S.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China;
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China;
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; (J.Z.); (J.W.); (W.L.); (J.S.)
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
24
|
Rondanelli M, Borromeo S, Cavioni A, Gasparri C, Gattone I, Genovese E, Lazzarotti A, Minonne L, Moroni A, Patelli Z, Razza C, Sivieri C, Valentini EM, Barrile GC. Therapeutic Strategies to Modulate Gut Microbial Health: Approaches for Chronic Metabolic Disorder Management. Metabolites 2025; 15:127. [PMID: 39997751 PMCID: PMC11857149 DOI: 10.3390/metabo15020127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Numerous recent studies have suggested that the composition of the intestinal microbiota can trigger metabolic disorders, such as diabetes, prediabetes, obesity, metabolic syndrome, sarcopenia, dyslipidemia, hyperhomocysteinemia, and non-alcoholic fatty liver disease. Since then, considerable effort has been made to understand the link between the composition of intestinal microbiota and metabolic disorders, as well as the role of probiotics in the modulation of the intestinal microbiota. The aim of this review was to summarize the reviews and individual articles on the state of the art regarding ideal therapy with probiotics and prebiotics in order to obtain the reversion of dysbiosis (alteration in microbiota) to eubiosis during metabolic diseases, such as diabetes, prediabetes, obesity, hyperhomocysteinemia, dyslipidemia, sarcopenia, and non-alcoholic fatty liver diseases. This review includes 245 eligible studies. In conclusion, a condition of dysbiosis, or in general, alteration of the intestinal microbiota, could be implicated in the development of metabolic disorders through different mechanisms, mainly linked to the release of pro-inflammatory factors. Several studies have already demonstrated the potential of using probiotics and prebiotics in the treatment of this condition, detecting significant improvements in the specific symptoms of metabolic diseases. These findings reinforce the hypothesis that a condition of dysbiosis can lead to a generalized inflammatory picture with negative consequences on different organs and systems. Moreover, this review confirms that the beneficial effects of probiotics on metabolic diseases are promising, but more research is needed to determine the optimal probiotic strains, doses, and administration forms for specific metabolic conditions.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Sara Borromeo
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Cavioni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Ilaria Gattone
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Elisa Genovese
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Lazzarotti
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Leonardo Minonne
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessia Moroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Zaira Patelli
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Razza
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Sivieri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Eugenio Marzio Valentini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Gaetan Claude Barrile
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| |
Collapse
|
25
|
Sun H, Qi L, Ming Y, Wang W, Hu M. Life's Simple 7 and its impact on chronic bowel disorders: a study on constipation and diarrhea in the U.S. adult population. Front Med (Lausanne) 2025; 12:1516210. [PMID: 40012973 PMCID: PMC11862994 DOI: 10.3389/fmed.2025.1516210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/03/2025] [Indexed: 02/28/2025] Open
Abstract
Background Chronic gastrointestinal disorders, such as chronic constipation and diarrhea, pose significant public health challenges, affecting quality of life and healthcare costs. Life's Simple 7 (LS7), established by the American Heart Association, encompasses essential health behaviors that may influence bowel health. Methods We utilized data from the National Health and Nutrition Examination Survey (NHANES) conducted between 2005 and 2010, focusing on adults aged 20 years and older. A total of 12,912 participants were included in the analysis. Bowel health was assessed through self-reported questionnaires, while LS7 was evaluated based on seven components: smoking status, physical activity, dietary quality, BMI, blood pressure, blood glucose, and blood cholesterol. Survey-weighted logistic regression models were employed to assess the associations between LS7 and chronic constipation and diarrhea, adjusting for various demographic and health-related covariates. Results Our findings revealed a significant inverse association between LS7 adherence and the prevalence of chronic constipation (OR: 0.914, 95% CI: 0.864-0.966, p = 0.003) and chronic diarrhea (OR: 0.883, 95% CI: 0.856-0.912, p < 0.0001). The protective effect of LS7 was more pronounced among males and individuals with a BMI under 30 kg/m2 for chronic constipation, and among younger adults and those without hypertension for chronic diarrhea. Restricted cubic spline analyses indicated a dose-response relationship, particularly for chronic diarrhea. Conclusion This study highlights the protective role of LS7 in promoting bowel health and preventing chronic constipation and diarrhea. Tailoring public health interventions based on demographic and health characteristics may enhance the effectiveness of strategies aimed at improving gastrointestinal health outcomes.
Collapse
Affiliation(s)
- Hongzhi Sun
- The Third People's Hospital of Hefei, Hefei, Anhui, China
- Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Lei Qi
- The Third People's Hospital of Hefei, Hefei, Anhui, China
- Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Yiwei Ming
- The Third People's Hospital of Hefei, Hefei, Anhui, China
- Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Weichen Wang
- The Second People's Hospital of Anhui Province, Hefei, Anhui, China
| | - Maoneng Hu
- The Third People's Hospital of Hefei, Hefei, Anhui, China
- Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
26
|
Li T, Liu Y, Duan T, Guo C, Liu B, Fu X, Wang L, Wang X, Dong X, Wang C, Lu Y, Wang Y, Shi L, Tian H, Yang X. Nondigestible stachyose binds membranous HSP90β on small intestinal epithelium to regulate the exosomal miRNAs: A new function and mechanism. Cell Metab 2025; 37:345-360.e6. [PMID: 39561765 DOI: 10.1016/j.cmet.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/16/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024]
Abstract
Oligosaccharides are conventionally recognized as "passersby" in the small intestine. However, our research has reframed this understanding by uncovering a new function of oligosaccharide stachyose, which binds hydrophobic residues of membranous HSP90β on small intestinal epithelial cells, thus reprograming the exosomal miRNA profile. CRISPR-Cas9-mediated HSP90β knockout abolished the accumulation of stachyose on cell membrane and its regulatory effects on these miRNAs. Notably, stachyose's regulation on these miRNAs is independent of its prebiotic role, as evidenced by the observation of stachyose-altered fecal miRNAs in pseudo-germ-free mice. These stachyose-altered miRNAs further shaped colonic microbiome, especially harboring Lactobacillus in mice. Thereinto, miR-30a-5p that was downregulated (Log2FC < -2) in both mice and human feces following stachyose treatment could specifically suppress the growth of Lactobacillus reuteri. These findings build a new regulatory axis of stachyose-intestinal miRNAs-gut microbiota and unveil a previously unknown mechanism underlying the direct "talk" of oligosaccharides to intestine epithelium via membranous HSP90β.
Collapse
Affiliation(s)
- Ting Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Yueyue Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Tianchi Duan
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Xiuqiong Fu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Lu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xiaoyuan Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xinyue Dong
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Chennan Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yalong Lu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Lin Shi
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
27
|
Levi-D'Ancona E, Walker EM, Zhu J, Deng Y, Sidarala V, Stendahl AM, Reck EC, Henry-Kanarek BA, Lietzke AC, Pasmooij MB, Hubers DL, Basrur V, Ghosh S, Stiles L, Nesvizhskii AI, Shirihai OS, Soleimanpour SA. TRAF6 integrates innate immune signals to regulate glucose homeostasis via Parkin-dependent and -independent mitophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635900. [PMID: 39974969 PMCID: PMC11838480 DOI: 10.1101/2025.01.31.635900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Activation of innate immune signaling occurs during the progression of immunometabolic diseases, including type 2 diabetes (T2D), yet the impact of innate immune signaling on glucose homeostasis is controversial. Here, we report that the E3 ubiquitin ligase TRAF6 integrates innate immune signals following diet-induced obesity to promote glucose homeostasis through the induction of mitophagy. Whereas TRAF6 was dispensable for glucose homeostasis and pancreatic β-cell function under basal conditions, TRAF6 was pivotal for insulin secretion, mitochondrial respiration, and increases in mitophagy following metabolic stress in both mouse and human islets. Indeed, TRAF6 was critical for the recruitment and function of machinery within both the ubiquitin-mediated (Parkin-dependent) and receptor-mediated (Parkin-independent) mitophagy pathways upon metabolic stress. Intriguingly, the effect of TRAF6 deficiency on glucose homeostasis and mitophagy was fully reversed by concomitant Parkin deficiency. Thus, our results implicate a role for TRAF6 in the cross-regulation of both ubiquitin- and receptor- mediated mitophagy through the restriction of Parkin. Together, we illustrate that β-cells engage innate immune signaling to adaptively respond to a diabetogenic environment.
Collapse
|
28
|
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
|
29
|
Adams MS, Enichen E, Demmig-Adams B. Reframing Diabetes Prevention: From Body Shaming to Metabolic Reprogramming. Am J Lifestyle Med 2025; 19:168-191. [PMID: 39981552 PMCID: PMC11836583 DOI: 10.1177/15598276231182655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025] Open
Abstract
This review integrates new developments in psychology with updated physiological insight on the complex relationships among chronic psychological stress (arising from weight stigmatization and body shaming), food composition, physical activity and metabolic health for the example of diabetes. We address how visual measures of health, such as body mass index (BMI) and waist-to-hip ratio, do not adequately capture metabolic health and can instead contribute to weight stigmatization, chronic stress, and system-wide impairment of metabolic health. We also emphasize the importance of food composition over calorie counting. We summarize how chronic stress interacts with nutritional deficiencies and physical inactivity to disrupt the stress response, immune response, gut microbiome, and function of fat depots. We specifically address how interactions among lifestyle factors and the gut microbiome regulate whether fat stored around the waist has a negative or positive effect on metabolic health. We aim to provide a resource and updated framework for diabetes prevention and health promotion by (i) highlighting metabolic imbalances triggered by lifestyle changes during the transition to industrialized society and (ii) detailing the potential to support metabolic health through access to modest, but comprehensive lifestyle adjustments.
Collapse
Affiliation(s)
- Melanie S Adams
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, CO, USA
| | | | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
30
|
de Cuevillas B, Riezu-Boj JI, Milagro FI, Galera Alquegui S, Babio N, Pastor-Villaescusa B, Gil-Campos M, Leis R, De Miguel-Etayo P, Moreno LA, Salas-Salvadó J, Martínez JA, Navas-Carretero S. Parent-child microbiota relationships involved in childhood obesity: A CORALS ancillary study. Nutrition 2025; 130:112603. [PMID: 39550838 DOI: 10.1016/j.nut.2024.112603] [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: 06/28/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVES Childhood obesity continues to rise worldwide. Family gut microorganisms may be associated with childhood obesity. The aim of the study was to analyze bacterial similarities in fecal microbiota composition between parent-offspring pairs as linked to body weight. METHODS A total of 146 father/mother and offspring pairs were categorized into four groups according to the weight status of the parent-child pair as follows: group 1, parent and child with normal weight; group 2, parent and child with overweight/obesity; group 3, parent with normal weight and child with overweight/obesity; group 4, parent with overweight/obesity and child with normal weight. Anthropometric measurements and lifestyle assessments were performed in all participants. Microbiota characteristics were determined by 16S ribosomal RNA gene sequencing. Logistic regression models were performed to determine whether the abundance of any bacteria was able to predict childhood obesity. Moreover, receiver operating characteristic curves were fitted to define the relative diagnostic strength of bacterial taxa for the correct identification of childhood obesity. RESULTS The absence/abundance of Catenibacterium mitsuokai, Prevotella stercorea, Desulfovibrio piger, Massiliprevotella massiliensis, and Phascolarctobacterium succinatutens was involved in body weight family associations. A positive relationship between P. succinatutens richness from parents and M. massiliensis from children was observed with regard to body weight status (odds ratio, 1.14, P = 0.013). CONCLUSIONS This study describes five potential gut bacteria that may be putatively involved in family weight status relationships and appear to be useful for predicting obesity.
Collapse
Affiliation(s)
- Begoña de Cuevillas
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Jose I Riezu-Boj
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain
| | - Fermín I Milagro
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Galera Alquegui
- Department of Personalized Medicine, Navarra Services and Technologies, Government of Navarra, Pamplona, Spain
| | - Nancy Babio
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Food, Nutrition, Development and Mental Health Research Group, Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Belén Pastor-Villaescusa
- Metabolism and Investigation Unit, Maimónides Institute of Biomedicine Research of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Mercedes Gil-Campos
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Metabolism and Investigation Unit, Maimónides Institute of Biomedicine Research of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Rosaura Leis
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Pediatric Service, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; Pediatric Nutrition Research Group, Unit of Investigation in Nutrition, Growth and Human Development of Galicia-USC, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Pilar De Miguel-Etayo
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, Instituto de Investigación Sanitaria Aragón, University of Zaragoza, Zaragoza, Spain
| | - Luis A Moreno
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, Instituto de Investigación Sanitaria Aragón, University of Zaragoza, Zaragoza, Spain
| | - Jordi Salas-Salvadó
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Food, Nutrition, Development and Mental Health Research Group, Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - J Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Navas-Carretero
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
31
|
Wankhede NL, Kale MB, Kyada A, M RM, Chaudhary K, Naidu KS, Rahangdale S, Shende PV, Taksande BG, Khalid M, Gulati M, Umekar MJ, Fareed M, Kopalli SR, Koppula S. Sleep deprivation-induced shifts in gut microbiota: Implications for neurological disorders. Neuroscience 2025; 565:99-116. [PMID: 39622383 DOI: 10.1016/j.neuroscience.2024.11.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sleep deprivation is a prevalent issue in contemporary society, with significant ramifications for both physical and mental well-being. Emerging scientific evidence illuminates its intricate interplay with the gut-brain axis, a vital determinant of neurological function. Disruptions in sleep patterns disturb the delicate equilibrium of the gut microbiota, resulting in dysbiosis characterized by alterations in microbial composition and function. This dysbiosis contributes to the exacerbation of neurological disorders such as depression, anxiety, and cognitive decline through multifaceted mechanisms, including heightened neuroinflammation, disturbances in neurotransmitter signalling, and compromised integrity of the gut barrier. In response to these challenges, there is a burgeoning interest in therapeutic interventions aimed at restoring gut microbial balance and alleviating neurological symptoms precipitated by sleep deprivation. Probiotics, dietary modifications, and behavioural strategies represent promising avenues for modulating the gut microbiota and mitigating the adverse effects of sleep disturbances on neurological health. Moreover, the advent of personalized interventions guided by advanced omics technologies holds considerable potential for tailoring treatments to individualized needs and optimizing therapeutic outcomes. Interdisciplinary collaboration and concerted research efforts are imperative for elucidating the underlying mechanisms linking sleep, gut microbiota, and neurological function. Longitudinal studies, translational research endeavours, and advancements in technology are pivotal for unravelling the complex interplay between these intricate systems.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences Marwadi University, Rajkot 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
32
|
Garutti M, Sirico M, Noto C, Foffano L, Hopkins M, Puglisi F. Hallmarks of Appetite: A Comprehensive Review of Hunger, Appetite, Satiation, and Satiety. Curr Obes Rep 2025; 14:12. [PMID: 39849268 DOI: 10.1007/s13679-024-00604-w] [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] [Accepted: 12/18/2024] [Indexed: 01/25/2025]
Abstract
PURPOSE OF REVIEW The present review describes the available literature on the physiologic mechanisms that modulate hunger, appetite, satiation, and satiety with a particular focus on well-established and emerging factors involved in the classic satiety cascade model. RECENT FINDING Obesity is a significant risk factor for numerous chronic conditions like cancer, cardiovascular diseases, and diabetes. As excess energy intake is considered by some to be the primary driver of weight gain, tremendous collective effort should be directed toward reducing excessive feeding at the individual and population levels. From this perspective, detailed understanding of physiologic mechanisms that control appetite, and in turn, the design of effective interventions to manage appetite, may represent key strategies in controlling the obesity epidemic. With the obesity's prevalence on the rise worldwide, research on hunger, appetite, satiation and satiety is more relevant than ever. This research aims to provide practical insights for medical practitioners, nutrition professionals, and the broader scientific community in the fight against this global health challenge.
Collapse
Affiliation(s)
- Mattia Garutti
- CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy.
| | - Marianna Sirico
- Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo Per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Forli-Cesena, Italy
| | - Claudia Noto
- Medical Oncology, Azienda Sanitaria Universitaria Integrata Di Trieste, Ospedale Maggiore, Piazza Dell'Ospitale 1, 34125, Trieste, Italy
| | - Lorenzo Foffano
- CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Mark Hopkins
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Fabio Puglisi
- CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine, University of Udine, 33100, Udine, Italy
| |
Collapse
|
33
|
Xiang M, Qiao L, Han Q, Zha Y, Sui X, Wang Q. Effects of Supplementation With Different Specificities of Dietary Fiber on Health-Related Indicators in Adults With Overweight or Obesity: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Nutr Rev 2025:nuae193. [PMID: 39821284 DOI: 10.1093/nutrit/nuae193] [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] [Indexed: 01/19/2025] Open
Abstract
CONTEXT Dietary fiber (DF) exhibits variations in its chemical and physical complexity, as well as in its utilization by the gut microbiota. However, the impact of these differences on the health status of adults with overweight or obesity remains unclear. OBJECTIVE This meta-analysis aimed to explore the varying effects of supplementing with different specificities of DF on the health of adults with overweight or obesity, providing guidance on selecting DF supplementation to improve health status. DATA SOURCES The literature search encompassed 4 electronic databases-PubMed, Cochrane Library, Web of Science, and EMBASE-and was conducted between January 1, 2012, and November 10, 2023. Randomized controlled trials comparing DF with placebo treatment, without energy restriction, were included. DATA EXTRACTION Two independent reviewers extracted data using a standardized form, resolving discrepancies through discussion. The data included study characteristics, participant demographics, DF specifications, and outcome measures. DATA ANALYSIS Random-effects models and the generic inverse variance method were used to analyze data, assuming varying outcomes based on DF specificity. Meta-regression assessed the impact of population, duration, and dosage. Publication bias was evaluated using funnel plots and Egger's and Begg's tests. The analysis included 34 trials (n = 1804) examining DF supplementation at 1.5 to 40 g/day for 3 to 16 weeks. DF supplementation significantly reduced glycated hemoglobin (HbA1c) by 0.13%, fasting insulin by 0.82 μIU/mL, and homeostatic model assessment of insulin resistance (HOMA-IR) by 0.33 in adults with overweight or obesity. Subgroup analyses based on DF specificity revealed differences in effects on HbA1c, fasting insulin, and systolic blood pressure. The low-specificity subgroup showed significant heterogeneity in body weight, body mass index, HbA1c, fasting insulin, and HOMA-IR, with a decrease in fasting insulin by 1.09 μIU/mL. The low-to-intermediate-specificity subgroup had reductions in HbA1c by 0.8%, fasting insulin by 2.08 μIU/mL, and HOMA-IR by 0.61. The intermediate-specificity subgroup experienced a 2.85-kg decrease in body weight and a 9.03-mg/dL increase in LDL cholesterol. The mixed subgroup showed an increase in systolic blood pressure by 3.85 mmHg. CONCLUSION Supplementing with different specificities of DF may have distinct effects on health-related indicators in adults with overweight or obesity. Considering individuals' gut microbiota composition and specific health goals is recommended when selecting DF supplementation for adults with overweight or obesity. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023432920.
Collapse
Affiliation(s)
- Mai Xiang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
- College of Exercise Science, Beijing Sport University, Beijing 100084, China
| | - Li Qiao
- Beijing Competitor Sports Nutrition Research Institute, Beijing 100029, China
| | - Qi Han
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
| | - Yu Zha
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xuemei Sui
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Qirong Wang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
| |
Collapse
|
34
|
Govender P, Ghai M. Population-specific differences in the human microbiome: Factors defining the diversity. Gene 2025; 933:148923. [PMID: 39244168 DOI: 10.1016/j.gene.2024.148923] [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: 05/24/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Differences in microbial communities at different body habitats define the microbiome composition of the human body. The gut, oral, skin vaginal fluid and tissue microbiome, are pivotal for human development and immune response and cross talk between these microbiomes is evident. Population studies reveal that various factors, such as host genetics, diet, lifestyle, aging, and geographical location are strongly associated with population-specific microbiome differences. The present review discusses the factors that shape microbiome diversity in humans, and microbiome differences in African, Asian and Caucasian populations. Gut microbiome studies show that microbial species Bacteroides is commonly found in individuals living in Western countries (Caucasian populations), while Prevotella is prevalent in non-Western countries (African and Asian populations). This association is mainly due to the high carbohydrate, high fat diet in western countries in contrast to high fibre, low fat diets in African/ Asian regions. Majority of the microbiome studies focus on the bacteriome component; however, interesting findings reveal that increased bacteriophage richness, which makes up the virome component, correlates with decreased bacterial diversity, and causes microbiome dysbiosis. An increase of Caudovirales (bacteriophages) is associated with a decrease in enteric bacteria in inflammatory bowel diseases. Future microbiome studies should evaluate the interrelation between bacteriome and virome to fully understand their significance in the pathogenesis and progression of human diseases. With ethnic health disparities becoming increasingly apparent, studies need to emphasize on the association of population-specific microbiome differences and human diseases, to develop microbiome-based therapeutics. Additionally, targeted phage therapy is emerging as an attractive alternative to antibiotics for bacterial infections. With rapid rise in microbiome research, focus should be on standardizing protocols, advanced bioinformatics tools, and reducing sequencing platform related biases. Ultimately, integration of multi-omics data (genomics, transcriptomics, proteomics and metabolomics) will lead to precision models for personalized microbiome therapeutics advancement.
Collapse
Affiliation(s)
- Priyanka Govender
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Meenu Ghai
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa.
| |
Collapse
|
35
|
Guidi L, Martinez-Tellez B, Ortega Santos CP. Obesity, gut bacteria, and the epigenetic control of metabolic disease. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:333-368. [DOI: 10.1016/b978-0-443-18979-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Hamamah S, Iatcu OC, Covasa M. Dietary Influences on Gut Microbiota and Their Role in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Nutrients 2024; 17:143. [PMID: 39796579 PMCID: PMC11722922 DOI: 10.3390/nu17010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major contributor to liver-related morbidity, cardiovascular disease, and metabolic complications. Lifestyle interventions, including diet and exercise, are first line in treating MASLD. Dietary approaches such as the low-glycemic-index Mediterranean diet, the ketogenic diet, intermittent fasting, and high fiber diets have demonstrated potential in addressing the metabolic dysfunction underlying this condition. The development and progression of MASLD are closely associated with taxonomic shifts in gut microbial communities, a relationship well-documented in the literature. Given the importance of diet as a primary treatment for MASLD, it is important to understand how gut microbiota and their metabolic byproducts mediate favorable outcomes induced by healthy dietary patterns. Conversely, microbiota changes conferred by unhealthy dietary patterns such as the Western diet may induce dysbiosis and influence steatotic liver disease through promoting hepatic inflammation, up-regulating lipogenesis, dysregulating bile acid metabolism, increasing insulin resistance, and causing oxidative damage in hepatocytes. Although emerging evidence has identified links between diet, microbiota, and development of MASLD, significant gaps remain in understanding specific microbial roles, metabolite pathways, host interactions, and causal relationships. Therefore, this review aims to provide mechanistic insights into the role of microbiota-mediated processes through the analysis of both healthy and unhealthy dietary patterns and their contribution to MASLD pathophysiology. By better elucidating the interplay between dietary nutrients, microbiota-mediated processes, and the onset and progression of steatotic liver disease, this work aims to identify new opportunities for targeted dietary interventions to treat MASLD efficiently.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA;
| | - Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| |
Collapse
|
37
|
Ha S, Wong VWS, Zhang X, Yu J. Interplay between gut microbiome, host genetic and epigenetic modifications in MASLD and MASLD-related hepatocellular carcinoma. Gut 2024; 74:141-152. [PMID: 38950910 PMCID: PMC11671994 DOI: 10.1136/gutjnl-2024-332398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a wide spectrum of liver injuries, ranging from hepatic steatosis, metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis to MASLD-associated hepatocellular carcinoma (MASLD-HCC). Recent studies have highlighted the bidirectional impacts between host genetics/epigenetics and the gut microbial community. Host genetics influence the composition of gut microbiome, while the gut microbiota and their derived metabolites can induce host epigenetic modifications to affect the development of MASLD. The exploration of the intricate relationship between the gut microbiome and the genetic/epigenetic makeup of the host is anticipated to yield promising avenues for therapeutic interventions targeting MASLD and its associated conditions. In this review, we summarise the effects of gut microbiome, host genetics and epigenetic alterations in MASLD and MASLD-HCC. We further discuss research findings demonstrating the bidirectional impacts between gut microbiome and host genetics/epigenetics, emphasising the significance of this interconnection in MASLD prevention and treatment.
Collapse
Affiliation(s)
- Suki Ha
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
38
|
Arellano-García LI, Portillo MP, Martínez JA, Courtois A, Milton-Laskibar I. Postbiotics for the management of obesity, insulin resistance/type 2 diabetes and NAFLD. Beyond microbial viability. Crit Rev Food Sci Nutr 2024:1-24. [PMID: 39644489 DOI: 10.1080/10408398.2024.2437143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Highly prevalent comorbidities associated with metabolic syndrome, such as abdominal obesity, nonalcoholic fatty liver disease (NAFLD) and insulin-resistance/Type 2 diabetes (IR/T2D) share alterations in gut microbiota composition as a potential triggering factor. Recent studies put the attention in the potential usage of postbiotics (inactivated probiotics) on these metabolic alterations. This review summarizes the current evidence regarding the efficacy of postbiotic administration in both, preclinical and clinical studies, for the management of obesity, NAFLD and IR/T2D. Data from preclinical studies (rodents) suggest that postbiotic administration effectively prevents obesity, whereas clinical studies corroborate these benefits also in overweight/obese subjects receiving inactivated bacteria. As for NAFLD, although preclinical studies indicate that postbiotic administration improves different liver markers, no data obtained in humans have been published so far since all the studies are ongoing clinical trials. Finally, while the administration of inactivated bacteria demonstrated to be a promising approach for the management of IR/T2D in rodents, data from clinical trials indicates that in humans, this approach is more effective on IR than in T2D. In conclusion, the available scientific data indicate that postbiotic administration not only is safer, but also as effective as probiotic administration for the management of obesity associated prevalent metabolic alterations.
Collapse
Affiliation(s)
- Laura Isabel Arellano-García
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - María P Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
- BIOARABA Health Research Institute, Vitoria-Gasteiz, Spain
| | - J Alfredo Martínez
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health 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
| | - Arnaud Courtois
- Univ. Bordeaux, Bordeaux INP, INRAE, OENO, UMR 1366, ISVV, Villenave d'Ornon, France
- Bordeaux Sciences Agro, Bordeaux INP, INRAE, OENO, UMR 1366, ISVV, Gradignan, France
- Centre Antipoison de Nouvelle Aquitaine, CHU de Bordeaux, Bordeaux, France
| | - Iñaki Milton-Laskibar
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Centre, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
- BIOARABA Health Research Institute, Vitoria-Gasteiz, Spain
| |
Collapse
|
39
|
Han YH, Cui XW, Li YX, Chen X, Zhang H, Zhang Y, Wang SS, Li M. Bacterial cellulose is a desirable biological macromolecule that can prevent obesity via modulating lipid metabolism and gut microbiota. Int J Biol Macromol 2024; 283:137522. [PMID: 39537056 DOI: 10.1016/j.ijbiomac.2024.137522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/02/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Obesity has attracted great concern because of its undesirable effects on our life quality. Bacterial cellulose (BC) is a biological macromolecule that can improve gut homeostasis and lipid metabolism. However, its potential role in preventing obesity and associated mechanisms is still poorly understood. Herein, a supplement of BC was used to fully evaluate how it prevents obesity based on physio-biochemical and gut microbial analyses. Results showed that BC consumption helped decrease body and liver weight, and fat accumulation in kidney and epididymis. Correspondingly, glucose concentrations, total triglycerides, total cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol were reversed to the control levels. Consuming BC also improved liver fat metabolism and intestinal function, and alleviated ileum and epididymis inflammation. High-throughput sequencing suggested that a high-fat diet significantly decreased gut microbiota diversity, which could be reversed by consuming BC. A decreased Firmicutes and Proteobacteria and an increased Bacteroidetes following BC consumption were observed. The OTU-based analysis identified that Lachnospiraceae, Desulfovibrio, Lachnoclostridium, Blautia, Anaerotruncus, Bacteroides, Faecalibaculum, Bacteroidales S24-7 group, Prevotellaceae UCG-001 group, and Alloprevotella might be involved in obesity development or prevention. Our data suggest that BC is a good insoluble dietary fiber to prevent obesity via regulating lipid metabolism and gut microbiota.
Collapse
Affiliation(s)
- Yong-He Han
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Xi-Wen Cui
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yi-Xi Li
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Xian Chen
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hong Zhang
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhang
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Shan-Shan Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; College of Life Science, Fujian Normal University, Fuzhou 350117, China.
| | - Min Li
- College of Life Science, Fujian Normal University, Fuzhou 350117, China.
| |
Collapse
|
40
|
Mu R, Fu Y, Li J, Xie Q, Ma W. Effects of different milk powders on the growth and intestinal flora in weaned rats: Comparison of special formula milk powder with ordinary milk powder. Food Sci Nutr 2024; 12:10448-10462. [PMID: 39723089 PMCID: PMC11666978 DOI: 10.1002/fsn3.4387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 12/28/2024] Open
Abstract
The objective of this investigation was to examine the effects of distinct dosages of infant formula and diverse formula constituents on the growth and development of weaned rats. Fifty specific pathogen-free (SPF) male Sprague-Dawley (SD) rats aged 3 weeks were divided into the basic diet group, 20% ordinary milk powder group, 20% special formula milk powder group, 30% ordinary milk powder group, and 30% special formula milk powder group randomly. After 28 days of feeding, compared with the basic diet group, the body mass and brain/body weight of rats in the 30% ordinary and special formula milk powder groups were decreased. At the Genus level, Bacteroides in the group supplemented with 20% special formula milk powder was significantly lower than that in the basic diet group, and Parabacteroides was significantly lower than that in the 20% ordinary milk powder group. Lactobacillus was significantly higher than those in the basic diet group and the 20% ordinary milk powder group, and Blautia was significantly higher than those in the basic diet group and the 20% and 30% ordinary milk powder groups, and UBA1819 was significantly higher than those in the other groups. The abundance of Parasutterella in the basic diet group was significantly higher than those in the groups supplemented with 20% ordinary milk powder, 20% special formula milk powder, and 30% ordinary milk powder. This study found that different doses and different formula components of infant milk powder could affect body mass and intestinal flora in Sprague-Dawley (SD) rats, and the addition of low-dose (20%) special formula infant milk powder can increase the beneficial bacteria in the intestinal flora of rats and may reduce the pathogenic bacteria.
Collapse
Affiliation(s)
- Ruiqi Mu
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| | - Yu Fu
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| | - Jufang Li
- Feihe Research InstituteHeilongjiang Feihe Dairy Co., Ltd.BeijingChina
| | - Qinggang Xie
- Feihe Research InstituteHeilongjiang Feihe Dairy Co., Ltd.BeijingChina
| | - Weiwei Ma
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| |
Collapse
|
41
|
Sezer B, Albers J, Meisters R, Schram MT, Köhler S, Stehouwer CDA, Koster A, Bosma H. Do poor psychosocial resources mediate health inequalities in type 2 diabetes mellitus? Findings from the Maastricht study. Eur J Public Health 2024; 34:1029-1035. [PMID: 39254600 PMCID: PMC11631483 DOI: 10.1093/eurpub/ckae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
Low socioeconomic position (SEP) has been identified as a risk factor for type 2 diabetes mellitus (T2DM), and psychosocial resources might be on the pathway in this association. We examined two poor psychosocial resources, low control beliefs and inferiority beliefs, that might link low SEP with T2DM. 8292 participants aged 40-75 living in Southern Netherlands participated in The Maastricht Study starting from September 2010 to October 2020 and were followed up to 10 years with annual questionnaires. SEP (education, income, occupation), low control beliefs, inferiority beliefs, and (pre)diabetes by oral glucose tolerance test were measured at baseline. Incident T2DM was self-reported per annum. We analysed the mediating roles of poor psychosocial resources by using counterfactual mediation analysis. People with low SEP had more often prevalent and incident T2DM (e.g. low education: HR = 2.13, 95%CI: 1.53-2.97). Low control beliefs and high inferiority beliefs were more common among people with low SEP. Moreover, low control beliefs and high inferiority beliefs were risk factors for T2DM (e.g. low control beliefs: HR = 1.50, 95%CI: 1.08-2.09). The relationship between SEP and T2DM was partially mediated by control beliefs (8.0-13.6%) and inferiority beliefs (2.2-4.5%). We conclude that poor psychosocial resources are important in socioeconomic inequalities in diabetes. Researchers and practitioners should consider the psychosocial profile of people with lower SEP, as such a profile might interfere with the development, treatment, and prevention of T2DM. Further research should explore how poor psychosocial resources interact with chronic stress in relation to socioeconomic health inequalities.
Collapse
Affiliation(s)
- Bengisu Sezer
- Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Jeroen Albers
- Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Rachelle Meisters
- Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Miranda T Schram
- Department of Internal Medicine, Maastricht University Medical Center+, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- MHeNS School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Heart and Vascular Center, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Sebastian Köhler
- MHeNS School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Alzheimer Centrum Limburg, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center+, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Annemarie Koster
- Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Hans Bosma
- Department of Social Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
42
|
Ramos-Lopez O, Assmann TS, Astudillo Muñoz EY, Baquerizo-Sedano L, Barrón-Cabrera E, Bernal CA, Bressan J, Cuevas-Sierra A, Dávalos A, De la Cruz-Mosso U, De la Garza AL, De Luis DA, Díaz de la Garza RI, Dos Santos K, Fernández-Condori RC, Fernández-Quintela A, Garcia Diaz DF, Gonzalez-Becerra K, Lopes Rosado E, López de Las Hazas MC, Marín Alejandre BA, Angel Martin A, Martinez-Lopez E, Martínez-Urbistondo D, Milagro FI, Hermsdorff HHM, Muguerza B, Nicoletti CF, Obregón Rivas AM, Parra-Rojas I, Portillo MP, Santos JL, Steemburgo T, Tejero ME, Terán AC, Treviño V, Vizmanos B, Martinez JA. Guidance and Position of RINN22 regarding Precision Nutrition and Nutriomics. Lifestyle Genom 2024; 18:1-19. [PMID: 39617000 PMCID: PMC11844698 DOI: 10.1159/000542789] [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: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Precision nutrition is based on the integration of individual's phenotypical and biological characteristics including genetic variants, epigenetic marks, gut microbiota profiles, and metabolite fingerprints as well as medical history, lifestyle practices, and environmental and cultural factors. Thus, nutriomics areas including nutrigenomics, nutrigenetics, nutriepigenetics, nutrimetabolomics, and nutrimetagenomics have emerged to comprehensively understand the complex interactions between nutrients, diet, and the human body's molecular processes through precision nutrition. SUMMARY This document from the Ibero-American Network of Nutriomics and Precision Nutrition (RINN22; https://rinn22.com/) provides a comprehensive overview of the concepts of precision nutrition approaches to guide their application in clinical and public health as well as establish the position of RINN22 regarding the current and future state of precision nutrition. KEY MESSAGES The progress and participation of nutriomics to precision nutrition is an essential pillar for addressing diet-related diseases and developing innovative managing strategies, which will be promoted by advances in bioinformatics, machine learning, and integrative software, as well as the description of specific novel biomarkers. In this context, synthesizing and critically evaluating the latest developments, potential applications, and future needs in the field of nutrition is necessary with a holistic perspective, incorporating progress in omics technologies aimed at precision nutrition interventions. This approach must address and confront healthy, social, food security, physically active lifestyle, sanitation, and sustainability challenges with preventive, participatory, and predictive strategies of personalized, population, and planetary nutrition for a precision tailored health.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana, Mexico
| | - Taís Silveira Assmann
- Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Elcy Yaned Astudillo Muñoz
- Grupo de Investigación Gerencia del Cuidado, Facultad de Ciencias de la Salud, Universidad Libre Pereira, Pereira, Colombia
| | | | - Elisa Barrón-Cabrera
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacan, Mexico
| | - Claudio Adrián Bernal
- Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Amanda Cuevas-Sierra
- Precision Nutrition and Cardiometabolic Health, IMDEA-Alimentacion Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI), UAM+CSIC, Madrid, Spain
| | - Alberto Dávalos
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, Madrid, Spain
| | - Ulises De la Cruz-Mosso
- Red de Inmunonutrición y Genómica Nutricional en las Enfermedades Autoinmunes, Departamento de Neurociencias, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ana Laura De la Garza
- Universidad Autónoma de Nuevo León, Facultad de Salud Pública y Nutrición, Centro de Investigación en Nutrición y Salud Pública, Monterrey, Mexico
| | - Daniel A. De Luis
- Center of Investigation of Endocrinology and Nutrition, Medicine School and Department of Endocrinology and Investigation, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | | | - Karina Dos Santos
- Graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro (PPGBMC/UNIRIO), Rio de Janeiro, Brazil
| | | | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Vitoria-Gasteiz, Spain
| | - Diego F. Garcia Diaz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Karina Gonzalez-Becerra
- Instituto de Investigación en Genética Molecular, Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega, Universidad de Guadalajara, Ocotlán, Mexico
| | - Eliane Lopes Rosado
- Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - María-Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, Madrid, Spain
| | | | - Alberto Angel Martin
- Escuela de Nutrición y Dietética, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Erika Martinez-Lopez
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Diego Martínez-Urbistondo
- Departamento de Medicina Interna, Area de Medicina Vascular-Madrid, Clinica Universidad de Navarra, Madrid, Spain
| | - Fermin I. Milagro
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | | | - Begoña Muguerza
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Nutrigenomics Research Group, Tarragona, Spain
| | | | - Ana Maria Obregón Rivas
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de Los Bravo, Mexico
| | - Maria Puy Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Center, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Vitoria-Gasteiz, Spain
| | - José L. Santos
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Thais Steemburgo
- Graduate Program in Food, Nutrition, and Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Elizabeth Tejero
- Laboratorio de Nutrigenómica y Nutrigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Anny Cristina Terán
- Hospital Verdi Cevallos Balda, Ministerio de Salud Pública del Ecuador, Portoviejo, Ecuador
| | - Victor Treviño
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, Mexico
| | - Bárbara Vizmanos
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - J. Alfredo Martinez
- Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Centre of Medicine and Endocrinology, University of Valladolid, Valladolid, Spain
| |
Collapse
|
43
|
Ponce Martínez C, Murcia García E, Pérez Sánchez H, Milagro FI, Riezu-Boj JI, Ramos Molina B, Gómez Gallego M, Zamora S, Cañavate Cutillas R, Hernández Morante JJ. Effect of Silibinin on Human Pancreatic Lipase Inhibition and Gut Microbiota in Healthy Volunteers: A Randomized Controlled Trial. Int J Mol Sci 2024; 25:12853. [PMID: 39684564 PMCID: PMC11640983 DOI: 10.3390/ijms252312853] [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: 10/21/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Thistle (Onopordum acanthium) has been traditionally employed for liver protection. However, we recently identified silibinin, the main bioactive compound of thistle extract, as an in vitro pancreatic lipase inhibitor, which suggested a potential role as an anti-obesity agent. This study aimed to assess, in vivo, the efficacy, safety, and effects of silibinin on human lipase. As a secondary objective, we evaluated potential changes in gut microbiota after silibinin treatment. A randomized trial comparing 150 mg/silibinin, 300 mg/silibinin, and a thistle extract (equivalent to 150 mg/silibinin) with placebo and orlistat/120 mg was conducted. Fecal fat excretion, clinical parameters, and microbiota changes were analyzed. Orlistat showed the highest fecal fat excretion, although thistle extract had similar results (p = 0.582). The 150 mg/silibinin group reported the fewest adverse effects. Both silibinin and orlistat reduced plasma triglycerides (p = 0.016) and waist circumference (p = 0.001). Specific microbiota changes, such as increases in Mycobacteriaceae and Veillonellaceae, were associated with higher fat excretion. Although the present work was conducted in the short term and in people of normal weight, our results suggest that silibinin may be safe and effective for obesity, with minimal adverse effects and no significant changes in microbiota diversity. Further studies are needed to explore its microbiota-related benefits.
Collapse
Affiliation(s)
- Cristina Ponce Martínez
- Unidad de Investigación de Trastornos de la Alimentación, Facultad de Enfermería, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain; (C.P.M.); (E.M.G.); (R.C.C.)
| | - Elena Murcia García
- Unidad de Investigación de Trastornos de la Alimentación, Facultad de Enfermería, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain; (C.P.M.); (E.M.G.); (R.C.C.)
- Bioinformatics and High Performance Computing Group, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain;
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain;
| | - Horacio Pérez Sánchez
- Bioinformatics and High Performance Computing Group, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain;
| | - Fermín I. Milagro
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, Universidad de Navarra, C/Irunlarrea, 1, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea, 3, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José I. Riezu-Boj
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea, 3, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Bruno Ramos Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain;
| | - María Gómez Gallego
- Facultad de Ciencias Sociosanitarias, Campus de Lorca, Av. de las Fuerzas Armadas, 0, Lorca, 30800 Murcia, Spain;
| | - Salvador Zamora
- Departamento de Fisiología, Facultad de Biología, Universidad de Murcia, C/Campus Universitario, 5, 30100 Murcia, Spain;
| | - Rubén Cañavate Cutillas
- Unidad de Investigación de Trastornos de la Alimentación, Facultad de Enfermería, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain; (C.P.M.); (E.M.G.); (R.C.C.)
| | - Juan José Hernández Morante
- Unidad de Investigación de Trastornos de la Alimentación, Facultad de Enfermería, Universidad Católica de Murcia, Campus de Guadalupe, Avda. de Los Jerónimos, s/n, 30107 Murcia, Spain; (C.P.M.); (E.M.G.); (R.C.C.)
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain;
| |
Collapse
|
44
|
Yang C, Liu L, Du Y, Zhao L, Liu L, Yang X, Zhao Y. Summer-autumn tea promotes adipocyte browning and thermogenesis in association with gut microbiota regulation in high-fat diet-fed mice. Food Funct 2024; 15:11458-11471. [PMID: 39479981 DOI: 10.1039/d4fo03826f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
This study revealed for the first time the anti-obesity effect of summer-autumn tea aqueous extract (SATE) and its underlying mechanism. High-fat diet (HFD)-fed C57BL/6J mice were treated with or without 400 mg kg-1 SATE for 12 weeks, and administration of SATE significantly ameliorated glucolipid metabolism disorder and induced beige-fat development and brown adipose tissue (BAT)-derived non-shivering thermogenesis via the AMPK-PGC-1α-UCP1 signal axis in HFD-fed mice. 16S rDNA-based microbiota and targeted metabolomics analyses indicated that SATE improved intestinal microbiota dysbiosis and microbial metabolism abnormality caused by HFD, reflected by a dramatic increase in the relative abundance of Muribaculaceae, Bifidobacterium and Odoribacter and production of short-chain fatty acids (SCFAs). Interestingly, SATE-induced thermogenesis was highly correlated with the reconstruction of the gut microbiome and the formation of SCFAs. These findings suggest that SATE has the potential to alleviate obesity by activating adipose browning and thermogenesis in association with the reconstruction of the gut microbiota and its metabolites, providing a theoretical foundation for summer-autumn tea as a functional tea to prevent obesity.
Collapse
Affiliation(s)
- Chengcheng Yang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Luyao Liu
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yao Du
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Lu Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Lu Liu
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Xingbin Yang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
45
|
Patloka O, Komprda T, Franke G. Review of the Relationships Between Human Gut Microbiome, Diet, and Obesity. Nutrients 2024; 16:3996. [PMID: 39683390 DOI: 10.3390/nu16233996] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is a complex disease that increases the risk of other pathologies. Its prevention and long-term weight loss maintenance are problematic. Gut microbiome is considered a potential obesity modulator. The objective of the present study was to summarize recent findings regarding the relationships between obesity, gut microbiota, and diet (vegetable/animal proteins, high-fat diets, restriction of carbohydrates), with an emphasis on dietary fiber and resistant starch. The composition of the human gut microbiome and the methods of its quantification are described. Products of the gut microbiome metabolism, such as short-chain fatty acids and secondary bile acids, and their effects on the gut microbiota, intestinal barrier function and immune homeostasis are discussed in the context of obesity. The importance of dietary fiber and resistant starch is emphasized as far as effects of the host diet on the composition and function of the gut microbiome are concerned. The complex relationships between human gut microbiome and obesity are finally summarized.
Collapse
Affiliation(s)
- Ondřej Patloka
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Tomáš Komprda
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Gabriela Franke
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| |
Collapse
|
46
|
Zhao J, Zhao W, Dong J, Zhang H, Yang K, Gao S, Feng W, Song Y, Qi M, He X. Integrative analysis of metabolites and microbial diversity revealed metabolic mechanism of coarse feeding tolerance in Songliao Black sows during gestation. Front Microbiol 2024; 15:1484134. [PMID: 39629212 PMCID: PMC11611567 DOI: 10.3389/fmicb.2024.1484134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/30/2024] [Indexed: 12/07/2024] Open
Abstract
Dietary fiber is a key nutritional regulatory factor that has been studied intensively for its role in improving reproduction in sows during gestation. However, the metabolic mechanism underlying the effect of interactions between metabolites and gut microbes on coarse feeding tolerance in indigenous sows remains to be elucidated. Therefore, the present study aimed to investigate the effects of dietary supplementation with alfalfa at different content ratios on the reproductive performance of pregnant Songliao Black sows. In total, 40 Songliao Black sows at 30 days of gestation were allocated to four treatments, which received the following diets: (1) a corn-soybean meal basal diet with no alfalfa meal (CON group), (2) a corn-soybean meal basal diet +10% alfalfa meal (Treatment 1 group), (3) a corn-soybean meal basal diet +20% alfalfa meal (Treatment 2 group), and (4) a corn-soybean meal basal diet +30% alfalfa meal (Treatment 3 group). Untargeted metabolomics, 16S rDNA sequencing, and enzyme-linked immunosorbent assay (ELISA) were performed to determine the possible effects of metabolites, the microbial communities in fecal samples and their functional potential, and the effects of dietary fiber on serum biochemical parameters, oxidative stress, and reproductive hormones in Songliao Black sows during gestation. The results revealed that the meals with 10 and 20% alfalfa had a beneficial effect on sows in terms of improving the reproductive performance of these sows. Bacterial 16S rDNA sequencing of the fecal samples revealed that the 10% alfalfa meal group had a higher α-diversity and higher abundance of probiotics. Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria were revealed as the most abundant groups at the phylum level and Lactobacillus, Prevotella, Ruminococcus, Streptococcus, and Clostridium were the most abundant at the genus level in the sows fed with diets containing higher fiber levels. A total of 239 differential metabolites were identified in the sows fed with alfalfa meals. These metabolites were enriched mainly in the cAMP signaling pathway, biosynthesis of amino acids, and steroid biosynthesis. Pearson correlation analysis revealed significant positive correlations between Blautia and Daizein, Fibrobacter and 5-alpha-Cholestanone, Sphaerochaeta, Sutterella, and Metaraminol. Negative correlations were revealed between Sphaerochaeta and Erucic acid, Prevotellaceae and Harmaline, and Streptococcus and 5-alpha-Cholestanone. Collectively, these findings provide novel insights into the application of dietary fiber in sow diets.
Collapse
Affiliation(s)
- Jinbo Zhao
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Wenjiang Zhao
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Jiaqiang Dong
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Hong Zhang
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Kun Yang
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Shengyue Gao
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Wanyu Feng
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Yan Song
- Branch of Animal Husbandry and Veterinary, Heilongjiang Academy of Agricultural Sciences, Qiqihar, China
| | - Meiyu Qi
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Haerbin, China
| | - Xinmiao He
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Haerbin, China
| |
Collapse
|
47
|
Enache RM, Profir M, Roşu OA, Creţoiu SM, Gaspar BS. The Role of Gut Microbiota in the Onset and Progression of Obesity and Associated Comorbidities. Int J Mol Sci 2024; 25:12321. [PMID: 39596385 PMCID: PMC11595101 DOI: 10.3390/ijms252212321] [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: 10/05/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity, a global public health problem, is constantly increasing, so the concerns in preventing and combating it are increasingly focused on the intestinal microbiota. It was found that the microbiota is different in lean people compared to obese individuals, but the exact mechanisms by which energy homeostasis is influenced are still incompletely known. Numerous studies show the involvement of certain bacterial species in promoting obesity and associated diseases such as diabetes, hypertension, cancer, etc. Our aim is to summarize the main findings regarding the influence of several factors such as lifestyle changes, including diet and bariatric surgery, on the diversity of the gut microbiota in obese individuals. The second purpose of this paper is to investigate the potential effect of various microbiota modulation techniques on ameliorating obesity and its comorbidities. A literature search was conducted using the PubMed database, identifying articles published between 2019 and 2024. Most studies identified suggest that obesity is generally associated with alterations of the gut microbiome such as decreased microbial diversity, an increased Firmicutes-to-Bacteroidetes ratio, and increased SCFAs levels. Our findings also indicate that gut microbiota modulation techniques could represent a novel strategy in treating obesity and related metabolic diseases. Although some mechanisms (e.g., inflammation or hormonal regulation) are already considered a powerful connection between gut microbiota and obesity development, further research is needed to enhance the knowledge on this particular topic.
Collapse
Affiliation(s)
- Robert-Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
48
|
Pannerchelvan S, Rios-Solis L, Wasoh H, Sobri MZM, Faizal Wong FW, Mohamed MS, Mohamad R, Halim M. Functional yogurt: a comprehensive review of its nutritional composition and health benefits. Food Funct 2024; 15:10927-10955. [PMID: 39446126 DOI: 10.1039/d4fo03671a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Functional yogurt, renowned for its enhanced nutritional profile and potential health benefits, has emerged as a promising functional food. This review meticulously examines the nutritional composition of functional yogurt, highlighting its enriched content of probiotics, prebiotics, synbiotics, antioxidants, vitamins, minerals, proteins, and other bioactive compounds, which contribute to its health-promoting properties. Functional yogurt has positively affected digestive health, immune function, metabolic health, and mental well-being. It benefits digestive health by alleviating diarrhoeal symptoms, constipation, colon cancer, irritable bowel syndrome (IBS), Helicobacter pylori infection, and digestive-related allergies. Moreover, the immune-boosting properties of functional yogurt play a pivotal role in reducing the risk of infections and inflammation. In addition, functional yogurt has the potential to improve metabolic health, leading to decreased cholesterol levels and enhanced blood sugar regulation. Emerging research also suggests functional yogurt may positively influence mood, behavior, and cognitive function. Functional yogurt is a valuable addition to the human diet, holding significant implications for public health. In addition to its numerous health benefits, functional yogurt also faces limitations, such as the stability of functional compounds, sensory alterations, potential digestive discomfort, and inconsistent efficacy across populations, highlighting the need for further research and optimization.
Collapse
Affiliation(s)
- Sangkaran Pannerchelvan
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Leonardo Rios-Solis
- Department of Biochemical Engineering, University College London, Gower Street, Bernard Katz Building, 6.07, WC1E 6BT, United Kingdom
| | - Helmi Wasoh
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohamad Zulfazli Mohd Sobri
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Fadzlie Wong Faizal Wong
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohd Shamzi Mohamed
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Rosfarizan Mohamad
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Murni Halim
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
49
|
Shang D, Zhao S. Molecular mechanisms of obesity predisposes to atopic dermatitis. Front Immunol 2024; 15:1473105. [PMID: 39564133 PMCID: PMC11574713 DOI: 10.3389/fimmu.2024.1473105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
Obesity is a prevalent metabolic disease that reduces bacterial diversity, colonizes the epidermis with lipophilic bacteria, and increases intestinal pro-inflammatory species, all of which lead to impaired epithelial barriers. Adipose tissue secretes immunomodulatory molecules, such as adipokines, leptin, and adiponectin, which alters the morphology of adipocytes and macrophages as well as modulates T cell differentiation and peripheral Th2-dominated immune responses. Atopic dermatitis (AD) and obesity have similar pathological manifestations, including inflammation as well as insulin and leptin resistance. This review examines the major mechanisms between obesity and AD, which focus on the effect on skin and gut microbiota, immune responses mediated by the toll like receptor (TLR) signaling pathway, and changes in cytokine levels (TNF-a, IL-6, IL-4, and IL13). Moreover, we describe the potential effects of adipokines on AD and finally mechanisms by which PPAR-γ suppresses and regulates type 2 immunity.
Collapse
Affiliation(s)
- Dajin Shang
- School of China Medical University, Shenyang, Liaoning, China
| | - Shengnan Zhao
- School of China Medical University, Shenyang, Liaoning, China
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Chen D, Wang A, Lv J, Peng Y, Zheng Y, Zuo J, Kan J, Zong S, Zeng X, Liu J. Tea (Camellia sinensis L.) flower polysaccharide attenuates metabolic syndrome in high-fat diet induced mice in association with modulation of gut microbiota. Int J Biol Macromol 2024; 279:135340. [PMID: 39255891 DOI: 10.1016/j.ijbiomac.2024.135340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/14/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
There is a growing body of evidence suggesting that dietary polysaccharides play a crucial role in preventing metabolic syndrome (MetS) through their interaction with gut microbes. Tea (Camellia sinensis L.) flower polysacchride (TFPS) is a novel functional compound known for its diverse beneficial effects in both vivo and vitro. To further investigate the effects of TFPS on MetS and gut microbiota, and the possible association between gut microbiota and their activities, this study was carried out on mice that were fed a high-fat diet (HFD) and given oral TFPS at a dose of 400 and 800 mg/kg·body weight (BW)/d, respectively. TFPS treatment significantly mitigated HFD-induced MetS, evidenced by reductions in body weight, fat accumulation, plasma levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and pro-inflammatory cytokines tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and IL-1β, along with an increase in plasma IL-10 levels. Furthermore, TFPS induced alterations in the diversity and composition of HFD-induced gut microbiota. Specifically, TFPS influenced the relative abundance of 11 genera, including Lactobacillus and Lactococcus, which showed strong correlations with metabolic improvements and likely contributed to the amelioration of MetS. In conclusion, TFPS exhibits promising prebiotic properties in preventing MetS and regulating gut microbiota.
Collapse
Affiliation(s)
- Dan Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Ao Wang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Jialiang Lv
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Yiling Peng
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Yunqing Zheng
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Jiayu Zuo
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Shuai Zong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China.
| |
Collapse
|