1
|
Macur K, Roszkowska A, Czaplewska P, Miękus-Purwin N, Klejbor I, Moryś J, Bączek T. Pressure Cycling Technology Combined With MicroLC-SWATH Mass Spectrometry for the Analysis of Sex-Related Differences Between Male and Female Cerebella: A Promising Approach to Investigating Proteomics Differences in Psychiatric and Neurodegenerative Diseases. Proteomics Clin Appl 2024:e202400001. [PMID: 39205462 DOI: 10.1002/prca.202400001] [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: 01/03/2024] [Revised: 07/19/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Pressure cycling technology (PCT) coupled with data-independent sequential window acquisition of all theoretical mass spectra (SWATH-MS) can be a powerful tool for identifying and quantifying biomarkers (e.g., proteins) in complex biological samples. Mouse models are frequently used in brain studies, including those focusing on different neurodevelopmental and psychiatric disorders. More and more pieces of evidence have suggested that sex-related differences in the brain impact the rates, clinical manifestations, and therapy outcomes of these disorders. However, sex-based differences in the proteomic profiles of mouse cerebella have not been widely investigated. EXPERIMENTAL DESIGN In this pilot study, we evaluate the applicability of coupling PCT sample preparation with microLC-SWATH-MS analysis to map and identify differences in the proteomes of two female and two male mice cerebellum samples. RESULTS We identified and quantified 174 proteins in mice cerebella. A comparison of the proteomic profiles revealed that the levels of 11 proteins in the female and male mice cerebella varied significantly. CONCLUSIONS AND CLINICAL RELEVANCE Although this study utilizes a small sample, our results indicate that the studied male and female mice cerebella possessed differing proteome compositions, mainly with respect to energy metabolism processes.
Collapse
Affiliation(s)
- Katarzyna Macur
- Core Facility Laboratories, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Roszkowska
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Paulina Czaplewska
- Core Facility Laboratories, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Miękus-Purwin
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Ilona Klejbor
- Department of Anatomy, Institute of Medical Sciences, Jan Kochanowski University, Kielce, Poland
| | - Janusz Moryś
- Department of Normal Anatomy, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
- Department of Nursing and Medical Rescue, Institute of Health Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| |
Collapse
|
2
|
Hadzi-Petrushev N, Stojchevski R, Jakimovska A, Stamenkovska M, Josifovska S, Stamatoski A, Sazdova I, Sopi R, Kamkin A, Gagov H, Mladenov M, Avtanski D. GLUT5-overexpression-related tumorigenic implications. Mol Med 2024; 30:114. [PMID: 39107723 PMCID: PMC11304774 DOI: 10.1186/s10020-024-00879-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Glucose transporter 5 (GLUT5) overexpression has gained increasing attention due to its profound implications for tumorigenesis. This manuscript provides a comprehensive overview of the key findings and implications associated with GLUT5 overexpression in cancer. GLUT5 has been found to be upregulated in various cancer types, leading to alterations in fructose metabolism and enhanced glycolysis, even in the presence of oxygen, a hallmark of cancer cells. This metabolic shift provides cancer cells with an alternative energy source and contributes to their uncontrolled growth and survival. Beyond its metabolic roles, recent research has unveiled additional aspects of GLUT5 in cancer biology. GLUT5 overexpression appears to play a critical role in immune evasion mechanisms, which further worsens tumor progression and complicates therapeutic interventions. This dual role of GLUT5 in both metabolic reprogramming and immune modulation highlights its significance as a potential diagnostic marker and therapeutic target. Understanding the molecular mechanisms driving GLUT5 overexpression is crucial for developing targeted therapeutic strategies that can disrupt the unique vulnerabilities of GLUT5-overexpressing cancer cells. This review emphasizes the complexities surrounding GLUT5's involvement in cancer and underscores the pressing need for continued research to unlock its potential as a diagnostic biomarker and therapeutic target, ultimately improving cancer management and patient outcomes.
Collapse
Affiliation(s)
- Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY, 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Anastasija Jakimovska
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
| | - Mimoza Stamenkovska
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
| | - Slavica Josifovska
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
| | - Aleksandar Stamatoski
- Faculty of Dental Medicine, University Clinic for Maxillofacial Surgery in Skopje, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
| | - Iliyana Sazdova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University 'St. Kliment Ohridski', Sofia, 1504, Bulgaria
| | - Ramadan Sopi
- Faculty of Medicine, University of Prishtina, Prishtina, 10 000, Kosovo
| | - Andre Kamkin
- Institute of Physiology of the Federal State Autonomous Educational Institution of Higher Education "N.I. Pirogov Russian National Research Medical University" Ministry of Health, Moscow, Russian Federation
| | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University 'St. Kliment Ohridski', Sofia, 1504, Bulgaria
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, Skopje, 1000, North Macedonia
- Institute of Physiology of the Federal State Autonomous Educational Institution of Higher Education "N.I. Pirogov Russian National Research Medical University" Ministry of Health, Moscow, Russian Federation
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY, 10022, USA.
- Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
3
|
Li Y, Jiang T, Du M, He S, Huang N, Cheng B, Yan C, Tang W, Gao W, Guo H, Li Q, Wang Q. Ketohexokinase-dependent metabolism of cerebral endogenous fructose in microglia drives diabetes-associated cognitive dysfunction. Exp Mol Med 2023; 55:2417-2432. [PMID: 37907746 PMCID: PMC10689812 DOI: 10.1038/s12276-023-01112-y] [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: 02/21/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 11/02/2023] Open
Abstract
Dementia, as an advanced diabetes-associated cognitive dysfunction (DACD), has become the second leading cause of death among diabetes patients. Given that little guidance is currently available to address the DACD process, it is imperative to understand the underlying mechanisms and screen out specific therapeutic targets. The excessive endogenous fructose produced under high glucose conditions can lead to metabolic syndrome and peripheral organ damage. Although generated by the brain, the role of endogenous fructose in the exacerbation of cognitive dysfunction is still unclear. Here, we performed a comprehensive study on leptin receptor-deficient T2DM mice and their littermate m/m mice and revealed that 24-week-old db/db mice had cognitive dysfunction and excessive endogenous fructose metabolism in the hippocampus by multiomics analysis and further experimental validation. We found that the rate-limiting enzyme of fructose metabolism, ketohexokinase, is primarily localized in microglia. It is upregulated in the hippocampus of db/db mice, which enhances mitochondrial damage and reactive oxygen species production by promoting nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) expression and mitochondrial translocation. Inhibiting fructose metabolism via ketohexokinase depletion reduces microglial activation, leading to the restoration of mitochondrial homeostasis, recovery of structural synaptic plasticity, improvement of CA1 pyramidal neuron electrophysiology and alleviation of cognitive dysfunction. Our findings demonstrated that enhanced endogenous fructose metabolism in microglia plays a dominant role in diabetes-associated cognitive dysfunction and could become a potential target for DACD.
Collapse
Affiliation(s)
- Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Tao Jiang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Shuxuan He
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Ning Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Wenxin Tang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Hongyan Guo
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qiao Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Wang Z, Lipshutz A, Liu ZL, Trzeciak AJ, Miranda IC, Martínez de la Torre C, Schild T, Lazarov T, Rojas WS, Saavedra PHV, Romero-Pichardo JE, Baako A, Geissmann F, Faraco G, Gan L, Etchegaray JI, Lucas CD, Parkhurst CN, Zeng MY, Keshari KR, Perry JSA. Early life high fructose exposure disrupts microglia function and impedes neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553242. [PMID: 37645894 PMCID: PMC10462086 DOI: 10.1101/2023.08.14.553242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Despite the success of fructose as a low-cost food additive, recent epidemiological evidence suggests that high fructose consumption by pregnant mothers or during adolescence is associated with disrupted neurodevelopment 1-7 . An essential step in appropriate mammalian neurodevelopment is the synaptic pruning and elimination of newly-formed neurons by microglia, the central nervous system's (CNS) resident professional phagocyte 8-10 . Whether early life high fructose consumption affects microglia function and if this directly impacts neurodevelopment remains unknown. Here, we show that both offspring born to dams fed a high fructose diet and neonates exposed to high fructose exhibit decreased microglial density, increased uncleared apoptotic cells, and decreased synaptic pruning in vivo . Importantly, deletion of the high affinity fructose transporter SLC2A5 (GLUT5) in neonates completely reversed microglia dysfunction, suggesting that high fructose directly affects neonatal development. Mechanistically, we found that high fructose treatment of both mouse and human microglia suppresses synaptic pruning and phagocytosis capacity which is fully reversed in GLUT5-deficient microglia. Using a combination of in vivo and in vitro nuclear magnetic resonance- and mass spectrometry-based fructose tracing, we found that high fructose drives significant GLUT5-dependent fructose uptake and catabolism, rewiring microglia metabolism towards a hypo-phagocytic state. Importantly, mice exposed to high fructose as neonates exhibited cognitive defects and developed anxiety-like behavior which were rescued in GLUT5-deficient animals. Our findings provide a mechanistic explanation for the epidemiological observation that early life high fructose exposure is associated with increased prevalence of adolescent anxiety disorders.
Collapse
|
5
|
Cargnin-Carvalho A, da Silva MR, Costa AB, Engel NA, Farias BX, Bressan JB, Backes KM, de Souza F, da Rosa N, de Oliveira Junior AN, Goldim MPDS, Correa MEAB, Venturini LM, Fortunato JJ, Prophiro JS, Petronilho F, Silveira PCL, Ferreira GK, Rezin GT. High concentrations of fructose cause brain damage in mice. Biochem Cell Biol 2023; 101:313-325. [PMID: 36947832 DOI: 10.1139/bcb-2022-0088] [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: 03/24/2023] Open
Abstract
Excessive fructose consumption is associated with the incidence of obesity and systemic inflammation, resulting in increased oxidative damage and failure to the function of brain structures. Thus, we hypothesized that fructose consumption will significantly increase inflammation, oxidative damage, and mitochondrial dysfunction in the mouse brain and, consequently, memory damage. The effects of different fructose concentrations on inflammatory and biochemical parameters in the mouse brain were evaluated. Male Swiss mice were randomized into four groups: control, with exclusive water intake, 5%, 10%, and 20% fructose group. The 10% and 20% fructose groups showed an increase in epididymal fat, in addition to higher food consumption. Inflammatory markers were increased in epididymal fat and in some brain structures. In the evaluation of oxidative damage, it was possible to observe significant increases in the hypothalamus, prefrontal cortex, and hippocampus. In the epididymal fat and in the prefrontal cortex, there was a decrease in the activity of the mitochondrial respiratory chain complexes and an increase in the striatum. Furthermore, short memory was impaired in the 10% and 20% groups but not long memory. In conclusion, excess fructose consumption can cause fat accumulation, inflammation, oxidative damage, and mitochondrial dysfunction, which can damage brain structures and consequently memory.
Collapse
Affiliation(s)
- Anderson Cargnin-Carvalho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Ana Beatriz Costa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Nicole Alessandra Engel
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Bianca Xavier Farias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Joice Benedet Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Kassiane Mathiola Backes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Francielly de Souza
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Naiana da Rosa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Aloir Neri de Oliveira Junior
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariana Pereira de Souza Goldim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | | | - Ligia Milanez Venturini
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | - Jucélia Jeremias Fortunato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Josiane Somariva Prophiro
- Immunoparasitology Research Group, Postgraduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Fabrícia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | | | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| |
Collapse
|
6
|
Johnson RJ, Lee SMK, Sánchez-Lozada LG, Kanbay M, Bansal A, Tolan DR, Bjornstad P, Lanaspa MA, Maesaka J. Fructose: A New Variable to Consider in SIADH and the Hyponatremia Associated With Long-Distance Running? Am J Kidney Dis 2023; 82:105-112. [PMID: 36940740 PMCID: PMC10330032 DOI: 10.1053/j.ajkd.2023.01.443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/01/2023] [Indexed: 03/23/2023]
Abstract
Fructose has recently been proposed to stimulate vasopressin secretion in humans. Fructose-induced vasopressin secretion is not only postulated to result from ingestion of fructose-containing drinks but may also occur from endogenous fructose production via activation of the polyol pathway. This raises the question of whether fructose might be involved in some cases of vasopressin-induced hyponatremia, especially in situations where the cause is not fully known such as in the syndrome of inappropriate secretion of diuretic hormone (SIADH) and exercise-associated hyponatremia, which has been observed in marathon runners. Here we discuss the new science of fructose and vasopressin, and how it may play a role in some of these conditions, as well as in the complications associated with rapid treatment (such as the osmotic demyelination syndrome). Studies to test the role of fructose could provide new pathophysiologic insights as well as novel potential treatment strategies for these common conditions.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado.
| | | | | | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, School of Medicine, Koc University, Istanbul, Turkey
| | - Anip Bansal
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Dean R Tolan
- Biology Department, Boston University, Boston Massachusetts
| | - Petter Bjornstad
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado; Section of Endocrinology, Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - John Maesaka
- Department of Medicine and Division of Nephrology and Hypertension, NYU Langone Hospitals, Mineola, New York
| |
Collapse
|
7
|
Johnson RJ, Tolan DR, Bredesen D, Nagel M, Sánchez-Lozada LG, Fini M, Burtis S, Lanaspa MA, Perlmutter D. Could Alzheimer's disease be a maladaptation of an evolutionary survival pathway mediated by intracerebral fructose and uric acid metabolism? Am J Clin Nutr 2023; 117:455-466. [PMID: 36774227 PMCID: PMC10196606 DOI: 10.1016/j.ajcnut.2023.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
An important aspect of survival is to assure enough food, water, and oxygen. Here, we describe a recently discovered response that favors survival in times of scarcity, and it is initiated by either ingestion or production of fructose. Unlike glucose, which is a source for immediate energy needs, fructose metabolism results in an orchestrated response to encourage food and water intake, reduce resting metabolism, stimulate fat and glycogen accumulation, and induce insulin resistance as a means to reduce metabolism and preserve glucose supply for the brain. How this survival mechanism affects brain metabolism, which in a resting human amounts to 20% of the overall energy demand, is only beginning to be understood. Here, we review and extend a previous hypothesis that this survival mechanism has a major role in the development of Alzheimer's disease and may account for many of the early features, including cerebral glucose hypometabolism, mitochondrial dysfunction, and neuroinflammation. We propose that the pathway can be engaged in multiple ways, including diets high in sugar, high glycemic carbohydrates, and salt. In summary, we propose that Alzheimer's disease may be the consequence of a maladaptation to an evolutionary-based survival pathway and what had served to enhance survival acutely becomes injurious when engaged for extensive periods. Although more studies are needed on the role of fructose metabolism and its metabolite, uric acid, in Alzheimer's disease, we suggest that both dietary and pharmacologic trials to reduce fructose exposure or block fructose metabolism should be performed to determine whether there is potential benefit in the prevention, management, or treatment of this disease.
Collapse
Affiliation(s)
- Richard J Johnson
- Department of Medicine, Rocky Mountain VA Medical Center, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| | - Dean R Tolan
- Biology Department, Boston University, Boston, MA, USA
| | - Dale Bredesen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Maria Nagel
- Department of Neurology, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Laura G Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Mehdi Fini
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | | - Miguel A Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | |
Collapse
|
8
|
Vratarić M, Šenk V, Bursać B, Gligorovska L, Ignjatović D, Kovačević S, Veličković N, Djordjevic A. Fructose diet ameliorate effects of macrophage migration inhibitory factor deficiency on prefrontal cortex inflammation, neural plasticity, and behavior in male mice. Biofactors 2023; 49:90-107. [PMID: 34767656 DOI: 10.1002/biof.1802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that represents a link between diet-induced inflammation and insulin resistance. Our aim was to examine whether fructose diet affects inflammation and insulin signaling in the prefrontal cortex (PFC) of Mif knockout mice (MIF-KO), and their possible link to neural plasticity and behavior. We analyzed nuclear factor κB (NF-κB) and glucocorticoid signaling, expression of F4/80, IL-1β, TNF-α, TLR-4, MyD88, arginase 1 (Arg-1), mannose receptor (Mrc-1), and leukemia inhibitory factor (Lif) to assess inflammation in the PFC of C57/BL6J and MIF-KO mice consuming 20% fructose solution for 9 weeks. Insulin receptor (IR), IRS-1 serine phosphorylations (307 and 1101) and activity of PKCα, Akt, GSK-3β and AMPKα were used to analyze insulin signaling. Brain-derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1) mRNA levels, together with synapthophysin and PSD-95 protein level and calcium calmodulin-dependent kinase 2 (CaMKII) activity, were used as plasticity markers. Behavior was examined in elevated plus maze, light dark box and novel object recognition test. The results showed concomitant increase of Tnf-α, Tlr-4, MyD88 and M2 microglia markers (Arg-1, Mrc-1, Lif) in the PFC of MIF-KO, paralleled with unchanged glucocorticoid and insulin signaling. Increase of BDNF and IGF-1 was paralleled with increased CaMKII activity, decreased PSD-95 protein level, anxiogenic behavior, and impaired memory in MIF-KO mice. Fructose feeding restored these parameters in the PFC to the control level and mitigated behavioral changes, suggesting that ameliorating effects of fructose on neuroinflammation and behavior depend on the presence of MIF.
Collapse
Affiliation(s)
- Miloš Vratarić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vladimir Šenk
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Biljana Bursać
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ljupka Gligorovska
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Djurdjica Ignjatović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
9
|
Boyle AJ, Murrell E, Tong J, Schifani C, Narvaez A, Wuest M, West F, Wuest F, Vasdev N. PET Imaging of Fructose Metabolism in a Rodent Model of Neuroinflammation with 6-[ 18F]fluoro-6-deoxy-D-fructose. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238529. [PMID: 36500626 PMCID: PMC9736258 DOI: 10.3390/molecules27238529] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Fluorine-18 labeled 6-fluoro-6-deoxy-D-fructose (6-[18F]FDF) targets the fructose-preferred facilitative hexose transporter GLUT5, which is expressed predominantly in brain microglia and activated in response to inflammatory stimuli. We hypothesize that 6-[18F]FDF will specifically image microglia following neuroinflammatory insult. 6-[18F]FDF and, for comparison, [18F]FDG were evaluated in unilateral intra-striatal lipopolysaccharide (LPS)-injected male and female rats (50 µg/animal) by longitudinal dynamic PET imaging in vivo. In LPS-injected rats, increased accumulation of 6-[18F]FDF was observed at 48 h post-LPS injection, with plateaued uptake (60-120 min) that was significantly higher in the ipsilateral vs. contralateral striatum (0.985 ± 0.047 and 0.819 ± 0.033 SUV, respectively; p = 0.002, n = 4M/3F). The ipsilateral-contralateral difference in striatal 6-[18F]FDF uptake expressed as binding potential (BPSRTM) peaked at 48 h (0.19 ± 0.11) and was significantly decreased at one and two weeks. In contrast, increased [18F]FDG uptake in the ipsilateral striatum was highest at one week post-LPS injection (BPSRTM = 0.25 ± 0.06, n = 4M). Iba-1 and GFAP immunohistochemistry confirmed LPS-induced activation of microglia and astrocytes, respectively, in ipsilateral striatum. This proof-of-concept study revealed an early response of 6-[18F]FDF to neuroinflammatory stimuli in rat brain. 6-[18F]FDF represents a potential PET radiotracer for imaging microglial GLUT5 density in brain with applications in neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Amanda J. Boyle
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Christin Schifani
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Andrea Narvaez
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Melinda Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
| | - Frederick West
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| |
Collapse
|
10
|
Tigchelaar C, van Zuylen ML, Hulst AH, Preckel B, van Beek AP, Kema IP, Hermanides J, Absalom AR. Elevated cerebrospinal fluid glucose levels and diabetes mellitus are associated with activation of the neurotoxic polyol pathway. Diabetologia 2022; 65:1098-1107. [PMID: 35380232 PMCID: PMC9174140 DOI: 10.1007/s00125-022-05693-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/17/2022] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS During hyperglycaemia, some glucose bypasses glycolysis and is metabolised via the potentially neurotoxic polyol pathway, in which glucose is metabolised to sorbitol and fructose. Increased polyol concentrations have been demonstrated in the cerebrospinal fluid (CSF) of neurological patients with and without diabetes mellitus. However, polyol levels in patients without evident neurological abnormalities have not been investigated so far. The aim of this study was to determine CSF polyol concentrations in patients without major neurological disease with normal or elevated CSF glucose concentrations. METHODS This observational cohort study used CSF and plasma analyses, as well as clinical data, from 30 participants of the Anaesthetic Biobank of Cerebrospinal Fluid study. Biomaterial was collected from adult patients scheduled for elective surgery under spinal anaesthesia. CSF polyol concentrations were measured by GC/flame ionisation detector in ten patients with normal CSF glucose levels (group 1), ten patients with elevated CSF glucose levels (group 2) and ten patients with elevated CSF glucose levels and type 2 diabetes (group 3). We compared the concentrations of plasma glucose, CSF glucose, sorbitol and fructose, and CSF polyol/glucose ratios between the three groups, and determined the correlation between plasma glucose levels and CSF glucose, sorbitol and fructose levels. RESULTS Groups 2 and 3 had significantly higher CSF fructose levels compared with group 1 (p=0.036 and p<0.001, respectively). Group 3 showed significant differences compared with groups 1 and 2 for CSF sorbitol (p<0.001 and 0.036, respectively). Moreover, patients with diabetes had a significantly higher CSF sorbitol/glucose ratio compared with patients without diabetes. There was a strong positive correlation between plasma glucose and CSF glucose, sorbitol and fructose. Finally, age, sex, CSF/plasma albumin ratio and preoperative cognitive function scores were significantly correlated with plasma glucose and CSF glucose, sorbitol and fructose levels. CONCLUSIONS/INTERPRETATION Hyperglycaemia causes a proportional increase in polyol concentrations in CSF of patients without major neurological disease. Furthermore, this study provides the first indication of upregulation of the cerebral polyol pathway in patients with diabetes without evident neurological abnormalities.
Collapse
Affiliation(s)
- Celien Tigchelaar
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Mark L van Zuylen
- Department of Anaesthesiology, Amsterdam UMC - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham H Hulst
- Department of Anaesthesiology, Amsterdam UMC - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Intensive Care, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Benedikt Preckel
- Department of Anaesthesiology, Amsterdam UMC - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - André P van Beek
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jeroen Hermanides
- Department of Anaesthesiology, Amsterdam UMC - Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anthony R Absalom
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
11
|
Kageyama I, Yamada H, Munetsuna E, Yamazaki M, Ando Y, Mizuno G, Fujii R, Nouchi Y, Wakasugi T, Sakakibara T, Teshigawara A, Ishikawa H, Shimono Y, Suzuki K, Hashimoto S, Ohashi K. Differential effects of excess high-fructose corn syrup on the DNA methylation of hippocampal neurotrophic factor in childhood and adolescence. PLoS One 2022; 17:e0270144. [PMID: 35714129 PMCID: PMC9205497 DOI: 10.1371/journal.pone.0270144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Consumption of fructose-containing beverages such as high-fructose corn syrup (HFCS) is increasing, raising concerns about the negative effects of excessive fructose intake. A recent report indicated that excess HFCS intake impairs hippocampal function. In this study, we focused on neurotrophic factors (NFs) in the hippocampus from the viewpoint of epigenetics to clarify the adverse effects of fructose. We analyzed the effects of HFCS intake on hippocampal function in three age categories: childhood and adolescence (postnatal day (PD) 21–60), young adulthood (PD60-100), and late adulthood (PD100-140). For the experiments, male Sprague-Dawley rats were divided into three age categories, the control group was received distilled water and the HFCS group was received 20% HFCS solution for 40 days in each period. We analyzed mRNA and protein levels for qPCR and western blotting, respectively, of a hippocampal NF, brain-derived neurotrophic factor (Bdnf). HFCS consumption reduced hippocampal Bdnf mRNA and protein expressions in childhood and adolescence. Moreover, pyrosequencing assays revealed increased DNA methylation at the Bdnf promoter in childhood and adolescence. This Bdnf levels reduction may be due to hypermethylation of the promoter regions. It should be noted that this phenomenon was observed only in childhood and adolescence fructose consumption. Our results indicate that the sensitivity of the hippocampus to fructose may vary with age. This study provides insight into the adverse effects of excessive HFCS consumption on the hippocampus in children.
Collapse
Affiliation(s)
- Itsuki Kageyama
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- * E-mail:
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Mirai Yamazaki
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Takamatsu, Kagawa, Japan
| | - Yoshitaka Ando
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Genki Mizuno
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yuki Nouchi
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Takuya Wakasugi
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tomohide Sakakibara
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Atsushi Teshigawara
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroaki Ishikawa
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yohei Shimono
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Shuji Hashimoto
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Koji Ohashi
- Department of Informative Clinical Medicine, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| |
Collapse
|
12
|
Parıldı E, Kola O, Özcan BD, Akkaya MR, Dikkaya E. Recombinant D‐tagatose 3‐epimerase production and converting fructose into allulose. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.15508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Erva Parıldı
- Department of Food Engineering, Faculty of Engineering Adana Alparslan Türkeş Science and Technology University Sarıçam Turkey
| | - Osman Kola
- Department of Food Engineering, Faculty of Engineering Adana Alparslan Türkeş Science and Technology University Sarıçam Turkey
| | - Bahri Devrim Özcan
- Department of Animal Science, Faculty of Agriculture Çukurova University Sarıçam Turkey
| | - Murat Reis Akkaya
- Department of Food Engineering, Faculty of Engineering Adana Alparslan Türkeş Science and Technology University Sarıçam Turkey
| | - Elif Dikkaya
- Department of Animal Science, Faculty of Agriculture Çukurova University Sarıçam Turkey
| |
Collapse
|
13
|
Metabolites and Biomarker Compounds of Neurodegenerative Diseases in Cerebrospinal Fluid. Metabolites 2022; 12:metabo12040343. [PMID: 35448530 PMCID: PMC9031591 DOI: 10.3390/metabo12040343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 04/11/2022] [Indexed: 12/25/2022] Open
Abstract
Despite recent advances in diagnostic procedures for neurological disorders, it is still difficult to definitively diagnose some neurodegenerative diseases without neuropathological examination of autopsied brain tissue. As pathological processes in the brain are frequently reflected in the components of cerebrospinal fluid (CSF), CSF samples are sometimes useful for diagnosis. After CSF is secreted from the choroid plexus epithelial cells in the ventricles, some flows in the brain, some is mixed with intracerebral interstitial fluid, and some is excreted through two major drainage pathways, i.e., the intravascular periarterial drainage pathway and the glymphatic system. Accordingly, substances produced by metabolic and pathological processes in the brain may be detectable in CSF. Many papers have reported changes in the concentration of substances in the CSF of patients with metabolic and neurological disorders, some of which can be useful biomarkers of the disorders. In this paper, we show the significance of glucose- and neurotransmitter-related CSF metabolites, considering their transporters in the choroid plexus; summarize the reported candidates of CSF biomarkers for neurodegenerative diseases, including amyloid-β, tau, α-synuclein, microRNAs, and mitochondrial DNA; and evaluate their potential as efficient diagnostic tools.
Collapse
|
14
|
Kareva I, Brown JS. Estrogen as an Essential Resource and the Coexistence of ER+ and ER– Cancer Cells. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.673082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diagnosis of estrogen sensitivity in breast cancer is largely predicated on the ratio of ER+ and ER– cancer cells obtained from biopsies. Estrogen is a growth factor necessary for cell survival and division. It can also be thought of as an essential resource that can act in association with other nutrients, glucose, glutamine, fatty acids, amino acids, etc. All of these nutrients, collectively or individually, may limit the growth of the cancer cells (Liebig’s Law of the Minimum). Here we model estrogen susceptibility in breast cancer as a consumer-resource interaction: ER+ cells require both estrogen and glucose as essential resources, whereas ER– only require the general resource. The model predicts that when estrogen is the limiting factor, other nutrients may go unconsumed and available at higher levels, thus permitting the invasion of ER– cells. Conversely, when ER– cells are less efficient on glucose than ER+ cells, then ER– cells limited by glucose may be susceptible to invasion by ER+ cells, provided that sufficient levels of estrogen are available. ER+ cells will outcompete ER– cells when estrogen is abundant, resulting in low concentrations of interstitial glucose within the tumor. In the absence of estrogen, ER– cells will outcompete ER+ cells, leaving a higher concentration of interstitial glucose. At intermediate delivery rates of estrogen and glucose, ER+ and ER– cells are predicted to coexist. In modeling the dynamics of cells in the same tumor with different resource requirements, we can apply concepts and terms familiar to many ecologists. These include: resource supply points, R∗, ZNGI (zero net growth isoclines), resource depletion, and resource uptake rates. Based on the circumstances favoring ER+ vs. ER– breast cancer, we use the model to explore the consequences of therapeutic regimens that may include hormonal therapies, possible roles of diet in changing cancer cell composition, and potential for evolutionarily informed therapies. More generally, the model invites the viewpoint that cancer’s eco-evolutionary dynamics are a consumer-resource interaction, and that other growth factors such as EGFR or androgens may be best viewed as essential resources within these dynamics.
Collapse
|
15
|
Neural mechanisms underlying the role of fructose in overfeeding. Neurosci Biobehav Rev 2021; 128:346-357. [PMID: 34182019 DOI: 10.1016/j.neubiorev.2021.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022]
Abstract
Fructose consumption has been linked with metabolic syndrome and obesity. Fructose-based sweeteners like high fructose corn syrup taste sweeter, improve food palatability, and are increasingly prevalent in our diet. The increase in fructose consumption precedes the rise in obesity and is a contributing driver to the obesity epidemic worldwide. The role of dietary fructose in obesity can be multifactorial by promoting visceral adiposity, hypertension, and insulin resistance. Interestingly, one emergent finding from human and animal studies is that dietary fructose promotes overfeeding. As the brain is a critical regulator of food intake, we reviewed the evidence that fructose can act in the brain and elucidated the major brain systems underlying fructose-induced overfeeding. We found that fructose acts on multiple interdependent brain systems to increase orexigenic drive and the incentive salience of food while decreasing the latency between food bouts and reducing cognitive control to disinhibit feeding. We concluded that the collective actions of fructose may promote feeding behavior by producing a hunger-like state in the brain.
Collapse
|
16
|
Shi YN, Liu YJ, Xie Z, Zhang WJ. Fructose and metabolic diseases: too much to be good. Chin Med J (Engl) 2021; 134:1276-1285. [PMID: 34010200 PMCID: PMC8183764 DOI: 10.1097/cm9.0000000000001545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Excessive consumption of fructose, the sweetest of all naturally occurring carbohydrates, has been linked to worldwide epidemics of metabolic diseases in humans, and it is considered an independent risk factor for cardiovascular diseases. We provide an overview about the features of fructose metabolism, as well as potential mechanisms by which excessive fructose intake is associated with the pathogenesis of metabolic diseases both in humans and rodents. To accomplish this aim, we focus on illuminating the cellular and molecular mechanisms of fructose metabolism as well as its signaling effects on metabolic and cardiovascular homeostasis in health and disease, highlighting the role of carbohydrate-responsive element-binding protein in regulating fructose metabolism.
Collapse
Affiliation(s)
- Ya-Nan Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Weiping J. Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
17
|
Liang RJ, Taylor S, Nahiyaan N, Song J, Murphy CJ, Dantas E, Cheng S, Hsu TW, Ramsamooj S, Grover R, Hwang SK, Ngo B, Cantley LC, Rhee KY, Goncalves MD. GLUT5 (SLC2A5) enables fructose-mediated proliferation independent of ketohexokinase. Cancer Metab 2021; 9:12. [PMID: 33762003 PMCID: PMC7992954 DOI: 10.1186/s40170-021-00246-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Fructose is an abundant source of carbon and energy for cells to use for metabolism, but only certain cell types use fructose to proliferate. Tumor cells that acquire the ability to metabolize fructose have a fitness advantage over their neighboring cells, but the proteins that mediate fructose metabolism in this context are unknown. Here, we investigated the determinants of fructose-mediated cell proliferation. METHODS Live cell imaging and crystal violet assays were used to characterize the ability of several cell lines (RKO, H508, HepG2, Huh7, HEK293T (293T), A172, U118-MG, U87, MCF-7, MDA-MB-468, PC3, DLD1 HCT116, and 22RV1) to proliferate in fructose (i.e., the fructolytic ability). Fructose metabolism gene expression was determined by RT-qPCR and western blot for each cell line. A positive selection approach was used to "train" non-fructolytic PC3 cells to utilize fructose for proliferation. RNA-seq was performed on parental and trained PC3 cells to find key transcripts associated with fructolytic ability. A CRISPR-cas9 plasmid containing KHK-specific sgRNA was transfected in 293T cells to generate KHK-/- cells. Lentiviral transduction was used to overexpress empty vector, KHK, or GLUT5 in cells. Metabolic profiling was done with seahorse metabolic flux analysis as well as LC/MS metabolomics. Cell Titer Glo was used to determine cell sensitivity to 2-deoxyglucose in media containing either fructose or glucose. RESULTS We found that neither the tissue of origin nor expression level of any single gene related to fructose catabolism determine the fructolytic ability. However, cells cultured chronically in fructose can develop fructolytic ability. SLC2A5, encoding the fructose transporter, GLUT5, was specifically upregulated in these cells. Overexpression of GLUT5 in non-fructolytic cells enabled growth in fructose-containing media across cells of different origins. GLUT5 permitted fructose to flux through glycolysis using hexokinase (HK) and not ketohexokinase (KHK). CONCLUSIONS We show that GLUT5 is a robust and generalizable driver of fructose-dependent cell proliferation. This indicates that fructose uptake is the limiting factor for fructose-mediated cell proliferation. We further demonstrate that cellular proliferation with fructose is independent of KHK.
Collapse
Affiliation(s)
- Roger J Liang
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Samuel Taylor
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-I MD-PhD program, New York, NY, 10065, USA
| | - Navid Nahiyaan
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Junho Song
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Charles J Murphy
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shuyuan Cheng
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ting-Wei Hsu
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shakti Ramsamooj
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Rahul Grover
- Weill Cornell Medical College, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Seo-Kyoung Hwang
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Bryan Ngo
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kyu Y Rhee
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
18
|
Mazzoli A, Spagnuolo MS, Nazzaro M, Gatto C, Iossa S, Cigliano L. Fructose Removal from the Diet Reverses Inflammation, Mitochondrial Dysfunction, and Oxidative Stress in Hippocampus. Antioxidants (Basel) 2021; 10:487. [PMID: 33804637 PMCID: PMC8003595 DOI: 10.3390/antiox10030487] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 11/16/2022] Open
Abstract
Young age is often characterized by high consumption of processed foods and fruit juices rich in fructose, which, besides inducing a tendency to become overweight, can promote alterations in brain function. The aim of this study was therefore to (a) clarify brain effects resulting from fructose consumption in juvenile age, a critical phase for brain development, and (b) verify whether these alterations can be rescued after removing fructose from the diet. Young rats were fed a fructose-rich or control diet for 3 weeks. Fructose-fed rats were then fed a control diet for a further 3 weeks. We evaluated mitochondrial bioenergetics by high-resolution respirometry in the hippocampus, a brain area that is critically involved in learning and memory. Glucose transporter-5, fructose and uric acid levels, oxidative status, and inflammatory and synaptic markers were investigated by Western blotting and spectrophotometric or enzyme-linked immunosorbent assays. A short-term fructose-rich diet induced mitochondrial dysfunction and oxidative stress, associated with an increased concentration of inflammatory markers and decreased Neurofilament-M and post-synaptic density protein 95. These alterations, except for increases in haptoglobin and nitrotyrosine, were recovered by returning to a control diet. Overall, our results point to the dangerous effects of excessive consumption of fructose in young age but also highlight the effect of partial recovery by switching back to a control diet.
Collapse
Affiliation(s)
- Arianna Mazzoli
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Naples, Italy; (A.M.); (M.N.); (C.G.); (S.I.)
| | - Maria Stefania Spagnuolo
- Department of Bio-Agrofood Science, Institute for the Animal Production System, National Research Council, 80147 Naples, Italy;
| | - Martina Nazzaro
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Naples, Italy; (A.M.); (M.N.); (C.G.); (S.I.)
| | - Cristina Gatto
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Naples, Italy; (A.M.); (M.N.); (C.G.); (S.I.)
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Naples, Italy; (A.M.); (M.N.); (C.G.); (S.I.)
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, 80126 Naples, Italy; (A.M.); (M.N.); (C.G.); (S.I.)
| |
Collapse
|
19
|
Gomez-Pinilla F, Cipolat RP, Royes LFF. Dietary fructose as a model to explore the influence of peripheral metabolism on brain function and plasticity. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166036. [PMID: 33508421 DOI: 10.1016/j.bbadis.2020.166036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
High consumption of fructose has paralleled an explosion in metabolic disorders including obesity and type 2 diabetes. Even more problematic, sustained consumption of fructose is perceived as a threat for brain function and development of neurological disorders. The action of fructose on peripheral organs is an excellent model to understand how systemic physiology impacts the brain. Given the recognized action of fructose on liver metabolism, here we discuss mechanisms by which fructose can impact the brain by interacting with liver and other organs. The interaction between peripheral and central mechanisms is a suitable target to reduce the pathophysiological consequences of neurological disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Neurosurgery, UCLA Brain Injury Research Center, University of California Los Angeles, USA; Department of Integrative Biology and Physiology, UCLA Brain Injury Research Center, University of California Los Angeles, USA.
| | - Rafael Parcianello Cipolat
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Luiz Fernando Freire Royes
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| |
Collapse
|
20
|
Song S, Niu M, Liang Q, Wang L, Min H, Peng Y, Wang H, Gao Q. Statin Treatment Induced a Lipogenic Expression Hierarchical Network Centered by SREBF2 in the Liver. Front Endocrinol (Lausanne) 2021; 12:573824. [PMID: 34349727 PMCID: PMC8326809 DOI: 10.3389/fendo.2021.573824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Statin treatment is a major prevention treatment for hypercholesterolemia and the management of patients with increased risk of cardiovascular disease (CVD) due to its protective effects. However, its long-term safety was questioned regarding its potential role in new-onset type 2 diabetes mellitus, and its effect on gene regulation in the liver is not yet fully understood. By reanalyzing the transcriptome of the livers of patients with obesity and hypercholesterolemia, this study shows a multiple module organization that is related to various clinical metabolic parameters and identified an expression hierarchical network involving cholesterol and fatty acid syntheses in the liver of statin-treated patients. The key genes of the network were validated by QPCR in the hepatocytes upon statin treatment. The upregulation of the key enzymes involving the synthesis of Acetyl-CoA and the induction of gentle global acetylation of pan-protein and histone H4 in hepatocytes were observed. The study provides an overall view of the statin effect on transcriptional and post-transcriptional regulation of genes in the liver.
Collapse
Affiliation(s)
- Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Mengyuan Niu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Lei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Haiyan Min
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yuming Peng
- Department of General Practice of Central Hospital of Karamay, Karamay, China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Qian Gao, ; Hongwei Wang,
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Qian Gao, ; Hongwei Wang,
| |
Collapse
|
21
|
Sweet but Bitter: Focus on Fructose Impact on Brain Function in Rodent Models. Nutrients 2020; 13:nu13010001. [PMID: 33374894 PMCID: PMC7821920 DOI: 10.3390/nu13010001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Fructose consumption has drastically increased during the last decades due to the extensive commercial use of high-fructose corn syrup as a sweetener for beverages, snacks and baked goods. Fructose overconsumption is known to induce obesity, dyslipidemia, insulin resistance and inflammation, and its metabolism is considered partially responsible for its role in several metabolic diseases. Indeed, the primary metabolites and by-products of gut and hepatic fructolysis may impair the functions of extrahepatic tissues and organs. However, fructose itself causes an adenosine triphosphate (ATP) depletion that triggers inflammation and oxidative stress. Many studies have dealt with the effects of this sugar on various organs, while the impact of fructose on brain function is, to date, less explored, despite the relevance of this issue. Notably, fructose transporters and fructose metabolizing enzymes are present in brain cells. In addition, it has emerged that fructose consumption, even in the short term, can adversely influence brain health by promoting neuroinflammation, brain mitochondrial dysfunction and oxidative stress, as well as insulin resistance. Fructose influence on synaptic plasticity and cognition, with a major impact on critical regions for learning and memory, was also reported. In this review, we discuss emerging data about fructose effects on brain health in rodent models, with special reference to the regulation of food intake, inflammation, mitochondrial function and oxidative stress, insulin signaling and cognitive function.
Collapse
|
22
|
Hadj-Moussa H, Pamenter ME, Storey KB. Hypoxic naked mole-rat brains use microRNA to coordinate hypometabolic fuels and neuroprotective defenses. J Cell Physiol 2020; 236:5080-5097. [PMID: 33305831 DOI: 10.1002/jcp.30216] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/19/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022]
Abstract
Naked mole-rats are among the mammalian champions of hypoxia tolerance. They evolved adaptations centered around reducing metabolic rate to overcome the challenges experienced in their underground burrows. In this study, we used next-generation sequencing to investigate one of the factors likely supporting hypoxia tolerance in naked mole-rat brains, posttranscriptional microRNAs (miRNAs). Of the 212 conserved miRNAs identified using small RNA sequencing, 18 displayed significant differential expression during hypoxia. Bioinformatic enrichment revealed that hypoxia-mediated miRNAs were suppressing energy expensive processes including de novo protein translation and cellular proliferation. This suppression occurred alongside the activation of neuroprotective and neuroinflammatory pathways, and the induction of central signal transduction pathways including HIF-1α and NFκB via miR-335, miR-101, and miR-155. MiRNAs also coordinated anaerobic glycolytic fuel sources, where hypoxia-upregulated miR-365 likely suppressed protein levels of ketohexokinase, the enzyme responsible for catalyzing the first committed step of fructose catabolism. This was further supported by a hypoxia-mediated reduction in glucose transporter 5 proteins that import fructose into the cell. Yet, messenger RNA and protein levels of lactate dehydrogenase, which converts pyruvate to lactate in the absence of oxygen, were elevated during hypoxia. Together, this demonstrated the induction of anaerobic glycolysis despite a lack of reliance on fructose as the primary fuel source, suggesting that hypoxic brains are metabolically different than anoxic naked mole-rat brains that were previously found to shift to fructose-based glycolysis. Our findings contribute to the growing body of oxygen-responsive miRNAs "OxymiRs" that facilitate natural miRNA-mediated mechanisms for successful hypoxic exposures.
Collapse
Affiliation(s)
| | - Matthew E Pamenter
- Biology Department, University of Ottawa, Ottawa, Ontario, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Kenneth B Storey
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
23
|
Helsley RN, Moreau F, Gupta MK, Radulescu A, DeBosch B, Softic S. Tissue-Specific Fructose Metabolism in Obesity and Diabetes. Curr Diab Rep 2020; 20:64. [PMID: 33057854 DOI: 10.1007/s11892-020-01342-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to provide up-to-date and comprehensive discussion of tissue-specific fructose metabolism in the context of diabetes, dyslipidemia, and nonalcoholic fatty liver disease (NAFLD). RECENT FINDINGS Increased intake of dietary fructose is a risk factor for a myriad of metabolic complications. Tissue-specific fructose metabolism has not been well delineated in terms of its contribution to detrimental health effects associated with fructose intake. Since inhibitors targeting fructose metabolism are being developed for the management of NAFLD and diabetes, it is essential to recognize how inability of one tissue to metabolize fructose may affect metabolism in the other tissues. The primary sites of fructose metabolism are the liver, intestine, and kidney. Skeletal muscle and adipose tissue can also metabolize a large portion of fructose load, especially in the setting of ketohexokinase deficiency, the rate-limiting enzyme of fructose metabolism. Fructose can also be sensed by the pancreas and the brain, where it can influence essential functions involved in energy homeostasis. Lastly, fructose is metabolized by the testes, red blood cells, and lens of the eye where it may contribute to infertility, advanced glycation end products, and cataracts, respectively. An increase in sugar intake, particularly fructose, has been associated with the development of obesity and its complications. Inhibition of fructose utilization in tissues primary responsible for its metabolism alters consumption in other tissues, which have not been traditionally regarded as important depots of fructose metabolism.
Collapse
Affiliation(s)
- Robert N Helsley
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Francois Moreau
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Medicine, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Aurelia Radulescu
- Department of Pediatrics, University of Kentucky College of Medicine and Kentucky Children's Hospital, Lexington, KY, 40536, USA
| | - Brian DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63131, USA
| | - Samir Softic
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, 138 Leader Ave, Lexington, KY, 40506, USA.
| |
Collapse
|
24
|
Chiba Y, Murakami R, Matsumoto K, Wakamatsu K, Nonaka W, Uemura N, Yanase K, Kamada M, Ueno M. Glucose, Fructose, and Urate Transporters in the Choroid Plexus Epithelium. Int J Mol Sci 2020; 21:E7230. [PMID: 33008107 PMCID: PMC7582461 DOI: 10.3390/ijms21197230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
The choroid plexus plays a central role in the regulation of the microenvironment of the central nervous system by secreting the majority of the cerebrospinal fluid and controlling its composition, despite that it only represents approximately 1% of the total brain weight. In addition to a variety of transporter and channel proteins for solutes and water, the choroid plexus epithelial cells are equipped with glucose, fructose, and urate transporters that are used as energy sources or antioxidative neuroprotective substrates. This review focuses on the recent advances in the understanding of the transporters of the SLC2A and SLC5A families (GLUT1, SGLT2, GLUT5, GLUT8, and GLUT9), as well as on the urate-transporting URAT1 and BCRP/ABCG2, which are expressed in choroid plexus epithelial cells. The glucose, fructose, and urate transporters repertoire in the choroid plexus epithelium share similar features with the renal proximal tubular epithelium, although some of these transporters exhibit inversely polarized submembrane localization. Since choroid plexus epithelial cells have high energy demands for proper functioning, a decline in the expression and function of these transporters can contribute to the process of age-associated brain impairment and pathophysiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| | - Wakako Nonaka
- Department of Supportive and Promotive Medicine of the Municipal Hospital, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (N.U.); (K.Y.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (N.U.); (K.Y.)
| | - Masaki Kamada
- Department of Neurological Intractable Disease Research, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (K.M.); (K.W.)
| |
Collapse
|
25
|
Johnson RJ, Gomez-Pinilla F, Nagel M, Nakagawa T, Rodriguez-Iturbe B, Sanchez-Lozada LG, Tolan DR, Lanaspa MA. Cerebral Fructose Metabolism as a Potential Mechanism Driving Alzheimer's Disease. Front Aging Neurosci 2020; 12:560865. [PMID: 33024433 PMCID: PMC7516162 DOI: 10.3389/fnagi.2020.560865] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
The loss of cognitive function in Alzheimer's disease is pathologically linked with neurofibrillary tangles, amyloid deposition, and loss of neuronal communication. Cerebral insulin resistance and mitochondrial dysfunction have emerged as important contributors to pathogenesis supporting our hypothesis that cerebral fructose metabolism is a key initiating pathway for Alzheimer's disease. Fructose is unique among nutrients because it activates a survival pathway to protect animals from starvation by lowering energy in cells in association with adenosine monophosphate degradation to uric acid. The fall in energy from fructose metabolism stimulates foraging and food intake while reducing energy and oxygen needs by decreasing mitochondrial function, stimulating glycolysis, and inducing insulin resistance. When fructose metabolism is overactivated systemically, such as from excessive fructose intake, this can lead to obesity and diabetes. Herein, we present evidence that Alzheimer's disease may be driven by overactivation of cerebral fructose metabolism, in which the source of fructose is largely from endogenous production in the brain. Thus, the reduction in mitochondrial energy production is hampered by neuronal glycolysis that is inadequate, resulting in progressive loss of cerebral energy levels required for neurons to remain functional and viable. In essence, we propose that Alzheimer's disease is a modern disease driven by changes in dietary lifestyle in which fructose can disrupt cerebral metabolism and neuronal function. Inhibition of intracerebral fructose metabolism could provide a novel way to prevent and treat this disease.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maria Nagel
- Departments of Neurology and Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | - Bernardo Rodriguez-Iturbe
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Laura G Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, United States
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
26
|
Glucose transporters in brain in health and disease. Pflugers Arch 2020; 472:1299-1343. [PMID: 32789766 PMCID: PMC7462931 DOI: 10.1007/s00424-020-02441-x] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
Energy demand of neurons in brain that is covered by glucose supply from the blood is ensured by glucose transporters in capillaries and brain cells. In brain, the facilitative diffusion glucose transporters GLUT1-6 and GLUT8, and the Na+-d-glucose cotransporters SGLT1 are expressed. The glucose transporters mediate uptake of d-glucose across the blood-brain barrier and delivery of d-glucose to astrocytes and neurons. They are critically involved in regulatory adaptations to varying energy demands in response to differing neuronal activities and glucose supply. In this review, a comprehensive overview about verified and proposed roles of cerebral glucose transporters during health and diseases is presented. Our current knowledge is mainly based on experiments performed in rodents. First, the functional properties of human glucose transporters expressed in brain and their cerebral locations are described. Thereafter, proposed physiological functions of GLUT1, GLUT2, GLUT3, GLUT4, and SGLT1 for energy supply to neurons, glucose sensing, central regulation of glucohomeostasis, and feeding behavior are compiled, and their roles in learning and memory formation are discussed. In addition, diseases are described in which functional changes of cerebral glucose transporters are relevant. These are GLUT1 deficiency syndrome (GLUT1-SD), diabetes mellitus, Alzheimer’s disease (AD), stroke, and traumatic brain injury (TBI). GLUT1-SD is caused by defect mutations in GLUT1. Diabetes and AD are associated with changed expression of glucose transporters in brain, and transporter-related energy deficiency of neurons may contribute to pathogenesis of AD. Stroke and TBI are associated with changes of glucose transporter expression that influence clinical outcome.
Collapse
|
27
|
Nakagawa T, Lanaspa MA, Millan IS, Fini M, Rivard CJ, Sanchez-Lozada LG, Andres-Hernando A, Tolan DR, Johnson RJ. Fructose contributes to the Warburg effect for cancer growth. Cancer Metab 2020; 8:16. [PMID: 32670573 PMCID: PMC7350662 DOI: 10.1186/s40170-020-00222-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity and metabolic syndrome are strongly associated with cancer, and these disorders may share a common mechanism. Recently, fructose has emerged as a driving force to develop obesity and metabolic syndrome. Thus, we assume that fructose may be the mechanism to explain why obesity and metabolic syndrome are linked with cancer. Clinical and experimental evidence showed that fructose intake was associated with cancer growth and that fructose transporters are upregulated in various malignant tumors. Interestingly, fructose metabolism can be driven under low oxygen conditions, accelerates glucose utilization, and exhibits distinct effects as compared to glucose, including production of uric acid and lactate as major byproducts. Fructose promotes the Warburg effect to preferentially downregulate mitochondrial respiration and increases aerobic glycolysis that may aid metastases that initially have low oxygen supply. In the process, uric acid may facilitate carcinogenesis by inhibiting the TCA cycle, stimulating cell proliferation by mitochondrial ROS, and blocking fatty acid oxidation. Lactate may also contribute to cancer growth by suppressing fat oxidation and inducing oncogene expression. The ability of fructose metabolism to directly stimulate the glycolytic pathway may have been protective for animals living with limited access to oxygen, but may be deleterious toward stimulating cancer growth and metastasis for humans in modern society. Blocking fructose metabolism may be a novel approach for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, 2 Otowa-Chinji-cho, Yamashina-ku, Kyoto, Japan
- Department of Stem Cell Biology & Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Inigo San Millan
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, USA
| | - Mehdi Fini
- University of Colorado Cancer Center, Aurora, CO USA
| | | | - Laura G. Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, 14080 Mexico City, CP Mexico
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Dean R. Tolan
- Department of Biology, Boston University, Boston, MA USA
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| |
Collapse
|
28
|
Cargnin-Carvalho A, de Mello AH, Bressan JB, Backes KM, Uberti MF, Fogaça JB, da Rosa Turatti C, Cavalheiro EKFF, Vilela TC, Rezin GT. Can fructose influence the development of obesity mediated through hypothalamic alterations? J Neurosci Res 2020; 98:1662-1668. [PMID: 32524664 DOI: 10.1002/jnr.24628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
Epidemiological data from the last decades point to an exponential growth in the number of obese people. Different behavioral factors, mainly associated with food consumption, appear to contribute significantly to its development. Concomitant with increased obesity rates, an increase in the consumption of fructose has been observed; therefore, fructose consumption has been implicated as an important obesogenic factor. However, changes in brain activity due to fructose consumption are possible, especially in relation to hypothalamic satiety mechanisms. In addition, the obese state may provide an environment of chronic inflammation and further contribute to the discontinuation of satiety mechanisms in the hypothalamus. We briefly review the intrinsic alterations to the increased adipose tissue, its connections with the hypothalamus in the control of energy signaling mechanisms and, consequently, the participation of fructose as a co-adjuvant or trigger. Presenting the current context with clinical trials involving human and animal studies, we seek to contribute to a better understanding of the role of fructose in the progression of obesity.
Collapse
Affiliation(s)
- Anderson Cargnin-Carvalho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Aline Haas de Mello
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Joice Benedet Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Kassiane Mathiola Backes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Marcela Fornari Uberti
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Jéssica Benedet Fogaça
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Cristini da Rosa Turatti
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Eulla Keimili Fernandes Ferreira Cavalheiro
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Thais Ceresér Vilela
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina at Tubarão, Santa Catarina, Brazil
| |
Collapse
|
29
|
Buziau AM, Schalkwijk CG, Stehouwer CDA, Tolan DR, Brouwers MCGJ. Recent advances in the pathogenesis of hereditary fructose intolerance: implications for its treatment and the understanding of fructose-induced non-alcoholic fatty liver disease. Cell Mol Life Sci 2020; 77:1709-1719. [PMID: 31713637 PMCID: PMC11105038 DOI: 10.1007/s00018-019-03348-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/31/2022]
Abstract
Hereditary fructose intolerance (HFI) is a rare inborn disease characterized by a deficiency in aldolase B, which catalyzes the cleavage of fructose 1,6-bisphosphate and fructose 1-phosphate (Fru 1P) to triose molecules. In patients with HFI, ingestion of fructose results in accumulation of Fru 1P and depletion of ATP, which are believed to cause symptoms, such as nausea, vomiting, hypoglycemia, and liver and kidney failure. These sequelae can be prevented by a fructose-restricted diet. Recent studies in aldolase B-deficient mice and HFI patients have provided more insight into the pathogenesis of HFI, in particular the liver phenotype. Both aldolase B-deficient mice (fed a very low fructose diet) and HFI patients (treated with a fructose-restricted diet) displayed greater intrahepatic fat content when compared to controls. The liver phenotype in aldolase B-deficient mice was prevented by reduction in intrahepatic Fru 1P concentrations by crossing these mice with mice deficient for ketohexokinase, the enzyme that catalyzes the synthesis of Fru 1P. These new findings not only provide a potential novel treatment for HFI, but lend insight into the pathogenesis of fructose-induced non-alcoholic fatty liver disease (NAFLD), which has raised to epidemic proportions in Western society. This narrative review summarizes the most recent advances in the pathogenesis of HFI and discusses the implications for the understanding and treatment of fructose-induced NAFLD.
Collapse
Affiliation(s)
- Amée M Buziau
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA.
| | - Martijn C G J Brouwers
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands.
| |
Collapse
|
30
|
Johnson RJ, Stenvinkel P, Andrews P, Sánchez-Lozada LG, Nakagawa T, Gaucher E, Andres-Hernando A, Rodriguez-Iturbe B, Jimenez CR, Garcia G, Kang DH, Tolan DR, Lanaspa MA. Fructose metabolism as a common evolutionary pathway of survival associated with climate change, food shortage and droughts. J Intern Med 2020; 287:252-262. [PMID: 31621967 PMCID: PMC10917390 DOI: 10.1111/joim.12993] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
Mass extinctions occur frequently in natural history. While studies of animals that became extinct can be informative, it is the survivors that provide clues for mechanisms of adaptation when conditions are adverse. Here, we describe a survival pathway used by many species as a means for providing adequate fuel and water, while also providing protection from a decrease in oxygen availability. Fructose, whether supplied in the diet (primarily fruits and honey), or endogenously (via activation of the polyol pathway), preferentially shifts the organism towards the storing of fuel (fat, glycogen) that can be used to provide energy and water at a later date. Fructose causes sodium retention and raises blood pressure and likely helped survival in the setting of dehydration or salt deprivation. By shifting energy production from the mitochondria to glycolysis, fructose reduced oxygen demands to aid survival in situations where oxygen availability is low. The actions of fructose are driven in part by vasopressin and the generation of uric acid. Twice in history, mutations occurred during periods of mass extinction that enhanced the activity of fructose to generate fat, with the first being a mutation in vitamin C metabolism during the Cretaceous-Paleogene extinction (65 million years ago) and the second being a mutation in uricase that occurred during the Middle Miocene disruption (12-14 million years ago). Today, the excessive intake of fructose due to the availability of refined sugar and high-fructose corn syrup is driving 'burden of life style' diseases, including obesity, diabetes and high blood pressure.
Collapse
Affiliation(s)
- R J Johnson
- From the, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - P Stenvinkel
- Division of Renal Diseases, Karolinska Institute, Stockholm, Sweden
| | - P Andrews
- Museum of Natural History, London, UK
| | | | - T Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - E Gaucher
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - A Andres-Hernando
- From the, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - C R Jimenez
- From the, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - G Garcia
- From the, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - D-H Kang
- Division of Renal Diseases, Ewha University, Seoul, Korea
| | - D R Tolan
- Department of Biology, Boston University, Boson, MA, USA
| | - M A Lanaspa
- From the, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
31
|
Abstract
The adult brain consumes glucose for energy needs and stores glucose as glycogen mainly in astrocytes. Schulz et al. (2020. J. Cell Biol.https://doi.org/10.1083/jcb.201807127) identify the stress-regulated metabolic enzyme GDPGP1 that promotes neuronal survival likely through glycogen reserves in mouse and C. elegans neurons.
Collapse
Affiliation(s)
- Neel S. Singhal
- Department of Neurology, University of California, San Francisco, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Evan M. Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Physiology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
32
|
Insulin deficiency promotes formation of toxic amyloid-β42 conformer co-aggregating with hyper-phosphorylated tau oligomer in an Alzheimer's disease model. Neurobiol Dis 2020; 137:104739. [PMID: 31927145 DOI: 10.1016/j.nbd.2020.104739] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/27/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
The toxic conformer of amyloid β-protein (Aβ) ending at 42 (Aβ42), which contains a unique turn conformation at amino acid residue positions 22 and 23 and tends to form oligomers that are neurotoxic, was reported to play a critical role in the pathomechanisms of Alzheimer's disease (AD), in which diabetes mellitus (DM)-like mechanisms are also suggested to be operative. It remains to be established whether the attenuation of insulin signaling is involved in an increase of toxic Aβ42 conformer levels. The present study investigated the association between impaired insulin metabolism and formation of toxic Aβ42 conformers in the brains of an AD mouse model. In particular, we studied whether insulin deficiency or resistance affected the formation of toxic Aβ42 conformers in vivo. We induced insulin deficiency and resistance in 3xTg-AD mice, a mouse AD model harboring two familial AD-mutant APP (KM670/671NL) and PS1 (M146 V) genes and a mutant TAU (P301L) gene, by streptozotocin (STZ) injection and a high fructose diet (HFuD), respectively. Cognitive impairment was significantly worsened by STZ injection but not by HFuD. Dot blot analysis revealed significant increases in total Aβ42 levels and the ratio of toxic Aβ42 conformer/total Aβ42 in STZ-treated mice compared with control and HFuD-fed mice. Immunostaining showed the accumulation of toxic Aβ42 conformers and hyper-phosphorylated tau protein (p-tau), which was more prominent in the cortical and hippocampal neurons of STZ-treated mice compared with HFuD-fed and control mice. HFuD-fed mice showed only a mild-to-moderate increase of these proteins compared with controls. Toxic Aβ42 conformers were co-localized with p-tau oligomers (Pearson's correlation coefficient = 0.62) in the hippocampus, indicating their co-aggregation. Toxic Aβ42 conformer levels were inversely correlated with pancreatic insulin secretion capacity as shown by fasting immunoreactive insulin levels in STZ-treated mice (correlation coefficient = -0.5879, p = .04441), but not HFuD-fed mice, suggesting a decrease in serum insulin levels correlates with toxic Aβ42 conformer formation. Levels of p-Akt and phosphorylated glycogen synthase kinase-3β measured by a homogeneous time-resolved fluorescence assay were significantly lower in STZ-treated mice than in HFuD-fed mice, suggesting a greater inhibition of brain insulin signaling by STZ than HFuD, although both levels were significantly decreased in these groups compared with controls. Iba1-positive and NOS2-positive areas in the cortex and hippocampus were significantly increased in STZ-treated mice and to a lesser extent in HFuD-fed mice compared with controls. These findings suggest that insulin deficiency rather than insulin resistance and the resultant impairment of brain insulin signaling facilitates the formation of toxic Aβ42 conformer and its co-aggregation with p-tau oligomers, and that insulin deficiency is an important pathogenic factor in the progression of AD.
Collapse
|
33
|
Merino B, Fernández-Díaz CM, Cózar-Castellano I, Perdomo G. Intestinal Fructose and Glucose Metabolism in Health and Disease. Nutrients 2019; 12:E94. [PMID: 31905727 PMCID: PMC7019254 DOI: 10.3390/nu12010094] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemics of obesity and diabetes have been linked to increased sugar consumption in humans. Here, we review fructose and glucose metabolism, as well as potential molecular mechanisms by which excessive sugar consumption is associated to metabolic diseases and insulin resistance in humans. To this end, we focus on understanding molecular and cellular mechanisms of fructose and glucose transport and sensing in the intestine, the intracellular signaling effects of dietary sugar metabolism, and its impact on glucose homeostasis in health and disease. Finally, the peripheral and central effects of dietary sugars on the gut-brain axis will be reviewed.
Collapse
Affiliation(s)
- Beatriz Merino
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Cristina M. Fernández-Díaz
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Irene Cózar-Castellano
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - German Perdomo
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Departamento de Ciencias de la Salud, Universidad de Burgos, Burgos 09001, Spain
| |
Collapse
|
34
|
Enrichment of Aldolase C Correlates with Low Non-Mutated IDH1 Expression and Predicts a Favorable Prognosis in Glioblastomas. Cancers (Basel) 2019; 11:cancers11091238. [PMID: 31450822 PMCID: PMC6770576 DOI: 10.3390/cancers11091238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022] Open
Abstract
The aldolases family is one of the main enzymes involved in the process of glycolysis. Aldolase C (ALDOC), which belongs to the aldolase family, is found in normal brain tissue and is responsible for the repair of injured tissue. However, the role of ALDOC in glioblastoma remains unclear. In this study, we data-mined in silico databases to evaluate aldolase family members’ mRNA expression in glioblastoma patient cohorts for determining its prognostic values. After that, we also performed immunohistochemical stain (IHC) analysis to evaluate protein expression levels of ALDOC in glioblastoma tissues. From The Cancer Genome Atlas (TCGA) database analyses, higher mRNA expression levels in normal brain tissue compared to glioblastoma was observed. In addition, compared to low-grade glioma, ALDOC expression was significantly downregulated in high-grade glioblastoma. Besides, the expression level of ALDOC was associated with molecular subtypes of glioblastomas and recurrent status in several data sets. In contrast, aldolase A (ALDOA) and aldolase B (ALDOB) revealed no significant prognostic impacts in the glioblastoma cohorts. Furthermore, we also proved that ALDOC mRNA and protein expression inversely correlated with non-mutated IDH1 expressions in glioblastoma patient cohorts. Additionally, the concordance of low ALDOC and high non-mutated IDH1 expressions predicted a stronger poor prognosis in glioblastoma patients compared to each of above tests presented alone. The plausible ALDOC and IDH1 regulatory mechanism was further elucidated. Our results support high ALDOC expression in glioblastomas that might imply the mutated status of IDH1, less possibility of mesenchymal subtype, and predict a favorable prognosis.
Collapse
|
35
|
Kroemer G, López-Otín C, Madeo F, de Cabo R. Carbotoxicity-Noxious Effects of Carbohydrates. Cell 2019; 175:605-614. [PMID: 30340032 DOI: 10.1016/j.cell.2018.07.044] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/18/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Modern nutrition is often characterized by the excessive intake of different types of carbohydrates ranging from digestible polysaccharides to refined sugars that collectively mediate noxious effects on human health, a phenomenon that we refer to as "carbotoxicity." Epidemiological and experimental evidence combined with clinical intervention trials underscore the negative impact of excessive carbohydrate uptake, as well as the beneficial effects of reducing carbs in the diet. We discuss the molecular, cellular, and neuroendocrine mechanisms that link exaggerated carbohydrate intake to disease and accelerated aging as we outline dietary and pharmacologic strategies to combat carbotoxicity.
Collapse
Affiliation(s)
- Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| |
Collapse
|
36
|
Andres-Hernando A, Johnson RJ, Lanaspa MA. Endogenous fructose production: what do we know and how relevant is it? Curr Opin Clin Nutr Metab Care 2019; 22:289-294. [PMID: 31166222 PMCID: PMC6684314 DOI: 10.1097/mco.0000000000000573] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Excessive sugar and particularly fructose consumption has been proposed to be a key player in the pathogenesis of metabolic syndrome and kidney disease in humans and animal models. However, besides its dietary source, fructose can be endogenously produced in the body from glucose via the activation of the polyol pathway. In this review, we aim to describe the most recent findings and current knowledge on the potential role of endogenous fructose production and metabolism in disease. RECENT FINDINGS Over the recent years, the activation of the polyol pathway and endogenous fructose production has been observed in multiple tissues including the liver, renal cortex, and hypothalamic areas of the brain. The activation occurs during the development and progression of metabolic syndrome and kidney disease and results from different stimuli including osmotic effects, diabetes, and ischemia. Even though the potential toxicity of the activation of the polyol pathway can be attributed to several intermediate products, the blockade of endogenous fructose metabolism either by using fructokinase deficient mice or specific inhibitors resulted in marked amelioration of multiple metabolic diseases. SUMMARY New findings suggest that fructose can be produced in the body and that the blockade of tis metabolism could be clinically relevant for the prevention and treatment of metabolic syndrome and kidney disease.
Collapse
|
37
|
Myrka AM, Welch KC. Evidence of high transport and phosphorylation capacity for both glucose and fructose in the ruby-throated hummingbird (Archilochus colubris). Comp Biochem Physiol B Biochem Mol Biol 2018; 224:253-261. [DOI: 10.1016/j.cbpb.2017.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023]
|
38
|
Strawbridge RJ, Ward J, Lyall LM, Tunbridge EM, Cullen B, Graham N, Ferguson A, Johnston KJA, Lyall DM, Mackay D, Cavanagh J, Howard DM, Adams MJ, Deary I, Escott-Price V, O'Donovan M, McIntosh AM, Bailey MES, Pell JP, Harrison PJ, Smith DJ. Genetics of self-reported risk-taking behaviour, trans-ethnic consistency and relevance to brain gene expression. Transl Psychiatry 2018; 8:178. [PMID: 30181555 PMCID: PMC6123450 DOI: 10.1038/s41398-018-0236-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/05/2018] [Indexed: 12/25/2022] Open
Abstract
Risk-taking behaviour is an important component of several psychiatric disorders, including attention-deficit hyperactivity disorder, schizophrenia and bipolar disorder. Previously, two genetic loci have been associated with self-reported risk taking and significant genetic overlap with psychiatric disorders was identified within a subsample of UK Biobank. Using the white British participants of the full UK Biobank cohort (n = 83,677 risk takers versus 244,662 controls) for our primary analysis, we conducted a genome-wide association study of self-reported risk-taking behaviour. In secondary analyses, we assessed sex-specific effects, trans-ethnic heterogeneity and genetic overlap with psychiatric traits. We also investigated the impact of risk-taking-associated SNPs on both gene expression and structural brain imaging. We identified 10 independent loci for risk-taking behaviour, of which eight were novel and two replicated previous findings. In addition, we found two further sex-specific risk-taking loci. There were strong positive genetic correlations between risk-taking and attention-deficit hyperactivity disorder, bipolar disorder and schizophrenia. Index genetic variants demonstrated effects generally consistent with the discovery analysis in individuals of non-British White, South Asian, African-Caribbean or mixed ethnicity. Polygenic risk scores comprising alleles associated with increased risk taking were associated with lower white matter integrity. Genotype-specific expression pattern analyses highlighted DPYSL5, CGREF1 and C15orf59 as plausible candidate genes. Overall, our findings substantially advance our understanding of the biology of risk-taking behaviour, including the possibility of sex-specific contributions, and reveal consistency across ethnicities. We further highlight several putative novel candidate genes, which may mediate these genetic effects.
Collapse
Affiliation(s)
- Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden.
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Keira J A Johnston
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Division of Psychiatry, College of Medicine, University of Edinburgh, Edinburgh, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel Mackay
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - David M Howard
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF, UK
| | - Ian Deary
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9YL, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9YL, UK
| | | | - Michael O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF, UK
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| |
Collapse
|
39
|
Allen RJ, Musante CJ. A mathematical analysis of adaptations to the metabolic fate of fructose in essential fructosuria subjects. Am J Physiol Endocrinol Metab 2018; 315:E394-E403. [PMID: 29664676 DOI: 10.1152/ajpendo.00317.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Fructose is a major component of Western diets and is implicated in the pathogenesis of obesity and type 2 diabetes. In response to an oral challenge, the majority of fructose is cleared during "first-pass" liver metabolism, primarily via phosphorylation by ketohexokinase (KHK). A rare benign genetic deficiency in KHK, called essential fructosuria (EF), leads to altered fructose metabolism. The only reported symptom of EF is the appearance of fructose in the urine following either oral or intravenous fructose administration. Here we develop and use a mathematical model to investigate the adaptations to altered fructose metabolism in people with EF. First, the model is calibrated to fit available data in normal healthy subjects. Then, to mathematically represent EF subjects, we systematically implement metabolic adaptations such that model simulations match available data for this phenotype. We hypothesize that these modifications represent the major metabolic adaptations present in these subjects. This modeling approach suggests that several other aspects of fructose metabolism, beyond hepatic KHK deficiency, are altered and contribute to the etiology of this benign condition. Specifically, we predict that fructose absorption into the portal vein is altered, peripheral metabolism is slowed, renal reabsorption of fructose is mostly ablated, and alternate pathways for hepatic metabolism of fructose are upregulated. Moreover, these findings have implications for drug discovery and development, suggesting that the therapeutic targeting of fructose metabolism could lead to unexpected metabolic adaptations, potentially due to a physiological response to high-fructose conditions.
Collapse
Affiliation(s)
- Richard J Allen
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Cynthia J Musante
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| |
Collapse
|
40
|
Fuhr L, El-Athman R, Scrima R, Cela O, Carbone A, Knoop H, Li Y, Hoffmann K, Laukkanen MO, Corcione F, Steuer R, Meyer TF, Mazzoccoli G, Capitanio N, Relógio A. The Circadian Clock Regulates Metabolic Phenotype Rewiring Via HKDC1 and Modulates Tumor Progression and Drug Response in Colorectal Cancer. EBioMedicine 2018; 33:105-121. [PMID: 30005951 PMCID: PMC6085544 DOI: 10.1016/j.ebiom.2018.07.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022] Open
Abstract
An endogenous molecular clockwork drives various cellular pathways including metabolism and the cell cycle. Its dysregulation is able to prompt pathological phenotypes including cancer. Besides dramatic metabolic alterations, cancer cells display severe changes in the clock phenotype with likely consequences in tumor progression and treatment response. In this study, we use a comprehensive systems-driven approach to investigate the effect of clock disruption on metabolic pathways and its impact on drug response in a cellular model of colon cancer progression. We identified distinctive time-related transcriptomic and metabolic features of a primary tumor and its metastatic counterpart. A mapping of the expression data to a comprehensive genome-scale reconstruction of human metabolism allowed for the in-depth functional characterization of 24 h-oscillating transcripts and pointed to a clock-driven metabolic reprogramming in tumorigenesis. In particular, we identified a set of five clock-regulated glycolysis genes, ALDH3A2, ALDOC, HKDC1, PCK2, and PDHB with differential temporal expression patterns. These findings were validated in organoids and in primary fibroblasts isolated from normal colon and colon adenocarcinoma from the same patient. We further identified a reciprocal connection of HKDC1 to the clock in the primary tumor, which is lost in the metastatic cells. Interestingly, a disruption of the core-clock gene BMAL1 impacts on HKDC1 and leads to a time-dependent rewiring of metabolism, namely an increase in glycolytic activity, as well as changes in treatment response. This work provides novel evidence regarding the complex interplay between the circadian clock and metabolic alterations in carcinogenesis and identifies new connections between both systems with pivotal roles in cancer progression and response to therapy.
Collapse
Affiliation(s)
- Luise Fuhr
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Rukeia El-Athman
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Annalucia Carbone
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Henning Knoop
- Institute for Theoretical Biology, Institut für Biologie, Humboldt-Universität zu Berlin, Germany
| | - Yin Li
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany
| | - Karen Hoffmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology Berlin, Germany
| | | | - Francesco Corcione
- Department of General, Laparoscopic and Robotic Surgery, Azienda Ospedaliera Specialistica dei Colli, Monaldi Hospital, Via Leonardo Bianchi, 80131 Naples, Italy
| | - Ralf Steuer
- Institute for Theoretical Biology, Institut für Biologie, Humboldt-Universität zu Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology Berlin, Germany
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Angela Relógio
- Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Institute for Theoretical Biology, Germany; Charité - Universitätsmedizin Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Germany.
| |
Collapse
|
41
|
Weng Y, Zhu J, Chen Z, Fu J, Zhang F. Fructose fuels lung adenocarcinoma through GLUT5. Cell Death Dis 2018; 9:557. [PMID: 29748554 PMCID: PMC5945656 DOI: 10.1038/s41419-018-0630-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 04/21/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Yuanyuan Weng
- Core Facility, Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Jin Zhu
- Core Facility, Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Zhenhong Chen
- Department of Oncology, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Feng Zhang
- Core Facility, Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, Zhejiang, China.
| |
Collapse
|
42
|
Tappy L. Fructose metabolism and noncommunicable diseases: recent findings and new research perspectives. Curr Opin Clin Nutr Metab Care 2018; 21:214-222. [PMID: 29406418 DOI: 10.1097/mco.0000000000000460] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW There is increasing concern that dietary fructose may contribute to the development of noncommunicable diseases. This review identifies major new findings related to fructose's physiological or adverse effects. RECENT FINDINGS Fructose is mainly processed in splanchnic organs (gut, liver, kidneys) to glucose, lactate, and fatty acids, which can then be oxidized in extrasplanchnic organs and tissues. There is growing evidence that splanchnic lactate production, linked to extrasplanchnic lactate metabolism, represents a major fructose disposal pathway during and after exercise. Chronic excess fructose intake can be directly responsible for an increase in intrahepatic fat concentration and for the development of hepatic, but not muscle insulin resistance. Although it has long been thought that fructose was exclusively metabolized in splanchnic organs, several recent reports provide indirect that some fructose may also be metabolized in extrasplanchnic cells, such as adipocytes, muscle, or brain cells; the quantity of fructose directly metabolized in extrasplanchnic cells, and its physiological consequences, remain however unknown. There is also growing evidence that endogenous fructose production from glucose occurs in humans and may have important physiological functions, but may also be associated with adverse health effects. SUMMARY Fructose is a physiological nutrient which, when consumed in excess, may have adverse metabolic effects, mainly in the liver (hepatic insulin resistance and fat storage). There is also concern that exogenous or endogenously produced fructose may be directly metabolized in extrasplanchnic cells in which it may exert adverse metabolic effects.
Collapse
Affiliation(s)
- Luc Tappy
- Physiology Department, Faculty of Biology and Medicine, University of Lausanne, Lausanne
- Metabolic Center, Hôpital Intercantonal de la Broye, Estavayer-le-lac, Switzerland
| |
Collapse
|
43
|
Su C, Li H, Gao W. GLUT5 increases fructose utilization and promotes tumor progression in glioma. Biochem Biophys Res Commun 2018; 500:462-469. [PMID: 29660339 DOI: 10.1016/j.bbrc.2018.04.103] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/13/2018] [Indexed: 12/20/2022]
Abstract
Fructose is now such an important component of human diets, and several studies have found that some cancer cells could utilize fructose to overcome low glucose micro-environment, but the study on the role of fructose in glioma is rare. To explore the role of fructose in glioma, we detected the proliferation and colony formation ability of glioma cells in fructose medium, and found that the abilities of proliferation and colony formation of glioma cells in fructose medium were similar with abilities in glucose medium, however, fructose just partly restored proliferation ability of normal glial cells. To explore the mechanism, we compared the expression level of GLUT5 (Glucose transporter type 5) in these cell lines, and the results showed that glioma cell lines had higher GLUT5 expression than normal glial cell lines. And knockdown of GLUT5 could significantly inhabit cell proliferation of glioma cells in fructose medium. Furthermore, we found that GLUT5 was also higher expressed in glioma tissues, and GLUT5 expression correlated significantly with glioma malignancy and poor survival of glioma patients (p < 0.01). In addition, we also demonstrated that knockdown of GLUT5 could significantly inhabit tumor proliferation in vivo, and intake fructose could increase tumor volume prominently. Taken together, our data show that fructose can be used by glioma cells, and restrict the fructose intake or targeting GLUT5 could be efficacious strategies in glioma.
Collapse
Affiliation(s)
- Chunhai Su
- Department of Neurosurgery, Jining NO.1 People's Hospital, Jining 272011, Shandong, China.
| | - Hui Li
- Department of Nursing, Jining Medical University, Jining 272011, Shandong, China.
| | - Wenbo Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou 256600, Shandong, China.
| |
Collapse
|
44
|
Juszczak GR, Stankiewicz AM. Glucocorticoids, genes and brain function. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:136-168. [PMID: 29180230 DOI: 10.1016/j.pnpbp.2017.11.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 01/02/2023]
Abstract
The identification of key genes in transcriptomic data constitutes a huge challenge. Our review of microarray reports revealed 88 genes whose transcription is consistently regulated by glucocorticoids (GCs), such as cortisol, corticosterone and dexamethasone, in the brain. Replicable transcriptomic data were combined with biochemical and physiological data to create an integrated view of the effects induced by GCs. The most frequently reported genes were Errfi1 and Ddit4. Their up-regulation was associated with the altered transcription of genes regulating growth factor and mTORC1 signaling (Gab1, Tsc22d3, Dusp1, Ndrg2, Ppp5c and Sesn1) and progression of the cell cycle (Ccnd1, Cdkn1a and Cables1). The GC-induced reprogramming of cell function involves changes in the mRNA level of genes responsible for the regulation of transcription (Klf9, Bcl6, Klf15, Tle3, Cxxc5, Litaf, Tle4, Jun, Sox4, Sox2, Sox9, Irf1, Sall2, Nfkbia and Id1) and the selective degradation of mRNA (Tob2). Other genes are involved in the regulation of metabolism (Gpd1, Aldoc and Pdk4), actin cytoskeleton (Myh2, Nedd9, Mical2, Rhou, Arl4d, Osbpl3, Arhgef3, Sdc4, Rdx, Wipf3, Chst1 and Hepacam), autophagy (Eva1a and Plekhf1), vesicular transport (Rhob, Ehd3, Vps37b and Scamp2), gap junctions (Gjb6), immune response (Tiparp, Mertk, Lyve1 and Il6r), signaling mediated by thyroid hormones (Thra and Sult1a1), calcium (Calm2), adrenaline/noradrenaline (Adcy9 and Adra1d), neuropeptide Y (Npy1r) and histamine (Hdc). GCs also affected genes involved in the synthesis of polyamines (Azin1) and taurine (Cdo1). The actions of GCs are restrained by feedback mechanisms depending on the transcription of Sgk1, Fkbp5 and Nr3c1. A side effect induced by GCs is increased production of reactive oxygen species. Available data show that the brain's response to GCs is part of an emergency mode characterized by inactivation of non-core activities, restrained inflammation, restriction of investments (growth), improved efficiency of energy production and the removal of unnecessary or malfunctioning cellular components to conserve energy and maintain nutrient supply during the stress response.
Collapse
Affiliation(s)
- Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland.
| | - Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
45
|
High fructose diet-induced metabolic syndrome: Pathophysiological mechanism and treatment by traditional Chinese medicine. Pharmacol Res 2018; 130:438-450. [PMID: 29471102 DOI: 10.1016/j.phrs.2018.02.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/09/2018] [Accepted: 02/14/2018] [Indexed: 02/08/2023]
Abstract
Fructose is a natural monosaccharide broadly used in modern society. Over the past few decades, epidemiological studies have demonstrated that high fructose intake is an etiological factor of metabolic syndrome (MetS). This review highlights research advances on fructose-induced MetS, especially the underlying pathophysiological mechanism as well as pharmacotherapy by traditional Chinese medicine (TCM), using the PubMed, Web of science, China National Knowledge Infrastructure, China Science and Technology Journal and Wanfang Data. This review focuses on de novo lipogenesis (DNL) and uric acid (UA) production, two unique features of fructolysis different from glucose glycolysis. High level of DNL and UA production can result in insulin resistance, the key pathological event in developing MetS, mostly through oxidative stress and inflammation. Some other pathologies like the disturbance in brain and gut microbiota in the development of fructose-induced MetS in the past years, are also discussed. In management of MetS, TCM is an excellent representative in alternative and complementary medicine with a complete theory system and substantial herbal remedies. TCMs against MetS or MetS components, including Chinese patent medicines, TCM compound formulas, single TCM herbs and active compounds of TCM herbs, are reviewed on their effects and molecular mechanisms. TCMs with hypouricemic activity, which specially target fructose-induced MetS, are highlighted. And new technologies and strategies (such as high-throughput assay and systems biology) in this field are further discussed. In summary, fructose-induced MetS is a multifactorial disorder with the underlying complex mechanisms. Current clinical and pre-clinical evidence supports the potential of TCMs in management of MetS. Additionally, TCMs may show some advantages against complex MetS as their holistic feature through multiple target actions. However, further work is needed to confirm the effectivity and safety of TCMs by high-standard clinical trials, clarify the molecular mechanisms, and develop new anti-MetS drugs by development and application of optimized and feasible strategies and methods.
Collapse
|
46
|
Moulin S, Seematter G, Seyssel K. Fructose use in clinical nutrition: metabolic effects and potential consequences. Curr Opin Clin Nutr Metab Care 2017; 20:272-278. [PMID: 28383298 DOI: 10.1097/mco.0000000000000376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW The current article presents recent findings on the metabolic effects of fructose. RECENT FINDINGS Fructose has always been considered as a simple 'caloric' hexose only metabolized by splanchnic tissues. Nevertheless, there is growing evidence that fructose acts as a second messenger and induces effects throughout the human body. SUMMARY Recent discoveries made possible with the evolution of technology have highlighted that fructose induces pleiotropic effects on different tissues. The fact that all these tissues express the specific fructose carrier GLUT5 let us reconsider that fructose is not only a caloric hexose, but could also be a potential actor of some behaviors and metabolic pathways. The physiological relevance of fructose as a metabolic driver is pertinent regarding recent scientific literature.
Collapse
Affiliation(s)
- Sandra Moulin
- aDepartment of Critical Care Medicine, Hôpital cantonal de Fribourg, Fribourg bDepartment of Anaesthesia, Hôpital Riviera-Chablais, Montreux cDepartment of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
47
|
Zhang DM, Jiao RQ, Kong LD. High Dietary Fructose: Direct or Indirect Dangerous Factors Disturbing Tissue and Organ Functions. Nutrients 2017; 9:E335. [PMID: 28353649 PMCID: PMC5409674 DOI: 10.3390/nu9040335] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 02/06/2023] Open
Abstract
High dietary fructose is a major contributor to insulin resistance and metabolic syndrome, disturbing tissue and organ functions. Fructose is mainly absorbed into systemic circulation by glucose transporter 2 (GLUT2) and GLUT5, and metabolized in liver to produce glucose, lactate, triglyceride (TG), free fatty acid (FFA), uric acid (UA) and methylglyoxal (MG). Its extrahepatic absorption and metabolism also take place. High levels of these metabolites are the direct dangerous factors. During fructose metabolism, ATP depletion occurs and induces oxidative stress and inflammatory response, disturbing functions of local tissues and organs to overproduce inflammatory cytokine, adiponectin, leptin and endotoxin, which act as indirect dangerous factors. Fructose and its metabolites directly and/or indirectly cause oxidative stress, chronic inflammation, endothelial dysfunction, autophagy and increased intestinal permeability, and then further aggravate the metabolic syndrome with tissue and organ dysfunctions. Therefore, this review addresses fructose-induced metabolic syndrome, and the disturbance effects of direct and/or indirect dangerous factors on the functions of liver, adipose, pancreas islet, skeletal muscle, kidney, heart, brain and small intestine. It is important to find the potential correlations between direct and/or indirect risk factors and healthy problems under excess dietary fructose consumption.
Collapse
Affiliation(s)
- Dong-Mei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China.
| | - Rui-Qing Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China.
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
48
|
Akgun B, Sarı A, Ozturk S, Erol FS, Ozercan IH, Ulu R. Effects of Mucuna pruriens on Free Fatty Acid Levels and Histopathological Changes in the Brains of Rats Fed a High Fructose Diet. Med Princ Pract 2017; 26:561-566. [PMID: 28898884 PMCID: PMC5848471 DOI: 10.1159/000481402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 09/11/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To investigate free fatty acid levels and histopathological changes in the brain of rats fed a high fructose diet (HFrD) and to evaluate the effects of Mucuna pruriens, known to have antidiabetic activity, on these changes. MATERIALS AND METHODS The study comprised 28 mature female Wistar rats. The rats were divided into 4 groups, each included 7 rats. Group 1: control; group 2: fed an HFrD; group 3: fed normal rat chow and M. pruriens; group 4: fed an HFrD and M. pruriens for 6 weeks. At the end of 6 weeks, the rats were decapitated, blood and brain tissues were obtained. Serum glucose and triglyceride levels were measured. Free fatty acid levels were measured in 1 cerebral hemisphere of each rat and histopathological changes in the other. The Mann-Whitney U test was used to compare quantitative continuous data between 2 independent groups, and the Kruskal-Wallis test was used to compare quantitative continuous data between more than 2 independent groups. RESULTS Arachidonic acid and docosahexaenoic acid levels were significantly higher in group 2 than in group 1 (p < 0.05). Free arachidonic acid and docosahexaenoic acid levels in group 4 were significantly less than in group 2 (p < 0.05). Histopathological examination of group 2 revealed extensive gliosis, neuronal hydropic degeneration, and edema. In group 4, gliosis was much lighter than in group 2, and edema was not observed. Neuronal structures in group 4 were similar to those in group 1. CONCLUSIONS The HFrD increased the levels of free arachidonic acid and docosahexaenoic acid probably due to membrane degradation resulting from possible oxidative stress and inflammation in the brain. The HFrD also caused extensive gliosis, neuronal hydropic degeneration, and edema. Hence, M. pruriens could have therapeutic effects on free fatty acid metabolism and local inflammatory responses in the brains of rats fed an HFrD.
Collapse
Affiliation(s)
- Bekir Akgun
- Department of Neurosurgery, Firat University, Elazig, Turkey
- *Bekir Akgun, MD, Department of Neurosurgery, Firat University Hospital, TR-23119 Elazig (Turkey), E-Mail
| | - Aysel Sarı
- Department of Chemistry/Biochemistry, Firat University, Elazig, Turkey
| | - Sait Ozturk
- Department of Neurosurgery, Firat University, Elazig, Turkey
| | | | | | - Ramazan Ulu
- Department of Internal Medicine, Firat University, Elazig, Turkey
| |
Collapse
|