1
|
Nie P, Hu L, Feng X, Xu H. Gut Microbiota Disorders and Metabolic Syndrome: Tales of a Crosstalk Process. Nutr Rev 2025; 83:908-924. [PMID: 39504479 DOI: 10.1093/nutrit/nuae157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
The microbiota in humans consists of trillions of microorganisms that are involved in the regulation of the gastrointestinal tract and immune and metabolic homeostasis. The gut microbiota (GM) has a prominent impact on the pathogenesis of metabolic syndrome (MetS). This process is reciprocal, constituting a crosstalk process between the GM and MetS. In this review, GM directly or indirectly inducing MetS via the host-microbial metabolic axis has been systematically reviewed. Additionally, the specifically altered GM in MetS are detailed in this review. Moreover, short-chain fatty acids (SCFAs), as unique gut microbial metabolites, have a remarkable effect on MetS, and the role of SCFAs in MetS-related diseases is highlighted to supplement the gaps in this area. Finally, the existing therapeutics are outlined, and the superiority and shortcomings of different therapeutic approaches are discussed, in hopes that this review can contribute to the development of potential treatment strategies.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation Co., Ltd, Nanchang University, Nanchang 330200, China
| |
Collapse
|
2
|
Shi L, Wang W, Jing C, Hu J, Liao X. Berberine and health outcomes: an overview of systematic reviews. BMC Complement Med Ther 2025; 25:147. [PMID: 40269802 PMCID: PMC12016319 DOI: 10.1186/s12906-025-04872-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Berberine is an isoquinoline alkaloid isolated from Chinese herb coptis chinensis and other berberis plants which can be used to treat a wide range of chronic diseases. However, the current research evidence on the therapeutic effects of berberine has not been summarized. We aimed to synthesize the current evidence on the systematic review (SRs) of berberine for the treatment of diverse conditions. METHODS A comprehensive search of the Cochrane Library, PubMed, EMBASE, Web of Science, CNKI, Wanfang, VIP, and SinoMed was performed from the database inception to April 11, 2024. SRs on berberine were included and evaluated. The methodological quality and the reporting quality of each SR were assessed using the AMSTAR-2 tool and PRISMA checklist, respectively. The quality of evidence was appraised based on the GRADE. RESULTS Fifty-four SRs were included and analyzed. Overall, associations were found between berberine and 70 health outcomes concerned with 9 diseases. Berberine has improved most outcomes of these diseases: 78% (25/32) cardiovascular disease outcomes, 92.59% (25/27) type 2 diabetes mellitus outcomes, 94.74% (18/19) gastrointestinal disorders outcomes, 72.22% (13/18) polycystic ovary syndrome (PCOS) outcomes, 86.67% (13/15) non-alcoholic fatty liver disease (NAFLD) outcomes, 92.31% (12/13) schizophrenia outcomes, 90.91% (10/11) metabolic syndrome outcomes, 57.14% (4/7) obesity outcomes, and 100.00% (6/6) dyslipidemia outcomes. There was a high overlap of primary studies (CCA > 15%) in the SRs of PCOS, NAFLD, obesity, and schizophrenia. Only one SR was rated as high quality while eight SRs were rated as low quality and forty-five SRs as very low quality according to AMSTAR-2. Regarding the reporting quality, Item 14, 15, 21, and 22 were poorly reported for the included SRs in terms of PRSMA assessment. For GRADE, eight outcomes were rated as high quality evidence, twenty-two outcomes were rated as moderate quality, and 110 outcomes were rated as low quality. CONCLUSION Current evidence suggests that berberine has beneficial effects on a range of health outcomes for people with chronic diseases. Specifically, berberine significantly improves type 2 diabetes, gastrointestinal disorders, schizophrenia, metabolic syndrome, and dyslipidemia outcomes. However, caution is needed considering the shortcomings in the quality of the relevant system reviews included.
Collapse
Affiliation(s)
- Lanjun Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Wenya Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Chengyang Jing
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Jing Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, China.
| | - Xing Liao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Science, Beijing, China.
| |
Collapse
|
3
|
Rabiu L, Zhang P, Liu Z, Tang Y, Gidado KI, Ibrahim A, Saliu MA, Tariq HK, Wan X, Xu S, Xu Z, Zhang G. TIPE2 deficiency amplifies inflammation and immune dysregulation in MASH through modulating hepatic lipid metabolism and immune cell function. Inflamm Res 2025; 74:65. [PMID: 40244311 DOI: 10.1007/s00011-025-02031-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Metabolic Dysfunction-Associated Steatohepatitis (MASH) affects nearly 25% of the global population, yet there are no effective pharmacological treatments. Tumor necrosis factor α-induced protein 8-like 2 (TIPE2) is expressed in various immune cells and is crucial for regulating both innate and adaptive immune responses. However, its role in MASH development and the underlying mechanisms remain unclear. METHOD In this study, the role of TIPE2 in MASH was investigated using TIPE2 knockout (KO) mice and human hepatic LO2 cells. Immune cell infiltration, cytokine levels, and gene expression were analyzed. Techniques included flow cytometry for immune cell profiling, cytokine analysis, RNA sequencing, and quantitative PCR (qPCR) for validating gene expression changes. RESULTS TIPE2 was identified as a key regulator in MASH, influencing immune modulation and metabolic processes. TIPE2 KO mice exhibited increased infiltration and activation of natural killer (NK) cells, M1 macrophages, and myeloid-derived suppressor cells (MDSCs), along with elevated pro-inflammatory cytokines such as IFN-gamma, TNF-alpha, IL- 1 beta, and IL- 6. MDSCs from TIPE2 KO mice demonstrated enhanced PD-L1 expression, contributing to chronic liver inflammation through T cell suppression. RNA sequencing revealed that TIPE2 overexpression in human hepatic LO2 cells upregulated genes associated with amino acid biosynthesis, carbon metabolism, lipid regulation, glycolysis, and gluconeogenesis. These findings were supported by qPCR analyses of liver samples from mice, confirming TIPE2's role in maintaining lipid homeostasis and modulating immune responses. CONCLUSION The study highlights the pivotal role of TIPE2 in immune regulation and its influence on immune cell activation and inflammatory responses, which are critical in MASH progression. By exploring TIPE2-mediated immune regulation and its impact on the interplay between immune cell dynamics and liver metabolism, this research underscores TIPE2's central role in linking immune dysfunction to metabolic disturbances in MASH.
Collapse
Affiliation(s)
- Lawan Rabiu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Pengchao Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhongming Liu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Yexiao Tang
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China
| | - Khalid I Gidado
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Abdulrahman Ibrahim
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Muhammad A Saliu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Hafiza Kashaf Tariq
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Shu Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China.
| | - Zhiming Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, PR China.
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
4
|
Di Fulvio M, Rathod YD, Khader S. Diuretics: a review of the pharmacology and effects on glucose homeostasis. Front Pharmacol 2025; 16:1513125. [PMID: 40223924 PMCID: PMC11985539 DOI: 10.3389/fphar.2025.1513125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Thiazides, thiazide-like and loop diuretics are commonly prescribed to manage hypertension and heart failure. The main mechanism of action of these diuretics involve inhibition of Na+ reabsorption in the kidneys, leading to increased urine production. While effective, diuretics, particularly hydrochlorothiazide, have been linked to altered glucose metabolism and other metabolic issues. These disruptions in fuel homeostasis are not clearly related to their primary action of fluid management, raising concerns for patients with metabolic syndrome, in which high blood pressure coexists with obesity, insulin resistance, glucose intolerance and dyslipidemia. In this review, we conducted an extensive examination of existing literature on these classes of diuretics, covering publications from the late 1950s to the present. Our objective was to investigate the origins, development and current understanding of the widely recognized association between the use of diuretics in general and their potential negative impact on glucose homeostasis. We focused on the clinical and experimental evidence of the most commonly prescribed diuretics: hydrochlorothiazide, chlorthalidone, bumetanide and furosemide. On one hand, the clinical evidence supports the hypothesis that the metabolic effects on glucose homeostasis are primarily linked to hydrochlorothiazide, with little, if any impact observed in other diuretics. In addition, these metabolic effects do not appear to be related to their diuretic action or intended pharmacological targets, raising concerns about the long-term metabolic impact of specific diuretics, particularly in vulnerable populations, including those with metabolic syndrome. On the other hand, the experimental evidence using animal models suggest variable effects of diuretics in insulin secretion and general glucose metabolism. Although the mechanisms involved are not clearly understood, further research is needed to uncover the molecular mechanisms by which certain diuretics disrupt fuel metabolism and contribute to metabolic disturbances.
Collapse
Affiliation(s)
- Mauricio Di Fulvio
- Department of Pharmacology and Toxicology, School of Medicine, Wright State University, Dayton, OH, United States
| | | | | |
Collapse
|
5
|
Vily-Petit J, Gautier-Stein A, Mithieux G. [Intestinal gluconeogenesis : When the intestine produces glucose to prevent obesity and hepatic steatosis]. Med Sci (Paris) 2025; 41:246-252. [PMID: 40117549 DOI: 10.1051/medsci/2025023] [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: 03/23/2025] Open
Abstract
Intestinal gluconeogenesis refers to the ability of the gut to produce glucose outside of meals. By initiating a gut-brain neural axis, its activation by dietary fiber or protein improves the regulation of energy balance. Recently, the creation of a genetic activation model of intestinal gluconeogenesis has demonstrated its anti-obesity, anti-diabetes and anti-hepatic steatosis effects. Interestingly, it increases thermogenesis in brown adipose tissue, thereby promoting energy expenditure and contributing to the fight against obesity. Therefore, targeting intestinal gluconeogenesis could be an innovative strategy to address metabolic diseases such as hepatic steatosis and diabetes, paving the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Justine Vily-Petit
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Amandine Gautier-Stein
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Gilles Mithieux
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| |
Collapse
|
6
|
Ma YX, Han YQ, Wang PZ, Wang MY, Yang GY, Li JL, Wang J, Chu BB. Porcine reproductive and respiratory syndrome virus activates lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs axis. Int J Biol Macromol 2024; 282:136720. [PMID: 39433189 DOI: 10.1016/j.ijbiomac.2024.136720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is a highly contagious pathogen in pigs. This study aimed to investigate the impact of PRRSV infection on cellular metabolism, particularly focusing on lipid metabolism to understand its role in promoting viral replication. We conducted a metabolic analysis on MARC-145 cells before and after PRRSV infection. Our results demonstrated that the most significant alterations in cellular metabolism, accounting for 40.8 % of total changes, were related to lipid metabolism. These changes were primarily driven by the activation of sterol regulatory-element binding proteins (SREBPs), critical regulators of lipid biosynthesis. To understand the mechanisms behind SREBPs activation by PRRSV, we investigated the involvement of upstream effectors, specifically protein kinase B (AKT) and phosphoenolpyruvate carboxykinase 1 (PCK1). Our findings indicated that PRRSV infection triggered AKT activation, leading to the subsequent activation of PCK1. Activated PCK1 then phosphorylated insulin-induced genes (INSIGs), resulting in their degradation. This degradation facilitated the translocation of SREBPs from the endoplasmic reticulum to the nucleus. Additionally, we observed that PRRSV infection stimulated the production of reactive oxygen species (ROS), which played a critical role in activating AKT. Collectively, our findings demonstrate that PRRSV enhances lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs signaling axis, which provides new insights into the metabolic strategies employed by PRRSV.
Collapse
Affiliation(s)
- Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ya-Qi Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Pei-Zhu Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ming-Yang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Jian-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China.
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Longhu Advanced Immunization Laboratory, Zhengzhou 450046, Henan Province, China; International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
7
|
Estrada-Meza J, Videlo J, Bron C, Duchampt A, Saint-Béat C, Zergane M, Silva M, Rajas F, Bouret SG, Mithieux G, Gautier-Stein A. Intestinal gluconeogenesis controls the neonatal development of hypothalamic feeding circuits. Mol Metab 2024; 89:102036. [PMID: 39304064 PMCID: PMC11470480 DOI: 10.1016/j.molmet.2024.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE Intestinal gluconeogenesis (IGN) regulates adult energy homeostasis in part by controlling the same hypothalamic targets as leptin. In neonates, leptin exhibits a neonatal surge controlling axonal outgrowth between the different hypothalamic nuclei involved in feeding circuits and autonomic innervation of peripheral tissues involved in energy and glucose homeostasis. Interestingly, IGN is induced during this specific time-window. We hypothesized that the neonatal pic of IGN also regulates the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues. METHODS We genetically induced neonatal IGN by overexpressing G6pc1 the catalytic subunit of glucose-6-phosphatase (the mandatory enzyme of IGN) at birth or at twelve days after birth. The neonatal development of hypothalamic feeding circuits was studied by measuring Agouti-related protein (AgRP) and Pro-opiomelanocortin (POMC) fiber density in hypothalamic nuclei of 20-day-old pups. The effect of the neonatal induction of intestinal G6pc1 on sympathetic innervation of the adipose tissues was studied via tyrosine hydroxylase (TH) quantification. The metabolic consequences of the neonatal induction of intestinal G6pc1 were studied in adult mice challenged with a high-fat/high-sucrose (HFHS) diet for 2 months. RESULTS Induction of intestinal G6pc1 at birth caused a neonatal reorganization of AgRP and POMC fiber density in the paraventricular nucleus of the hypothalamus, increased brown adipose tissue tyrosine hydroxylase levels, and protected against high-fat feeding-induced metabolic disorders. In contrast, inducing intestinal G6pc1 12 days after birth did not impact AgRP/POMC fiber densities, adipose tissue innervation or adult metabolism. CONCLUSION These findings reveal that IGN at birth but not later during postnatal life controls the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues, promoting a better management of metabolism in adulthood.
Collapse
Affiliation(s)
| | - Jasmine Videlo
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Clara Bron
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Adeline Duchampt
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Mickael Zergane
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Marine Silva
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Fabienne Rajas
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Sebastien G Bouret
- University Lille, Inserm, CHU Lille, Laboratory of development and plasticity of the Neuroendocrine brain, Lille Neuroscience & Cognition, Inserm UMR-S1172, Lille, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
8
|
Schwärzler J, Mayr L, Grabherr F, Tilg H, Adolph TE. Epithelial metabolism as a rheostat for intestinal inflammation and malignancy. Trends Cell Biol 2024; 34:913-927. [PMID: 38341347 DOI: 10.1016/j.tcb.2024.01.004] [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: 11/20/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
The gut epithelium protects the host from a potentially hostile environment while allowing nutrient uptake that is vital for the organism. To maintain this delicate task, the gut epithelium has evolved multilayered cellular functions ranging from mucus production to hormone release and orchestration of mucosal immunity. Here, we review the execution of intestinal epithelial metabolism in health and illustrate how perturbation of epithelial metabolism affects experimental gut inflammation and tumorigenesis. We also discuss the impact of environmental factors and host-microbe interactions on epithelial metabolism in the context of inflammatory bowel disease and colorectal cancer. Insights into epithelial metabolism hold promise to unravel mechanisms of organismal health that may be therapeutically exploited in humans in the future.
Collapse
Affiliation(s)
- Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
9
|
Gautier-Stein A, Vily-Petit J, Rajas F, Mithieux G. Intestinal gluconeogenesis: A translator of nutritional information needed for glycemic and emotional balance. Biochimie 2024; 223:206-214. [PMID: 38040189 DOI: 10.1016/j.biochi.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
At the interface between the outside world and the self, the intestine is the first organ receiving nutritional information. One intestinal function, gluconeogenesis, is activated by various nutrients, particularly diets enriched in fiber or protein, and thus results in glucose production in the portal vein in the post-absorptive period. The detection of portal glucose induces a nervous signal controlling the activity of the central nuclei involved in the regulation of metabolism and emotional behavior. Induction of intestinal gluconeogenesis is necessary for the beneficial effects of fiber or protein-enriched diets on metabolism and emotional behavior. Through its ability to translate nutritional information from the diet to the brain's regulatory centers, intestinal gluconeogenesis plays an essential role in maintaining physiological balance.
Collapse
Affiliation(s)
- Amandine Gautier-Stein
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France.
| | - Justine Vily-Petit
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Fabienne Rajas
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Gilles Mithieux
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| |
Collapse
|
10
|
Vily-Petit J, Taki A, Sinet F, Soty M, Guiard B, Zemdegs J, Malleret G, Stefanutti A, Mithieux G, Gautier-Stein A. Absence of the Peptide Transporter 1 Induces a Prediabetic and Depressive-Like Phenotype in Mice. Neuroendocrinology 2024; 115:226-241. [PMID: 38852578 DOI: 10.1159/000539499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Protein-enriched diets improve glycemic control in diabetes or emotional behavior in depressive patients. In mice, these benefits depend on intestinal gluconeogenesis activation by di-/tripeptides. Intestinal di-/tripeptides absorption is carried out by the peptide transporter 1, PEPT1. The lack of PEPT1 might thus alter glucose and emotional balance. METHODS To determine the effects of PEPT1 deficiency under standard dietary conditions or during a dietary challenge known to promote both metabolic and cognitive dysfunction, insulin sensitivity, anxiety, and depressive-like traits, hippocampal serotonin (5-HT) and insulin signaling pathway were measured in wild-type (WT) and Pept1-/- mice fed either a chow or a high-fat high-sucrose (HF-HS) diet. RESULTS Pept1-/- mice exhibited slight defects in insulin sensitivity and emotional behavior, which were aggravated by an HF-HS diet. Pept1-/- mice fed a chow diet had lower hippocampal 5-HT levels and exhibited cerebral insulin resistance under HF-HS diet. These defects were independent of intestinal gluconeogenesis but might be linked to increased plasma amino acids levels. CONCLUSION Pept1-/- mice develop prediabetic and depressive-like traits and could thus be used to develop strategies to prevent or cure both diseases.
Collapse
Affiliation(s)
| | - Amelie Taki
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Flore Sinet
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Maud Soty
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Bruno Guiard
- CRCA - UMR - Université Paul Sabatier, Toulouse, France
| | | | - Gael Malleret
- Forgetting and Cortical Dynamics, Lyon Neuroscience Research Center, Lyon, France
| | - Anne Stefanutti
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France,
| | | |
Collapse
|
11
|
Fu Y, Wang Z, Qin H. Examining the Pathogenesis of MAFLD and the Medicinal Properties of Natural Products from a Metabolic Perspective. Metabolites 2024; 14:218. [PMID: 38668346 PMCID: PMC11052500 DOI: 10.3390/metabo14040218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), characterized primarily by hepatic steatosis, has become the most prevalent liver disease worldwide, affecting approximately two-fifths of the global population. The pathogenesis of MAFLD is extremely complex, and to date, there are no approved therapeutic drugs for clinical use. Considerable evidence indicates that various metabolic disorders play a pivotal role in the progression of MAFLD, including lipids, carbohydrates, amino acids, and micronutrients. In recent years, the medicinal properties of natural products have attracted widespread attention, and numerous studies have reported their efficacy in ameliorating metabolic disorders and subsequently alleviating MAFLD. This review aims to summarize the metabolic-associated pathological mechanisms of MAFLD, as well as the natural products that regulate metabolic pathways to alleviate MAFLD.
Collapse
Affiliation(s)
| | | | - Hong Qin
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha 410006, China; (Y.F.); (Z.W.)
| |
Collapse
|
12
|
Akter R, Awais M, Boopathi V, Ahn JC, Yang DC, Kang SC, Yang DU, Jung SK. Inversion of the Warburg Effect: Unraveling the Metabolic Nexus between Obesity and Cancer. ACS Pharmacol Transl Sci 2024; 7:560-569. [PMID: 38481689 PMCID: PMC10928896 DOI: 10.1021/acsptsci.3c00301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 02/04/2025]
Abstract
Obesity is a well-established risk factor for cancer, significantly impacting both cancer incidence and mortality. However, the intricate molecular mechanisms connecting adipose tissue to cancer cell metabolism are not fully understood. This Review explores the historical context of tumor energy metabolism research, tracing its origins to Otto Warburg's pioneering work in 1920. Warburg's discovery of the "Warburg effect", wherein cancer cells prefer anaerobic glycolysis even in the presence of oxygen, laid the foundation for understanding cancer metabolism. Building upon this foundation, the "reverse Warburg effect" emerged in 2009, elucidating the role of aerobic glycolysis in cancer-associated fibroblasts (CAFs) and its contribution to lactate accumulation in the tumor microenvironment, subsequently serving as a metabolic substrate for cancer cells. In contrast, within high-adiposity contexts, cancer cells exhibit a unique metabolic shift termed the "inversion of the Warburg effect". This phenomenon, distinct from the stromal-dependent reverse Warburg effect, relies on increased nutrient abundance in obesity environments, leading to the generation of glucose from lactate as a metabolic substrate. This Review underscores the heightened tumor proliferation and aggressiveness associated with obesity, introducing the "inversion of the Warburg effect" as a novel mechanism rooted in the altered metabolic landscape within an obese milieu. The insights presented here open promising avenues for therapeutic exploration, offering fresh perspectives and opportunities for the development of innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Reshmi Akter
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Muhammad Awais
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Vinothini Boopathi
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Jong Chan Ahn
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Deok Chun Yang
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Se Chan Kang
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Dong Uk Yang
- AIBIOME, 6, Jeonmin-ro 30beon-gil, Yuseong-gu 34214, Daejeon, Republic of Korea
| | - Seok-Kyu Jung
- Department
of Horticulture, Kongju National University, Yesan 32588,Chungcheongnam-do, Republic of Korea
| |
Collapse
|
13
|
Tian S, Chu Q, Ma S, Ma H, Song H. Dietary Fiber and Its Potential Role in Obesity: A Focus on Modulating the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14853-14869. [PMID: 37815013 DOI: 10.1021/acs.jafc.3c03923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Dietary fiber is a carbohydrate polymer with ten or more monomeric units that are resistant to digestion by human digestive enzymes, and it has gained widespread attention due to its significant role in health improvement through regulating gut microbiota. In this review, we summarized the interaction between dietary fiber, gut microbiota, and obesity, and the beneficial effects of dietary fiber on obesity through the modulation of microbiota, such as modifying selective microbial composition, producing starch-degrading enzymes, improving gut barrier function, reducing the inflammatory response, reducing trimethylamine N-oxide, and promoting the production of gut microbial metabolites (e.g., short chain fatty acids, bile acids, ferulic acid, and succinate). In addition, factors affecting the gut microbiota composition and metabolites by dietary fiber (length of the chain, monosaccharide composition, glycosidic bonds) were also concluded. Moreover, strategies for enhancing the biological activity of dietary fiber (fermentation technology, ultrasonic modification, nanotechnology, and microfluidization) were subsequently discussed. This review may provide clues for deeply exploring the structure-activity relationship between dietary fiber and antiobesity properties by targeting specific gut microbiota.
Collapse
Affiliation(s)
- Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Qiang Chu
- Tea Research Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, PR China
| | - Shaotong Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Huan Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| |
Collapse
|
14
|
Carlsen H, Pajari AM. Dietary fiber - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2023; 67:9979. [PMID: 37920675 PMCID: PMC10619389 DOI: 10.29219/fnr.v67.9979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 11/04/2023] Open
Abstract
Dietary fiber is a term crudely defined as carbohydrates (CHOs) that escape digestion and uptake in the small intestine. Lignin, which is not a CHO, is also a part of the dietary fiber definition. Dietary fibers come in different sizes and forms, with a variety of combinations of monomeric units. Health authorities worldwide have for many years recommended a diet rich in dietary fibers based on consistent findings that dietary fibers are associated with reduced incidences of major non-communicable diseases, including obesity, type 2 diabetes, cardiovascular disease, and colorectal cancer. Most fibers come from common edible foods from the plant kingdom, but fibers are also found in food additives, supplements, and breast milk. The recommended intake in Nordic Nutrition Recommendations 2012 (NNR2012) is 25 g/d for women and 35 g/d for men, whereas the actual intake is significantly lower, ranging from 16 g/d to 22 g/d in women and 18 g/d to 26 g/d in men. New studies since NNR2012 confirm the current view that dietary fiber is beneficial for health, advocating intakes of at least 25 g/day.
Collapse
Affiliation(s)
- Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Anne-Maria Pajari
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Li HY, Huang SY, Zhou DD, Xiong RG, Luo M, Saimaiti A, Han MK, Gan RY, Zhu HL, Li HB. Theabrownin inhibits obesity and non-alcoholic fatty liver disease in mice via serotonin-related signaling pathways and gut-liver axis. J Adv Res 2023; 52:59-72. [PMID: 36639024 PMCID: PMC10555776 DOI: 10.1016/j.jare.2023.01.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/30/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) with obesity seriously threats public health. Our previous studies showed that dark tea had more potential on regulating lipid metabolism than other teas, and theabrownin (TB) was considered to be a main contributor to the bioactivity of dark tea. OBJECTIVES This in vivo study aims to reveal the effects and molecular mechanisms of TB on NAFLD and obesity, and the role of the gut-liver axis is explored. METHODS The histopathological examinations, biochemical tests, and nuclear magnetic resonance were applied to evaluate the effects of TB on NAFLD and obesity. The untargeted metabolomics was used to find the key molecule for further exploration of molecular mechanisms. The 16S rRNA gene sequencing was used to assess the changes in gut microbiota. The antibiotic cocktail and fecal microbiota transplant were used to clarify the role of gut microbiota. RESULTS TB markedly reduced body weight gain (67.01%), body fat rate (62.81%), and hepatic TG level (51.35%) in the preventive experiment. Especially, TB decreased body weight (32.16%), body fat rate (42.56%), and hepatic TG level (42.86%) in the therapeutic experiment. The mechanisms of action could be the improvement of fatty acid oxidation, lipolysis, and oxidative stress via the regulation of serotonin-related signaling pathways. Also, TB increased the abundance of serotonin-related gut microbiota, such as Akkermansia, Bacteroides and Parabacteroides. Antibiotics-induced gut bacterial dysbiosis disrupted the regulation of TB on serotonin-related signaling pathways in liver, whereas the beneficial regulation of TB on target proteins was regained with the restoration of gut microbiota. CONCLUSION We find that TB has markedly preventive and therapeutic effects on NAFLD and obesity by regulating serotonin level and related signaling pathways through gut microbiota. Furthermore, gut microbiota and TB co-contribute to alleviating NAFLD and obesity. TB could be a promising medicine for NAFLD and obesity.
Collapse
Affiliation(s)
- Hang-Yu Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Si-Yu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dan-Dan Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ruo-Gu Xiong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Min Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Adila Saimaiti
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Mu-Ke Han
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, National Agricultural Science & Technology Center, Chengdu 610213, China.
| | - Hui-Lian Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
16
|
Sun M, Wan Y, Shi M, Meng ZX, Zeng W. Neural innervation in adipose tissue, gut, pancreas, and liver. LIFE METABOLISM 2023; 2:load022. [PMID: 39872245 PMCID: PMC11749697 DOI: 10.1093/lifemeta/load022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 01/30/2025]
Abstract
Efficient communication between the brain and peripheral organs is indispensable for regulating physiological function and maintaining energy homeostasis. The peripheral nervous system (PNS) in vertebrates, consisting of the autonomic and somatic nervous systems, bridges the peripheral organs and the central nervous system (CNS). Metabolic signals are processed by both vagal sensory nerves and somatosensory nerves. The CNS receives sensory inputs via ascending nerves, serves as the coordination and integration center, and subsequently controls internal organs and glands via descending nerves. The autonomic nervous system consists of sympathetic and parasympathetic branches that project peripheral nerves into various anatomical locations to regulate the energy balance. Sympathetic and parasympathetic nerves typically control the reflexive and involuntary functions in organs. In this review article, we outline the innervation of adipose tissue, gut, pancreas, and liver, to illustrate the neurobiological basis of central-peripheral interactions. We emphasize the importance of understanding the functional atlas of neural control of energy metabolism, and more importantly, provide potential avenues for further research in this area.
Collapse
Affiliation(s)
- Mengxue Sun
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yongwen Wan
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Mengjie Shi
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
17
|
Wu KK. Extracellular Succinate: A Physiological Messenger and a Pathological Trigger. Int J Mol Sci 2023; 24:11165. [PMID: 37446354 DOI: 10.3390/ijms241311165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
When tissues are under physiological stresses, such as vigorous exercise and cold exposure, skeletal muscle cells secrete succinate into the extracellular space for adaptation and survival. By contrast, environmental toxins and injurious agents induce cellular secretion of succinate to damage tissues, trigger inflammation, and induce tissue fibrosis. Extracellular succinate induces cellular changes and tissue adaptation or damage by ligating cell surface succinate receptor-1 (SUCNR-1) and activating downstream signaling pathways and transcriptional programs. Since SUCNR-1 mediates not only pathological processes but also physiological functions, targeting it for drug development is hampered by incomplete knowledge about the characteristics of its physiological vs. pathological actions. This review summarizes the current status of extracellular succinate in health and disease and discusses the underlying mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Institute of Biotechnology, College of Life Science, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
18
|
Vily-Petit J, Soty M, Silva M, Micoud M, Bron C, Guérin-Deremaux L, Mithieux G. Improvement of energy metabolism associated with NUTRIOSE® soluble fiber, a dietary ingredient exhibiting prebiotic properties, requires intestinal gluconeogenesis. Food Res Int 2023; 167:112723. [PMID: 37087279 DOI: 10.1016/j.foodres.2023.112723] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 03/29/2023]
Abstract
While the prevalence of obesity progresses worldwide, the consumption of sugars and dietary fiber increases and decreases, respectively. In this context, NUTRIOSE® soluble fiber is a plant-based food ingredient with beneficial effects in Humans. Here, we studied in mice the mechanisms involved, particularly the involvement of intestinal gluconeogenesis (IGN), the essential function in the beneficial effects of dietary fibers. To determine whether NUTRIOSE® exerts its beneficial effects via the activation of IGN, we studied the effects of dietary NUTRIOSE® on the development of obesity, diabetes and non-alcoholic fatty liver disease (NAFLD), which IGN is able to prevent. To assert the role of IGN in the observed effects, we studied wild-type (WT) and IGN-deficient mice. In line with our hypothesis, NUTRIOSE® exerts metabolic benefits in WT mice, but not in IGN-deficient mice. Indeed, WT mice are protected from body weight gain and NAFLD induced by a high calorie diet. In addition, our data suggests that NUTRIOSE® may improve energy balance by activating a browning process in subcutaneous white adipose tissue. While the gut microbiota composition changes with NUTRIOSE®, this is not sufficient in itself to account for the benefits observed. On the contrary, IGN is obligatory in the NUTRIOSE® benefits, since no benefit take place in absence of IGN. In conclusion, IGN plays a crucial and essential role in the set-up of the beneficial effects of NUTRIOSE®, highlighting the interest of the supplementation of food with healthy ingredients in the context of the current obesity epidemic.
Collapse
Affiliation(s)
- Justine Vily-Petit
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France
| | - Maud Soty
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France
| | - Marine Silva
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France
| | - Manon Micoud
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France
| | - Clara Bron
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France
| | | | - Gilles Mithieux
- Institut de la Santé et de la Recherche Médicale, U1213 Lyon, France; Université Claude Bernard Lyon1, Villeurbanne, France; Université de Lyon, Lyon, France.
| |
Collapse
|
19
|
Ye Q, Liu Y, Zhang G, Deng H, Wang X, Tuo L, Chen C, Pan X, Wu K, Fan J, Pan Q, Wang K, Huang A, Tang N. Deficiency of gluconeogenic enzyme PCK1 promotes metabolic-associated fatty liver disease through PI3K/AKT/PDGF axis activation in male mice. Nat Commun 2023; 14:1402. [PMID: 36918564 PMCID: PMC10015095 DOI: 10.1038/s41467-023-37142-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Metabolic associated fatty liver disease (MAFLD) encompasses a broad spectrum of hepatic disorders, including steatosis, nonalcoholic steatohepatitis (NASH) and fibrosis. We demonstrated that phosphoenolpyruvate carboxykinase 1 (PCK1) plays a central role in MAFLD progression. Male mice with liver Pck1 deficiency fed a normal diet displayed hepatic lipid disorder and liver injury, whereas fibrosis and inflammation were aggravated in mice fed a high-fat diet with drinking water containing fructose and glucose (HFCD-HF/G). Forced expression of hepatic PCK1 by adeno-associated virus ameliorated MAFLD in male mice. PCK1 deficiency stimulated lipogenic gene expression and lipid synthesis. Moreover, loss of hepatic PCK1 activated the RhoA/PI3K/AKT pathway by increasing intracellular GTP levels, increasing secretion of platelet-derived growth factor-AA (PDGF-AA), and promoting hepatic stellate cell activation. Treatment with RhoA and AKT inhibitors or gene silencing of RhoA or AKT1 alleviated MAFLD progression in vivo. Hepatic PCK1 deficiency may be important in hepatic steatosis and fibrosis development through paracrine secretion of PDGF-AA in male mice, highlighting a potential therapeutic strategy for MAFLD.
Collapse
Affiliation(s)
- Qian Ye
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yi Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guiji Zhang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haijun Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaojun Wang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Tuo
- Department of Infectious Disease, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, China
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xuanming Pan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Kang Wu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jiangao Fan
- Department of Gastroenterology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Pan
- Department of Gastroenterology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
20
|
Intestinal gluconeogenesis: metabolic benefits make sense in the light of evolution. Nat Rev Gastroenterol Hepatol 2023; 20:183-194. [PMID: 36470967 DOI: 10.1038/s41575-022-00707-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 03/02/2023]
Abstract
The intestine, like the liver and kidney, in various vertebrates and humans is able to carry out gluconeogenesis and release glucose into the blood. In the fed post-absorptive state, intestinal glucose is sensed by the gastrointestinal nervous system. The latter initiates a signal to the brain regions controlling energy homeostasis and stress-related behaviour. Intestinal gluconeogenesis (IGN) is activated by several complementary mechanisms, in particular nutritional situations (for example, when food is enriched in protein or fermentable fibre and after gastric bypass surgery in obesity). In these situations, IGN has several metabolic and behavioural benefits. As IGN is activated by nutrients capable of fuelling systemic gluconeogenesis, IGN could be a signal to the brain that food previously ingested is suitable for maintaining plasma glucose for a while. This process might account for the benefits observed. Finally, in this Perspective, we discuss how the benefits of IGN in fasting and fed states could explain why IGN emerged and was maintained in vertebrates by natural selection.
Collapse
|
21
|
Karlson O, Arnell H, Gudjonsdottir AH, Agardh D, Torinsson Naluai Å. Intestinal gluconeogenesis is downregulated in pediatric patients with celiac disease. BMC Med 2022; 20:440. [PMID: 36369023 PMCID: PMC9652951 DOI: 10.1186/s12916-022-02635-3] [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: 04/07/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Untreated celiac disease (CD) patients have increased levels of blood glutamine and a lower duodenal expression of glutaminase (GLS). Intestinal gluconeogenesis (IGN) is a process through which glutamine is turned into glucose in the small intestine, for which GLS is crucial. Animal studies suggest impaired IGN may have long-term effects on metabolic control and be associated with the development of type 2 diabetes and non-alcoholic fatty liver disease (NAFLD). The aim of this study was to thoroughly investigate IGN at the gene expression level in children with untreated celiac disease. METHODS Quantitative polymerase chain reaction (qPCR) was used to quantify the expression of 11 target genes related to IGN using the delta-delta Ct method with three reference genes (GUSB, IPO8, and YWHAZ) in duodenal biopsies collected from 84 children with untreated celiac disease and 58 disease controls. RESULTS Significantly lower expression of nine target genes involved in IGN was seen in duodenal biopsies from CD patients compared with controls: FBP1, G6PC, GLS, GPT1, PCK1, PPARGC1A, SLC2A2, SLC5A1, and SLC6A19. No significant difference in the expression was observed for G6PC3 or GOT1. CONCLUSIONS Children with untreated celiac disease have lower expression of genes important for IGN. Further studies are warranted to disentangle whether this is a consequence of intestinal inflammation or due to an impaired metabolic pathway shared with other chronic metabolic diseases. Impaired IGN could be a mechanism behind the increased risk of NAFLD seen in CD patients.
Collapse
Affiliation(s)
- Olof Karlson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Arnell
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Audur H Gudjonsdottir
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Daniel Agardh
- Department of Clinical Sciences, Unit of Celiac Disease and Diabetes, Lund University, Malmö, Sweden
| | - Åsa Torinsson Naluai
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
22
|
Stojanović O, Miguel-Aliaga I, Trajkovski M. Intestinal plasticity and metabolism as regulators of organismal energy homeostasis. Nat Metab 2022; 4:1444-1458. [PMID: 36396854 DOI: 10.1038/s42255-022-00679-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 10/06/2022] [Indexed: 11/18/2022]
Abstract
The small intestine displays marked anatomical and functional plasticity that includes adaptive alterations in adult gut morphology, enteroendocrine cell profile and their hormone secretion, as well as nutrient utilization and storage. In this Perspective, we examine how shifts in dietary and environmental conditions bring about changes in gut size, and describe how the intestine adapts to changes in internal state, bowel resection and gastric bypass surgery. We highlight the critical importance of these intestinal remodelling processes in maintaining energy balance of the organism, and in protecting the metabolism of other organs. The intestinal resizing is supported by changes in the microbiota composition, and by activation of carbohydrate and fatty acid metabolism, which govern the intestinal stem cell proliferation, intestinal cell fate, as well as survivability of differentiated epithelial cells. The discovery that intestinal remodelling is part of the normal physiological adaptation to various triggers, and the potential for harnessing the reversible gut plasticity, in our view, holds extraordinary promise for developing therapeutic approaches against metabolic and inflammatory diseases.
Collapse
Affiliation(s)
- Ozren Stojanović
- Department of Cell Physiology and Metabolism, Centre Medical Universitaire (CMU), Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Centre, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK.
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, Centre Medical Universitaire (CMU), Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Diabetes Centre, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
23
|
Mithieux G. [Intestinal gluconeogenesis: an insulin-mimetic function]. Biol Aujourdhui 2022; 216:37-39. [PMID: 35876519 DOI: 10.1051/jbio/2022003] [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: 01/16/2022] [Indexed: 06/15/2023]
Abstract
Intestinal gluconeogenesis (IGN) is a regulatory function of energy homeostasis. IGN-produced glucose is sensed by the gastrointestinal nervous system and sends a signal to regions of the brain regulating food intake and glucose control. IGN is activated by dietary protein and dietary fibre, and by gastric bypass surgery of obesity. Glutamine, propionate and succinate are the main substrates used for glucose production by IGN. Activation of IGN accounts for the well-known satiety effect of protein-enriched diets and the anti-obesity and anti-diabetes effects associated with fibre feeding and gastric bypass surgery. Genetic activation of IGN in mice shows the same beneficial effects, independently of any nutritional manipulation, including a marked prevention of hepatic steatosis under hypercaloric feeding. The activation of IGN could thus be the basis for new approaches to prevent or correct metabolic diseases in humans.
Collapse
Affiliation(s)
- Gilles Mithieux
- UMR-S Inserm 1213-UCB Lyon 1 « Nutrition, Diabète et Cerveau », Faculté Laennec-Lyon-Est, Rue Guillaume Paradin, 69372 Lyon cedex 8, France
| |
Collapse
|
24
|
Ding Q, Lu C, Hao Q, Zhang Q, Yang Y, Olsen RE, Ringo E, Ran C, Zhang Z, Zhou Z. Dietary Succinate Impacts the Nutritional Metabolism, Protein Succinylation and Gut Microbiota of Zebrafish. Front Nutr 2022; 9:894278. [PMID: 35685883 PMCID: PMC9171437 DOI: 10.3389/fnut.2022.894278] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Succinate is widely used in the food and feed industry as an acidulant, flavoring additive, and antimicrobial agent. This study investigated the effects of dietary succinate on growth, energy budget, nutritional metabolism, protein succinylation, and gut microbiota composition of zebrafish. Zebrafish were fed a control-check (0% succinate) or four succinate-supplemented diets (0.05, 0.10, 0.15, and 0.2%) for 4 weeks. The results showed that dietary succinate at the 0.15% additive amount (S0.15) can optimally promote weight gain and feed intake. Whole body protein, fat, and energy deposition increased in the S0.15 group. Fasting plasma glucose level decreased in fish fed the S0.15 diet, along with improved glucose tolerance. Lipid synthesis in the intestine, liver, and muscle increased with S0.15 feeding. Diet with 0.15% succinate inhibited intestinal gluconeogenesis but promoted hepatic gluconeogenesis. Glycogen synthesis increased in the liver and muscle of S0.15-fed fish. Glycolysis was increased in the muscle of S0.15-fed fish. In addition, 0.15% succinate-supplemented diet inhibited protein degradation in the intestine, liver, and muscle. Interestingly, different protein succinylation patterns in the intestine and liver were observed in fish fed the S0.15 diet. Intestinal proteins with increased succinylation levels were enriched in the tricarboxylic acid cycle while proteins with decreased succinylation levels were enriched in pathways related to fatty acid and amino acid degradation. Hepatic proteins with increased succinylation levels were enriched in oxidative phosphorylation while proteins with decreased succinylation levels were enriched in the processes of protein processing and transport in the endoplasmic reticulum. Finally, fish fed the S0.15 diet had a higher abundance of Proteobacteria but a lower abundance of Fusobacteria and Cetobacterium. In conclusion, dietary succinate could promote growth and feed intake, promote lipid anabolism, improve glucose homeostasis, and spare protein. The effects of succinate on nutritional metabolism are associated with alterations in the levels of metabolic intermediates, transcriptional regulation, and protein succinylation levels. However, hepatic fat accumulation and gut microbiota dysbiosis induced by dietary succinate suggest potential risks of succinate application as a feed additive for fish. This study would be beneficial in understanding the application of succinate as an aquatic feed additive.
Collapse
Affiliation(s)
- Qianwen Ding
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chenyao Lu
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiang Hao
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingshuang Zhang
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Rolf Erik Olsen
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Ringo
- Norwegian College of Fishery Science, Faculty of Bioscience, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Zhen Zhang,
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Zhigang Zhou,
| |
Collapse
|
25
|
Boscaini S, Leigh SJ, Lavelle A, García-Cabrerizo R, Lipuma T, Clarke G, Schellekens H, Cryan JF. Microbiota and body weight control: Weight watchers within? Mol Metab 2022; 57:101427. [PMID: 34973469 PMCID: PMC8829807 DOI: 10.1016/j.molmet.2021.101427] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite several decades of research, managing body weight remains an unsolved clinical problem. Health problems associated with dysregulated body weight, such as obesity and cachexia, exhibit several gut microbiota alterations. There is an increased interest in utilising the gut microbiota for body weight control, as it responds to intervention and plays an important role in energy extraction from food, as well as biotransformation of nutrients. SCOPE OF THE REVIEW This review provides an overview of the role of the gut microbiota in the physiological and metabolic alterations observed in two body weight dysregulation-related disorders, namely obesity and cachexia. Second, we assess the available evidence for different strategies, including caloric restriction, intermittent fasting, ketogenic diet, bariatric surgery, probiotics, prebiotics, synbiotics, high-fibre diet, and fermented foods - effects on body weight and gut microbiota composition. This approach was used to give insights into the possible link between body weight control and gut microbiota configuration. MAJOR CONCLUSIONS Despite extensive associations between body weight and gut microbiota composition, limited success could be achieved in the translation of microbiota-related interventions for body weight control in humans. Manipulation of the gut microbiota alone is insufficient to alter body weight and future research is needed with a combination of strategies to enhance the effects of lifestyle interventions.
Collapse
Affiliation(s)
- Serena Boscaini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Timothy Lipuma
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
26
|
Tang C, Cao G, Zhao W, Bie X, Lu F, Lu Z, Lu Y. Lactobacillus acidophilus NX2-6 Improved High-Fat Diet-Induced Glucose Metabolism Disorder Independent of Promotion of Insulin Secretion in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:15598-15610. [PMID: 34788040 DOI: 10.1021/acs.jafc.1c05948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
High-fat diet (HFD) contributes to metabolic inflammation and glucose metabolism disorder, thereby resulting in the pathogenesis of metabolic syndrome. Accumulating evidence has revealed that some probiotics could improve HFD-induced metabolic inflammation and glucose metabolism disorder. Our previous study has discovered that Lactobacillus acidophilus NX2-6 exhibited in vitro lipid-lowering, antioxidative, and anti-inflammatory activities. This study mainly investigated whether L. acidophilus NX2-6 improved HFD-induced glucose metabolism disorder. The results exhibited that L. acidophilus NX2-6 effectively reduced blood glucose levels and improved glucose tolerance by activating the insulin signaling pathway, promoting glucose uptake, glycolysis, and intestinal gluconeogenesis and suppressing hepatic gluconeogenesis, independent of regulation of glycogen synthesis in the liver and muscle. Enhanced insulin sensitivity was associated with L. acidophilus NX2-6-mediated suppression of inflammatory cascades in the target organs. Meanwhile, L. acidophilus NX2-6 also improved hepatic energy metabolism via the FGF21/AMPKα/PGC-1α/NRF1 pathway. However, L. acidophilus NX2-6 did not affect apoptosis, pyroptosis, inflammation, and endoplasmic reticulum stress in the pancreas of HFD-fed mice. In conclusion, our results indicated that L. acidophilus NX2-6 improved glucose metabolism disorder through enhancing insulin sensitivity, suppressing metabolic inflammation, and promoting energy expenditure.
Collapse
Affiliation(s)
- Chao Tang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Gang Cao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wen Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Xiaomei Bie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Fengxia Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
27
|
Li Y, Cao H, Wang X, Guo L, Ding X, Zhao W, Zhang F. Diet-mediated metaorganismal relay biotransformation: health effects and pathways. Crit Rev Food Sci Nutr 2021:1-19. [PMID: 34802351 DOI: 10.1080/10408398.2021.2004993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In recent years, the concept of metaorganism expands our insight into how diet-microbe-host interactions contribute to human health and diseases. We realized that many biological metabolic processes in the host can be summarized into metaorganismal relay pathways, in which metabolites such as trimethylamine-N‑oxide, short-chain fatty acids and bile acids act as double-edged swords (beneficial or harmful effects) in the initiation and progression of diseases. Pleiotropic effects of metabolites are derived from several influencing factors including dose level, targeted organ of effect, action duration and species of these metabolites. Based on the pleiotropic effects of metabolites, personalized therapeutic approaches including microecological agents, enzymatic regulators and changes in dietary habits to govern related metabolite production may provide a new insight in promoting human health. In this review, we summarize our current knowledge of metaorganismal relay pathways and elaborate on the pleiotropic effects of metabolites in these pathways, with special emphasis on related therapeutic nutritional interventions.
Collapse
Affiliation(s)
- Yanmin Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hong Cao
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoqian Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Lichun Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoying Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Feng Zhang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
28
|
Abstract
The reactions of the tricarboxylic acid (TCA) cycle allow the controlled combustion of fat and carbohydrate. In principle, TCA cycle intermediates are regenerated on every turn and can facilitate the oxidation of an infinite number of nutrient molecules. However, TCA cycle intermediates can be lost to cataplerotic pathways that provide precursors for biosynthesis, and they must be replaced by anaplerotic pathways that regenerate these intermediates. Together, anaplerosis and cataplerosis help regulate rates of biosynthesis by dictating precursor supply, and they play underappreciated roles in catabolism and cellular energy status. They facilitate recycling pathways and nitrogen trafficking necessary for catabolism, and they influence redox state and oxidative capacity by altering TCA cycle intermediate concentrations. These functions vary widely by tissue and play emerging roles in disease. This article reviews the roles of anaplerosis and cataplerosis in various tissues and discusses how they alter carbon transitions, and highlights their contribution to mechanisms of disease. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Melissa Inigo
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Stanisław Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; .,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
29
|
Sauter ER, Heckman-Stoddard B. Metabolic Surgery and Cancer Risk: An Opportunity for Mechanistic Research. Cancers (Basel) 2021; 13:cancers13133183. [PMID: 34202319 PMCID: PMC8268861 DOI: 10.3390/cancers13133183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Metabolic (bariatric) surgery (MBS) provides the greatest maximum and sustained weight loss among individuals who are morbidly obese. It is more effective than lifestyle interventions in improving or eliminating type 2 diabetes mellitus (T2DM) and in decreasing cardiovascular (CV) risk. Preclinical studies have been conducted to investigate the mechanisms by which MBS leads to the benefits in T2DM and CV risk. In this review, we describe the emerging evidence that MBS may also impact cancer risk and mortality, and whom may benefit most. We describe the long term involvement and commitment of the National Institutes of Health in obesity research in general and MBS in particular. We outline the need for additional research to understand the mechanism(s) by which MBS may influence cancer, since these mechanism(s) are currently unknown. Abstract Metabolic (bariatric) surgery (MBS) is recommended for individuals with a BMI > 40 kg/m2 or those with a BMI 35–40 kg/m2 who have one or more obesity related comorbidities. MBS leads to greater initial and sustained weight loss than nonsurgical weight loss approaches. MBS provides dramatic improvement in metabolic function, associated with a reduction in type 2 diabetes mellitus and cardiovascular risk. While the number of MBS procedures performed in the U.S. and worldwide continues to increase, they are still only performed on one percent of the affected population. MBS also appears to reduce the risk of certain obesity related cancers, although which cancers are favorably impacted vary by study, who benefits most is uncertain, and the mechanism(s) driving this risk reduction are mostly speculative. The goal of this manuscript is to highlight (1) emerging evidence that MBS influences cancer risk, and that the potential benefit appears to vary based on cancer, gender, surgical procedure, and likely other variables; (2) the role of the NIH in MBS research in T2DM and CV risk for many years, and more recently in cancer; and (3) the opportunity for research to understand the mechanism(s) by which MBS influences cancer. There is evidence that women benefit more from MBS than men, that MBS may actually increase the risk of colorectal cancer in both women and men, and there is speculation that the benefit in cancer risk reduction may vary according to which MBS procedure an individual undergoes. Herein, we review what is currently known, the historical role of government, especially the National Institutes of Health (NIH), in driving this research, and provide suggestions that we believe could lead to a better understanding of whether and how MBS impacts cancer risk, which cancers are impacted either favorably or unfavorably, the role of the NIH and other research agencies, and key questions to address that will help us to move the science forward.
Collapse
|
30
|
Sun EW, Martin AM, de Fontgalland D, Sposato L, Rabbitt P, Hollington P, Wattchow DA, Colella AD, Chataway T, Wewer Albrechtsen NJ, Spencer NJ, Young RL, Keating DJ. Evidence for Glucagon Secretion and Function Within the Human Gut. Endocrinology 2021; 162:6127286. [PMID: 33534908 DOI: 10.1210/endocr/bqab022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Indexed: 11/19/2022]
Abstract
Glucagon is secreted by pancreatic α cells in response to hypoglycemia and increases hepatic glucose output through hepatic glucagon receptors (GCGRs). There is evidence supporting the notion of extrapancreatic glucagon but its source and physiological functions remain elusive. Intestinal tissue samples were obtained from patients undergoing surgical resection of cancer. Mass spectrometry analysis was used to detect glucagon from mucosal lysate. Static incubations of mucosal tissue were performed to assess glucagon secretory response. Glucagon concentration was quantitated using a highly specific sandwich enzyme-linked immunosorbent assay. A cholesterol uptake assay and an isolated murine colonic motility assay were used to assess the physiological functions of intestinal GCGRs. Fully processed glucagon was detected by mass spectrometry in human intestinal mucosal lysate. High glucose evoked significant glucagon secretion from human ileal tissue independent of sodium glucose cotransporter and KATP channels, contrasting glucose-induced glucagon-like peptide 1 (GLP-1) secretion. The GLP-1 receptor agonist Exendin-4 attenuated glucose-induced glucagon secretion from the human ileum. GCGR blockade significantly increased cholesterol uptake in human ileal crypt culture and markedly slowed ex vivo colonic motility. Our findings describe the human gut as a potential source of extrapancreatic glucagon and demonstrate a novel enteric glucagon/GCGR circuit with important physiological functions beyond glycemic regulation.
Collapse
Affiliation(s)
- Emily W Sun
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Alyce M Martin
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | - Luigi Sposato
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Philippa Rabbitt
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Paul Hollington
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Alexander D Colella
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Tim Chataway
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | - Nick J Spencer
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Richard L Young
- Adelaide Medical School and NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
- Nutrition, Diabetes and Metabolism, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, SA, Australia
| | - Damien J Keating
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
31
|
Al-Yousif N, Rawal S, Jurczak M, Mahmud H, Shah FA. Endogenous Glucose Production in Critical Illness. Nutr Clin Pract 2021; 36:344-359. [PMID: 33682953 DOI: 10.1002/ncp.10646] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regulation of endogenous glucose production (EGP) by hormonal, neuronal, and metabolic signaling pathways contributes to the maintenance of euglycemia under normal physiologic conditions. EGP is defined by the generation of glucose from substrates through glycogenolysis and gluconeogenesis, usually in fasted states, for local and systemic use. Abnormal increases in EGP are noted in patients with diabetes mellitus type 2, and elevated EGP may also impact the pathogenesis of nonalcoholic fatty liver disease and congestive heart failure. In this narrative review, we performed a literature search in PubMed to identify recently published English language articles characterizing EGP in critical illness. Evidence from preclinical and clinical studies demonstrates that critical illness can disrupt EGP through multiple mechanisms including increased systemic inflammation, counterregulatory hormone and catecholamine release, alterations in the hypothalamic-pituitary axis, insulin resistance, lactic acidosis, and iatrogenic insults such as vasopressors and glucocorticoids administered as part of clinical care. EGP contributes to hyperglycemia in critical illness when abnormally elevated and to hypoglycemia when abnormally depressed, each of which has been independently associated with increased mortality. Increased EGP may also promote protein catabolism that could worsen critical illness myopathy and impede recovery. Better understanding of the mechanisms and factors contributing to dysregulated EGP in critical illness may help in the development of therapeutic strategies that promote euglycemia, reduce intensive care unit-associated catabolism, and improve patient outcomes.
Collapse
Affiliation(s)
- Nameer Al-Yousif
- Department of Internal Medicine, UPMC Mercy Hospital, Pittsburgh, Pennsylvania, USA
| | - Sagar Rawal
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Jurczak
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hussain Mahmud
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Faraaz Ali Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
- Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Wang Z, Zeng M, Wang Z, Qin F, Chen J, He Z. Dietary Polyphenols to Combat Nonalcoholic Fatty Liver Disease via the Gut-Brain-Liver Axis: A Review of Possible Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3585-3600. [PMID: 33729777 DOI: 10.1021/acs.jafc.1c00751] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Polyphenols are a group of micronutrients widely existing in plant foods including fruits, vegetables, and teas that can improve nonalcoholic fatty liver disease (NAFLD). In this review, the existing knowledge of dietary polyphenols for the development of NAFLD regulated by intestinal microecology is discussed. Polyphenols can influence the vagal afferent pathway in the central and enteric nervous system to control NAFLD via gut-brain-liver cross-talk. The possible mechanisms involve in the alteration of microbial community structure, effects of gut metabolites (short-chain fatty acids (SCFAs), bile acids (BAs), endogenous ethanol (EnEth)), and stimulation of gut-derived hormones (ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and leptin) based on the targets excavated from the gut-brain-liver axis. Consequently, the communication among the intestine, brain, and liver paves the way for new approaches to understand the underlying roles and mechanisms of dietary polyphenols in NAFLD pathology.
Collapse
Affiliation(s)
- Zhenyu Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jie Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
33
|
Bioinformatis analysis reveals possible molecular mechanism of PXR on regulating ulcerative colitis. Sci Rep 2021; 11:5428. [PMID: 33686088 PMCID: PMC7940411 DOI: 10.1038/s41598-021-83742-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease of the gastrointestinal (GI) tract. Ulcerative colitis (UC) is a type of IBD. Pregnane X Receptor (PXR) is a member of the nuclear receptor superfamily. In order to deepen understanding and exploration of the molecular mechanism of regulation roles of PXR on UC, biological informatics analysis was performed. First, 878 overlapping differentially expressed genes (DEGs) between UC and normal samples were obtained from the Gene Expression Omnibus (GEO) database (GSE59071 and GSE38713) by using the "limma" R language package. Then WGCNA analysis was performed by 878 DEGs to obtain co-expression modules that were positively and negatively correlated with clinical traits. GSEA analysis of PXR results obtained the signal pathways enriched in the PXR high and low expression group and the active genes of each signal pathway. Then the association of PXR with genes that are both active in high expression group and negatively related to diseases (gene set 1), or both active in low expression group and negatively related to diseases (gene set 2) was analyzed by String database. Finally, carboxylesterase 2 (CES2), ATP binding cassette subfamily G member 2 (ABCG2), phosphoenolpyruvate carboxykinase (PCK1), PPARG coactivator 1 alpha (PPARGC1A), cytochrome P450 family 2 subfamily B member 6 (CYP2B6) from gene set 1 and C-X-C motif chemokine ligand 8 (CXCL8) from gene set 2 were screened out. After the above analysis and reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) verification, we speculated that PXR may exert a protective role on UC by promoting CES2, ABCG2, PCK1, PPARGC1A, CYP2B6 expression and inhibiting CXCL8 expression in their corresponding signal pathway in intestinal tissue.
Collapse
|
34
|
Zhang L, Liu C, Jiang Q, Yin Y. Butyrate in Energy Metabolism: There Is Still More to Learn. Trends Endocrinol Metab 2021; 32:159-169. [PMID: 33461886 DOI: 10.1016/j.tem.2020.12.003] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/25/2022]
Abstract
Butyrate, a main product of gut microbial fermentation, has been recognized as an important mediator of gut microbiota regulation in whole body energy homeostasis. However, the mechanisms of butyrate metabolic control remain unclear. This review summarizes studies that directly examined the effects of butyrate on metabolic health. The effects of butyrate on metabolic functions, including thermogenesis, lipid and glucose metabolism, appetite, inflammation, and influence on gut microbiota, are described. The effects of butyrate on cellular systems via G protein-coupled receptors (GPRs), as a histone deacetylase inhibitor, and as a substrate that is metabolized intercellularly, are also discussed. Hopefully, a better understanding of butyrate metabolic regulation may provide new perspectives for the nutritional prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Lin Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Chudan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| |
Collapse
|
35
|
Shcherbakova ES, Sall TS, Sitkin SI, Vakhitov TY, Demyanova EV. The role of bacterial metabolites derived from aromatic amino acids in non-alcoholic fatty liver disease. ALMANAC OF CLINICAL MEDICINE 2020; 48:375-386. [DOI: 10.18786/2072-0505-2020-48-066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The review deals with the role of aromatic amino acids and their microbial metabolites in the development and progression of non-alcoholic fatty liver disease (NAFLD). Pathological changes typical for NAFLD, as well as abnormal composition and/or functional activity of gut microbiota, results in abnormal aromatic amino acid metabolism. The authors discuss the potential of these amino acids and their bacterial metabolites to produce both negative and positive impact on the main steps of NAFLD pathophysiology, such as lipogenesis and inflammation, as well as on the liver functions through regulation of the intestinal barrier and microbiota-gut-liver axis signaling. The review gives detailed description of the mechanism of biological activity of tryptophan and its derivatives (indole, tryptamine, indole-lactic, indole-propyonic, indole-acetic acids, and indole-3-aldehyde) through the activation of aryl hydrocarbon receptor (AhR), preventing the development of liver steatosis. Bacteria-produced phenyl-alanine metabolites could promote liver steatosis (phenyl acetic and phenyl lactic acids) or, on the contrary, could reduce liver inflammation and increase insulin sensitivity (phenyl propionic acid). Tyramine, para-cumarate, 4-hydroxyphenylacetic acids, being by-products of bacterial catabolism of tyrosine, can prevent NAFLD, whereas para-cresol and phenol accelerate the progression of NAFLD by damaging the barrier properties of intestinal epithelium. Abnormalities in bacterial catabolism of tyrosine, leading to its excess, stimulate fatty acid synthesis and promote lipid infiltration of the liver. The authors emphasize a close interplay between bacterial metabolism of aromatic amino acids by gut microbiota and the functioning of the human body. They hypothesize that microbial metabolites of aromatic amino acids may represent not only therapeutic targets or non-invasive biomarkers, but also serve as bioactive agents for NAFLD treatment and prevention.
Collapse
Affiliation(s)
| | - T. S. Sall
- State Research Institute of Especially Purified Bioproducts
| | - S. I. Sitkin
- State Research Institute of Especially Purified Bioproducts;
North Western State Medical University named after I.I. Mechnikov
| | | | | |
Collapse
|
36
|
Guerra S, Gastaldelli A. The role of the liver in the modulation of glucose and insulin in non alcoholic fatty liver disease and type 2 diabetes. Curr Opin Pharmacol 2020; 55:165-174. [PMID: 33278735 DOI: 10.1016/j.coph.2020.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022]
Abstract
In this review we have discussed how the liver plays a central role in the regulation of glucose metabolism and in insulin clearance. Both non-alcoholic fatty liver disease (NAFLD) and diabetes (T2D) are characterized by high plasma insulin concentrations, hepatic insulin resistance, high hepatic glucose production (HGP), in particular gluconeogenesis (GNG), that are increased proportionally to fasting hyperglycemia, while postprandial hyperglycemia is due to impaired suppression of HGP by insulin, and reduced hepatic glycogen storage. The liver acts also as a modulator of peripheral insulin since most of insulin secreted by the pancreas is cleared by the liver during the first pass. Hepatokines and hepatic lipids can act in either autocrine or paracrine way and can be responsible of the changes in insulin sensitivity and alterations in glucose metabolism.
Collapse
Affiliation(s)
- Sara Guerra
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy.
| |
Collapse
|
37
|
|
38
|
Fujii H, Kawada N. The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2020; 21:ijms21113863. [PMID: 32485838 PMCID: PMC7312931 DOI: 10.3390/ijms21113863] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) consists of the entire spectrum of fatty liver disease in patients without significant alcohol consumption, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) to cirrhosis, with NASH recently shown as an important cause of hepatocellular carcinoma (HCC). There is a close relationship between insulin resistance (IR) and NAFLD, with a five-fold higher prevalence of NAFLD in patients with type 2 diabetes (T2DM) compared to that in patients without T2DM. IR is involved in the progression of disease conditions such as steatosis and NASH, as well as hepatic fibrosis progression. The mechanisms underlying these processes involve genetic factors, hepatic fat accumulation, alterations in energy metabolism, and inflammatory signals derived from various cell types including immune cells. In NASH-associated fibrosis, the principal cell type responsible for extracellular matrix production is the hepatic stellate cell (HSC). HSC activation by IR involves “direct” and “indirect” pathways. This review will describe the molecular mechanisms of inflammation and hepatic fibrosis in IR, the relationship between T2DM and hepatic fibrosis, and the relationship between T2DM and HCC in patients with NAFLD.
Collapse
Affiliation(s)
- Hideki Fujii
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan;
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
- Correspondence: ; Tel.: +81-6-6645-3897
| | | |
Collapse
|