1
|
Chen W, Xie C, He W. Tandem mass tag-based proteomics analysis to reveal growth stage-dependent pathways in Dendrobium nobile Lindl. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1261:124647. [PMID: 40393349 DOI: 10.1016/j.jchromb.2025.124647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE This study aimed to analyze differentially proteins at various growth stages of Dendrobium nobile Lindl and attempt to find some possible functional proteins related to plant growth or pharmacological activity. METHODS The samples were grouped into two categories based on their growth periods as 2-3 years (S1) and 7-8 years (S2), respectively. Tandem Mass Tag(TMT) labeling-based quantitative proteomics combined LC-MS/MS was applied to identify differential protein expression at various growth stages of Dendrobium nobile Lindl. Those proteins were analyzed by various bioinformatics methods. The activities of several antioxidant enzymes including superoxide dismutase(SOD), peroxidase (POD) and catalase (CAT) in the plant were specifically examined using commercially available assay kits. RESULTS A total of 6032 proteins were identified with 283 differential proteins related to 168 up-regulated and 115 down-regulated. KEGG pathway enrichment analysis revealed that proteins involved in heat shock response, phenylpropanoid biosynthesis, and antioxidant activity showed significant changes across the growth stages. Specifically, a decrease in small heat shock proteins (sHSPs) was observed in older plants, potentially reducing their stress resilience, while proteins involved in lignin biosynthesis, such as SOD, CAT, and POD, were upregulated, suggesting improved stress response. CONCLUSION There are 283 differential proteins with diversiform function between different growth stages. Some upregulated antioxidant enzymes contribute to Dendrobium's ability to combat oxidative stress in older plants. These findings provide insights into how protein expression varies with growth stage, offering scientific support for determining optimal harvesting periods or pharmacological mechanism for Dendrobium nobile Lindl.
Collapse
Affiliation(s)
- Wanhui Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education and Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Cong Xie
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education and Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China; Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Wenying He
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education and Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| |
Collapse
|
2
|
Kabonick SG, Verma K, Modesto JL, Pearce VH, Winokur KM, Groisman EA, Townsend GE. Hierarchical glycolytic pathways control the carbohydrate utilization regulator in human gut Bacteroides. Nat Commun 2025; 16:4488. [PMID: 40368925 PMCID: PMC12078602 DOI: 10.1038/s41467-025-59704-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
Human dietary choices control the gut microbiome. Industrialized populations consume abundant amounts of glucose and fructose, resulting in microbe-dependent intestinal disorders. Simple sugars inhibit the carbohydrate utilization regulator (Cur), a transcription factor in members of the prominent gut bacterial phylum, Bacteroidetes. Cur controls products necessary for carbohydrate utilization, host immunomodulation, and intestinal colonization. Here, we demonstrate how simple sugars decrease Cur activity in the mammalian gut. Our findings in two Bacteroides species show that ATP-dependent fructose-1,6-bisphosphate (FBP) synthesis is necessary for glucose or fructose to inhibit Cur, but dispensable for growth because of an essential pyrophosphate (PPi)-dependent enzyme. Furthermore, we show that ATP-dependent FBP synthesis is required to regulate Cur in the gut but does not contribute to fitness when cur is absent, indicating PPi is sufficient to drive glycolysis in these bacteria. Our findings reveal how sugar-rich diets inhibit Cur, thereby disrupting Bacteroides fitness and diminishing products that are beneficial to the host.
Collapse
Affiliation(s)
- Seth G Kabonick
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Kamalesh Verma
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Jennifer L Modesto
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Victoria H Pearce
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Kailyn M Winokur
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | | | - Guy E Townsend
- Penn State College of Medicine, Hershey, PA, USA.
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA.
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA.
| |
Collapse
|
3
|
Chen YW, Ahn IS, Wang SSM, Majid S, Diamante G, Cely I, Zhang G, Cabanayan A, Komzyuk S, Bonnett J, Arneson D, Yang X. Multitissue single-cell analysis reveals differential cellular and molecular sensitivity between fructose and high-fat high-sucrose diets. Cell Rep 2025; 44:115690. [PMID: 40349341 DOI: 10.1016/j.celrep.2025.115690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/03/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic syndrome (MetS), a conglomerate of dysregulated metabolic traits that vary between individuals, is partially driven by modern diets high in fat, sucrose, or fructose and their interactions with host genes in metabolic tissues. To elucidate the roles of individual tissues and cell types in diet-induced MetS, we performed single-cell RNA sequencing on the hypothalamus, liver, adipose tissue, and small intestine of mice fed high-fat high-sucrose (HFHS) or fructose diets. We found that hypothalamic neurons were sensitive to fructose, while adipose progenitor cells and macrophages were responsive to HFHS. Ligand-receptor analysis revealed lipid metabolism and inflammation networks among peripheral tissues driven by HFHS, while both diets stimulated synaptic remodeling within the hypothalamus. mt-Rnr2, a top responder to both diets, mitigated diet-induced MetS by stimulating thermogenesis. Our study demonstrates that HFHS and fructose diets have differential cell type and network targets but also share regulators such as mt-Rnr2 to affect MetS risk.
Collapse
Affiliation(s)
- Yen-Wei Chen
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susanna Sue-Ming Wang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sana Majid
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ingrid Cely
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Guanglin Zhang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Angelus Cabanayan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sergey Komzyuk
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jack Bonnett
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Douglas Arneson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Ramirez CB, Ahn IS, Rubtsova VI, Cely I, Le J, Kim J, Jung S, Kelly ME, Kim Y, Bae H, Song WS, Alam YH, Zhang G, Diamante G, Chao A, Hoffner L, Anica A, Le I, Lopez ML, Tamburini IJ, Moyer EM, Tsai A, Yang Q, Dai X, Piomelli D, Lee G, Yang X, Jang C. Circulating glycerate predicts resilience to fructose-induced hepatic steatosis. Cell Metab 2025; 37:1223-1234.e5. [PMID: 40267913 PMCID: PMC12058382 DOI: 10.1016/j.cmet.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/18/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
Excessive intake of dietary fructose increases the risk of metabolic-dysfunction-associated steatotic liver disease (MASLD), cirrhosis, and cancers. However, what host factors determine disease vulnerability is incompletely understood. Here, we leverage genetically divergent mouse strains, mass spectrometry-based metabolomics, and in vivo isotope tracing, identifying circulating glycerate as a biomarker that predicts resilience to fructose-induced hepatic steatosis in both sexes. We found that the surge of circulating glycerate after an oral fructose provision reflects strong small-intestinal fructose catabolism. Such fructose clearance by the small intestine is linked to a weaker induction of hepatic de novo lipogenesis and steatosis upon chronic fructose exposure across strains. These data indicate the potential utility of an oral fructose tolerance test and circulating glycerate measurements to predict an individual's susceptibility to fructose-elicited steatotic liver and provide personalized dietary recommendations.
Collapse
Affiliation(s)
- Cuauhtemoc B Ramirez
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Varvara I Rubtsova
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Ingrid Cely
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Joohwan Kim
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Miranda E Kelly
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Yeojin Kim
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Hosung Bae
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Won-Suk Song
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Yasmine H Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alina Chao
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Lauren Hoffner
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Alexis Anica
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Izabelle Le
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Miranda L Lopez
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Ian J Tamburini
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Elena M Moyer
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Ariel Tsai
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Qin Yang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Xing Dai
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Daniele Piomelli
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Gina Lee
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA; Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Liu Y, Han F, Ma X, Yang L, Shi Z. Organic acids from ice wine ameliorate fructose-induced disorders of glycolipid metabolism in C57BL/6J mice. Food Funct 2025; 16:3296-3307. [PMID: 40079381 DOI: 10.1039/d4fo05580b] [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: 03/15/2025]
Abstract
Excessive intake of fructose has been widely reported to cause glycolipid metabolism disorders, and it is unclear whether long-term consumption of ice wine, a sweet wine with high sugar content, is beneficial for health. In this study, 6-week-old male C57BL/6J mice were divided into pure water, ice wine, fructose, fructose + succinic acid, fructose + malic acid and fructose + alcohol groups to study the effects and mechanisms of organic acids on glycolipid metabolism. The results indicated that long-term consumption of ice wine did not lead to disorders of glycolipid metabolism, and organic acids inhibited the negative effects of fructose and reduced hepatic fat synthesis by decreasing the mRNA expression of hepatic ACC1, SREBP-1c, and ChREBP-β, as well as controlling the protein expression of KHK-C. These findings provide a theoretical basis for the healthy consumption of ice wine, helping consumers enjoy wine more scientifically and promoting the high-quality development of the industry.
Collapse
Affiliation(s)
- Yang Liu
- College of Enology, Northwest A&F University, Yangling 712100, China.
| | - Fuliang Han
- College of Enology, Northwest A&F University, Yangling 712100, China.
- Shaanxi Engineering Research Center for Viti-Viniculture, Northwest A&F University, Yangling 712100, China
- Viti-Viniculture Key Laboratory of China General Chamber of Commerce, Northwest A&F University, Yangling 712100, China
| | - Xinyuan Ma
- College of Enology, Northwest A&F University, Yangling 712100, China.
| | - Luye Yang
- College of Enology, Northwest A&F University, Yangling 712100, China.
| | - Zhenan Shi
- College of Enology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
6
|
Yu Z, Wang JR, Shi YN, Zhang W, Liu Y. KHK in leptin receptor-expressing neurons is dispensable for fructose preference and energy homeostasis. Biochem Biophys Res Commun 2025; 761:151710. [PMID: 40186919 DOI: 10.1016/j.bbrc.2025.151710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Fructose metabolism in the central nervous system not only affects local neuronal function but also plays a critical role in regulating appetite and energy metabolism. However, the precise mechanisms underlying these effects are still not well understood. The leptin receptor (LepR) plays a crucial role in modulating neuronal signaling pathways that control food intake and energy expenditure, integrating peripheral metabolic signals to maintain physiological balance. This study aimed to investigate the potential impact of fructose metabolism in central nervous system on energy metabolism and glucose homeostasis. To this end, we generated a leptin receptor cell-specific Khk knockout mouse model (LepR-Cre;Khkfl/fl, KhkΔLepR) and assessed body weight and blood glucose under normal chow conditions. Fructose preference was evaluated using a two-bottle drinking test. Following prolonged fructose consumption, energy metabolism and fructose metabolism were monitored. Our results showed no significant changes in body weight, blood glucose, or fructose preference in KhkΔLepR mice under normal chow conditions. Moreover, after long-term fructose drinking intervention, there were no notable differences in energy expenditure or fructose metabolism. Taken together, these findings suggest that KHK-mediated fructose metabolism in leptin receptor-expressing neurons is not essential for fructose preference or energy homeostasis in mice.
Collapse
Affiliation(s)
- Zhengzhen Yu
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, China
| | - Jue-Rui Wang
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, China
| | - Ya-Nan Shi
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, China
| | - Weiping Zhang
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, China.
| | - Yajin Liu
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, China.
| |
Collapse
|
7
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [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: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
8
|
Flores Monar GV, Sanchez Cruz C, Calderon Martinez E. Mindful Eating: A Deep Insight Into Fructose Metabolism and Its Effects on Appetite Regulation and Brain Function. J Nutr Metab 2025; 2025:5571686. [PMID: 40297675 PMCID: PMC12037248 DOI: 10.1155/jnme/5571686] [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: 09/30/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fructose, a common sweetener in modern diets, has profound effects on both metabolism and brain function, primarily due to its distinct metabolic pathways. Unlike glucose, fructose bypasses critical regulatory steps in metabolism, particularly the phosphofructokinase-1 (PFK-1) feedback inhibition, leading to uncontrolled metabolism and increased fat storage. This review delves into the metabolic consequences of fructose consumption, including its limited role in directly stimulating insulin secretion, which affects satiety signaling and contributes to increased food intake. The small intestine initially helps metabolize ingested fructose, shielding the liver and brain from excessive exposure. However, when consumed in excess, particularly in diets high in processed foods, this protective mechanism becomes overwhelmed, contributing to metabolic disorders such as insulin resistance, obesity, and fatty liver disease. The review also explores fructose's impact on the brain, with a focus on the hippocampus, a key region for memory and learning. Chronic high fructose intake has been linked to mitochondrial dysfunction, increased production of reactive oxygen species (ROS), and neuroinflammation, all of which contribute to cognitive decline and impairments in memory and learning. Additionally, fructose-induced alterations in insulin signaling in the brain are associated with increased risk for neurodegenerative diseases. These findings underscore the potential long-term neurological consequences of excessive fructose intake and highlight the need for further human studies to assess the full spectrum of its effects on brain health. Addressing the rising consumption of fructose, particularly in processed foods, is essential for developing targeted strategies to mitigate its adverse metabolic and cognitive outcomes.
Collapse
Affiliation(s)
| | - Camila Sanchez Cruz
- Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, Mexico
| | | |
Collapse
|
9
|
Hetta HF, Sirag N, Elfadil H, Salama A, Aljadrawi SF, Alfaifi AJ, Alwabisi AN, AbuAlhasan BM, Alanazi LS, Aljohani YA, Ramadan YN, Abd Ellah NH, Algammal AM. Artificial Sweeteners: A Double-Edged Sword for Gut Microbiome. Diseases 2025; 13:115. [PMID: 40277825 PMCID: PMC12025785 DOI: 10.3390/diseases13040115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
Background and Aim: The human gut microbiome plays a crucial role in maintaining health. Artificial sweeteners, also known as non-nutritive sweeteners (NNS), have garnered attention for their potential to disrupt the balance of the gut microbiome. This review explores the complex relationship between NNS and the gut microbiome, highlighting their potential benefits and risks. By synthesizing current evidence, we aim to provide a balanced perspective on the role of AS in dietary practices and health outcomes, emphasizing the need for targeted research to guide their safe and effective use. Methods: A comprehensive literature review was conducted through searches in PubMed and Google Scholar, focusing on the effects of artificial sweeteners on gut microbiota. The search utilized key terms including "Gut Microbiome", "gut microbiota", "Eubiosis", "Dysbiosis", "Artificial Sweeteners", and "Nonnutritive Sweeteners". Results: NNS may alter the gut microbiome, but findings remain inconsistent. Animal studies often report a decrease in beneficial bacteria like Bifidobacterium and Lactobacillus, and an increase in harmful strains such as Clostridium difficile and E. coli, potentially leading to inflammation and gut imbalance. Disruptions in short-chain fatty acid (SCFA) production and gut hormone signaling have also been observed. However, human studies generally show milder or no significant changes, highlighting the limitations in translating animal model findings directly to humans. Differences in study design, dosage, exposure time, and sweetener type likely contribute to these varied outcomes. Conclusions: While NNS offer certain benefits, including reduced caloric intake and improved blood sugar regulation, their impact on gut microbiome health raises important concerns. The observed reduction in beneficial bacteria and the rise in pathogenic strains underscore the need for caution in NNS consumption. Furthermore, the disruption of SCFA production and metabolic pathways illustrates the intricate relationship between diet and gut health.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Nizar Sirag
- Division of Pharmacognosy, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Ayman Salama
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Sara F. Aljadrawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Amani J. Alfaifi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Asma N. Alwabisi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Bothinah M. AbuAlhasan
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Layan S. Alanazi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Yara A. Aljohani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.F.A.); (A.J.A.); (A.N.A.); (B.M.A.); (L.S.A.); (Y.A.A.)
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt;
| | - Noura H. Abd Ellah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Badr University in Assiut, Naser City 2014101, Assiut, Egypt;
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Abdelazeem M. Algammal
- Department of Bacteriology, Immunology and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
10
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
11
|
Kabonick SG, Verma K, Modesto JL, Pearce VH, Winokur KM, Groisman EA, Townsend GE. Hierarchical glycolytic pathways control the carbohydrate utilization regulator in human gut Bacteroides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.13.623061. [PMID: 39605394 PMCID: PMC11601483 DOI: 10.1101/2024.11.13.623061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Human dietary choices control the gut microbiome. Industrialized populations consume abundant amounts of glucose and fructose, resulting in microbe-dependent intestinal disorders. Simple sugars inhibit the carbohydrate utilization regulator (Cur), a transcription factor in members of the prominent gut bacterial phylum, Bacteroidetes. Cur controls products necessary for carbohydrate utilization, host immunomodulation, and intestinal colonization. Here, we demonstrate how simple sugars decrease Cur activity in the mammalian gut. Our findings in two Bacteroides species show that ATP-dependent fructose-1,6-bisphosphate (FBP) synthesis is necessary for glucose or fructose to inhibit Cur, but dispensable for growth because of an essential pyrophosphate (PPi)-dependent enzyme. Furthermore, we show that ATP-dependent FBP synthesis is required to regulate Cur in the gut but does not contribute to fitness when cur is absent, indicating PPi is sufficient to drive glycolysis in these bacteria. Our findings reveal how sugar-rich diets inhibit Cur, thereby disrupting Bacteroides fitness and diminishing products that are beneficial to the host.
Collapse
Affiliation(s)
- Seth G. Kabonick
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Kamalesh Verma
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Jennifer L. Modesto
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Victoria H. Pearce
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | - Kailyn M. Winokur
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| | | | - Guy E. Townsend
- Penn State College of Medicine, Hershey, PA, USA
- Penn State One Health Microbiome Center, Pennsylvania State University, State College, PA, USA
- Center for Molecular Carcinogenesis and Toxicology, Pennsylvania State University, State College, PA, USA
| |
Collapse
|
12
|
Lien EC, Vu N, Westermark AM, Danai LV, Lau AN, Gültekin Y, Kukurugya MA, Bennett BD, Vander Heiden MG. Effects of Aging on Glucose and Lipid Metabolism in Mice. Aging Cell 2025; 24:e14462. [PMID: 39731205 PMCID: PMC11984682 DOI: 10.1111/acel.14462] [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: 02/01/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Aging is accompanied by multiple molecular changes that contribute to aging associated pathologies, such as accumulation of cellular damage and mitochondrial dysfunction. Tissue metabolism can also change with age, in part, because mitochondria are central to cellular metabolism. Moreover, the cofactor NAD+, which is reported to decline across multiple tissues during aging, plays a central role in metabolic pathways such as glycolysis, the tricarboxylic acid cycle, and the oxidative synthesis of nucleotides, amino acids, and lipids. To further characterize how tissue metabolism changes with age, we intravenously infused [U-13C]-glucose into young and old C57BL/6J, WSB/EiJ, and diversity outbred mice to trace glucose fate into downstream metabolites within plasma, liver, gastrocnemius muscle, and brain tissues. We found that glucose incorporation into central carbon and amino acid metabolism was robust during healthy aging across these different strains of mice. We also observed that levels of NAD+, NADH, and the NAD+/NADH ratio were unchanged in these tissues with healthy aging. However, aging tissues, particularly brain, exhibited evidence of upregulated fatty acid and sphingolipid metabolism reactions that regenerate NAD+ from NADH. These data suggest that NAD+-generating lipid metabolism reactions may help to maintain the NAD+/NADH ratio during healthy aging.
Collapse
Affiliation(s)
- Evan C. Lien
- Department of Metabolism and Nutritional ProgrammingVan Andel InstituteGrand RapidsMichiganUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ngoc Vu
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | - Anna M. Westermark
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Laura V. Danai
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Biochemistry and Molecular BiologyUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - Allison N. Lau
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yetiş Gültekin
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
13
|
Keung WS, Zhang WH, Luo HY, Chan KC, Chan YM, Xu J. Correlation between the structures of natural polysaccharides and their properties in regulating gut microbiota: Current understanding and beyond. Carbohydr Polym 2025; 352:123209. [PMID: 39843110 DOI: 10.1016/j.carbpol.2024.123209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Natural polysaccharides have complex structural properties and a wide range of health-promoting effects. Accumulating evidence suggests that the effects are significantly mediated through fermentation by gut microbiota. In recent years, the relationship between the structures of natural polysaccharides and their properties in regulating gut microbiota has garnered significant research attention as researchers attempt to precisely understand the role of gut microbiota in the bioactivities of natural polysaccharides. Progress in this niche, however, remains limited. In this review, we first provide an overview of current research investigating this structure-property relationship. We then present a detailed correlation analysis between the structural characteristics of 159 purified natural polysaccharides and their effects on gut microbiota reported over the past two decades. The analysis revealed that diverse gut bacteria show specific correlations with the molecular weight, glycosidic linkages, and monosaccharide composition of natural polysaccharides. Multifaceted molecular mechanisms, including carbohydrate binding, enzymatic degradation, and cross-feeding, were proposed to be collectively involved in these correlations. Finally, we offer our perspective on future studies to further improve our understanding of the relationship between polysaccharide structure and gut microbiota regulation.
Collapse
Affiliation(s)
- Wing-Shan Keung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Wei-Hao Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Han-Yan Luo
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Kam-Chun Chan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Yui-Man Chan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong.
| |
Collapse
|
14
|
Sturno AM, Hassell JE, Lanaspa MA, Bruce KD. Do microglia metabolize fructose in Alzheimer's disease? J Neuroinflammation 2025; 22:85. [PMID: 40089786 PMCID: PMC11910010 DOI: 10.1186/s12974-025-03401-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disorder with a complex etiology. While emerging AD therapeutics can slow cognitive decline, they may worsen dementia in certain groups of individuals. Therefore, alternative treatments are much needed. Microglia, the brain resident macrophages, have the potential to be novel therapeutic targets as they regulate many facets of AD, including lipid droplet (LD) accumulation, amyloid beta (Aβ) clearance, and neuroinflammation. To carry out such functions, microglia undergo phenotypic changes, which are linked to shifts in metabolism and substrate utilization. While homeostatic microglia are driven by oxidative phosphorylation (OXPHOS) and glycolysis, in aging and AD, microglia shift further towards glycolysis. Interestingly, this "metabolic reprogramming" may be linked to an increase in fructose metabolism. In the brain, microglia predominantly express the fructose transporter SLC2A5 (GLUT5), and enzymes involved in fructolysis and endogenous fructose production, with their expression being upregulated in aging and disease. Here, we review evidence for fructose uptake, breakdown, and production in microglia. We also evaluate emerging literature targeting fructose metabolism in the brain and periphery to assess its ability to modulate microglial function in AD. The ability of microglia to transport and utilize fructose, coupled with the well-established role of fructose in metabolic dysfunction, supports the notion that microglial fructose metabolism may be a novel potential therapeutic target for AD.
Collapse
Affiliation(s)
- Annalise M Sturno
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 17th Ave, Aurora, CO, 80045, USA
| | - James E Hassell
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 17th Ave, Aurora, CO, 80045, USA
| | - Miguel A Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 17th Ave, Aurora, CO, 80045, USA
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E. 17th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
Schild T, Wallisch P, Zhao Y, Wang YT, Haughton L, Chirayil R, Pierpont K, Chen K, Nunes-Violante S, Cross J, de Stanchina E, Thompson CB, Scheinberg DA, Perry JSA, Keshari KR. Metabolic engineering to facilitate anti-tumor immunity. Cancer Cell 2025; 43:552-562.e9. [PMID: 40020672 PMCID: PMC11929521 DOI: 10.1016/j.ccell.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/23/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Fructose consumption is elevated in western diets, but its impact on anti-tumor immunity is unclear. Fructose is metabolized in the liver and small intestine, where fructose transporters are highly expressed. Most tumors are unable to drive glycolytic flux using fructose, enriching fructose in the tumor microenvironment (TME). Excess fructose in the TME may be utilized by immune cells to enhance effector functions if engineered to express the fructose-specific transporter GLUT5. Here, we show that GLUT5-expressing CD8+ T cells, macrophages, and chimeric antigen receptor (CAR) T cells all demonstrate improved effector functions in glucose-limited conditions in vitro. GLUT5-expressing T cells show high fructolytic activity in vitro and higher anti-tumor efficacy in murine syngeneic and human xenograft models in vivo, especially following fructose supplementation. Together, our data demonstrates that metabolic engineering through GLUT5 enables immune cells to efficiently utilize fructose and boosts anti-tumor immunity in the glucose-limited TME.
Collapse
Affiliation(s)
- Tanya Schild
- Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Patrick Wallisch
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Yixuan Zhao
- Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ya-Ting Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lyric Haughton
- Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rachel Chirayil
- Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kaitlyn Pierpont
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Kevin Chen
- Anti-tumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sara Nunes-Violante
- Metabolism Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Justin Cross
- Metabolism Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa de Stanchina
- Anti-tumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Craig B Thompson
- Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David A Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Justin S A Perry
- Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kayvan R Keshari
- Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Gerstner Sloan Kettering School for Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
16
|
Delannoy P, Tolan DR, Lanaspa MA, San Millán I, Bae SY, Johnson RJ. Aldose reductase, fructose and fat production in the liver. Biochem J 2025; 482:295-307. [PMID: 40040471 DOI: 10.1042/bcj20240748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 03/06/2025]
Abstract
There is an increasing interest in the role of fructose as a major driver of non-alcoholic fatty liver disease (NAFLD), and it is linked closely with the intake of sugar. However, there has also been the recognition that fructose can be produced directly from intracellular glucose via the evolutionarily conserved polyol pathway whose access is governed by aldose reductase (AR). The purpose of this article is to review the biochemistry of AR and the role of the polyol pathway in opening fructose metabolism. This article provides a new perspective about AR and the other key enzymes surrounding the decision to divert glucose into the polyol pathway which suggests that the production of endogenous fructose may be of much greater significance than historically viewed. There are important aspects of the regulation of the polyol pathway and its committal step catalyzed by AR, which supports the notion that fructose-uric acid pathway is activated by elevated glucose with the downstream consequence of NAFLD and perhaps other chronic metabolic diseases.
Collapse
Affiliation(s)
- Peter Delannoy
- Orgins of Human Metabolic Disease, Phoneix, AZ,85016, U.S.A
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, U.S.A
| | - Miguel A Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, U.S.A
| | - Iñigo San Millán
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, U.S.A
| | - So Young Bae
- Molecular Biology, Cell Biology, and Biochemistry Program, Boston University, Boston, U.S.A
| | - Richard J Johnson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, U.S.A
| |
Collapse
|
17
|
Westerbeke FHM, Attaye I, Rios‐Morales M, Nieuwdorp M. Glycaemic sugar metabolism and the gut microbiota: past, present and future. FEBS J 2025; 292:1421-1436. [PMID: 39359099 PMCID: PMC11927047 DOI: 10.1111/febs.17293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 08/02/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Non-communicable diseases (NCDs), such as type 2 diabetes (T2D) and metabolic dysfunction-associated fatty liver disease, have reached epidemic proportions worldwide. The global increase in dietary sugar consumption, which is largely attributed to the production and widespread use of cheap alternatives such as high-fructose corn syrup, is a major driving factor of NCDs. Therefore, a comprehensive understanding of sugar metabolism and its impact on host health is imperative to rise to the challenge of reducing NCDs. Notably, fructose appears to exert more pronounced deleterious effects than glucose, as hepatic fructose metabolism induces de novo lipogenesis and insulin resistance through distinct mechanisms. Furthermore, recent studies have demonstrated an intricate relationship between sugar metabolism and the small intestinal microbiota (SIM). In contrast to the beneficial role of colonic microbiota in complex carbohydrate metabolism, sugar metabolism by the SIM appears to be less beneficial to the host as it can generate toxic metabolites. These fermentation products can serve as a substrate for fatty acid synthesis, imposing negative health effects on the host. Nevertheless, due to the challenging accessibility of the small intestine, our knowledge of the SIM and its involvement in sugar metabolism remains limited. This review presents an overview of the current knowledge in this field along with implications for future research, ultimately offering potential therapeutic avenues for addressing NCDs.
Collapse
Affiliation(s)
- Florine H. M. Westerbeke
- Department of Internal and Experimental Vascular MedicineAmsterdam University Medical Centers, location AMCThe Netherlands
| | - Ilias Attaye
- Department of Internal and Experimental Vascular MedicineAmsterdam University Medical Centers, location AMCThe Netherlands
| | - Melany Rios‐Morales
- Department of Internal and Experimental Vascular MedicineAmsterdam University Medical Centers, location AMCThe Netherlands
| | - Max Nieuwdorp
- Department of Internal and Experimental Vascular MedicineAmsterdam University Medical Centers, location AMCThe Netherlands
| |
Collapse
|
18
|
Sellami E, Evangelista-Silva PH, Jordão Teixeira C, Diop K, Mitchell P, Forato Anhê F. High fructose rewires gut glucose sensing via glucagon-like peptide 2 to impair metabolic regulation in mice. Mol Metab 2025; 93:102101. [PMID: 39855562 PMCID: PMC11830333 DOI: 10.1016/j.molmet.2025.102101] [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: 10/30/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Increased fructose consumption contributes to type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD), but the mechanisms are ill-defined. Gut nutrient sensing involves enterohormones like Glucagon-like peptide (Glp)2, which regulates the absorptive capacity of luminal nutrients. While glucose is the primary dietary energy source absorbed in the gut, it is unknown whether excess fructose alters gut glucose sensing to impair blood glucose regulation and liver homeostasis. METHODS Mice were fed diets where carbohydrates were either entirely glucose (70 %Kcal) or glucose partially replaced with fructose (8.5 %Kcal). Glp2 receptor (Glp2r) was inhibited with Glp2 (3-33) injections. Glucose tolerance, insulin sensitivity, and gut glucose absorption were concomitantly assessed, and enteric sugar transporters and absorptive surface were quantified by RT-qPCR and histological analysis, respectively. RESULTS High fructose feeding led to impairment of blood glucose disposal, ectopic fat accumulation in the liver, and hepatic (but not muscle or adipose tissue) insulin resistance independent of changes in fat mass. This was accompanied by increased gut glucose absorption, which preceded glucose intolerance and liver steatosis. Fructose upregulated glucose transporters and enlarged the gut surface, but these effects were prevented by Glp2r inhibition. Blocking Glp2r prevented fructose-induced impairments in glucose disposal and hepatic lipid handling. CONCLUSION Excess fructose impairs blood glucose and liver homeostasis by rewiring gut glucose sensing and exacerbating gut glucose absorption. Our findings are positioned to inform novel early diagnostic tools and treatments tailored to counter high fructose-induced metabolic derangements predisposing to T2D and MASLD.
Collapse
Affiliation(s)
- Eya Sellami
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Medicine, Faculty of Medicine, Université Laval - 1050, Av. de la Médecine, Québec, QC, G1V 0A6, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada
| | - Paulo Henrique Evangelista-Silva
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo - 1524, Av. Prof. Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Caio Jordão Teixeira
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo - 1524, Av. Prof. Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Khoudia Diop
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada
| | - Patricia Mitchell
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Fernando Forato Anhê
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Medicine, Faculty of Medicine, Université Laval - 1050, Av. de la Médecine, Québec, QC, G1V 0A6, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
19
|
Pranzini E, Ippolito L, Pardella E, Giannoni E, Chiarugi P. Adapt and shape: metabolic features within the metastatic niche. Trends Endocrinol Metab 2025; 36:205-218. [PMID: 39122599 DOI: 10.1016/j.tem.2024.07.016] [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: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
The success of disseminating cancer cells (DTCs) at specific metastatic sites is influenced by several metabolic factors. Even before DTCs arrival, metabolic conditioning from the primary tumor participates in creating a favorable premetastatic niche at distant organs. In addition, DTCs adjust their metabolism to better survive along the metastatic journey and successfully colonize their ultimate destination. However, the idea that the environment of the target organs may metabolically impact the metastatic fate is often underestimated. Here, we review the coexistence of two distinct strategies by which cancer cells shape and/or adapt to the metabolic profile of colonized tissues, ultimately creating a proper soil for their seeding and proliferation.
Collapse
Affiliation(s)
- Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy.
| |
Collapse
|
20
|
Hara H, Takayanagi K, Shimizu T, Iwashita T, Ikari A, Maeshima A, Hasegawa H. Possible involvement of up-regulated salt-dependent glucose transporter-5 (SGLT5) in high-fructose diet-induced hypertension. Hypertens Res 2025; 48:1068-1079. [PMID: 39706885 DOI: 10.1038/s41440-024-01915-0] [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: 02/26/2024] [Revised: 07/17/2024] [Accepted: 09/06/2024] [Indexed: 12/23/2024]
Abstract
Excessive fructose intake causes a variety of adverse conditions (e.g., obesity, hepatic steatosis, insulin resistance and uric acid overproduction). High fructose-induced hypertension is a particularly common and pathologically significant condition induced by excess fructose, but its underlying mechanisms remain unknown. We investigated these mechanisms in 7-week-old male Sprague-Dawley rats fed normal rat food or a diet containing 60% glucose (GLU group) or 60% fructose (FRU group) for 3, 6, or 12 weeks. Daily food consumption was measured to avoid between-group discrepancies in caloric/salt intake, adjusting for feeding amounts. The mean blood pressure of FRU rats was significantly higher (12 weeks GLU: 94.8 ± 3.4 mmHg vs. 12 weeks FRU: 103.7 ± 1.2 mmHg), and fractional sodium excretion was significantly lower (12 weeks GLU: 0.084 ± 0.011% vs. 12 weeks FRU: 0.059 ± 0.08%), indicating that the high-fructose diet caused salt retention. The kidney weight and glomerular surface area were greater in FRU rats (12 weeks GLU: 7495 ± 181 vs. 12 weeks FRU: 9831 ± 164 μm2), suggesting that the high-fructose diet induced an increase in extracellular fluid volume. The expressions of GLUT5 and ketohexokinase, an enzyme required for fructose metabolism, were up-regulated in the FRU group rats (GLUT5 12 weeks GLU: 104.7 ± 15.4% vs. 12 weeks FLU: 309.0 ± 99.9%, ketohexokinase 12 weeks GLU: 129.6 ± 3.5% vs. 12 weeks FLU: 163.9 ± 13.0%). Cortical ATP levels were significantly lower in FRU rats (12 weeks GLU: 9.82 ± 1.26 nmol/mg protein vs. 12 weeks FRU: 7.59 ± 1.68 nmol/mg protein), possibly indicating ATP consumption due to fructose metabolism. Unlike in previous reports the high-fructose diet did not affect NHE3 expression (12 weeks GLU: 166.1 ± 6.3% vs. 12 weeks FLU: 142.0 ± 5.9%). A gene chip analysis conducted to identify susceptible molecules revealed that only Slc5a10 (corresponding to SGLT5) showed >two-fold up-regulation in FRU versus GLU rats. RT-PCR and in situ hybridization confirmed the SGLT5 up-regulation (12 weeks GLU: 75.0 ± 5.8% vs. 12 weeks FLU: 230.1 ± 16.0%). Our findings may indicate that the high-fructose diet increased sodium reabsorption principally through up-regulated SGLT5, finally causing salt-sensitive hypertension.
Collapse
Affiliation(s)
- Hiroaki Hara
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan
| | - Kaori Takayanagi
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan
- Ishikawa Kinenkai Kawagoe Ekimae Clinic, Wakita-honmachi 16-23, Kawagoe, Saitama, 350-1123, Japan
| | - Taisuke Shimizu
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan
| | - Takatsugu Iwashita
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, Japan
| | - Akito Maeshima
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan
| | - Hajime Hasegawa
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 350-8550, Japan.
| |
Collapse
|
21
|
Devall MA, Eaton S, Hu G, Sun X, Jakum E, Venkatesh S, Powell SM, Yoshida C, Weisenberger DJ, Cooper GS, Willis J, Ebrahim S, Zoellner J, Casey G, Li L. Association between dietary fructose and human colon DNA methylation: implication for racial disparities in colorectal cancer risk using a cross-sectional study. Am J Clin Nutr 2025; 121:522-534. [PMID: 39788295 PMCID: PMC11923427 DOI: 10.1016/j.ajcnut.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND An increasing body of evidence has linked fructose intake to colorectal cancer (CRC). African-American (AA) adults consume greater quantities of fructose and are more likely to develop right-side colon cancer than European American (EA) adults. OBJECTIVES We examined the hypothesis that fructose consumption leads to epigenomic and transcriptomic differences associated with CRC tumor biology. METHODS Deoxyribonucleic acid methylation data from this cross-sectional study was obtained using the Illumina Infinium MethylationEPIC kit (GSE151732). Right and left colon differentially methylated regions (DMRs) were identified using DMRcate through analysis of Food Frequency Questionnaire data on fructose consumption in normal colon biopsies (n = 79) of AA adults undergoing screening colonoscopy. Secondary analysis of CRC tumors was carried out using data derived from The Cancer Genome Atlas Colon Adenocarcinoma, GSE101764, and GSE193535. Right colon organoids derived from AA (n = 5) and EA (n = 5) adults were exposed to 4.4 mM of fructose for 72 h. Differentially expressed genes (DEGs) were identified using DESeq2. RESULTS We identified 4263 right colon fructose-associated DMRs [false-discovery rates (FDR) < 0.05]. In contrast, only 24 DMRs survived multiple testing corrections (FDR < 0.05) in matched, left colon. Almost 50% of right colon fructose-associated DMRs overlapped regions implicated in CRC in ≥1 of 3 data sets. Highly significant enrichment was also observed between genes corresponding to right colon fructose-associated DMRs and DEGs associated with fructose exposure in right colon organoids of AA individuals (P = 3.28E-30). Overlapping and significant enrichments for fatty acid metabolism, glycolysis, and cell proliferation pathways were also found. Cross-referencing genes within these pathways to DEGs in CRC tumors reveal potential roles for ankyrin repeat domain containing protein 23 and phosphofructokinase, platelet in fructose-mediated CRC risk for AA individuals. CONCLUSIONS Our data support that dietary fructose exerts a greater CRC risk-related effect in the right than left colon among AA adults, alluding to its potential role in contributing to racial disparities in CRC.
Collapse
Affiliation(s)
- Matthew A Devall
- Department of Family Medicine, University of Virginia, Charlottesville, VA, United States; University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Stephen Eaton
- Department of Family Medicine, University of Virginia, Charlottesville, VA, United States
| | - Gaizun Hu
- Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
| | - Xiangqing Sun
- Department of Family Medicine, University of Virginia, Charlottesville, VA, United States
| | - Ethan Jakum
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | - Samyukta Venkatesh
- Department of Family Medicine, University of Virginia, Charlottesville, VA, United States
| | - Steven M Powell
- Digestive Health Center, University of Virginia, Charlottesville, VA, United States
| | - Cynthia Yoshida
- Digestive Health Center, University of Virginia, Charlottesville, VA, United States
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, United States
| | - Gregory S Cooper
- Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Joseph Willis
- Department of Pathology, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Seham Ebrahim
- Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
| | - Jamie Zoellner
- University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, United States; Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States; Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA, United States; University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
22
|
Shen Z, Liu Z, Wang H, Landrock D, Noh JY, Zang QS, Lee C, Farnell YZ, Chen Z, Sun Y. Fructose induces inflammatory activation in macrophages and microglia through the nutrient-sensing ghrelin receptor. FASEB J 2025; 39:e70412. [PMID: 39985299 PMCID: PMC11846021 DOI: 10.1096/fj.202402531r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
High fructose corn syrup (HFCS) is a commonly used sweetener in soft drinks and processed foods, and HFCS exacerbates inflammation when consumed in excess. Fructose, a primary component of HFCS; however, it is unclear whether fructose directly activates inflammatory signaling. Growth hormone secretagogue receptor (GHSR) is a receptor of the nutrient-sensing hormone ghrelin. We previously reported that GHSR ablation mitigates HFCS-induced inflammation in adipose tissue and liver, shifting macrophages toward an anti-inflammatory spectrum. Since inflammation is primarily governed by innate immune cells, such as macrophages in the peripheral tissues and microglia in the brain, this study aims to investigate whether GHSR autonomously regulates pro-inflammatory activation in macrophages and microglia upon fructose exposure. GHSR deletion mutants of RAW 264.7 macrophages and the immortalized microglial cell line (IMG) were generated using CRISPR-Cas9 gene editing. After treating the cells with equimolar concentrations of fructose or glucose for 24 h, fructose increased mRNA and protein expression of GHSR and pro-inflammatory cytokines (Il1β, Il6, and Tnfα) in both macrophages and microglia, suggesting that fructose activates Ghsr and induces inflammation directly in macrophages and microglia. Remarkably, GHSR deletion mutants (Ghsrmutant) of macrophages and microglia exhibited reduced inflammatory responses to fructose, indicating that GHSR mediates fructose-induced inflammation. Furthermore, we found that GHSR regulates fructose transport and fructose metabolism and mediates fructose-induced inflammatory activation through CREB-AKT-NF-κB and p38 MAPK signaling pathways. Our results underscore that fructose triggers inflammation, and reducing HFCS consumption would reduce disease risk. Moreover, these findings reveal for the first time that the nutrient-sensing receptor GHSR plays a crucial role in fructose-mediated inflammatory activation, suggesting that targeting GHSR may be a promising therapeutic approach to combat the immunotoxicity of foods that contain fructose.
Collapse
Affiliation(s)
- Zheng Shen
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Zeyu Liu
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Hongying Wang
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Danilo Landrock
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Ji Yeon Noh
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Qun Sophia Zang
- Department of SurgeryLoyola University Chicago Health Science CampusMaywoodIllinoisUSA
| | - Chih‐Hao Lee
- Genomics Research Center, Academia SinicaTaipeiTaiwan
| | - Yuhua Z. Farnell
- Department of Poultry ScienceTexas A&M UniversityCollege StationTexasUSA
| | - Zheng Chen
- Department of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yuxiang Sun
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
23
|
El-Kurjieh A, Al-Arab R, Hachem QA, Ibrahim JN, Kobeissy PH. ACSS2 and metabolic diseases: from lipid metabolism to therapeutic target. Lipids Health Dis 2025; 24:74. [PMID: 40001058 PMCID: PMC11853604 DOI: 10.1186/s12944-025-02491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Elevated incidence of metabolic disorders has been reported worldwide in the recent decade, highlighting the need for developing efficient therapies. These diseases result from a complex interplay of various factors that contribute to disease progression, complications, and resistance to current treatment options. Acetyl-CoA Synthetase Short Chain Family Member 2 (ACSS2) is a nucleo-cytosolic enzyme with both lipogenic and metabolic regulatory roles. Studies on ACSS2 have shown that it is involved in pathways commonly dysregulated in metabolic disorders, leading to fat deposition and disrupted cellular signaling. Although multiple studies have suggested a role of ACSS2 in the metabolic rewiring during tumorigenesis, few studies have examined its involvement in the pathophysiology of metabolic diseases. Recent evidence indicates that ACSS2 may contribute to the pathogenesis of various metabolic disorders making its examination of great interest and potentially aiding in the development of new therapeutic strategies. The objective of this review is to summarize the current understanding of ACSS2's role in metabolic disorders and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Alaa El-Kurjieh
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Reem Al-Arab
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Qamar Abou Hachem
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - José-Noel Ibrahim
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon.
| | - Philippe Hussein Kobeissy
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon.
| |
Collapse
|
24
|
Melino G, Bischof J, Chen WL, Jia W, Juhl H, Kopeina GS, Mauriello A, Novelli F, Scimeca M, Shi Y, Pirozzi BM, Sica G, Zamaraev AV, Zhivotovsky B. New hope for the world cancer day. Biol Direct 2025; 20:14. [PMID: 39901154 PMCID: PMC11792201 DOI: 10.1186/s13062-025-00608-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
Affiliation(s)
- Gerry Melino
- TOR, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Julia Bischof
- Indivumed GmbH, Falkenried, 88 Building D, 20251, Hamburg, Germany
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wei Jia
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| | - Harmut Juhl
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| | - Gelina S Kopeina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, 96816, USA
| | | | - Yufang Shi
- The Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | | | - Giuseppe Sica
- TOR, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Alexey V Zamaraev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Boris Zhivotovsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Lomonosov Moscow State University, Moscow, Russia.
- Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Hayes AMR, Swackhamer C, Quezada-Calvillo R, Butte NF, Sterchi EE, Nichols BL, Hamaker BR. Moderating carbohydrate digestion rate in mice promotes fat oxidation and metabolic flexibility revealed through a new approach to assess metabolic substrate utilization. Eur J Nutr 2025; 64:83. [PMID: 39904882 PMCID: PMC11908681 DOI: 10.1007/s00394-025-03585-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Superior metabolic flexibility, or the ability to efficiently switch between oxidation of carbohydrate and fat, is inversely associated with obesity and type 2 diabetes. The influence of dietary factors on metabolic flexibility is incompletely understood. This study examined the impact of dietary carbohydrate digestion rate on metabolic flexibility and metabolic substrate utilization. METHODS We employed percent relative cumulative frequency (PRCF) analyses coupled with a new application of modeling using the Mixed Weibull Cumulative Distribution function to examine respiratory exchange ratio (RER) data from adult wild-type mice and mice lacking the mucosal maltase-glucoamylase enzyme (Mgam) under different dietary carbohydrate conditions, with diets matched for total carbohydrate contents and containing different ratios of slowly digestible starch (SDS) and resistant starch (RS), or that were high in sucrose or fat. Fungal amyloglucosidase (AMG) was administered in drinking water to increase carbohydrate digestion rate. We devised a Metabolic Flexibility Factor (MFF) to quantitate metabolic flexibility for each dietary condition and mouse genotype, with higher MFF indicating higher metabolic flexibility. RESULTS Diets high in SDS exhibited lower average RER and higher metabolic flexibility (MFF) than diets high in resistant starch, sucrose, or fat. Diets containing high and intermediate amounts of SDS led to a more complete shift to fat oxidation. While mouse genotype had minimal effects on substrate oxidation and MFF, AMG supplementation shifted substrate utilization to carbohydrate oxidation and generally decreased MFF. CONCLUSIONS Consumption of slowly digestible carbohydrates improved measures of metabolic substrate utilization at the whole-body level in adult mice.
Collapse
Affiliation(s)
- Anna M R Hayes
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA.
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, 97331, USA.
| | - Clay Swackhamer
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Roberto Quezada-Calvillo
- Facultad de Ciencias Quimicas, Universidad Autonoma de San Luis Potosi, Zona Universitaria,, 78210, San Luis Potosí, S.L.P., Mexico
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA
| | - Nancy F Butte
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA
| | - Erwin E Sterchi
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012, Bern, Switzerland
| | - Buford L Nichols
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA.
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
26
|
Tero-Vescan A, Ștefănescu R, Istrate TI, Pușcaș A. Fructose-induced hyperuricaemia - protection factor or oxidative stress promoter? Nat Prod Res 2025; 39:948-960. [PMID: 38522076 DOI: 10.1080/14786419.2024.2327624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/26/2024]
Abstract
Accumulating evidence suggests that dietary fructose may play a role in the hyperuricaemia development, but the precise mechanism remains unclear. Hyperuricaemia is characterised by excessive production and deposition of urate crystals, and the metabolism of fructose has been implicated in the elevation of serum urate levels. The association between fructose intake and the risk of hyperuricaemia is explained by the metabolism of fructose in the liver, small intestine, and kidney. Many studies have confirmed the correlation between fructose consumption and an increased risk of developing hyperuricaemia, but more prospective studies to fully elucidate the role of fructose intake in the pathogenesis of hyperuricaemia are needed. It is important to note that maintaining a balanced diet, and lifestyle is crucial when considering fructose intake. Limiting the consumption of products high in added sugars and maintaining a healthy weight can contribute to reducing the risk of hyperuricaemia and associated health complications.
Collapse
Affiliation(s)
- Amelia Tero-Vescan
- Medical Chemistry and Biochemistry Department, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Hamburg, Germany
| | - Ruxandra Ștefănescu
- Department of Pharmacognosy and Phytotherapy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mures, Romania
| | - Tudor-Ionuț Istrate
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, Târgu Mures, Romania
| | - Amalia Pușcaș
- Biochemistry and Chemistry of the Environmental Factors Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|
27
|
Leven AS, Wagner N, Nienaber S, Messiha D, Tasdogan A, Ugurel S. Changes in tumor and cardiac metabolism upon immune checkpoint. Basic Res Cardiol 2025; 120:133-152. [PMID: 39658699 PMCID: PMC11790718 DOI: 10.1007/s00395-024-01092-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Cardiovascular disease and cancer are the leading causes of death in the Western world. The associated risk factors are increased by smoking, hypertension, diabetes, sedentary lifestyle, aging, unbalanced diet, and alcohol consumption. Therefore, the study of cellular metabolism has become of increasing importance, with current research focusing on the alterations and adjustments of the metabolism of cancer patients. This may also affect the efficacy and tolerability of anti-cancer therapies such as immune-checkpoint inhibition (ICI). This review will focus on metabolic adaptations and their consequences for various cell types, including cancer cells, cardiac myocytes, and immune cells. Focusing on ICI, we illustrate how anti-cancer therapies interact with metabolism. In addition to the desired tumor response, we highlight that ICI can also lead to a variety of side effects that may impact metabolism or vice versa. With regard to the cardiovascular system, ICI-induced cardiotoxicity is increasingly recognized as one of the most life-threatening adverse events with a mortality of up to 50%. As such, significant efforts are being made to assess the specific interactions and associated metabolic changes associated with ICIs to improve both efficacy and management of side effects.
Collapse
Affiliation(s)
- Anna-Sophia Leven
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Natalie Wagner
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Nienaber
- Clinic III for Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Daniel Messiha
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre, University of Duisburg-Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
28
|
Westerbeke FHM, Rios-Morales M, Attaye I, Nieuwdorp M. Fructose catabolism and its metabolic effects: Exploring host-microbiota interactions and the impact of ethnicity. J Physiol 2025. [PMID: 39805044 DOI: 10.1113/jp287316] [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: 09/24/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Important health disparities are observed in the prevalence of obesity and associated non-communicable diseases (NCDs), including type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD) among ethnic groups. Yet, the underlying factors accounting for these disparities remain poorly understood. Fructose has been widely proposed as a potential mediator of these NCDs, given that hepatic fructose catabolism can result in deleterious metabolic effects, including insulin resistance and hepatic steatosis. Moreover, the fermentation of fructose by the gut microbiota can produce metabolites such as ethanol and acetate, both which serve as potential substrates for de novo lipogenesis (DNL) and could therefore contribute to the development of these metabolic conditions. Significant inter-ethnic differences in gut microbiota composition have been observed. Moreover, fructose consumption varies across ethnic groups, and fructose intake has been demonstrated to significantly alter gut microbiota composition, which can influence its fermenting properties and metabolic effects. Therefore, ethnic differences in gut microbiota composition, which may be influenced by variations in fructose consumption, could contribute to the observed health disparities. This review provides an overview of the complex interactions between host and microbial fructose catabolism, the role of ethnicity in shaping these metabolic processes and their impact on host health. Understanding these interactions could provide insights into the mechanisms driving ethnic health disparities to improve personalized nutrition strategies. KEY POINTS: Dietary fructose consumption has increased substantially over recent decades, which has been associated with the rising prevalence of obesity and non-communicable diseases (NCDs) such as type 2 diabetes and metabolic dysfunction-associated steatotic liver disease. Pronounced disparities among different ethnic groups in NCD prevalence and dietary fructose consumption underscore the need to elucidate the underlying mechanisms of fructose catabolism and its health effects. Together with the well-known toxic effects of hepatic fructose catabolism, emerging evidence highlights a role for the small intestinal microbiota in fermenting sugars like fructose into various bacterial products with potential deleterious metabolic effects. There are significant ethnic differences in gut microbiota composition that, combined with varying fructose consumption, could mediate the observed health disparities. To comprehensively understand the role of the gut microbiota in mediating fructose-induced adverse metabolic effects, future research should focus on the small intestinal microbiota. Future research on fructose - microbiota - host interactions should account for ethnic differences in dietary habits and microbial composition to elucidate the potential role of the gut microbiota in driving the mentioned health disparities.
Collapse
Affiliation(s)
- Florine H M Westerbeke
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Melany Rios-Morales
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ilias Attaye
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Fan YH, Zhang S, Wang Y, Wang H, Li H, Bai L. Inter-organ metabolic interaction networks in non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2025; 15:1494560. [PMID: 39850476 PMCID: PMC11754069 DOI: 10.3389/fendo.2024.1494560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem metabolic disorder, marked by abnormal lipid accumulation and intricate inter-organ interactions, which contribute to systemic metabolic imbalances. NAFLD may progress through several stages, including simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and potentially liver cancer. This disease is closely associated with metabolic disorders driven by overnutrition, with key pathological processes including lipid dysregulation, impaired lipid autophagy, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and local inflammation. While hepatic lipid metabolism in NAFLD is well-documented, further research into inter-organ communication mechanisms is crucial for a deeper understanding of NAFLD progression. This review delves into intrahepatic networks and tissue-specific signaling mediators involved in NAFLD pathogenesis, emphasizing their impact on distal organs.
Collapse
Affiliation(s)
- Yu-Hong Fan
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Siyao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
30
|
Fikry H, Saleh LA, Sadek DR, Alkhalek HAA. The possible protective effect of luteolin on cardiovascular and hepatic changes in metabolic syndrome rat model. Cell Tissue Res 2025; 399:27-60. [PMID: 39514020 DOI: 10.1007/s00441-024-03927-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
The metabolic syndrome, or MetS, is currently a global health concern. The anti-inflammatory, anti-proliferative, and antioxidant properties of luteolin are some of its advantageous pharmacological characteristics. This research was designed to establish a MetS rat model and investigate the possible protective effect of luteolin on cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. Forty adult male albino rats were split into four groups: a negative control group, a group treated with luteolin, a group induced MetS (fed 20% fructose), and a group treated with luteolin (fed 20% fructose and given luteolin). Following the experiment after 8 weeks, biochemical, histological (light and electron), and immunohistochemistry analyses were performed on liver and heart tissues. Serum levels of cTnI, CK-MB, and LDH were significantly elevated in response to the cardiovascular effect of MetS. Furthermore, compared to the negative control group, the MetS group showed a marked increase in lipid peroxidation in the cardiac and hepatic tissues, as evidenced by elevated levels of MDA and a decline in the antioxidant defense system, as demonstrated by lower activities of GSH and SOD. The fatty liver-induced group exhibited histological alterations, including disrupted hepatic architecture, dilated and congested central veins, blood sinusoids, and portal veins. In addition to nuclear structural alterations, most hepatocytes displayed varying degrees of cytoplasmic vacuolation, mitochondrial alterations, and endoplasmic reticulum dilatation. These alterations were linked to inflammatory cellular infiltrations, collagen fiber deposition, active hepatic stellate cells, and scattered hypertrophied Kupffer cells, as demonstrated by electron microscopy and validated by immunohistochemical analysis. It is interesting to note that eosinophils were seen between the liver cells and in dilated blood sinusoids. Moreover, the biochemical (hepatic and cardiac) and histological (liver) changes were significantly less severe in luteolin-treated rat on a high-fructose diet. These results suggested that luteolin protects against a type of metabolic syndrome that is produced experimentally.
Collapse
Affiliation(s)
- Heba Fikry
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon St, Abbasiya Sq., Cairo, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon St, Abbasiya Sq., Cairo, Egypt
| | - Doaa Ramadan Sadek
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon St, Abbasiya Sq., Cairo, Egypt
| | - Hadwa Ali Abd Alkhalek
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon St, Abbasiya Sq., Cairo, Egypt
| |
Collapse
|
31
|
Wu PW, Chin YT, Lin WT, Tsai S, Lee CY, Tsai WC, Seal DW, Lee CH. Fructose intake, endogenous biomarkers and latent metabolic construct in adolescents: Exploring path associations and mediating effects. Pediatr Obes 2025; 20:e13176. [PMID: 39340256 DOI: 10.1111/ijpo.13176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Uric acid (UA) and homeostatic model assessment of insulin resistance (HOMA-IR) are endogenous biomarkers implicated in metabolic disorders and dysfunction. OBJECTIVES To investigate the structural associations between sugar-sweetened beverage intake (SSB), UA, HOMA-IR and adolescent latent MetS construct (MetsC) representing paediatric metabolic syndrome (MetS). METHODS A population-based representative adolescent cohort (n = 1454) was evaluated for risk profiles of MetS. Structural equation modelling was performed to identify multifactor structural associations between study parameters and evaluate mediating effects. RESULTS Adolescents had a single-factor latent construct representing MetS. Increased SSB intake was associated with higher UA and HOMA-IR levels, and the two biomarkers were positively associated with the MetsC score. UA and HOMA-IR exerted three mediating effects on the association between fructose-rich tea beverage (FTB) intake of >500 mL/day and MetsC: adjusted standardized coefficient and mediating effect (%), FTB → UA → MetsC: 0.071, 23.1%; FTB → HOMA-IR → MetsC: 0.034, 11.0%; FTB → UA → HOMA-IR → MetsC: 0.010, 3.1%. The UA-associated pathways accounted for 31.1% of the overall mediation on the association between bottled sugar-containing beverage intake and MetsC. After accounting for the UA- and HOMA-IR-derived detrimental effects, the fructose-rich tea beverage intake of >500 mL/day had a tea-related beneficial effect on MetsC, with an adjusted standardized coefficient of -0.103. CONCLUSIONS UA and HOMA-IR individually and jointly mediate the adverse effects of high fructose-rich SSB intake on the mechanisms underlying paediatric MetS. Fructose-free tea-based beverages may have a beneficial effect on latent MetS structure in adolescents.
Collapse
Affiliation(s)
- Pei-Wen Wu
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Ting Chin
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Ting Lin
- Department of Social, Behavioral, and Population Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Sharon Tsai
- Department of Laboratory Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
| | - Chun-Ying Lee
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Chung Tsai
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - David W Seal
- Department of Social, Behavioral, and Population Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Chien-Hung Lee
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
32
|
Fadhul T, Park SH, Ali H, Alsiraj Y, Wali JA, Simpson SJ, Softic S. Fructose-Induced Metabolic Dysfunction Is Dependent on the Baseline Diet, the Length of the Dietary Exposure, and Sex of the Mice. Nutrients 2024; 17:124. [PMID: 39796558 PMCID: PMC11722689 DOI: 10.3390/nu17010124] [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: 11/20/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: High sugar intake, particularly fructose, is implicated in obesity and metabolic complications. On the other hand, fructose from fruits and vegetables has undisputed benefits for metabolic health. This raises a paradoxical question-how the same fructose molecule can be associated with detrimental health effects in some studies and beneficial in others. This study investigates how diet and sex interact with fructose to modulate the metabolic outcomes. Methods: Male and female mice were fed different normal chow diets, Boston chow diet (BCD; 23% protein, 22% fat, 55% carbohydrates), Lexington chow diet (LXD; 24% protein, 18% fat, 58% carbohydrates), and low-fat diet (LFD; 20% protein, 10% fat, 70% carbohydrates), supplemented with 30% fructose in water. Results: Fructose-supplemented male mice on BCD gained weight and developed glucose intolerance and hepatic steatosis. Conversely, male mice given fructose on LXD did not gain weight, remained glucose-tolerant, and had normal hepatic lipid content. Furthermore, fructose-fed male mice on LFD did not gain weight. However, upon switching to BCD, they gained weight, exhibited worsening liver steatosis, and advanced hepatic insulin resistance. The effects of fructose are sex-dependent. Thus, female mice did not gain weight and remained insulin-sensitive with fructose supplementation on BCD, despite developing hepatic steatosis. These differences in metabolic outcomes correlate with the propensity of the baseline diet to suppress hepatic ketohexokinase expression and the de novo lipogenesis pathway. This is likely driven by the dietary fat-to-carbohydrate ratio. Conclusions: Metabolic dysfunction attributed to fructose intake is not a universal outcome. Instead, it depends on baseline diet, dietary exposure length, and sex.
Collapse
Affiliation(s)
- Taghreed Fadhul
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (T.F.); (S.-H.P.); (Y.A.)
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (T.F.); (S.-H.P.); (Y.A.)
- Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA;
| | - Heba Ali
- Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA;
| | - Yasir Alsiraj
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (T.F.); (S.-H.P.); (Y.A.)
- Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA;
| | - Jibran A. Wali
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Stephen J. Simpson
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Samir Softic
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (T.F.); (S.-H.P.); (Y.A.)
- Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY 40536, USA;
| |
Collapse
|
33
|
Park SH, Fadhul T, Conroy LR, Clarke HA, Sun RC, Wallenius K, Boucher J, O’Mahony G, Boianelli A, Persson M, Jung S, Jang C, Loria AS, Martinez GJ, Kipp ZA, Bates EA, Hinds TD, Divanovic S, Softic S. Knockdown of ketohexokinase versus inhibition of its kinase activity exert divergent effects on fructose metabolism. JCI Insight 2024; 9:e184396. [PMID: 39418102 PMCID: PMC11623947 DOI: 10.1172/jci.insight.184396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Excessive fructose intake is a risk factor for the development of obesity and its complications. Targeting ketohexokinase (KHK), the first enzyme of fructose metabolism, has been investigated for the management of metabolic dysfunction-associated steatotic liver disease (MASLD). We compared the effects of systemic, small molecule inhibitor of KHK enzymatic activity with hepatocyte-specific, N-acetylgalactosamine siRNA-mediated knockdown of KHK in mice on an HFD. We measured KHK enzymatic activity, extensively quantified glycogen accumulation, performed RNA-Seq analysis, and enumerated hepatic metabolites using mass spectrometry. Both KHK siRNA and KHK inhibitor led to an improvement in liver steatosis; however, via substantially different mechanisms, KHK knockdown decreased the de novo lipogenesis pathway, whereas the inhibitor increased the fatty acid oxidation pathway. Moreover, KHK knockdown completely prevented hepatic fructolysis and improved glucose tolerance. Conversely, the KHK inhibitor only partially reduced fructolysis, but it also targeted triokinase, mediating the third step of fructolysis. This led to the accumulation of fructose-1 phosphate, resulting in glycogen accumulation, hepatomegaly, and impaired glucose tolerance. Overexpression of wild-type, but not kinase-dead, KHK in cultured hepatocytes increased hepatocyte injury and glycogen accumulation after treatment with fructose. The differences between KHK inhibition and knockdown are, in part, explained by the kinase-dependent and -independent effects of KHK on hepatic metabolism.
Collapse
Affiliation(s)
- Se-Hyung Park
- Department of Pediatrics and Division of Pediatric Gastroenterology and
| | - Taghreed Fadhul
- Department of Pediatrics and Division of Pediatric Gastroenterology and
| | - Lindsey R. Conroy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Harrison A Clarke
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
- Department of Biochemistry & Molecular Biology, Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, USA
| | - Ramon C. Sun
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
- Department of Biochemistry & Molecular Biology, Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, USA
| | - Kristina Wallenius
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jeremie Boucher
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Alessandro Boianelli
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marie Persson
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sunhee Jung
- Department of Biological Chemistry, School of Medicine; and Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of Medicine; and Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Genesee J. Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Evelyn A. Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine; and Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Samir Softic
- Department of Pediatrics and Division of Pediatric Gastroenterology and
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Fort G, Arnold H, Camolotto S, Tariq R, Waters A, O'Toole K, Snyder EL. Opposing lineage specifiers induce a pro-tumor hybrid-identity state in lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626384. [PMID: 39677719 PMCID: PMC11642828 DOI: 10.1101/2024.12.02.626384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The ability of cancer cells to alter their identity, known as lineage plasticity, is crucial for tumor progression and therapy resistance. In lung adenocarcinoma (LUAD), tumor progression is characterized by a gradual loss of lineage fidelity and the emergence of non-pulmonary identity programs. This can lead to hybrid-identity (hybrid-ID) states in which developmentally incompatible identity programs are co-activated within individual cells. However, the molecular mechanisms underlying these identity shifts remain incompletely understood. Here, we identify the gastrointestinal (GI) transcriptional regulator HNF4α as a critical driver of tumor growth and proliferation in KRAS-driven LUAD. In LUAD cells that express the lung lineage specifier NKX2-1, HNF4α can induce a GI/liver-like state by directly binding and activating its canonical targets. HNF4α also forms an aberrant protein complex with NKX2-1, which disrupts NKX2-1 localization and dampens pulmonary identity within hybrid-ID LUAD. Sustained signaling through the RAS/MEK pathway is critical for maintaining the hybrid-ID state. Moreover, RAS/MEK inhibition augments NKX2-1 chromatin binding at pulmonary-specific genes and induces resistance-associated pulmonary signatures. Finally, we demonstrate that HNF4α depletion enhances sensitivity to pharmacologic KRAS G12D inhibition. Collectively, our data show that co-expression of opposing lineage specifiers leads to a hybrid identity state that can drive tumor progression and dictate response to targeted therapy in LUAD.
Collapse
|
35
|
Pouille CL, Dugardin C, Behra J, Tourret M, Molinié R, Fontaine JX, Mathiron D, Palaric C, Gagneul D, Ravallec R, Rambaud C, Hilbert JL, Lucau-Danila A, Cudennec B. Metabolomic monitoring of chicory during in vitro gastrointestinal digestion and correlation with bioactive properties. Food Chem 2024; 467:142344. [PMID: 39644662 DOI: 10.1016/j.foodchem.2024.142344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Chicory, recognized as a functional food, is increasingly becoming the focus of research. This study aimed to investigate the in vitro impact of gastrointestinal digestion on the composition and bioactive properties of chicory decoction. Chicory flour, derived from the roots, was transformed into an aqueous decoction and was subjected to simulated in vitro human gastrointestinal digestion (SGID). For the first time, the influence of the digestive process on specific classes of bioactive molecules was tracked across different digestive compartments (oral, gastric, and intestinal) using a metabolomic approach. Concurrently, the antioxidant, anti-inflammatory, and intestinal hormone regulation effects were assessed before and after SGID. The findings revealed that specific transformations of chlorogenic acid (CGA) and sesquiterpene lactones (STL) during SGID enhanced antioxidant and anti-inflammatory properties post-digestion. Quantitative results demonstrated a significant increase in ROS scavenging capacity and metabolite activity.
Collapse
Affiliation(s)
- Céline L Pouille
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | | | - Josette Behra
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France
| | | | - Roland Molinié
- UPJV, UMRT 1158 BioEcoAgro. BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France
| | - Jean-Xavier Fontaine
- UPJV, UMRT 1158 BioEcoAgro. BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France
| | - David Mathiron
- Plateforme Analytique UFR des Sciences, UPJV, Bâtiment Serres-Transfert Rue Dallery-Passage du Sourire d'Avril, 80039 Amiens, France
| | - Cécile Palaric
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - David Gagneul
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | | | - Caroline Rambaud
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - Jean-Louis Hilbert
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France
| | - Anca Lucau-Danila
- Univ. Lille, UMRT 1158 BioEcoAgro, F-59000 Lille, France; Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d'Ascq, France.
| | | |
Collapse
|
36
|
Xu M, Taylor MS, Hill BG, Li X, Rouchka EC, McClain CJ, Song M. Intestine epithelial-specific hypoxia-inducible factor-1α overexpression ameliorates western diet-induced MASLD. Hepatol Commun 2024; 8:e0572. [PMID: 39585307 PMCID: PMC11596589 DOI: 10.1097/hc9.0000000000000572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/15/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Intestine epithelial hypoxia-inducible factor-1α (HIF-1α) plays a critical role in maintaining gut barrier function. The aim of this study was to determine whether pharmacological or genetic activation of intestinal HIF-1α ameliorates western diet-induced metabolic dysfunction-associated steatotic liver disease. METHODS Metabolic effects of pharmacological activation of HIF-1α by dimethyloxalylglycine were evaluated in HIF-α luciferase reporter (ODD-luc) mice. Male and/or female intestinal epithelial-specific Hif1α overexpression mice (Hif1αLSL/LSL;VilERcre) and wild-type littermates (Hif1αLSL/LSL) were fed with regular chow diet, high fructose (HFr) or high-fat (60% Kcal) high-fructose diet (HFHFr) for 8 weeks. Metabolic phenotypes were profiled. RESULTS Dimethyloxalylglycine treatment led to increased intestine HIF-α luciferase activity and decreased blood glucose levels in HFr diet-fed male ODD-luc mice. Male Hif1αLSL/LSL;VilERcre mice exhibited markedly improved glucose tolerance compared to Hif1αLSL/LSL mice in response to HFr diet. Eight weeks HFHFr feeding led to obesity in both Hif1αLSL/LSL;VilERcre and Hif1αLSL/LSL mice. However, male Hif1αLSL/LSL;VilERcre mice exhibited markedly attenuated hepatic steatosis along with reduced liver size and liver weight compared to male Hif1αLSL/LSL mice. Moreover, HFHFr-induced systemic inflammatory responses were mitigated in male Hif1αLSL/LSL;VilERcre mice compared to male Hif1αLSL/LSL mice, and those responses were not evident in female mice. Ileum RNA-seq analysis revealed that glycolysis/gluconeogenesis was up in male Hif1αLSL/LSL;VilERcre mice, accompanied by increased epithelial cell proliferation. Moreover, an in vitro study showed that HIF stabilization enhances glycolysis in intestine organoids. CONCLUSIONS Our data provide evidence that pharmacological or genetic activation of intestinal HIF-1α markedly ameliorates western diet-induced metabolic dysfunction-associated steatotic liver disease in a sex-dependent manner. The underlying mechanism is likely attributed to HIF-1α activation-induced upregulation of glycolysis, which, in turn, leads to enhanced epithelial cell proliferation and augmented gut barrier function.
Collapse
Affiliation(s)
- Manman Xu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Madison S. Taylor
- Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Bradford G. Hill
- Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiaohong Li
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, Louisville, Kentucky, USA
| | - Eric C. Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, Louisville, Kentucky, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
37
|
Pinheiro DF, Maciel GM, Lima NP, Lima NF, Ribeiro IS, Haminiuk CWI. Impact of fruit consumption on gut microbiota: Benefits, contaminants, and implications for human health. Trends Food Sci Technol 2024; 154:104785. [DOI: 10.1016/j.tifs.2024.104785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
38
|
Fernandes-da-Silva A, Santana-Oliveira DA, Oliveira ASD, Ferreira TAM, Monteiro NC, Silva-Veiga FM, Martins FF, Cummins CL, Romeiro LAS, Souza-Mello V. LDT409 (pan-PPAR partial agonist) mitigates metabolic dysfunction-associated steatotic liver disease in high-fructose-fed mice. Mol Cell Endocrinol 2024; 594:112380. [PMID: 39332468 DOI: 10.1016/j.mce.2024.112380] [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: 08/11/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
AIM This study sought to evaluate the effects of LDT409, a pan-PPAR partial agonist obtained from the main industrial waste from cashew nut processing, on hepatic remodeling, highlighting energy metabolism and endoplasmic reticulum (ER) stress in high-fructose-fed mice. METHODS Male C57BL/6 mice received a control diet (C) or a high-fructose diet (HFRU) for ten weeks. Then, a five-week treatment started: C, C-LDT409, HFRU, and HFRU-LDT409. The LDT409 (40 mg/kg of body weight) was mixed with the diets. RESULTS The HFRU diet caused insulin resistance and endoplasmic reticulum (ER) stress. High Pparg and decreased Ppara expression increased steatosis and pro-fibrogenic gene expression in livers of HFRU-fed mice. Suppressed lipogenic factors, orchestrated by PPAR-gamma, and mitigated ER stress concomitant with the increase in beta-oxidation driven by PPAR-alpha mediated the LDT409 beneficial effects. CONCLUSIONS LDT409 may represent a potential low-cost approach to treat metabolic dysfunction-associated steatotic liver disease, which does not currently have a specific treatment.
Collapse
Affiliation(s)
- Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andressa S de Oliveira
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Thaís A M Ferreira
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Natália Cipriano Monteiro
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Luiz Antonio Soares Romeiro
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
39
|
Zhao Q, Han B, Wang L, Wu J, Wang S, Ren Z, Wang S, Yang H, Carbone M, Dong C, Melino G, Chen WL, Jia W. AKR1B1-dependent fructose metabolism enhances malignancy of cancer cells. Cell Death Differ 2024; 31:1611-1624. [PMID: 39406918 PMCID: PMC11618507 DOI: 10.1038/s41418-024-01393-4] [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: 05/12/2024] [Revised: 08/15/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
Fructose metabolism has emerged as a significant contributor to cancer cell proliferation, yet the underlying mechanisms and sources of fructose for cancer cells remain incompletely understood. In this study, we demonstrate that cancer cells can convert glucose into fructose through a process called the AKR1B1-mediated polyol pathway. Inhibiting the endogenous production of fructose through AKR1B1 deletion dramatically suppressed glycolysis, resulting in reduced cancer cell migration, inhibited growth, and the induction of apoptosis and cell cycle arrest. Conversely, the acceleration of endogenous fructose through AKR1B1 overexpression has been shown to significantly enhance cancer cell proliferation and migration with increased S cell cycle progression. Our findings highlight the crucial role of endogenous fructose in cancer cell malignancy and support the need for further investigation into AKR1B1 as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Qing Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Bing Han
- Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Lu Wang
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Jia Wu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Siliang Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhenxing Ren
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shouli Wang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haining Yang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Michele Carbone
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Changsheng Dong
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
40
|
Fowle-Grider R, Rowles JL, Shen I, Wang Y, Schwaiger-Haber M, Dunham AJ, Jayachandran K, Inkman M, Zahner M, Naser FJ, Jackstadt MM, Spalding JL, Chiang S, McCommis KS, Dolle RE, Kramer ET, Zimmerman SM, Souroullas GP, Finck BN, Shriver LP, Kaufman CK, Schwarz JK, Zhang J, Patti GJ. Dietary fructose enhances tumour growth indirectly via interorgan lipid transfer. Nature 2024; 636:737-744. [PMID: 39633044 DOI: 10.1038/s41586-024-08258-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Fructose consumption has increased considerably over the past five decades, largely due to the widespread use of high-fructose corn syrup as a sweetener1. It has been proposed that fructose promotes the growth of some tumours directly by serving as a fuel2,3. Here we show that fructose supplementation enhances tumour growth in animal models of melanoma, breast cancer and cervical cancer without causing weight gain or insulin resistance. The cancer cells themselves were unable to use fructose readily as a nutrient because they did not express ketohexokinase-C (KHK-C). Primary hepatocytes did express KHK-C, resulting in fructolysis and the excretion of a variety of lipid species, including lysophosphatidylcholines (LPCs). In co-culture experiments, hepatocyte-derived LPCs were consumed by cancer cells and used to generate phosphatidylcholines, the major phospholipid of cell membranes. In vivo, supplementation with high-fructose corn syrup increased several LPC species by more than sevenfold in the serum. Administration of LPCs to mice was sufficient to increase tumour growth. Pharmacological inhibition of ketohexokinase had no direct effect on cancer cells, but it decreased circulating LPC levels and prevented fructose-mediated tumour growth in vivo. These findings reveal that fructose supplementation increases circulating nutrients such as LPCs, which can enhance tumour growth through a cell non-autonomous mechanism.
Collapse
Affiliation(s)
- Ronald Fowle-Grider
- Department of Chemistry, Washington University, St Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Joe L Rowles
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Isabel Shen
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Yahui Wang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Alden J Dunham
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kay Jayachandran
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew Inkman
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael Zahner
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Fuad J Naser
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Madelyn M Jackstadt
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Jonathan L Spalding
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah Chiang
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Kyle S McCommis
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Eva T Kramer
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah M Zimmerman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - George P Souroullas
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Brian N Finck
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Leah P Shriver
- Department of Chemistry, Washington University, St Louis, MO, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA
| | - Charles K Kaufman
- Division of Medical Oncology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jin Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Institute for Informatics, Data Science & Biostatistics (I2DB), Washington University School of Medicine, St Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St Louis, MO, USA.
- Center for Mass Spectrometry and Metabolic Tracing, Washington University, St Louis, MO, USA.
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
41
|
Fadhul T, Park SH, Ali H, Alsiraj Y, Wali JA, Simpson SJ, Softic S. The propensity of fructose to induce metabolic dysfunction is dependent on the baseline diet, length of the dietary exposure, and sex of the mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625441. [PMID: 39651130 PMCID: PMC11623593 DOI: 10.1101/2024.11.26.625441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background/Objectives Numerous studies have implicated high intake of sugar, particularly fructose, with the development of obesity and metabolic complications. On the other hand, fructose from fruits and vegetables has undisputed benefits for metabolic health. This paradox questions how the same fructose molecule can be associated with detrimental health effects in some studies and beneficial in others. Methods To answer this question, male and female mice were fed different normal chow diets and provided 30% fructose solution in water. Results Fructose-supplemented male mice on the Boston Chow Diet (BCD=23% protein, 22% fat, 55% carbs) gained weight, developed glucose intolerance and hepatic steatosis. In contrast, male mice on the Lexington Chow Diet (LXD=24% protein, 18% fat, 58% carbs) did not gain weight, remained glucose tolerant, and had normal hepatic lipid content when supplemented with fructose. Furthermore, fructose-fed male mice on a Low-Fat Diet (LFD=20% protein, 10% fat, 70% carbs) didn't gain weight, but once switched to the BCD, they gained weight, exhibited worsening liver steatosis, and more advanced hepatic insulin resistance. The effects of fructose are sex-dependent, as female mice didn't gain weight and remained insulin-sensitive when given fructose on BCD, despite developing hepatic steatosis. Conclusions The differences in metabolic outcomes correlate with the propensity of the baseline diet to suppress hepatic ketohexokinase expression and the de novo lipogenesis pathway. This is likely driven by the dietary fat-to-carbohydrate ratio. Thus, metabolic dysfunction attributed to fructose intake is not a universal outcome; rather, it depends on the baseline diet, sex, and exposure length.
Collapse
|
42
|
Martínez-Carrillo BE, De Sales-Millán A, Aguirre-Garrido JF, Valdés-Ramos R, de María Cruz-Estrada F, Castillo-Cardiel JA. Changes in the Composition and Diversity of the Intestinal Microbiota Associated with Carbohydrate Consumption in Type 2 Diabetes Mellitus Patients. Int J Mol Sci 2024; 25:12359. [PMID: 39596424 PMCID: PMC11594722 DOI: 10.3390/ijms252212359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial disease, influenced by dietary and environmental factors that can modify the intestinal microbiota. The aim of this study was to evaluate changes in the composition and diversity of the intestinal microbiota associated with carbohydrate (CHO) consumption in T2DM patients. Forty patients participated, with and without T2DM. Fecal samples were collected for the characterization of microbial diversity from the massive sequencing of the 16S rRNA gene. Carbohydrate consumption was quantified using the Frequency Consumption Foods questionnaire (FCF), the groups were categorized according to Body Mass Index (BMI) and BMI + CHO consumption. The group without T2DM showed normal biochemical and anthropometric parameters, although they had a high carbohydrate consumption compared to the group with T2DM. At the phylum level, there were differences in relative abundance; the control overweight group (CL-OW > CHO) and T2DM-Normal Weight > CHO patients had increased Bacteroides and decreased Firmicutes. In contrast, the CL-OW > CHO and T2DM-OW < CHO patients, showed reduced Bacteroidetes and an elevated amount of Firmicutes. At the genus level, the differences were in the relative abundance of Roseburia, Clostridium_IV, Prevotella, and Sporobacter, associated with the consumption of carbohydrates. The groups that consumed high amounts of carbohydrates, regardless of whether they had diabetes mellitus or were overweight, had a significantly reduced proportion of Faecalibacterium, an altered proportion of Bacteroides. The high consumption of carbohydrates showed considerable modifications in the composition and diversity of the bacterial communities.
Collapse
Affiliation(s)
- Beatriz Elina Martínez-Carrillo
- Laboratorio de Investigación en Nutrición, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico; (A.D.S.-M.); (R.V.-R.); (F.d.M.C.-E.)
| | - Amapola De Sales-Millán
- Laboratorio de Investigación en Nutrición, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico; (A.D.S.-M.); (R.V.-R.); (F.d.M.C.-E.)
| | | | - Roxana Valdés-Ramos
- Laboratorio de Investigación en Nutrición, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico; (A.D.S.-M.); (R.V.-R.); (F.d.M.C.-E.)
| | - Flor de María Cruz-Estrada
- Laboratorio de Investigación en Nutrición, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico; (A.D.S.-M.); (R.V.-R.); (F.d.M.C.-E.)
| | - José Arturo Castillo-Cardiel
- Department of Research, Continuing Education and Distance Learning, Universidad Autónoma de Durango, Durango 34209, Mexico;
| |
Collapse
|
43
|
Kilic EB, Koksal E. The Interaction Between Attention Deficit and Hyperactivity Disorder and Nutrition. Curr Nutr Rep 2024; 14:1. [PMID: 39508912 DOI: 10.1007/s13668-024-00592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
PURPOSE OF REVIEW This review aims to explore the relationship between Attention Deficit Hyperactivity Disorder (ADHD) and nutrition. ADHD, a neurodevelopmental disorder, has been examined in relation to dietary factors through various metabolic pathways, with a focus on the role of nutrition in symptom management. Unhealthy dietary patterns, particularly those characteristics of Western diets, are believed to exacerbate ADHD symptoms through these mechanisms. In contrast, dietary interventions such as intermittent fasting, which offer greater flexibility in application, have been proposed as potential strategies to alleviate ADHD symptoms. While further research in this area is expected to contribute significantly to the field, this review also provides researchers with a brief perspective on the challenges and limitations associated with experimental ADHD studies. Therefore, this study aims to offer a comprehensive evaluation of the interaction between ADHD and nutrition, providing researchers with an integrative approach to the topic. RECENT FINDINGS Western dietary patterns have been found to negatively impact gut barrier integrity, synaptic plasticity, insulin resistance, and oxidative stress. On the other hand, the intermittent fasting diet model, which offers practical flexibility, is thought to be a potentially supportive treatment in managing ADHD. Furthermore, it has been concluded that various experimental models are available for ADHD research, and researchers must work within these limitations. Western diets, particularly in their negative impact on synaptic plasticity and other key metabolic pathways involved in ADHD, can worsen the disorder's symptoms. Intermittent fasting emerges as a promising dietary alternative that may mitigate these adverse effects.
Collapse
Affiliation(s)
- Enes Bahadir Kilic
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gaziantep University, Gaziantep, 27310, Turkey.
| | - Eda Koksal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, 06490, Turkey
| |
Collapse
|
44
|
Yan H, Wang Z, Teng D, Chen X, Zhu Z, Chen H, Wang W, Wei Z, Wu Z, Chai Q, Zhang F, Wang Y, Shu K, Li S, Shi G, Zhu M, Piao HL, Shen X, Bu P. Hexokinase 2 senses fructose in tumor-associated macrophages to promote colorectal cancer growth. Cell Metab 2024; 36:2449-2467.e6. [PMID: 39471815 DOI: 10.1016/j.cmet.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/14/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
Fructose is associated with colorectal cancer tumorigenesis and metastasis through ketohexokinase-mediated metabolism in the colorectal epithelium, yet its role in the tumor immune microenvironment remains largely unknown. Here, we show that a modest amount of fructose, without affecting obesity and associated complications, promotes colorectal cancer tumorigenesis and growth by suppressing the polarization of M1-like macrophages. Fructose inhibits M1-like macrophage polarization independently of fructose-mediated metabolism. Instead, it serves as a signal molecule to promote the interaction between hexokinase 2 and inositol 1,4,5-trisphophate receptor type 3, the predominant Ca2+ channel on the endoplasmic reticulum. The interaction reduces Ca2+ levels in cytosol and mitochondria, thereby suppressing the activation of mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 1 (STAT1) as well as NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Consequently, this impedes M1-like macrophage polarization. Our study highlights the critical role of fructose as a signaling molecule that impairs the polarization of M1-like macrophages for tumor growth.
Collapse
Affiliation(s)
- Huiwen Yan
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhi Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Da Teng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, China
| | - Xiaodong Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zijing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Chen
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Wen Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Ziyuan Wei
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenzhen Wu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Chai
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Youwang Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kaile Shu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaotang Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guizhi Shi
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingzhao Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Xian Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
45
|
Chen Y, Li Y, Wang W, Jiang L, Yin S, Guo Z, Wu W, Wang C, Lu S, Wang F, Chen X. A fluorescent NBD "turn-on" probe for the rapid and on-site analysis of fructose in food. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124612. [PMID: 38857548 DOI: 10.1016/j.saa.2024.124612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
High fructose intake is an important cause of metabolic disease. Due to the increasing prevalence of metabolic diseases worldwide, the development of an accurate and efficient tool for monitoring fructose in food is urgently needed to control the intake of fructose. Herein, a new fluorescent probe NBD-PQ-B with 7-nitrobenz-2-oxa-1, 3-diazole (NBD) as the fluorophore, piperazine (PQ) as the bridging group and phenylboronic acid (B) as the recognition receptor, was synthesized to detect fructose. The fluorescence of NBD-PQ-B increased linearly at 550 nm at an excitation wavelength of 497 nm with increasing fructose concentration from 0.1 to 20 mM. The limit of detection (LOD) of fructose was 40 μM. The pKa values of NBD-PQ-B and its fructose complexes were 4.1 and 10.0, respectively. In addition, NBD-PQ-B bound to fructose in a few seconds. The present technique was applied to determine the fructose content in beverages, honey, and watermelon with satisfactory results. Finally, the system could not only be applied in an aqueous solution with a spectrophotometer, but also be fabricated as a NBD-PQ-B/polyvinyl oxide (PEO) film by electrospinning for on-site food analysis simply with the assistance of a smartphone.
Collapse
Affiliation(s)
- Yanan Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Yajing Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Wenjing Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Long Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Shaojie Yin
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Ziwei Guo
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Wenyan Wu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Chongqing Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Sheng Lu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Fang Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China.
| | - Xiaoqiang Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
46
|
Hermanson JB, Tolba SA, Chrisler EA, Leone VA. Gut microbes, diet, and genetics as drivers of metabolic liver disease: a narrative review outlining implications for precision medicine. J Nutr Biochem 2024; 133:109704. [PMID: 39029595 PMCID: PMC11480923 DOI: 10.1016/j.jnutbio.2024.109704] [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: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing in prevalence, impacting over a third of the global population. The advanced form of MASLD, Metabolic dysfunction-associated steatohepatitis (MASH), is on track to become the number one indication for liver transplant. FDA-approved pharmacological agents are limited for MASH, despite over 400 ongoing clinical trials, with only a single drug (resmetirom) currently on the market. This is likely due to the heterogeneous nature of disease pathophysiology, which involves interactions between highly individualized genetic and environmental factors. To apply precision medicine approaches that overcome interpersonal variability, in-depth insights into interactions between genetics, nutrition, and the gut microbiome are needed, given that each have emerged as dynamic contributors to MASLD and MASH pathogenesis. Here, we discuss the associations and molecular underpinnings of several of these factors individually and outline their interactions in the context of both patient-based studies and preclinical animal model systems. Finally, we highlight gaps in knowledge that will require further investigation to aid in successfully implementing precision medicine to prevent and alleviate MASLD and MASH.
Collapse
Affiliation(s)
- Jake B Hermanson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Samar A Tolba
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Evan A Chrisler
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa A Leone
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
47
|
Namjud N, Senaprom S, Ondee T, Bumrungpert A, Heath J, Pongpirul K. Glycemic index and glycemic load of brief sugary sweets: randomized controlled trials of eight Thai desserts. Front Nutr 2024; 11:1452602. [PMID: 39539376 PMCID: PMC11557348 DOI: 10.3389/fnut.2024.1452602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Background Thai desserts, celebrated for their exquisite sweetness, are widely enjoyed for personal indulgence and as cherished souvenirs. However, their high sugar content raises concerns regarding health impacts. This study aimed to quantify the glycemic index (GI) and glycemic load (GL) in healthy volunteers following consumption of various Thai desserts, out of 10 renowned desserts from across Thailand, identified by the Tourism Authority of Thailand, characterized by differing sugar levels. Method Eight were selected based on the absence of preservatives and microbial or chemical contaminations. Each participant consumed a 50-g serving of available carbohydrate (50avCHO) from these desserts. Ninety-six healthy volunteers, with a mean age of 31.8 ± 5.7 years, a mean body weight of 57.2 ± 7.3 kg, and 63.5% women, were randomized into eight groups, with each group comprising 12 participants. Blood samples were collected pre-and post-consumption to assess GI and GL values following established protocols. Results The findings revealed that Phetchaburi's Custard Cake exhibited the lowest GI and GL values (53.4 and 26.7, respectively), with progressively higher values observed in Saraburi's Curry Puff (61.8 and 30.9), Nakhon Sawan's Mochi (68.9 and 34.4), Suphan Buri's Sponge Cake (75.9 and 38.0), Ayutthaya's Cotton Candy (81.4 and 40.7), Prachuap Khiri Khan's Pineapple Cheese Cake Biscuit (87.4 and 43.7), Chon Buri's Bamboo Sticky Rice (109.3 and 54.7), and Lampang's Crispy Rice Cracker (149.3 and 74.7), respectively. Conclusion The study demonstrates that while Thai desserts exhibit a range of GI values, their GL values are uniformly high. It underscores the importance of disseminating GI and GL information to consumers, enabling them to make informed dietary choices and moderate their intake of these sugary delicacies.
Collapse
Affiliation(s)
- Nuttaphat Namjud
- Center of Excellence in Preventive and Integrative Medicine and Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sayamon Senaprom
- Center of Excellence in Preventive and Integrative Medicine and Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thunnicha Ondee
- Center of Excellence in Preventive and Integrative Medicine and Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Julia Heath
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Krit Pongpirul
- Center of Excellence in Preventive and Integrative Medicine and Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Bumrungrad International Hospital, Bangkok, Thailand
- Department of Infection Biology & Microbiomes, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
48
|
Song Q, Kikumoto A, Sun S, Mochizuki S, Oda H. High fat intake aggravates hyperlipidemia and suppresses fatty liver symptoms induced by a high-sucrose diet in rats. Food Funct 2024; 15:10516-10526. [PMID: 39365248 DOI: 10.1039/d4fo00863d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Overconsumption of sucrose or fat is widely acknowledged as a prominent feature of unhealthy dietary patterns. Both factors commonly co-occur and are recognized as hallmarks of the Western diet, which is an important contributor to non-communicative diseases. In this study, we investigated the hazards of high sucrose or fat intake, either alone or in combination. Wistar rats were divided into four groups and fed a control starch diet, high-sucrose diet, high-fat diet, or high-sucrose/fat diet for 30 days. High fat intake increased body weight and visceral and subcutaneous adipose tissue weights. Both high-sucrose and -fat diets were associated with increased plasma triglyceride and glucose levels, and high sucrose also elevated plasma cholesterol levels. The combination of high sucrose and fat synergistically elevated plasma triglyceride levels. The high-sucrose diet increased liver weight and hepatic total lipid and triglyceride levels, whereas this increase was suppressed by the high-fat diet. The high sucrose increased the mRNA levels of hepatic genes involved in fatty acid synthesis and transport (ACLY, ACACA, FAS, ELOVL6, SCD1, SREBP1, and CD36), whereas the high fat suppressed the high sucrose-induced expression of these genes. We observed that high sucrose and fat contents differently exerted their effects on hyperlipidemia and fatty liver. Furthermore, high fat aggravated hyperlipidemia and suppressed fatty liver induced by high sucrose.
Collapse
Affiliation(s)
- Qi Song
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan.
| | - Akari Kikumoto
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan.
| | - Shumin Sun
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan.
| | | | - Hiroaki Oda
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
49
|
Fu Y, Araki Y, Saito S, Nishitani S, Nishimura N, Mochizuki S, Oda H. High sucrose diet-induced abnormal lipid metabolism in mice is related to the dysbiosis of gut microbiota. Clin Nutr ESPEN 2024; 63:491-500. [PMID: 39018242 DOI: 10.1016/j.clnesp.2024.06.042] [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: 07/28/2023] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND & AIMS Excess sucrose intake induces metabolic syndrome. In human, abnormal lipids metabolism like obesity, hyperlipidemia and fatty liver are induced. However, excess sucrose causes different phenotypes in different species. Based on our previous study, excess sucrose induced fatty liver and hyperlipidemia in rats. The phenotypes and mechanism of abnormal lipid metabolism in mice is unclear. We investigated the different phenotypes in 5 strains of mice and the relationship between gut microbiome and abnormal lipid metabolism in C57BL/6N mice. METHODS We examined the effect of a high sucrose diet in 5 different strains of mice. Besides, to find out the relationship between gut microbiome and metabolic disorder induced by excess sucrose, C57BL/6N mice were fed with a high sucrose diet with or without antibiotics cocktail. RESULTS A high sucrose diet induced obesity and fatty liver in inbred mice, whereas did not induce hyperlipidemia in all strains of mice. Moreover, a high sucrose diet changed the composition of gut microbiota in C57BL/6N mice. Antibiotics treatment alleviated the abnormal lipid metabolism induced by high sucrose diet by changing the composition of gut short chain fatty acids. CONCLUSIONS These results indicates that the phenotypes of metabolic syndrome are influenced by genetic factors. Furthermore, the dysbiosis of gut microbiome caused by excess sucrose may contribute to the development of abnormal lipid metabolism via its metabolites.
Collapse
Affiliation(s)
- Yiying Fu
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan
| | - Yuki Araki
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan
| | - Shiori Saito
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan
| | - Shiori Nishitani
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan
| | - Naomichi Nishimura
- Academic Institute, College of Agriculture, Shizuoka University, Shizuoka 422-8529, Japan
| | | | - Hiroaki Oda
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
50
|
Noè R, Carrer A. Diet predisposes to pancreatic cancer through cellular nutrient sensing pathways. FEBS Lett 2024; 598:2470-2481. [PMID: 38886112 DOI: 10.1002/1873-3468.14959] [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: 03/27/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Pancreatic cancer is a lethal disease with limited effective treatments. A deeper understanding of its molecular mechanisms is crucial to reduce incidence and mortality. Epidemiological evidence suggests a link between diet and disease risk, though dietary recommendations for at-risk individuals remain debated. Here, we propose that cell-intrinsic nutrient sensing pathways respond to specific diet-derived cues to facilitate oncogenic transformation of pancreatic epithelial cells. This review explores how diet influences pancreatic cancer predisposition through nutrient sensing and downstream consequences for (pre-)cancer cell biology. We also examine experimental evidence connecting specific food intake to pancreatic cancer progression, highlighting nutrient sensing as a promising target for therapeutic development to mitigate disease risk.
Collapse
Affiliation(s)
- Roberta Noè
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Biology, University of Padova, Padua, Italy
| | - Alessandro Carrer
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Biology, University of Padova, Padua, Italy
| |
Collapse
|