1
|
Mohsen E, Haffez H, Ahmed S, Hamed S, El-Mahdy TS. Multiple Sclerosis: A Story of the Interaction Between Gut Microbiome and Components of the Immune System. Mol Neurobiol 2025; 62:7762-7775. [PMID: 39934561 PMCID: PMC12078361 DOI: 10.1007/s12035-025-04728-5] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Multiple sclerosis (MS) is defined as an inflammatory disorder that chronically affects the central nervous system of young people mostly and is distributed globally. It is associated with degeneration and demyelination of the myelin sheath around the nerves, resulting in multiple neurological disability symptoms ranging from mild to severe cases that end with paralysis sometimes. MS is one of the rising diseases globally that is unfortunately associated with reduced quality of life and adding national economic burdens. The definite MS mechanism is not clearly defined; however, all the previous researches confirm the role of the immune system as the master contributor in the pathogenesis. Innate and adaptive immune cells are activated peripherally then attracted toward the central nervous system (CNS) due to the breakdown of the blood-brain barrier. Recently, the gut-brain axis was shown to depend on gut metabolites that are produced by different microorganisms in the colon. The difference in microbiota composition between individuals is responsible for diversity in secreted metabolites that affect immune responses locally in the gut or systemically when reach blood circulation to the brain. It may enhance or suppress immune responses in the central nervous system (CNS) (repeated short forms); consequently, it may exacerbate or ameliorate MS symptoms. Recent data showed that some metabolites can be used as adjuvant therapy in MS and other inflammatory diseases. This review sheds light on the nature of MS and the possible interaction between gut microbiota and immune system regulation through the gut-brain axis, hence contributing to MS pathogenesis.
Collapse
Affiliation(s)
- Esraa Mohsen
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
| | - Hesham Haffez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR), Helwan University, Cairo, 11795, Egypt
| | - Sandra Ahmed
- Department of Neurology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Selwan Hamed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt.
| | - Taghrid S El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| |
Collapse
|
2
|
Bacaloni S, Agrawal DK. Nutrition, Gut Microbiota, and Epigenetics in the Modulation of Immune Response and Metabolic Health. CARDIOLOGY AND CARDIOVASCULAR MEDICINE 2025; 9:111-124. [PMID: 40443829 PMCID: PMC12121961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Immune system function is intricately shaped by nutritional status, dietary patterns, and gut microbiota composition. Micronutrients such as vitamins A, C, D, E, B-complex, zinc, selenium, iron, and magnesium are critical for maintaining physical barriers, supporting immune cell proliferation, and regulating inflammation. Macronutrients-including proteins, fats, and carbohydrates-also modulate immune responses through their impact on immune metabolism and the gut-immune axis. Epigenetic mechanisms, including DNA methylation, histone modifications, and microRNA expression, mediate the long-term effects of diet on immune function and tolerance. Diet-induced alterations in gut microbiota further influence immune homeostasis via microbial metabolites like short-chain fatty acids. Imbalanced diets, particularly the Western diet, contribute to immune dysregulation, chronic inflammation, and the development of metabolic disorders such as obesity and type 2 diabetes. While plant-based and Mediterranean dietary patterns have shown anti-inflammatory and immunoregulatory benefits, gaps remain in understanding the long-term epigenetic impacts of these diets. This review integrates current knowledge on how nutrition and the microbiome regulate immunity, highlighting future directions for personalized dietary strategies in preventing chronic immune-related conditions.
Collapse
Affiliation(s)
- Sabrin Bacaloni
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| |
Collapse
|
3
|
Mafe AN, Büsselberg D. Could a Mediterranean Diet Modulate Alzheimer's Disease Progression? The Role of Gut Microbiota and Metabolite Signatures in Neurodegeneration. Foods 2025; 14:1559. [PMID: 40361641 PMCID: PMC12071848 DOI: 10.3390/foods14091559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), the most common form of dementia, represent a growing global health crisis, yet current treatment strategies remain primarily palliative. Recent studies have shown that neurodegeneration through complex interactions within the gut-brain axis largely depends on the gut microbiota and its metabolites. This review explores the intricate molecular mechanisms linking gut microbiota dysbiosis to cognitive decline, emphasizing the impact of microbial metabolites, including short-chain fatty acids (SCFAs), bile acids, and tryptophan metabolites, on neuroinflammation, blood-brain barrier (BBB) integrity, and amyloid-β and tau pathology. The paper highlights major microbiome signatures associated with Alzheimer's disease, detailing their metabolic pathways and inflammatory crosstalk. Dietary interventions have shown promise in modulating gut microbiota composition, potentially mitigating neurodegenerative processes. This review critically examines the influence of dietary patterns, such as the Mediterranean and Western diets, on microbiota-mediated neuroprotection. Bioactive compounds like prebiotics, omega-3 fatty acids, and polyphenols exhibit neuroprotective effects by modulating gut microbiota and reducing neuroinflammation. Furthermore, it discusses emerging microbiome-based therapeutic strategies, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), as potential interventions for slowing Alzheimer's progression. Despite these advances, several knowledge gaps remain, including interindividual variability in microbiome responses to dietary interventions and the need for large-scale, longitudinal studies. The study proposes an integrative, precision medicine approach, incorporating microbiome science into Alzheimer's treatment paradigms. Ultimately, cognizance of the gut-brain axis at a mechanistic level could unlock novel therapeutic avenues, offering a non-invasive, diet-based strategy for managing neurodegeneration and improving cognitive health.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Ar-Rayyan P.O. Box 22104, Qatar
| |
Collapse
|
4
|
Wang H, Liu Z, Yang H, Bai Y, Li Q, Qi X, Li D, Zhao X, Ma Y. Integrated transcriptomics and metabolomics reveal the molecular characteristics and metabolic regulatory mechanisms among different muscles in Minxian black fur sheep. BMC Genomics 2025; 26:412. [PMID: 40301745 PMCID: PMC12039146 DOI: 10.1186/s12864-025-11607-9] [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] [Received: 06/19/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Mammalian skeletal muscle is comprised of heterogeneous fibers with various contractile and metabolic properties that affect muscle flavor. Thus, it is of great significance to identify and characterize the potential molecular characteristics and metabolic regulatory mechanisms associated with muscle fiber properties. RESULTS In this study, the muscle samples (Biceps femoris; longissimus dorsi; and infraspinatus) from Minxian black fur sheep were used to perform transcriptome and metabolome analyses. Then, the key genes regulating the metabolism of important flavor precursors (amino acids and lipids) were explored by integrating transcriptome and metabolome. Consequently, we identified 432 differentially expressed genes, which were mainly involved in muscle development and function maintenance (e.g., myofibril, myocyte differentiation, etc.), metabolism (e.g., fatty acid metabolism, arachidonic acid metabolism, PPAR signaling pathway, and PI3K-Akt signaling pathway, etc.), and homeostasis (e.g., regulation of actin cytoskeleton, ECM-receptor interaction, calcium signaling pathway, etc.). A total of 58 key genes affecting muscle fiber properties, including MYL2, HOXA/C/D, MYBPH8, MYH8, etc., were screened, which characterized the molecular differences in muscle fiber metabolic properties between oxidative and glycolytic muscle. Meanwhile, we identified 463 differentially accumulated metabolites. Except for glycerophospholipids, most flavor metabolites were higher in oxidative muscle. Subsequently, key genes highly related to flavor amino acids were identified by weighted gene co-expression network analysis, such as ALDH6A1, BCKDHB, SLC16A7, LDHB, etc. Based on KEGG enrichment analysis, a regulatory network with both lipid metabolism and its crosstalk with other metabolic pathways was constructed. CONCLUSIONS In conclusion, this study provides an in-depth understanding of the molecular mechanism of differences in muscle fiber properties among different muscles of Minxian black fur sheep, and also lays a foundation for further exploration of the regulatory mechanism of key genes on flavor metabolites.
Collapse
Affiliation(s)
- Huihui Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Zilong Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Hai Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yaqin Bai
- Animal Husbandry Technology Extension Station of Gansu Province, Lanzhou, 730030, China
| | - Qiao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xingcai Qi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Dengpan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xingxu Zhao
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Youji Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China.
| |
Collapse
|
5
|
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
|
6
|
Thonusin C, Suparan K, Kunasol C, Lungruammit N, Nawara W, Arunsak B, Kerdphoo S, Kongkaew A, Songtrai S, Pintana H, Maneechote C, Pratchayasakul W, Kaewsuwan S, Chattipakorn N, Chattipakorn SC. Interruptins Extracted from Cyclosorus terminans Protect Gut Pathologies Induced by High-Fat Diet in Rats. Nutrients 2025; 17:1387. [PMID: 40284250 PMCID: PMC12030309 DOI: 10.3390/nu17081387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The fern "Cyclosorus terminans" (C. terminans) or "Maiden Fern" contains interruptin A and interruptin B. This plant could attenuate obesity, insulin resistance, and fatty liver in rats fed a high-fat/calorie diet. However, the benefits of C. terminans to the gut remain unknown. We investigated the protective effect of C. terminans extract against gut dysfunction in rats exposed to a high-fat/calorie diet. METHODS Male Wistar rats were assigned to receive either (1) a normal diet treated with vehicle, (2) a high-fat/calorie diet treated with vehicle, (3) a high-fat/calorie diet treated with 100 mg per kg per day (mg·kg-1·day-1) of C. terminans extract, or (4) a high-fat/calorie diet treated with 200 mg·kg-1·day-1 of C. terminans extract. The rats were euthanized after 12 weeks of treatment to enable feces and colon tissue collection. RESULTS Both 100 and 200 mg·kg-1·day-1 of C. terminans extract reduced body weight (-10.49%; p = 0.030 and -10.54%; p = 0.037, respectively) and ameliorated gut inflammation, gut barrier disruption, changes in short-chain fatty acid levels, and gut dysbiosis caused by high-fat/calorie diet. CONCLUSIONS C. terminans extract attenuated an increase in body weight and exerted prophylactic effects against gut pathologies induced by high-fat/calorie diet.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokphong Suparan
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanon Kunasol
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sujinda Songtrai
- Faculty of Medical Technology, Rangsit University, Pathumthani 12000, Thailand;
| | - Hiranya Pintana
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
7
|
Ferreira YAM, Estadella D, Pisani LP. Effect of Different Fatty Acid Types on Mitochondrial Dysfunction Associated With Brown and Beige Adipose Tissue. Nutr Rev 2025:nuaf048. [PMID: 40233210 DOI: 10.1093/nutrit/nuaf048] [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: 04/17/2025] Open
Abstract
Adipose tissue serves as a dynamic endocrine organ that is pivotal in metabolic regulation. Augmenting mitochondrial activity within this tissue holds promise in combating obesity. Mitochondrial function is intricately modulated by diverse fatty acid compositions. This comprehensive review aimed to elucidate the molecular mechanisms underlying mitochondrial dysfunction induced by various fatty acid profiles. While saturated fatty acids (SFAs) pose a threat to mitochondrial integrity, polyunsaturated fatty acids (PUFAs), notably n-3, mitigate SFA-induced damage, concurrently regulating thermogenic gene expression. With regard to monounsaturated fatty acids (MUFAs), their impact on mitochondrial function in adipose tissue remains relatively unexplored. Although human studies are imperative for comprehensive insights, prioritizing the consumption of n-3 fatty acids and MUFAs has emerged as a strategic approach, potentially enhancing mitochondrial biogenesis and metabolic pathways. This synthesis underscores the critical need for further investigation of the differential effects of fatty acid types on adipose tissue mitochondria, offering potential avenues for obesity intervention.
Collapse
Affiliation(s)
- Yasmin Alaby Martins Ferreira
- Department of Biosciences, Institute of Health and Science, Federal University of São Paulo (UNIFESP), Santos, SP 11015-020, Brazil
| | - Débora Estadella
- Department of Biosciences, Institute of Health and Science, Federal University of São Paulo (UNIFESP), Santos, SP 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Department of Biosciences, Institute of Health and Science, Federal University of São Paulo (UNIFESP), Santos, SP 11015-020, Brazil
| |
Collapse
|
8
|
de Oliveira KM, Soares GM, da Silva Junior JA, Alves BL, Freitas IN, Bem KCP, Mousovich-Neto F, Ribeiro RA, Carneiro EM. Prolonged postweaning protein restriction induces gut dysbiosis and colonic dysfunction in male mice. Am J Physiol Endocrinol Metab 2025; 328:E599-E610. [PMID: 40019118 DOI: 10.1152/ajpendo.00229.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/30/2024] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Insufficient or imbalanced protein can disrupt gut microbiota, potentially compromising gut barrier function and increasing health risks. Herein, we investigated the effects of protein restriction on cecal microbiota and colon morphofunction in male mice. From 30 to 120 days of age, C57Bl/6 mice were fed a control protein diet [14% protein, control (C) group] or a low-protein diet [6% protein, protein-restricted (R) group]. At the end of the experimental period, R mice exhibited typical features of undernutrition, such as reduced body weight, hypoalbuminemia, and hypoproteinemia. In addition, despite the hyperphagia displayed in the R group, these mice presented a decreased amount of excreted feces and less energy content in feces. Cecal microbiota analysis demonstrated that protein restriction led to reductions in Shannon and Simpson indices and, therefore, dysbiosis. This effect was accompanied by morphological modifications in the proximal colon of R mice, such as 1) reduction in the total area of neurons of myenteric plexus; 2) increased number of goblet cells, with mucin droplets less developed; 3) reductions in crypt depth and diameter; 4) decreases in gene expressions for mucins and in the tight junction proteins expression; 5) enhanced paracellular permeability and expression of pro-inflammatory cytokines (tumor necrosis factor α, toll-like receptor 4, interferon γ, interleukin 1β, and interleukin 6), decreased anti-inflammatory cytokines (interleukins 4 and 10) in the colon, and increased plasma LPS binding protein concentrations. Therefore, protein restriction induced gut dysbiosis and may result in structural and functional negative impacts on the proximal colon barrier against luminal bacteria.NEW & NOTEWORTHY Prolonged postweaning protein restriction induced gut dysbiosis and led to a reduced neuron area in the myenteric plexus, with increased but underdeveloped goblet cells. Protein restriction decreased colonic crypt depth and diameter, and increased paracellular permeability due to lower expression of mucin-related genes and tight junction proteins. The diminished barrier function resulted in systemic inflammation, evidenced by elevated plasma LPS-binding protein and pro-inflammatory markers in the colon.
Collapse
Affiliation(s)
- Kênia Moreno de Oliveira
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela Moreira Soares
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Joel Alves da Silva Junior
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Bruna Lourençoni Alves
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Israelle Netto Freitas
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Kelly Cristina Pereira Bem
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Felippe Mousovich-Neto
- Departments of Pediatrics, Cell and Developmental Biology, Drukier Institute for Children's Health and Meyer Cancer Center, Weill Cornell Medicine, New York, New York, United States
| | - Rosane Aparecida Ribeiro
- Departamento de Biologia Geral, Setor de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa (UEPG), Ponta Grossa, Brazil
| | - Everardo Magalhães Carneiro
- Obesity and Comorbidities Research Center, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
9
|
Jones D, Morrison DJ, Gray SR, Ozanne SE, Celis-Morales C, Jain M, Mattin LR, Gittins M, Alkhedhairi SAA, Dorling JL, Burden S. Dietary intake in healthy older individuals is associated with lipopolysaccharide binding protein a biomarker of gut function: an exploratory cross-sectional study. FRONTIERS IN AGING 2025; 6:1572867. [PMID: 40231185 PMCID: PMC11994966 DOI: 10.3389/fragi.2025.1572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025]
Abstract
Diet, physical function and gut health are important modifiable factors in ageing. However, it is unclear how ageing affects various domains of gut function. Aims of this cross-sectional study were to explore relationships between nutrient intake, physical function, and biomarkers of gut function in older individuals. Healthy participants (n = 94, mean age 71.1 years SD 5.10, 56% female) were recruited to investigate the relationship between nutrient intake (protein, fibre, carbohydrate, fat), physical function (chair rise time, handgrip strength) and lipopolysaccharide (LPS) binding protein (LBP); a marker of gut permeability. Linear regression models, adjusted for age, fat mass/fat free mass ratio, weight and gender, reported LBP changed by; -161.9 ng/mL (95% CI -323.0, -0.8) for every 1 g increase in daily fibre/1,000 kilocalories; 80.5 ng/mL (6.7, 154.2) for 1% increase in daily energy intake as fat; and -88.1 ng/mL (-146.7, -29.6) for 1% increase in daily energy as carbohydrates. When further adjusted for C-reactive protein (CRP), a marker of inflammation, LBP decreased by an additional 6.9 ng/mL for fibre, increased by an additional 4.0 ng/mL for fat and decreased by an additional 3.7 ng/mL for carbohydrate. These findings suggest that in healthy older adults' nutrient intake is associated with LBP, and CRP appears to slightly modify these associations. There were no associations between LBP and handgrip strength or chair rise time. Results suggest that fibre, fat, and carbohydrates are important for maintaining gut function, potentially mediated by inflammation in older adults, although further research is needed to explore the implications for physical function and CRP as a mediator.
Collapse
Affiliation(s)
- Debra Jones
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Douglas J. Morrison
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Stuart R. Gray
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Susan E. Ozanne
- Institute of Metabolic Science - Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Mahek Jain
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Lewis R. Mattin
- School of Life Sciences, University of Westminster, London, United Kingdom
| | - Matthew Gittins
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Saleh A. A. Alkhedhairi
- Department of Medical Biosciences, College of Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - James L. Dorling
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sorrel Burden
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
- Salford Care Organisation, Northern Care Alliance NHS Trust, Salford, United Kingdom
| |
Collapse
|
10
|
Li H, Zhang Y, Peh HY. Interferon regulatory factor 3 beyond innate immunity: Regulation in obesity and metabolic disorders. Semin Immunol 2025; 78:101948. [PMID: 40156960 DOI: 10.1016/j.smim.2025.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Interferon regulatory factor 3 (IRF3) is a transcription factor known primarily for its role in antiviral immunity via regulation of type I interferons (IFNs). Recent research has broadened its significance to encompass metabolic disorders, particularly obesity and diabetes. Obesity is characterized by chronic low-grade inflammation, insulin resistance, and metabolic dysfunction, all of which are increasingly found to be associated with immune signaling pathways. IRF3 has emerged as an important regulator in the development of obesity and type 2 diabetes (T2D), predominantly through its regulation of inflammatory cytokines production in various cells in adipose tissue. In obese individuals, IRF3 is activated in the adipocytes and adipose tissue macrophages, to promote the expression of inflammatory cytokines, thereby contributing to chronic inflammation and exacerbating insulin resistance. Moreover, IRF3 has been linked to mitochondrial dysfunction in hepatic disorders, further amplifying metabolic stress and imbalances associated with obesity. The growing evidence suggests that IRF3 is an important mediator in both immune and metabolic pathways, highlighting its potential as a target for the development of therapeutic interventions for obesity-related inflammation and metabolic dysfunction.
Collapse
Affiliation(s)
- Heng Li
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
| | - Hong Yong Peh
- NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Singapore Lipidomics Incubator, Life Science Institute, National University of Singapore, Singapore 117456, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
11
|
Ma B, Barathan M, Ng MH, Law JX. Oxidative Stress, Gut Microbiota, and Extracellular Vesicles: Interconnected Pathways and Therapeutic Potentials. Int J Mol Sci 2025; 26:3148. [PMID: 40243936 PMCID: PMC11989138 DOI: 10.3390/ijms26073148] [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: 01/15/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Oxidative stress (OS) and gut microbiota are crucial factors influencing human health, each playing a significant role in the development and progression of chronic diseases. This review provides a comprehensive analysis of the complex interplay between these two factors, focusing on how an imbalance between reactive oxygen species (ROS) and antioxidants leads to OS, disrupting cellular homeostasis and contributing to a range of conditions, including metabolic disorders, cardiovascular diseases, neurological diseases, and cancer. The gut microbiota, a diverse community of microorganisms residing in the gastrointestinal tract, is essential for regulating immune responses, metabolic pathways, and overall health. Dysbiosis, an imbalance in the gut microbiota composition, is closely associated with chronic inflammation, metabolic dysfunction, and various diseases. This review highlights how the gut microbiota influences and is influenced by OS, complicating the pathophysiology of many conditions. Furthermore, emerging evidence has identified extracellular vesicles (EVs) as critical facilitators of cellular crosstalk between the OS and gut microbiota. EVs also play a crucial role in signaling between the gut microbiota and host tissues, modulating immune responses, inflammation, and metabolic processes. The signaling function of EVs holds promise for the development of targeted therapies aimed at restoring microbial balance and mitigating OS. Personalized therapeutic approaches, including probiotics, antioxidants, and fecal microbiota transplantation-based strategies, can be used to address OS-related diseases and improve health outcomes. Nonetheless, further research is needed to study the molecular mechanisms underlying these interactions and the potential of innovative interventions to offer novel strategies for managing OS-related diseases and enhancing overall human health.
Collapse
Affiliation(s)
| | | | | | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (B.M.); (M.B.); (M.H.N.)
| |
Collapse
|
12
|
Mun D, Ryu S, Lee DJ, Kwak MJ, Choi H, Kang AN, Lim DH, Oh S, Kim Y. Bovine colostrum-derived extracellular vesicles protect against non-alcoholic steatohepatitis by modulating gut microbiota and enhancing gut barrier function. Curr Res Food Sci 2025; 10:101039. [PMID: 40231313 PMCID: PMC11995039 DOI: 10.1016/j.crfs.2025.101039] [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: 01/23/2025] [Revised: 03/05/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
Non-alcoholic steatohepatitis (NASH), characterized by severe fatty liver-associated inflammation and hepatocellular damage, is a major precursor to cirrhosis and hepatocellular carcinoma. While the exact pathogenesis of NASH remains unclear, gut microbiota dysbiosis has been implicated as a key factor contributing to endotoxin translocation and chronic liver inflammation. Recent studies have highlighted the therapeutic potential of bovine colostrum-derived extracellular vesicles (BCEVs) in modulating gut microbiota and enhancing gut barrier function, but their effects on NASH remain largely unexplored. To investigate the potential protective effects of BCEVs against NASH, 8-wk-old mice were fed a NASH-inducing diet for 3 wks while concurrently receiving oral BCEV administration. BCEV treatment markedly ameliorated hepatic steatosis, fibrosis, and inflammation. Transcriptomic analyses demonstrated a notable reduction in lipid metabolism, bacterial response, and inflammatory pathways in the intestine, as well as reduced expression of inflammation- and fibrosis-related pathways in the liver. Gut microbiota profiling revealed an increased abundance of Akkermansia, accompanied by enhanced cholesterol excretion. Furthermore, BCEV treatment promoted the production of tight junction proteins and mucin in the gut, reinforcing intestinal barrier integrity. These findings suggest that BCEVs promote the proliferation of Akkermansia, which in turn prevents endotoxin translocation to the liver. This reduction in endotoxin leakage alleviates hepatic inflammation and fibrosis. Overall, this study highlights the therapeutic potential of BCEVs as a novel strategy for managing NASH by targeting the gut-liver axis through the modulation of gut microbiota and barrier function.
Collapse
Affiliation(s)
- Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangdon Ryu
- Honam National Institute of Biological Resources, Mokpo, 58762, Republic of Korea
| | - Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyejin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Hyun Lim
- Dairy Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, South Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
13
|
Simancas-Racines D, Román-Galeano NM, Verde L, Annunziata G, Marchetti M, Matos A, Campuzano-Donoso M, Reytor-González C, Muscogiuri G, Barrea L, Frias-Toral E. Targeting Cytokine Dysregulation in Psoriasis: The Role of Dietary Interventions in Modulating the Immune Response. Int J Mol Sci 2025; 26:2895. [PMID: 40243475 PMCID: PMC11988797 DOI: 10.3390/ijms26072895] [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: 03/01/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Psoriasis is a chronic immune-mediated skin disease characterized by cytokine dysregulation. Pro-inflammatory mediators, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-17, and IL-23, play pivotal roles in the pathogenesis of psoriasis. Emerging evidence suggests that dietary interventions can modulate cytokine activity, providing a complementary approach to standard therapies. This narrative review examines the impact of various dietary strategies, including a Mediterranean diet, ketogenic diet, gluten-free diet, and fasting-mimicking diet, on cytokine profiles and clinical outcomes in psoriasis. Research insights reveal that dietary components such as omega-3 fatty acids, polyphenols, and short-chain fatty acids influence immune signaling pathways. These pathways include nuclear factor-kappa B (NF-κB) and Signal Transducer and Activator of Transcription 3 (STAT3). Additionally, these dietary components promote anti-inflammatory effects mediated by gut microbiota. Clinical studies demonstrate significant reductions in psoriasis severity, improved quality of life, and modulation of key cytokines associated with disease activity. Despite these advancements, significant challenges persist in effectively integrating these findings into clinical practice. These challenges include variability in patient responses, adherence issues, and the need for robust biomarkers to monitor efficacy. Future directions emphasize the potential of personalized nutrition and precision medicine approaches to optimize dietary interventions tailored to individual cytokine profiles and genetic predispositions. Integrating these strategies into psoriasis care could transform treatment paradigms by simultaneously addressing both systemic inflammation and comorbid conditions.
Collapse
Affiliation(s)
- Daniel Simancas-Racines
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito 170527, Ecuador; (D.S.-R.); (N.M.R.-G.); (M.C.-D.)
| | - Náthaly Mercedes Román-Galeano
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito 170527, Ecuador; (D.S.-R.); (N.M.R.-G.); (M.C.-D.)
| | - Ludovica Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Giuseppe Annunziata
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, 80143 Naples, Italy;
| | - Marco Marchetti
- Departmental Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, Via Di Sant’Alessandro 8, 00131 Rome, Italy;
| | - Andri Matos
- School of Allied Health, Eastwick College, Ramsey, NJ 07446, USA;
| | - Martín Campuzano-Donoso
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito 170527, Ecuador; (D.S.-R.); (N.M.R.-G.); (M.C.-D.)
| | - Claudia Reytor-González
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito 170527, Ecuador; (D.S.-R.); (N.M.R.-G.); (M.C.-D.)
| | - Giovanna Muscogiuri
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
- Cattedra Unesco “Educazione Alla Salute e Allo Sviluppo Sostenibile”, University Federico II, Corso Umberto I 40, 80131 Naples, Italy
| | - Luigi Barrea
- Dipartimento Psicologia e Scienze della Salute, Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, 80143 Naples, Italy;
| | - Evelyn Frias-Toral
- Escuela de Medicina, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
- Division of Research, Texas State University, 601 University Dr, San Marcos, TX 78666, USA
| |
Collapse
|
14
|
Lebovics N, Heering G, Frishman WH, Lebovics E. Lean MASLD and Cardiovascular Disease: A Review. Cardiol Rev 2025:00045415-990000000-00445. [PMID: 40116510 DOI: 10.1097/crd.0000000000000893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Metabolic-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease, is prevalent worldwide and is highly associated with cardiovascular disease (CVD). Lean MASLD is defined by hepatic steatosis and cardiometabolic risk factors in individuals with a body mass index below 25 in Western populations or below 23 in Asian populations. Paradoxically, some studies indicate that lean MASLD is associated with an elevated risk of cardiovascular (CV) disease and CV mortality compared with nonlean MASLD. Lean MASLD patients exhibit distinctive metabolic, genetic, and microbiome profiles contributing to increased visceral adiposity, sarcopenia, hepatic fibrosis, systemic inflammation, and endothelial dysfunction. This review examines the epidemiology, pathophysiology, and CV outcomes associated with lean MASLD, addressing discrepancies in the literature. Furthermore, it highlights current clinical guidelines, emphasizes lifestyle modifications, and discusses emerging pharmacotherapies as potential treatment options.
Collapse
Affiliation(s)
- Nachum Lebovics
- From the Department of Medicine, NYC Health & Hospitals/Jacobi Medical Center, New York, NY
| | - Gabriel Heering
- Department of Medicine, Westchester Medical Center Health Network, Valhalla, NY
| | - William H Frishman
- Department of Medicine, Westchester Medical Center Health Network, Valhalla, NY
| | - Edward Lebovics
- Department of Medicine, Westchester Medical Center Health Network, Valhalla, NY
| |
Collapse
|
15
|
Kim NH, Kim MY, Yang YM, Jeong WI, Lee HW, Kim W, Kang SG, Han YH. Bacterial components-driven intrahepatic CXCR5 hi B cells are important population for MASH progression through inducing inflammation. FASEB J 2025; 39:e70322. [PMID: 39812617 DOI: 10.1096/fj.202401256r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by severe liver inflammation and fibrosis due to an imbalanced immune response caused by enhanced bacterial components. The progression of MASH is closely linked to increased permeability of intestinal mucosal barrier facilitating enter of bacterial components into hepatic portal venous system. B cells are important immune cells for adaptive responses and enhance hepatic inflammation through cytokine production and T cell activation. B cells are influenced by gut microbiota, but the specific B cell populations in MASH and their pathologic mechanism remain obscure. Here, we found that the numbers of B cells highly expressing CXCR5, the receptor of CXCL13 chemokine, were increased in the livers of MASH. CXCR5 high B cells are non-proliferating naive B cells with inflammatory features mainly residing in hepatic parenchyma to affect liver pathology. Importantly, we revealed that CXCR5 high B cells were induced by bacterial components stimulating TLRs. These bacterial stimulator-induced CXCR5hi B cells highly express TNFα, CD80, and MHC class II, leading to T cell activation. Consistently, we confirmed that intravenous injection of CXCR5 high B cells enhanced hepatic inflammation in MASH model. Ultimately, this study elucidates the role and mechanisms of CXCR5 high B cells in advancing MASH progression.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Mi-Yeon Kim
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Yoon Mee Yang
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
- Multidimentional Genomics Research Center, Kangwon National University, Chuncheon, South Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Hye Won Lee
- Department of Internal Medicine Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, South Korea
| | - Seung Goo Kang
- Department of Molecular Bioscience/Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon, South Korea
| | - Yong-Hyun Han
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
- Multidimentional Genomics Research Center, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
16
|
Pasta A, Formisano E, Calabrese F, Marabotto E, Furnari M, Bodini G, Torres MCP, Pisciotta L, Giannini EG, Zentilin P. From Dysbiosis to Hepatic Inflammation: A Narrative Review on the Diet-Microbiota-Liver Axis in Steatotic Liver Disease. Microorganisms 2025; 13:241. [PMID: 40005608 PMCID: PMC11857840 DOI: 10.3390/microorganisms13020241] [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: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The gut microbiota has emerged as a critical player in metabolic and liver health, with its influence extending to the pathogenesis and progression of steatotic liver diseases. This review delves into the gut-liver axis, a dynamic communication network linking the gut microbiome and liver through metabolic, immunological, and inflammatory pathways. Dysbiosis, characterized by altered microbial composition, contributes significantly to the development of hepatic steatosis, inflammation, and fibrosis via mechanisms such as gut barrier dysfunction, microbial metabolite production, and systemic inflammation. Dietary patterns, including the Mediterranean diet, are highlighted for their role in modulating the gut microbiota, improving gut-liver axis integrity, and attenuating liver injury. Additionally, emerging microbiota-based interventions, such as fecal microbiota transplantation and bacteriophage therapy, show promise as therapeutic strategies for steatotic liver disease. However, challenges such as population heterogeneity, methodological variability, and knowledge gaps hinder the translational application of current findings. Addressing these barriers through standardized approaches and integrative research will pave the way for microbiota-targeted therapies to mitigate the global burden of steatotic liver disease.
Collapse
Affiliation(s)
- Andrea Pasta
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
| | - Elena Formisano
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesco Calabrese
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Manuele Furnari
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giorgia Bodini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Maria Corina Plaz Torres
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Livia Pisciotta
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Edoardo Giovanni Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Patrizia Zentilin
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
17
|
Arellano‐García LI, Milton‐Laskibar I, Martínez JA, Arán‐González M, Portillo MP. Comparative effects of viable Lactobacillus rhamnosus GG and its heat-inactivated paraprobiotic in the prevention of high-fat high-fructose diet-induced non-alcoholic fatty liver disease in rats. Biofactors 2025; 51:e2116. [PMID: 39135211 PMCID: PMC11680974 DOI: 10.1002/biof.2116] [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: 06/04/2024] [Accepted: 07/29/2024] [Indexed: 12/29/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver alterations worldwide, being gut microbiota dysbiosis one of the contributing factors to its development. The aim of this research is to compare the potential effects of a viable probiotic (Lactobacillus rhamnosus GG) with those exerted by its heat-inactivated paraprobiotic counterpart in a dietary rodent model of NAFLD. The probiotic administration effectively prevented the hepatic lipid accumulation induced by a high-fat high-fructose diet feeding, as demonstrated by chemical (lower TG content) and histological (lower steatosis grade and lobular inflammation) analyses. This effect was mainly mediated by the downregulation of lipid uptake (FATP2 protein expression) and upregulating liver TG release to bloodstream (MTTP activity) in rats receiving the probiotic. By contrast, the effect of the paraprobiotic preventing diet-induced liver lipid accumulation was milder, and mainly derived from the downregulation of hepatic de novo lipogenesis (SREBP-1c protein expression and FAS activity) and TG assembly (DGAT2 and AQP9 protein expression). The obtained results demonstrate that under these experimental conditions, the effects induced by the administration of viable L. rhamnosus GG preventing liver lipid accumulation in rats fed a diet rich in saturated fat and fructose differ from those induced by its heat-inactivated paraprobiotic counterpart.
Collapse
Affiliation(s)
- Laura Isabel Arellano‐García
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, Faculty of Pharmacy and Lucio Lascaray Research CentreUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos IIIMadridSpain
| | - Iñaki Milton‐Laskibar
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, Faculty of Pharmacy and Lucio Lascaray Research CentreUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos IIIMadridSpain
- BIOARABA Health Research InstituteVitoria‐GasteizSpain
| | - J. Alfredo Martínez
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos IIIMadridSpain
- Precision Nutrition and Cardiometabolic Health, IMDEA‐Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research CouncilMadridSpain
| | - Miguel Arán‐González
- Unidad de Gestión Clínica de Anatomía Patológica de GuipúzcoaHospital Universitario DonostiaSan SebastiánSpain
| | - María P. Portillo
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, Faculty of Pharmacy and Lucio Lascaray Research CentreUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos IIIMadridSpain
- BIOARABA Health Research InstituteVitoria‐GasteizSpain
| |
Collapse
|
18
|
Yun SW, Shin YJ, Ma X, Kim DH. Lactobacillus plantarum and Bifidobacterium longum Alleviate High-Fat Diet-Induced Obesity and Depression/Cognitive Impairment-like Behavior in Mice by Upregulating AMPK Activation and Downregulating Adipogenesis and Gut Dysbiosis. Nutrients 2024; 16:3810. [PMID: 39599597 PMCID: PMC11597813 DOI: 10.3390/nu16223810] [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: 10/16/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objective: Long-term intake of a high-fat diet (HFD) leads to obesity and gut dysbiosis. AMP-activated protein kinase (AMPK) is a key regulator of energy metabolism. Herein, we investigated the impacts of Lactobacillus (Lactiplantibacillus) plantarum P111 and Bifidobacterium longum P121, which suppressed dexamethasone-induced adipogenesis in 3T3 L1 cells and increased lipopolysaccharide-suppressed AMPK activation in HepG2 cells, on HFD-induced obesity, liver steatosis, gut inflammation and dysbiosis, and depression/cognitive impairment (DCi)-like behavior in mice. Methods: Obesity is induced in mice by feeding with HFD. Biomarker levels were measured using immunoblotting, enzyme-linked immunosorbent assay, and immunofluorescence staining. Results: Orally administered P111, P121, or their mix LpBl decreased HFD-induced body weight gain, epididymal fat pad weight, and triglyceride (TG), total cholesterol (TC), and lipopolysaccharide levels in the blood. Additionally, they downregulated HFD-increased NF-κB activation and TNF-α expression in the liver and colon, while HFD-decreased AMPK activation was upregulated. They also suppressed HFD-induced DCi-like behavior and hippocampal NF-κB activation, NF-κB-positive cell population, and IL-1β and TNF-α levels, while increasing the hippocampal BDNF-positive cell population and BDNF level. The combination of P111 and P122 (LpBl) also improved body weight gain, liver steatosis, and DCi-like behavior. LpBl also mitigated HFD-induced gut dysbiosis: it decreased Desulfovibrionaceae, Helicobacteriaceae, Coriobacteriaceae, and Streptococcaceae populations and lipopolysaccharide production, which were positively correlated with TNF-α expression; and increased Akkermansiaceae, Bifidobacteriaceae, and Prevotellaceae populations, which were positively correlated with the BDNF expression. Conclusions: P111 and/or P121 downregulated adipogenesis, gut dysbiosis, and NF-κB activation and upregulatde AMPK activation, leading to the alleviation of obesity, liver steatosis, and DCi.
Collapse
Affiliation(s)
- Soo-Won Yun
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-W.Y.); (Y.-J.S.); (X.M.)
| | - Yoon-Jung Shin
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-W.Y.); (Y.-J.S.); (X.M.)
| | - Xiaoyang Ma
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-W.Y.); (Y.-J.S.); (X.M.)
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-W.Y.); (Y.-J.S.); (X.M.)
- PBLbioLab, Inc., Seoul 03174, Republic of Korea
| |
Collapse
|
19
|
Brito ML, Coutinho-Wolino KS, Almeida PP, Trigueira PDC, Alves APDP, Magliano DC, Stockler-Pinto MB. Unstressing the Reticulum: Nutritional Strategies for Modulating Endoplasmic Reticulum Stress in Obesity. Mol Nutr Food Res 2024; 68:e2400361. [PMID: 39363792 DOI: 10.1002/mnfr.202400361] [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: 05/15/2024] [Revised: 09/03/2024] [Indexed: 10/05/2024]
Abstract
The progression of obesity involves several molecular mechanisms that are closely associated with the pathophysiological response of the disease. Endoplasmic reticulum (ER) stress is one such factor. Lipotoxicity disrupts endoplasmic reticulum homeostasis in the context of obesity. Furthermore, it induces ER stress by activating several signaling pathways via inflammatory responses and oxidative stress. ER performs crucial functions in protein synthesis and lipid metabolism; thus, triggers such as lipotoxicity can promote the accumulation of misfolded proteins in the organelle. The accumulation of these proteins can lead to metabolic disorders and chronic inflammation, resulting in cell death. Thus, alternatives, such as flavonoids, amino acids, and polyphenols that are associated with antioxidant and anti-inflammatory responses have been proposed to attenuate this response by modulating ER stress via the administration of nutrients and bioactive compounds. Decreasing inflammation and oxidative stress can reduce the expression of several ER stress markers and improve clinical outcomes through the management of obesity, including the control of body weight, visceral fat, and lipid accumulation. This review explores the metabolic changes resulting from ER stress and discusses the role of nutritional interventions in modulating the ER stress pathway in obesity.
Collapse
Affiliation(s)
- Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Karen Salve Coutinho-Wolino
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Patricia Pereira Almeida
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | | | - Ana Paula de Paula Alves
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
- Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, 24020-150, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Nutrition Sciences Postgraduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24020-140, Brazil
| |
Collapse
|
20
|
Shen Y, Wang Y, Lu J, Mo Y, Ma X, Hu G, Zhou J. Habitual use of glucosamine and adverse liver outcomes among patients with type 2 diabetes and MASLD. Liver Int 2024; 44:2359-2367. [PMID: 38842441 DOI: 10.1111/liv.16001] [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: 11/30/2023] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Glucosamine is a dietary supplement commonly used to support joint health. However, there has been interest in exploring other effects of glucosamine on health outcomes due to its ant-inflammation effect. OBJECTIVE This study compared the risks of major adverse liver outcomes (MALOs) between regular users and non-users of glucosamine among patients with type 2 diabetes and metabolic dysfunction associated steatotic liver disease (MASLD) using the data from a large prospective cohort study. METHODS Demographic, anthropometric, laboratory and medication prescription information among 18 753 patients with type 2 diabetes and MASLD was obtained from the UK Biobank. MASLD was identified based on hepatic steatosis defined by fatty liver index ≥60 plus the presence of any clues of metabolic dysregulation and cardio-metabolic risk factors, excluding patients with moderate to severe alcohol consumption. RESULTS During a mean follow-up of 11.4 years, 826 incident MALOs events were recorded. Patients not regularly using glucosamine compared with patients using glucosamine showed a significantly higher risk of the composite MALOs (HR 1.36, 95% confidence interval [CI] 1.09-1.69) as well as most individual MALOs except for ascites. The multivariable-adjusted HRs of MALOs within 3, 5 and 10 years among non-users of glucosamine compared with regular users were 1.79 (95% CI .69-2.03), 1.88 (95% CI 1.21-2.54) and 1.32 (95% CI 1.05-1.72), respectively. Further subgroup analyses in participants with different baseline characteristics and sensitivity analyses excluding participants who regularly took any other supplements and participants who used self-reports to diagnose diabetes confirmed the findings. CONCLUSIONS The present study indicated that habitual use of glucosamine was associated with a low risk of individual and composite MALOs among patients with type 2 diabetes and MASLD.
Collapse
Affiliation(s)
- Yun Shen
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
- Chronic Disease Epidemiology, Pennington Biomedical Researcher Center, Baton Rouge, Louisiana, USA
| | - Yaxin Wang
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
| | - Jingyi Lu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
| | - Yifei Mo
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
| | - Gang Hu
- Chronic Disease Epidemiology, Pennington Biomedical Researcher Center, Baton Rouge, Louisiana, USA
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai, China
| |
Collapse
|
21
|
Samsing F, Sullivan R, Truong H, Rombenso A, Sangster CR, Bannister J, Longshaw M, Becker JA. Replacement of fishmeal with a microbial single-cell protein induced enteropathy and poor growth outcomes in barramundi (Lates calcarifer) fry. JOURNAL OF FISH DISEASES 2024; 47:e13985. [PMID: 38923541 DOI: 10.1111/jfd.13985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
Fish meal (FM) replacement is essential for the sustainable expansion of aquaculture. This study focussed on the feasibility of replacing FM with a single-cell protein (SCP) derived from methanotrophic bacteria (Methylococcus capsulatus, Bath) in barramundi fry (Lates calcarifer). Three isonitrogenous and isoenergetic diets were formulated with 0%, 6.4% and 12.9% inclusion of the SCP, replacing FM by 0%, 25% and 50%. Barramundi fry (initial body weight 2.5 ± 0.1 g) were fed experimental diets for 21 days to assess growth performance, gut microbiome composition and gut histopathology. Our findings revealed that both levels of SCP inclusion induced detrimental effects in barramundi fry, including impaired growth and reduced survival compared with the control group (66.7% and 71.7% survival in diets replacing FM with SCP by 25% and 50%, respectively; p < .05). Both dietary treatments presented mild necrotizing enteritis with subepithelial oedema and accumulation of PAS positive, diastase resistant droplets within hepatocytes (ceroid hepatopathy) and pancreatic atrophy. Microbiome analysis revealed a marked shift in the gut microbial community with the expansion of potential opportunistic bacteria in the genus Aeromonas. Reduced overall performance in the highest inclusion level (50% SCP) was primarily associated with reduced feed intake, likely related to palatability issues, albeit pathological changes observed in gut and liver may also play a role. Our study highlights the importance of meticulous optimization of SCP inclusion levels in aquafeed formulations, and the need for species and life-stage specific assessments to ensure the health and welfare of fish in sustainable aquaculture practices.
Collapse
Affiliation(s)
- Francisca Samsing
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camden, New South Wales, Australia
| | - Roisin Sullivan
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camden, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camden, New South Wales, Australia
| | - Ha Truong
- CSIRO Agriculture and Food, Livestock & Aquaculture Program, Bribie Island, Queensland, Australia
| | - Artur Rombenso
- CSIRO Agriculture and Food, Livestock & Aquaculture Program, Bribie Island, Queensland, Australia
| | - Cheryl R Sangster
- Veterinary Pathology Diagnostic Services (VPDS), Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jo Bannister
- Department of Primary Industries and Regional Development, South Perth, Western Australia, Australia
| | | | - Joy A Becker
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camden, New South Wales, Australia
| |
Collapse
|
22
|
Alcaire F, Giménez A, Ares G. Food additives associated with gut dysbiosis in processed and ultra-processed products commercialized in the Uruguayan market. Food Res Int 2024; 191:114721. [PMID: 39059917 DOI: 10.1016/j.foodres.2024.114721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Recent empirical evidence suggests that gut dysbiosis is one of the negative health outcomes potentially associated with chronic consumption of some food additives. In this context, the present study aimed at analyzing the disclosure of food additives associated with gut dysbiosis in the labels of products commercialized in the Uruguayan market. A cross-sectional survey of packaged products commercialized in nine supermarkets was conducted between August and September 2021. All packaged processed and ultra-processed products available in each data collection site were surveyed using a cellphone app. The information available on the labels was manually extracted and the disclosure of food additives was analyzed using a computer assisted approach. Results showed that 38.1% of the products disclosed at least one food additive associated with gut dysbiosis. Disclosure was most frequent in ice-cream and popsicles, beverages, meat products and analogues, desserts, and fats, oils and fat and oil emulsions. Potassium sorbate was the individual additive associated with gut dysbiosis most frequently disclosed on the labels, followed by mono- and di-glycerides of fatty acid, sucralose, carboxymethylcellulose, acesulphame potassium, carrageenan, and sodium benzoate. These food additives frequently co-occurred and network analysis enabled the identification of patterns of co-occurrence. Taken together, results from the present work suggest the need to conduct additional research to assess the intake of food additives associated with gut dysbiosis at the population level, as well as to evaluate potential synergistic effects of food additives.
Collapse
Affiliation(s)
- Florencia Alcaire
- Sensometrics & Consumer Science, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República. By Pass de Rutas 8 y 101 s/n, CP 91000. Pando, Canelones, Uruguay.
| | - Ana Giménez
- Sensometrics & Consumer Science, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República. By Pass de Rutas 8 y 101 s/n, CP 91000. Pando, Canelones, Uruguay
| | - Gastón Ares
- Sensometrics & Consumer Science, Instituto Polo Tecnológico de Pando, Facultad de Química, Universidad de la República. By Pass de Rutas 8 y 101 s/n, CP 91000. Pando, Canelones, Uruguay
| |
Collapse
|
23
|
Chansa O, Shantavasinkul PC, Monsuwan W, Sirivarasai J. Association between Gut Microbiota Profiles, Dietary Intake, and Inflammatory Markers in Overweight and Obese Women. Foods 2024; 13:2592. [PMID: 39200519 PMCID: PMC11353678 DOI: 10.3390/foods13162592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Being overweight and obesity are significant global public health challenges due to their association with adipose tissue dysfunction, pro-inflammatory marker production, and alterations in gut microbiota composition. To explore the relationship between gut microbiota, dietary factors, and inflammatory markers in overweight or obese women, we conducted a cross-sectional study involving a healthy group (n = 20) and an overweight or obese group (n = 75). We collected data, including clinical, anthropometric, and dietary assessments, and carried out a blood biochemical analysis, the measurement of inflammatory biomarkers (hs-CRP, IL-6, and TNF-α), and the 16S rRNA gene sequencing of fecal samples. The gut microbiota analysis revealed notable differences in alpha and beta diversity between the two groups. Moreover, the abundance of gut microbiota in the overweight or obese group correlated positively with adiposity markers, blood pressure, lipid profiles, and inflammatory markers. These findings highlight significant changes in gut microbiota associated with obesity, potentially implicating pathways such as lipopolysaccharide biosynthesis. Understanding the role of the gut microbiome in obesity could reveal specific avenues for intervention.
Collapse
Affiliation(s)
- Orada Chansa
- Master of Science Program in Nutrition, Faculty of Medicine Ramathibodi Hospital, Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand;
| | | | - Wutarak Monsuwan
- Nutrition Unit, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Jintana Sirivarasai
- Nutrition Unit, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| |
Collapse
|
24
|
Luo XY, Ying SQ, Cao Y, Jin Y, Jin F, Zheng CX, Sui BD. Liver-based inter-organ communication: A disease perspective. Life Sci 2024; 351:122824. [PMID: 38862061 DOI: 10.1016/j.lfs.2024.122824] [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: 05/10/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024]
Abstract
Inter-organ communication through hormones, cytokines and extracellular vesicles (EVs) has emerged to contribute to the physiological states and pathological processes of the human body. Notably, the liver coordinates multiple tissues and organs to maintain homeostasis and maximize energy utilization, with the underlying mechanisms being unraveled in recent studies. Particularly, liver-derived EVs have been found to play a key role in regulating health and disease. As an endocrine organ, the liver has also been found to perform functions via the secretion of hepatokines. Investigating the multi-organ communication centered on the liver, especially in the manner of EVs and hepatokines, is of great importance to the diagnosis and treatment of liver-related diseases. This review summarizes the crosstalk between the liver and distant organs, including the brain, the bone, the adipose tissue and the intestine in noticeable situations. The discussion of these contents will add to a new dimension of organismal homeostasis and shed light on novel theranostics of pathologies.
Collapse
Affiliation(s)
- Xin-Yan Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China; School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Si-Qi Ying
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yuan Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China; Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yan Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Fang Jin
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China.
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
25
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
26
|
Vegda HS, Patel B, Girdhar GA, Pathan MSH, Ahmad R, Haque M, Sinha S, Kumar S. Role of Nonalcoholic Fatty Liver Disease in Periodontitis: A Bidirectional Relationship. Cureus 2024; 16:e63775. [PMID: 39100036 PMCID: PMC11297857 DOI: 10.7759/cureus.63775] [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: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and periodontitis share common risk factors such as obesity, insulin resistance (IR), and dyslipidemia, which contribute to systemic inflammation. It has been suggested that a bidirectional relationship exists between NAFLD and periodontitis, indicating that one condition may exacerbate the other. NAFLD is characterized by excessive fat deposition in the liver and is associated with low-grade chronic inflammation. There are several risk factors for the development of NAFLD, including gender, geriatric community, race, ethnicity, poor sleep quality and sleep deprivation, physical activity, nutritional status, dysbiosis gut microbiota, increased oxidative stress, overweight, obesity, higher body mass index (BMI), IR, type 2 diabetes mellitus (T2DM), metabolic syndrome (MetS), dyslipidemia (hypercholesterolemia), and sarcopenia (decreased skeletal muscle mass). This systemic inflammation can contribute to the progression of periodontitis by impairing immune responses and exacerbating the inflammatory processes in the periodontal tissues. Furthermore, individuals with NAFLD often exhibit altered lipid metabolism, which may affect oral microbiota composition, leading to dysbiosis and increased susceptibility to periodontal disease. Conversely, periodontitis has been linked to the progression of NAFLD through mechanisms involving systemic inflammation and oxidative stress. Chronic periodontal inflammation can release pro-inflammatory cytokines and bacterial toxins into the bloodstream, contributing to liver inflammation and exacerbating hepatic steatosis. Moreover, periodontitis-induced oxidative stress may promote hepatic lipid accumulation and IR, further aggravating NAFLD. The interplay between NAFLD and periodontitis underscores the importance of comprehensive management strategies targeting both conditions. Lifestyle modifications such as regular exercise, a healthy diet, and proper oral hygiene practices are crucial for preventing and managing these interconnected diseases. Additionally, interdisciplinary collaboration between hepatologists and periodontists is essential for optimizing patient care and improving outcomes in individuals with NAFLD and periodontitis.
Collapse
Affiliation(s)
- Hardika S Vegda
- Department of Periodontology and Implantology, School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Bhavin Patel
- Department of Periodontology and Implantology, School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Gaurav A Girdhar
- Department of Periodontology and Implantology, School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Mohd Shabankhan H Pathan
- Department of Periodontology and Implantology, School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka, BGD
| | - Mainul Haque
- Department of Research, Karnavati Scientific Research Center (KSRC) School of Dentistry, Karnavati University, Gandhinagar, IND
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| | - Susmita Sinha
- Department of Physiology, Enam Medical College and Hospital, Dhaka, BGD
| | - Santosh Kumar
- Department of Periodontology and Implantology, School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
27
|
Kim NH, Lee SJ, Lee KJ, Song AR, Park HJ, Kang JS, Cha JY, Han YH. The Root Extract of Rosa multiflora Ameliorates Nonalcoholic Steatohepatitis Development via Blockade of De Novo Lipogenesis and Inflammation. Curr Issues Mol Biol 2024; 46:5881-5893. [PMID: 38921022 PMCID: PMC11202599 DOI: 10.3390/cimb46060351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by severe inflammation and fibrosis due to an excessive accumulation of triglycerides (TGs) in the liver with a dysregulated de novo lipogenesis (DNL) pathway. In this study, we aimed to evaluate the effectiveness of YC-1102, an extract obtained from the roots of Rosa multiflora, as a nutritional supplement in a diet-induced NASH mouse model. C57BL/6 wild-type mice were fed a fructose, palmitate, and cholesterol (FPC)-containing diet for 16 weeks to induce experimental NASH. A daily oral gavage of YC-1102 and obetichoic acid (OCA) was conducted for 9 weeks. After sacrifice, disease parameters related to hepatic lipids, inflammation, and fibrosis were evaluated. The treatment with YC-1102 significantly decreased the liver/body weight ratio, epididymal fat weight, and plasma ALT and AST levels, which are indicators of NASH injuries. YC-1102 attenuated hepatic lipid accumulation by inhibiting the transcription of DNL genes in the livers exhibiting NASH. Additionally, we found that YC-1102 blocked the development of hepatic inflammation and fibrosis by directly disturbing macrophage activation, resulting in an amelioration of hepatic fibrosis. Our findings suggest that YC-1102 could ameliorate NASH progression by inhibiting uncontrolled DNL and inflammation.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Seung-Jin Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Kyeong-Jin Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Ae Ri Song
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Hyun-Je Park
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Jong Soo Kang
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Joo Young Cha
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
28
|
Sati P, Dhyani P, Sharma E, Attri DC, Jantwal A, Devi R, Calina D, Sharifi-Rad J. Gut Microbiota Targeted Approach by Natural Products in Diabetes Management: An Overview. Curr Nutr Rep 2024; 13:166-185. [PMID: 38498287 DOI: 10.1007/s13668-024-00523-1] [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] [Accepted: 02/16/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE OF REVIEW This review delves into the complex interplay between obesity-induced gut microbiota dysbiosis and the progression of type 2 diabetes mellitus (T2DM), highlighting the potential of natural products in mitigating these effects. By integrating recent epidemiological data, we aim to provide a nuanced understanding of how obesity exacerbates T2DM through gut flora alterations. RECENT FINDINGS Advances in research have underscored the significance of bioactive ingredients in natural foods, capable of restoring gut microbiota balance, thus offering a promising approach to manage diabetes in the context of obesity. These findings build upon the traditional use of medicinal plants in diabetes treatment, suggesting a deeper exploration of their mechanisms of action. This comprehensive manuscript underscores the critical role of targeting gut microbiota dysbiosis in obesity-related T2DM management and by bridging traditional knowledge with current scientific evidence; we highlighted the need for continued research into natural products as a complementary strategy for comprehensive diabetes care.
Collapse
Affiliation(s)
- Priyanka Sati
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Praveen Dhyani
- Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany
| | - Eshita Sharma
- Department of Biochemistry and Molecular Biology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Dharam Chand Attri
- Department of Botany, Central University of Jammu, Rahya-Suchani (Bagla), Jammu and Kashmir, India
| | - Arvind Jantwal
- Department of Pharmaceutical Sciences, Kumaun University, Bhimtal, Uttarakhand, India
| | - Rajni Devi
- Department of Microbiology, Punjab Agricultural University, Ludhiana-141004, Punjab, India
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | |
Collapse
|
29
|
Turner TA, Lehman P, Ghimire S, Shahi SK, Mangalam A. Game of microbes: the battle within - gut microbiota and multiple sclerosis. Gut Microbes 2024; 16:2387794. [PMID: 39114974 PMCID: PMC11313001 DOI: 10.1080/19490976.2024.2387794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/03/2024] [Accepted: 07/30/2024] [Indexed: 08/11/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic and progressive autoimmune disease of the central nervous system (CNS), with both genetic and environmental factors contributing to the pathobiology of the disease. While human leukocyte antigen (HLA) genes have emerged as the strongest genetic factor, consensus on environmental risk factors are lacking. Recently, trillions of microbes residing in our gut (microbiome) have emerged as a potential environmental factor linked with the pathobiology of MS as PwMS show gut microbial dysbiosis (altered gut microbiome). Thus, there has been a strong emphasis on understanding the factors (host and environmental) regulating the composition of the gut microbiota and the mechanism(s) through which gut microbes contribute to MS disease, especially through immune system modulation. A better understanding of these interactions will help harness the enormous potential of the gut microbiota as a therapeutic approach to treating MS.
Collapse
Affiliation(s)
- Ti-Ara Turner
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
- Iowa City VA Health Care System, Iowa City, IA, USA
| | - Peter Lehman
- Iowa City VA Health Care System, Iowa City, IA, USA
- Experimental Pathology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Sudeep Ghimire
- Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Shailesh K. Shahi
- Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ashutosh Mangalam
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
- Iowa City VA Health Care System, Iowa City, IA, USA
- Experimental Pathology Graduate Program, University of Iowa, Iowa City, IA, USA
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
30
|
Wouters F, van der Hilst J, Bogie J. Lipids in inflammasome activation and autoinflammatory disorders. J Allergy Clin Immunol 2024; 153:1-11. [PMID: 37871669 DOI: 10.1016/j.jaci.2023.10.008] [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: 05/16/2023] [Revised: 09/06/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Autoinflammatory diseases (AIDs) are a group of rare monogenetic disorders characterized by recurrent episodes of fever and systemic inflammation. A major pathologic hallmark of AIDs is excessive inflammasome assembly and activation, often the result of gain-of-function mutations in genes encoding core inflammasome components, including pyrin and cryopyrin. Recent advances in lipidomics have revealed that dysregulated metabolism of lipids such as cholesterol and fatty acids, especially in innate immune cells, exerts complex effects on inflammasome activation and the pathogenesis of AIDs. In this review, we summarize and discuss the impact of lipids and their metabolism on inflammasome activation and the disease pathogenesis of the most common AIDs, including familial Mediterranean fever, cryopyrin-associated periodic syndromes, and mevalonate kinase deficiency. We postulate that lipids hold diagnostic value in AIDs and that dietary and pharmacologic intervention studies could represent a promising approach to attenuate inflammasome activation and AID progression.
Collapse
Affiliation(s)
- Flore Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jeroen van der Hilst
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium; Department of Infectious Diseases and Immune Pathology, Jessa General Hospital and Limburg Clinical Research Center, Hasselt, Belgium
| | - Jeroen Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium; University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
31
|
Jayachandran M, Qu S. Non-alcoholic fatty liver disease and gut microbial dysbiosis- underlying mechanisms and gut microbiota mediated treatment strategies. Rev Endocr Metab Disord 2023; 24:1189-1204. [PMID: 37840104 DOI: 10.1007/s11154-023-09843-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is by far the most prevalent form of liver disease worldwide. It's also the leading cause of liver-related hospitalizations and deaths. Furthermore, there is a link between obesity and NAFLD risk. A projected 25% of the world's population grieves from NAFLD, making it the most common chronic liver disorder. Several factors, such as obesity, oxidative stress, and insulin resistance, typically accompany NAFLD. Weight loss, lipid-lowering agents, thiazolidinediones, and metformin help prominently control NAFLD. Interestingly, pre-clinical studies demonstrate gut microbiota's potential causal role in NAFLD. Increased intestinal permeability and unhindered transport of microbial metabolites into the liver are the major disruptions due to gut microbiome dysbiosis, contributing to the development of NAFLD by dysregulating the gut-liver axis. Hence, altering the pathogenic bacterial population using probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) could benefit patients with NAFLD. Therefore, it is crucial to acknowledge the importance of microbiota-mediated therapeutic approaches for NAFLD and comprehend the underlying mechanisms that establish a connection between NAFLD and gut microbiota. This review provides a comprehensive overview of the affiliation between dysbiosis of gut microbiota and the progress of NAFLD, as well as the potential benefits of prebiotic, probiotic, synbiotic supplementation, and FMT in obese individuals with NAFLD.
Collapse
Affiliation(s)
- Muthukumaran Jayachandran
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai center of Thyroid diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
32
|
Cardoso-Lezama I, Fuentes-Figueroa MÁ, Ramos-Tovar E, Márquez-Quiroga LV, Ortiz-Fernández A, Vargas-Pozada EE, Arellanes-Robledo J, Tsutsumi V, Muriel P. Nicotinic acid attenuates experimental non-alcoholic steatohepatitis by inhibiting the NLRP3 inflammasome/pyroptosis pathway. Biochem Pharmacol 2023; 216:115762. [PMID: 37604293 DOI: 10.1016/j.bcp.2023.115762] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/05/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a global public health concern that may progress into fibrosis, cirrhosis, and liver cancer, with limited curative treatment options. While the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is closely linked to NASH progression, nicotinic acid (NA), a vitamin used for the treatment of dyslipidemia, is an emerging pharmaceutical treatment for hepatic steatosis and fibrosis. Here, we investigated pharmacological effects of NA on experimental NASH and whether NLRP3 inflammasome/pyroptosis inhibition is an associated mechanism of action. Rats were fed a high-fat sucrose diet supplemented with cholesterol and a low dose of CCl4. NA significantly reduced inflammation by decreasing the protein levels of tumor necrosis factor-alpha and nuclear factor kappa B. Moreover, NA inhibited the formation of NLRP3- apoptosis-associated speck-like protein containing caspase recruitment domain-Caspase-1, decreasing interleukin-1beta, interleukin-18, and gasdermin D protein. In addition, NA reduced tumor growth factor-beta, alpha-smooth muscle actin, and hepatic levels of collagen-1, consequently decreasing extracellular matrix synthesis. Our results indicate that NA can inhibit NASH progression and encourage further basic and clinical studies on the use of NA for the treatment of human NASH.
Collapse
Affiliation(s)
- Irina Cardoso-Lezama
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México
| | - Miguel Á Fuentes-Figueroa
- Laboratorio de Biología Celular y Productos Naturales, Escuela Nacional de Medicina y Homeopatía-IPN, Apartado Postal, 07320, Guillermo Massieu Helguera, La Escalera, Ciudad de México, México
| | - Erika Ramos-Tovar
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Apartado Postal 11340, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| | - Linda V Márquez-Quiroga
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México
| | - Arturo Ortiz-Fernández
- Departamento de Biología Celular, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, Apartado Postal 14-740, Ciudad de México, México
| | - Eduardo E Vargas-Pozada
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México
| | - Jaime Arellanes-Robledo
- Laboratorio de Enfermedades Hepáticas, Instituto Nacional de Medicina Genómica, Ciudad de México, México; Dirección de Cátedras, Consejo Nacional de Ciencia y Tecnología, Ciudad de México, México
| | - Víctor Tsutsumi
- Departamento de Infectómica y Patología Experimental, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, Apartado Postal 14-740, Ciudad de México, México
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México.
| |
Collapse
|
33
|
Morais VND, Gomes MJC, Grancieri M, Moreira LDPD, Toledo RCL, Costa NMB, da Silva BP, Martino HSD. Chia (Salvia hispanica L.) flour modulates the intestinal microbiota in Wistar rats fed a high-fat and high-fructose diet. Food Res Int 2023; 172:113095. [PMID: 37689868 DOI: 10.1016/j.foodres.2023.113095] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/21/2023] [Accepted: 06/09/2023] [Indexed: 09/11/2023]
Abstract
A diet rich in sugar and fat can promote metabolic disorders development, especially in the intestine. Chia flour (Salvia hispanica. L) is a source of dietary fiber, alpha-linolenic fatty acid (ALA), bioactive peptides, and phenolics, promoting health benefits. This study aimed to analyze chia flour's effect on gut microbiota modulation and intestinal health in adult male Wistar rats fed a high-fat and high-fructose (HFHF) diet. Male Wistar rats (n = 10/group) were fed the diets standard (AIN-93M) or HFHF (31% saturated fat and 20% fructose) in the first phase to induce metabolic disorders. In the second phase, the rats were fed AIN-93M, HFHF, or HFHF plus 14.7% chia flour (HFHF + CF) for 10 weeks. The consumption of chia flour increased the ALA (3.24 ± 0.24) intake and significantly improved immunoglobulin A (IgA) levels (1126.00 ± 145.90), goblet cells number (24.57 ± 2.76), crypt thickness (34.37 ± 5.86), crypt depth (215.30 ± 23.19), the longitudinal muscle layer (48.11 ± 5.04), cecum weight (4.39 ± 0.71), Shannon index (p < 0.05), and significantly increased the production of acetic (20.56 ± 4.10) and butyric acids (5.96 ± 1.50), Monoglobus sp., Lachnospiraceae sp., and Prevotellaceae sp. abundance. Furthermore, chia significantly reduced the cecal pH content (7.54 ± 1.17), body mass index (0.62 ± 0.03) and weight (411.00 ± 28.58), and Simpson index (p < 0.05). Therefore, chia intake improved intestinal health parameters and functionality in rats with metabolic disorders, which demonstrates to be an effective strategy for gut microbiota modulation.
Collapse
Affiliation(s)
- Violeta Nunes de Morais
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | | | - Mariana Grancieri
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre 29500-000, ES, Brazil
| | - Luiza de Paula Dias Moreira
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Legnaro 16,35020, PD, Italy; Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås 1430, Viken, Norway
| | | | - Neuza Maria Brunoro Costa
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre 29500-000, ES, Brazil
| | | | | |
Collapse
|
34
|
Liu D, Wang S, Liu Y, Luo Y, Wen B, Wu W, Zeng H, Huang J, Liu Z. Fuzhuan brick tea ameliorates hepatic steatosis and steatohepatitis through gut microbiota-derived aryl hydrocarbon receptor ligands in high-fat diet-induced obese mice. Food Funct 2023; 14:8351-8368. [PMID: 37606634 DOI: 10.1039/d3fo01782f] [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: 08/23/2023]
Abstract
High-fat diet (HFD) induced obesity and its associated conditions, such as hepatic steatosis and steatohepatitis, are major health concerns worldwide. Previous studies have reported the excellent efficiency of Fuzhuan brick tea (FBT) in attenuating HFD-induced obesity and metabolic disorders. In this study, we investigated the effects of FBT on hepatic steatosis and simple steatohepatitis in HFD-induced obese mice, as well as the metabolic function of the gut microbiome using metagenomics and metabolomics. The results showed that FBT ameliorated dyslipidemia, hepatic steatosis and steatohepatitis in HFD-induced obese mice by normalizing the gut microbiota structure and tryptophan metabolism. FBT increased the cecal abundance of aryl hydrocarbon receptor (AhR)-ligand producing bacteria such as Lactobacillus_reuteri and Lactobacillus_johnsonii, at the expense of AhR-ligand consuming bacteria, such as Faecalibaculum_rodentium and Escherichia_coli, and elevated the cecal contents of AhR-ligands such as IAA, IPA, and KYNA. Furthermore, FBT regulated the expressions of AhR and its targeted lipometabolic genes such as Pemt, Fasn, and SREBP-1c, as well as other inflammatory genes including TNF-α, IL-6, and IL-1β in the liver of mice. Overall, these findings highlight the beneficial effects of FBT on obesity-related hepatic steatosis and steatohepatitis via microbiota-derived AhR signaling.
Collapse
Affiliation(s)
- Dongmin Liu
- Changsha University of Science & Technology, Changsha 410114, China
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
| | - Siyu Wang
- Changsha University of Science & Technology, Changsha 410114, China
| | - Yaqing Liu
- Changsha University of Science & Technology, Changsha 410114, China
| | - Yong Luo
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| | - Beibei Wen
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| | - Wenliang Wu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- Tea Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Hongliang Zeng
- Research Institute of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, China
| | - Jianan Huang
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| |
Collapse
|
35
|
Lee EJ, Edward OC, Seo EB, Mun EG, Jeong SJ, Ha G, Han A, Cha YS. Gochujang Ameliorates Hepatic Inflammation by Improving Dysbiosis of Gut Microbiota in High-Fat Diet-Induced Obese Mice. Microorganisms 2023; 11:microorganisms11040911. [PMID: 37110334 PMCID: PMC10141003 DOI: 10.3390/microorganisms11040911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Abnormal fat accumulation with gut microbiota dysbiosis results in hepatic inflammation by upregulating the release of lipopolysaccharide (LPS) and inflammatory cytokine. Gochujang, a traditional fermented condiment, has beneficial effects, such as anti-colonic inflammatory effects. However, Gochujang has been controversial because of its high salt content (the Korean Paradox). Thus, the present study aimed to investigate the preventative effects of Gochujang on hepatic inflammation and related gut microbiota through discussing the Korean Paradox. The mice were divided into groups including a normal diet (ND), high-fat diet (HD), HD with salt (SALT), HD with a high percentage of beneficial microbiota Gochujang (HBM), and HD with diverse beneficial microbiota Gochujang (DBM). Gochujang markedly reduced lipid accumulation, hepatic injury, and inflammation response. Furthermore, Gochujang attenuated protein expression involved in the JNK/IκB/NF-κB pathway. Additionally, Gochujang regulated the gut microbiota-derived LPS production and Firmicutes/Bacteroidetes ratio. Gochujang regulated the levels of gut microbiota such as Bacteroides, Muribaculum, Lactobacillus, and Enterorhabdus, which were correlated with hepatic inflammation. Salt did not have foregoing effects, meaning that the salt content in Gochujang did not affect its anti-inflammatory effect. In conclusion, Gochujang showed anti-hepatic inflammation effects via reduced lipid accumulation, hepatic injury, and inflammatory response together with reorganization of gut microbiota dysbiosis regardless of salt content and the difference of micro bacteria composition.
Collapse
Affiliation(s)
- Eun-Ji Lee
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Olivet Chiamaka Edward
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Eun-Bi Seo
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Eun-Gyung Mun
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Su-Ji Jeong
- Department of R & D, Microbial Institute for Fermentation Industry, Sunchang-gun 56000, Republic of Korea
| | - Gwangsu Ha
- Department of R & D, Microbial Institute for Fermentation Industry, Sunchang-gun 56000, Republic of Korea
| | - Anna Han
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|