1
|
Daida T, Shin BC, Cepeda C, Devaskar SU. Neurodevelopment Is Dependent on Maternal Diet: Placenta and Brain Glucose Transporters GLUT1 and GLUT3. Nutrients 2024; 16:2363. [PMID: 39064806 PMCID: PMC11279700 DOI: 10.3390/nu16142363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Glucose is the primary energy source for most mammalian cells and its transport is affected by a family of facilitative glucose transporters (GLUTs) encoded by the SLC2 gene. GLUT1 and GLUT3, highly expressed isoforms in the blood-brain barrier and neuronal membranes, respectively, are associated with multiple neurodevelopmental disorders including epilepsy, dyslexia, ADHD, and autism spectrum disorder (ASD). Dietary therapies, such as the ketogenic diet, are widely accepted treatments for patients with the GLUT1 deficiency syndrome, while ameliorating certain symptoms associated with GLUT3 deficiency in animal models. A ketogenic diet, high-fat diet, and calorie/energy restriction during prenatal and postnatal stages can also alter the placental and brain GLUTs expression with long-term consequences on neurobehavior. This review focuses primarily on the role of diet/energy perturbations upon GLUT isoform-mediated emergence of neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Tomoko Daida
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| | - Bo-Chul Shin
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sherin U. Devaskar
- Department of Pediatrics, Division of Neonatology and Developmental Biology and Neonatal Research Center, at the UCLA Children’s Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.D.); (B.-C.S.)
| |
Collapse
|
2
|
Echeverría CE, Oyarzún VI, López-Cortés A, Cancino J, Sotomayor PC, Goncalves MD, Godoy AS. Biological role of fructose in the male reproductive system: Potential implications for prostate cancer. Prostate 2024; 84:8-24. [PMID: 37888416 PMCID: PMC10872645 DOI: 10.1002/pros.24631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Over the last 20 years, fructose has gradually emerged as a potential metabolic substrate capable of promoting the growth and progression of various cancers, including prostate cancer (PCa). The biological and molecular mechanisms that underlie the effects of fructose on cancer are beginning to be elucidated. METHODS This review summarizes the biological function of fructose as a potential carbon source for PCa cells and its role in the functionality of the male reproductive tract under normal conditions. RESULTS The most recent biological advances related to fructose transport and metabolism as well as their implications in PCa growth and progression suggest that fructose represent a potential carbon source for PCa cells. Consequently, fructose derivatives may represent efficient radiotracers for obtaining PCa images via positron emission tomography and fructose transporters/fructose-metabolizing enzymes could be utilized as potential diagnostic and/or predictive biomarkers for PCa. CONCLUSION The existing data suggest that restriction of fructose from the diet could be a useful therapeutic strategy for patients with PCa.
Collapse
Affiliation(s)
- Carolina E. Echeverría
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Vanessa I. Oyarzún
- Laboratory of Ocular and Systemic Autoimmune Diseases, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Jorge Cancino
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula C. Sotomayor
- Departamento de Urología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Alejandro S. Godoy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo New York, USA
| |
Collapse
|
3
|
Geiser A, Foylan S, Tinning PW, Bryant NJ, Gould GW. GLUT4 dispersal at the plasma membrane of adipocytes: a super-resolved journey. Biosci Rep 2023; 43:BSR20230946. [PMID: 37791639 PMCID: PMC10600063 DOI: 10.1042/bsr20230946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/05/2023] Open
Abstract
In adipose tissue, insulin stimulates glucose uptake by mediating the translocation of GLUT4 from intracellular vesicles to the plasma membrane. In 2010, insulin was revealed to also have a fundamental impact on the spatial distribution of GLUT4 within the plasma membrane, with the existence of two GLUT4 populations at the plasma membrane being defined: (1) as stationary clusters and (2) as diffusible monomers. In this model, in the absence of insulin, plasma membrane-fused GLUT4 are found to behave as clusters. These clusters are thought to arise from exocytic events that retain GLUT4 at their fusion sites; this has been proposed to function as an intermediate hub between GLUT4 exocytosis and re-internalisation. By contrast, insulin stimulation induces the dispersal of GLUT4 clusters into monomers and favours a distinct type of GLUT4-vesicle fusion event, known as fusion-with-release exocytosis. Here, we review how super-resolution microscopy approaches have allowed investigation of the characteristics of plasma membrane-fused GLUT4 and further discuss regulatory step(s) involved in the GLUT4 dispersal machinery, introducing the scaffold protein EFR3 which facilitates localisation of phosphatidylinositol 4-kinase type IIIα (PI4KIIIα) to the cell surface. We consider how dispersal may be linked to the control of transporter activity, consider whether macro-organisation may be a widely used phenomenon to control proteins within the plasma membrane, and speculate on the origin of different forms of GLUT4-vesicle exocytosis.
Collapse
Affiliation(s)
- Angéline Geiser
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Shannan Foylan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Peter W Tinning
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Nia J Bryant
- Department of Biology, University of York, Heslington, York, U.K
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| |
Collapse
|
4
|
Song A, Mao Y, Wei H. GLUT5: structure, functions, diseases and potential applications. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1519-1538. [PMID: 37674366 PMCID: PMC10582729 DOI: 10.3724/abbs.2023158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/19/2023] [Indexed: 09/08/2023] Open
Abstract
Glucose transporter 5 (GLUT5) is a membrane transporter that specifically transports fructose and plays a key role in dietary fructose uptake and metabolism. In recent years, a high fructose diet has occupied an important position in the daily intake of human beings, resulting in a significant increase in the incidence of obesity and metabolic diseases worldwide. Over the past few decades, GLUT5 has been well understood to play a significant role in the pathogenesis of human digestive diseases. Recently, the role of GLUT5 in human cancer has received widespread attention, and a large number of studies have focused on exploring the effects of changes in GLUT5 expression levels on cancer cell survival, metabolism and metastasis. However, due to various difficulties and shortcomings, the molecular structure and mechanism of GLUT5 have not been fully elucidated, which to some extent prevents us from revealing the relationship between GLUT5 expression and cell carcinogenesis at the protein molecular level. In this review, we summarize the current understanding of the structure and function of mammalian GLUT5 and its relationship to intestinal diseases and cancer and suggest that GLUT5 may be an important target for cancer therapy.
Collapse
Affiliation(s)
- Aqian Song
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Yuanpeng Mao
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| | - Hongshan Wei
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| |
Collapse
|
5
|
McComas SE, Reichenbach T, Mitrovic D, Alleva C, Bonaccorsi M, Delemotte L, Drew D. Determinants of sugar-induced influx in the mammalian fructose transporter GLUT5. eLife 2023; 12:e84808. [PMID: 37405832 PMCID: PMC10322154 DOI: 10.7554/elife.84808] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
In mammals, glucose transporters (GLUT) control organism-wide blood-glucose homeostasis. In human, this is accomplished by 14 different GLUT isoforms, that transport glucose and other monosaccharides with varying substrate preferences and kinetics. Nevertheless, there is little difference between the sugar-coordinating residues in the GLUT proteins and even the malarial Plasmodium falciparum transporter PfHT1, which is uniquely able to transport a wide range of different sugars. PfHT1 was captured in an intermediate 'occluded' state, revealing how the extracellular gating helix TM7b has moved to break and occlude the sugar-binding site. Sequence difference and kinetics indicated that the TM7b gating helix dynamics and interactions likely evolved to enable substrate promiscuity in PfHT1, rather than the sugar-binding site itself. It was unclear, however, if the TM7b structural transitions observed in PfHT1 would be similar in the other GLUT proteins. Here, using enhanced sampling molecular dynamics simulations, we show that the fructose transporter GLUT5 spontaneously transitions through an occluded state that closely resembles PfHT1. The coordination of D-fructose lowers the energetic barriers between the outward- and inward-facing states, and the observed binding mode for D-fructose is consistent with biochemical analysis. Rather than a substrate-binding site that achieves strict specificity by having a high affinity for the substrate, we conclude GLUT proteins have allosterically coupled sugar binding with an extracellular gate that forms the high-affinity transition-state instead. This substrate-coupling pathway presumably enables the catalysis of fast sugar flux at physiological relevant blood-glucose concentrations.
Collapse
Affiliation(s)
- Sarah E McComas
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm UniversityStockholmSweden
| | - Tom Reichenbach
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm UniversityStockholmSweden
| | - Darko Mitrovic
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Claudia Alleva
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm UniversityStockholmSweden
| | - Marta Bonaccorsi
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm UniversityStockholmSweden
| | - Lucie Delemotte
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - David Drew
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm UniversityStockholmSweden
| |
Collapse
|
6
|
Chałaśkiewicz K, Karaś K, Zakłos-Szyda M, Karwaciak I, Pastwińska J, Koziołkiewicz M, Ratajewski M. Trichostatin a inhibits expression of the human SLC2A5 gene via SNAI1/SNAI2 transcription factors and sensitizes colon cancer cells to platinum compounds. Eur J Pharmacol 2023; 949:175728. [PMID: 37062501 DOI: 10.1016/j.ejphar.2023.175728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
GLUT5, a key protein encoded by the SLC2A5 gene, is involved in the uptake of fructose from the intestine. Currently, with the increased consumption of this sugar and the associated increased incidence of obesity, diabetes and cancer, GLUT5 may represent an important molecular target in the prevention and treatment of these diseases. Here, we demonstrate that overexpression of the SNAI1 and SNAI2 transcription factors in cells expressing high levels of SLC2A5 mRNA reduced SLC2A5 gene expression. Furthermore, a histone deacetylase inhibitor, trichostatin A, which induces SNAI1 and SNAI2 expression, inhibits SLC2A5/GLUT5 expression and sensitizes colon cancer cells to cisplatin and oxaliplatin. This finding might have potential relevance for the development of therapeutic treatments aimed at modulating fructose transport or genes involved in this process for use with certain cancers.
Collapse
Affiliation(s)
- Katarzyna Chałaśkiewicz
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland; Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Małgorzata Zakłos-Szyda
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Maria Koziołkiewicz
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.
| |
Collapse
|
7
|
Gyimesi G, Hediger MA. Transporter-Mediated Drug Delivery. Molecules 2023; 28:molecules28031151. [PMID: 36770817 PMCID: PMC9919865 DOI: 10.3390/molecules28031151] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Transmembrane transport of small organic and inorganic molecules is one of the cornerstones of cellular metabolism. Among transmembrane transporters, solute carrier (SLC) proteins form the largest, albeit very diverse, superfamily with over 400 members. It was recognized early on that xenobiotics can directly interact with SLCs and that this interaction can fundamentally determine their efficacy, including bioavailability and intertissue distribution. Apart from the well-established prodrug strategy, the chemical ligation of transporter substrates to nanoparticles of various chemical compositions has recently been used as a means to enhance their targeting and absorption. In this review, we summarize efforts in drug design exploiting interactions with specific SLC transporters to optimize their therapeutic effects. Furthermore, we describe current and future challenges as well as new directions for the advanced development of therapeutics that target SLC transporters.
Collapse
|
8
|
Integrative transcriptome analysis of SARS-CoV-2 human-infected cells combined with deep learning algorithms identifies two potential cellular targets for the treatment of coronavirus disease. Braz J Microbiol 2022; 54:53-68. [PMID: 36435956 PMCID: PMC9702651 DOI: 10.1007/s42770-022-00875-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) quickly spread worldwide, leading coronavirus disease 2019 (COVID-19) to hit pandemic level less than 4 months after the first official cases. Hence, the search for drugs and vaccines that could prevent or treat infections by SARS-CoV-2 began, intending to reduce a possible collapse of health systems. After 2 years, efforts to find therapies to treat COVID-19 continue. However, there is still much to be understood about the virus' pathology. Tools such as transcriptomics have been used to understand the impact of SARS-CoV-2 on different cells isolated from various tissues, leaving datasets in the databases that integrate genes and differentially expressed pathways during SARS-CoV-2 infection. After retrieving transcriptome datasets from different human cells infected with SARS-CoV-2 available in the database, we performed an integrative analysis associated with deep learning algorithms to determine differentially expressed targets mainly after infection. The targets found represented a fructose transporter (GLUT5) and a component of proteasome 26s. These targets were then molecularly modeled, followed by molecular docking that identified potential inhibitors for both structures. Once the inhibition of structures that have the expression increased by the virus can represent a strategy for reducing the viral replication by selecting infected cells, associating these bioinformatics tools, therefore, can be helpful in the screening of molecules being tested for new uses, saving financial resources, time, and making a personalized screening for each infectious disease.
Collapse
|
9
|
Huang C, Shi C, Li Z, Wang W, Ming D, Gao Y, Liu H, Ma X, Wang F. Pyrroloquinoline quinone regulates glycolipid metabolism in the jejunum via inhibiting AMPK phosphorylation of weaned pigs. Food Funct 2022; 13:9610-9621. [PMID: 36004536 DOI: 10.1039/d2fo00281g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Maintenance of intestinal metabolic function is important for optimal growth performance in post-weaning pigs. This study aimed to evaluate the effect of pyrroloquinoline quinone (PQQ) on maintaining intestinal glycolipid metabolism in weaned pigs. Seventy-two Duroc × Landrace × Yorkshire crossbred pigs were divided into two groups: pigs fed a basal diet (CTRL group) and pigs fed a basal diet supplemented with 3.0 mg kg-1 PQQ (PQQ group). On d 14, serum was harvested from six pigs per group and the pigs were slaughtered to sample jejunal tissue. Compared with the CTRL group, pigs in the PQQ group had increased average daily gain (P < 0.05), decreased feed : gain (P < 0.05) and tended to have a reduced diarrhea ratio (P = 0.057). Jejunal villus height and villus height/crypt depth ratio were increased, and the crypt depth was decreased in the PQQ group (P < 0.01). The proteomics results showed that PQQ supplementation acted on three metabolic pathways, type I diabetes mellitus, the pancreatic secretion pathway and immune-related signalling. Compared with the CTRL group, PQQ supplementation increased (P < 0.05) serum insulin and jejunal mucosal pyruvate, triglyceride, total cholesterol and low-density lipoprotein cholesterol in the pigs. Jejunal mucosal lactic dehydrogenase and high-density lipoprotein cholesterol levels in the pigs were decreased by PQQ supplementation (P < 0.05). In addition, PQQ supplementation reduced glucose transporter 5 and phosphorylated-AMP-activated protein kinase expression in the jejunal mucosa of the pigs (P < 0.05). In conclusion, dietary supplementation with PQQ improved the growth performance and jejunal morphology and regulated glycolipid metabolism via inhibiting AMPK phosphorylation in weaned pigs.
Collapse
Affiliation(s)
- Caiyun Huang
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Chenyu Shi
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Zhe Li
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Wenhui Wang
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Dongxu Ming
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Youjun Gao
- Changmao Biochemical Engineering Company, Changzhou 213000, China
| | - Hu Liu
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Xi Ma
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| | - Fenglai Wang
- State Key Lab of Animal Nutrition, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
10
|
Xie J, Shi S, Liu Y, Wang S, Rajput SA, Song T. Fructose metabolism and its role in pig production: A mini-review. Front Nutr 2022; 9:922051. [PMID: 35967778 PMCID: PMC9373593 DOI: 10.3389/fnut.2022.922051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022] Open
Abstract
Epidemiological studies have shown that excessive intake of fructose is largely responsible for the increasing incidence of non-alcoholic fatty liver, obesity, and diabetes. However, depending on the amount of fructose consumption from diet, the metabolic role of fructose is controversial. Recently, there have been increasing studies reporting that diets low in fructose expand the surface area of the gut and increase nutrient absorption in mouse model, which is widely used in fructose-related studies. However, excessive fructose consumption spills over from the small intestine into the liver for steatosis and increases the risk of colon cancer. Therefore, suitable animal models may be needed to study fructose-induced metabolic changes. Along with its use in global meat production, pig is well-known as a biomedical model with an advantage over murine and other animal models as it has similar nutrition and metabolism to human in anatomical and physiological aspects. Here, we review the characteristics and metabolism of fructose and summarize observations of fructose in pig reproduction, growth, and development as well as acting as a human biomedical model. This review highlights fructose metabolism from the intestine to the blood cycle and presents the critical role of fructose in pig, which could provide new strategies for curbing human metabolic diseases and promoting pig production.
Collapse
Affiliation(s)
- Jiahao Xie
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shiyi Shi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yucheng Liu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shaoshuai Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shahid Ali Rajput
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture Multan, Multan, Pakistan
| | - Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
11
|
Szablewski L. Glucose transporters as markers of diagnosis and prognosis in cancer diseases. Oncol Rev 2022; 16:561. [PMID: 35340885 PMCID: PMC8941341 DOI: 10.4081/oncol.2022.561] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/24/2021] [Indexed: 11/22/2022] Open
Abstract
The primary metabolic substrate for cells is glucose, which acts as both a source of energy and a substrate in several processes. However, being lipophilic, the cell membrane is impermeable to glucose and specific carrier proteins are needed to allow transport. In contrast to normal cells, cancer cells are more likely to generate energy by glycolysis; as this process generates fewer molecules of adenosine triphosphate (ATP) than complete oxidative breakdown, more glucose molecules are needed. The increased demand for glucose in cancer cells is satisfied by overexpression of a number of glucose transporters, and decreased levels of others. As specific correlations have been observed between the occurrence of cancer and the expression of glucose carrier proteins, the presence of changes in expression of glucose transporters may be treated as a marker of diagnosis and/or prognosis for cancer patients.
Collapse
|
12
|
Chandris P, Giannouli CC, Panayotou G. Imaging Approaches for the Study of Metabolism in Real Time Using Genetically Encoded Reporters. Front Cell Dev Biol 2022; 9:725114. [PMID: 35118062 PMCID: PMC8804523 DOI: 10.3389/fcell.2021.725114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Metabolism comprises of two axes in order to serve homeostasis: anabolism and catabolism. Both axes are interbranched with the so-called bioenergetics aspect of metabolism. There is a plethora of analytical biochemical methods to monitor metabolites and reactions in lysates, yet there is a rising need to monitor, quantify and elucidate in real time the spatiotemporal orchestration of complex biochemical reactions in living systems and furthermore to analyze the metabolic effect of chemical compounds that are destined for the clinic. The ongoing technological burst in the field of imaging creates opportunities to establish new tools that will allow investigators to monitor dynamics of biochemical reactions and kinetics of metabolites at a resolution that ranges from subcellular organelle to whole system for some key metabolites. This article provides a mini review of available toolkits to achieve this goal but also presents a perspective on the open space that can be exploited to develop novel methodologies that will merge classic biochemistry of metabolism with advanced imaging. In other words, a perspective of "watching metabolism in real time."
Collapse
Affiliation(s)
- Panagiotis Chandris
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
| | | | - George Panayotou
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
| |
Collapse
|
13
|
Vallon V, Nakagawa T. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021; 12:2995-3044. [PMID: 34964123 PMCID: PMC9832976 DOI: 10.1002/cphy.c210030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The proximal tubule of the kidney is programmed to reabsorb all filtered glucose and fructose. Glucose is taken up by apical sodium-glucose cotransporters SGLT2 and SGLT1 whereas SGLT5 and potentially SGLT4 and GLUT5 have been implicated in apical fructose uptake. The glucose taken up by the proximal tubule is typically not metabolized but leaves via the basolateral facilitative glucose transporter GLUT2 and is returned to the systemic circulation or used as an energy source by distal tubular segments after basolateral uptake via GLUT1. The proximal tubule generates new glucose in metabolic acidosis and the postabsorptive phase, and fructose serves as an important substrate. In fact, under physiological conditions and intake, fructose taken up by proximal tubules is primarily utilized for gluconeogenesis. In the diabetic kidney, glucose is retained and gluconeogenesis enhanced, the latter in part driven by fructose. This is maladaptive as it sustains hyperglycemia. Moreover, renal glucose retention is coupled to sodium retention through SGLT2 and SGLT1, which induces secondary deleterious effects. SGLT2 inhibitors are new anti-hyperglycemic drugs that can protect the kidneys and heart from failing independent of kidney function and diabetes. Dietary excess of fructose also induces tubular injury. This can be magnified by kidney formation of fructose under pathological conditions. Fructose metabolism is linked to urate formation, which partially accounts for fructose-induced tubular injury, inflammation, and hemodynamic alterations. Fructose metabolism favors glycolysis over mitochondrial respiration as urate suppresses aconitase in the tricarboxylic acid cycle, and has been linked to potentially detrimental aerobic glycolysis (Warburg effect). © 2022 American Physiological Society. Compr Physiol 12:2995-3044, 2022.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, USA,Department of Pharmacology, University of California San Diego, La Jolla, California, USA,VA San Diego Healthcare System, San Diego, California, USA,Correspondence to and
| | - Takahiko Nakagawa
- Division of Nephrology, Rakuwakai-Otowa Hospital, Kyoto, Japan,Correspondence to and
| |
Collapse
|
14
|
Skagen C, Nyman TA, Peng XR, O'Mahony G, Kase ET, Rustan AC, Thoresen GH. Chronic treatment with terbutaline increases glucose and oleic acid oxidation and protein synthesis in cultured human myotubes. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100039. [PMID: 34909668 PMCID: PMC8663959 DOI: 10.1016/j.crphar.2021.100039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/04/2022] Open
Abstract
Objective In vivo studies have reported several beneficial metabolic effects of β-adrenergic receptor agonist administration in skeletal muscle, including increased glucose uptake, fatty acid metabolism, lipolysis and mitochondrial biogenesis. Although these effects have been widely studied in vivo, the in vitro data are limited to mouse and rat cell lines. Therefore, we sought to discover the effects of the β2-adrenergic receptor agonist terbutaline on metabolism and protein synthesis in human primary skeletal muscle cells. Methods Human cultured myotubes were exposed to terbutaline in various concentrations (0.01–30 μM) for 4 or 96 h. Thereafter uptake of [14C]deoxy-D-glucose, oxydation of [14C]glucose and [14C]oleic acid were measured. Incorporation of [14C]leucine, gene expression by qPCR and proteomics analyses by mass spectrometry by the STAGE-TIP method were performed after 96 h exposure to 1 and 10 μM of terbutaline. Results The results showed that 4 h treatment with terbutaline in concentrations up to 1 μM increased glucose uptake in human myotubes, but also decreased both glucose and oleic acid oxidation along with oleic acid uptake in concentrations of 10–30 μM. Moreover, administration of terbutaline for 96 h increased glucose uptake (in terbutaline concentrations up to 1 μM) and oxidation (1 μM), as well as oleic acid oxidation (0.1–30 μM), leucine incorporation into cellular protein (1–10 μM) and upregulated several pathways related to mitochondrial metabolism (1 μM). Data are available via ProteomeXchange with identifier PXD024063. Conclusion These results suggest that β2-adrenergic receptor have direct effects in human skeletal muscle affecting fuel metabolism and net protein synthesis, effects that might be favourable for both type 2 diabetes and muscle wasting disorders. The metabolic effects of terbutaline were studied in human primary myotubes. Acute treatment with terbutaline increased glucose uptake. Chronic treatment with terbutaline increased glucose and oleic acid oxidation. Chronic treatment with terbutaline increased protein synthesis. Proteomics analysis revealed an increase in mitochondrial proteins.
Collapse
Affiliation(s)
- Christine Skagen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Norway
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gavin O'Mahony
- Medicinal Chemsitry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Arild Chr Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
15
|
Herman MA, Birnbaum MJ. Molecular aspects of fructose metabolism and metabolic disease. Cell Metab 2021; 33:2329-2354. [PMID: 34619074 PMCID: PMC8665132 DOI: 10.1016/j.cmet.2021.09.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023]
Abstract
Excessive sugar consumption is increasingly considered as a contributor to the emerging epidemics of obesity and the associated cardiometabolic disease. Sugar is added to the diet in the form of sucrose or high-fructose corn syrup, both of which comprise nearly equal amounts of glucose and fructose. The unique aspects of fructose metabolism and properties of fructose-derived metabolites allow for fructose to serve as a physiological signal of normal dietary sugar consumption. However, when fructose is consumed in excess, these unique properties may contribute to the pathogenesis of cardiometabolic disease. Here, we review the biochemistry, genetics, and physiology of fructose metabolism and consider mechanisms by which excessive fructose consumption may contribute to metabolic disease. Lastly, we consider new therapeutic options for the treatment of metabolic disease based upon this knowledge.
Collapse
Affiliation(s)
- Mark A Herman
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | | |
Collapse
|
16
|
Rawat A, Morrison BM. Metabolic Transporters in the Peripheral Nerve-What, Where, and Why? Neurotherapeutics 2021; 18:2185-2199. [PMID: 34773210 PMCID: PMC8804006 DOI: 10.1007/s13311-021-01150-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
Cellular metabolism is critical not only for cell survival, but also for cell fate, function, and intercellular communication. There are several different metabolic transporters expressed in the peripheral nervous system, and they each play important roles in maintaining cellular energy. The major source of energy in the peripheral nervous system is glucose, and glucose transporters 1 and 3 are expressed and allow blood glucose to be imported and utilized by peripheral nerves. There is also increasing evidence that other sources of energy, particularly monocarboxylates such as lactate that are transported primarily by monocarboxylate transporters 1 and 2 in peripheral nerves, can be efficiently utilized by peripheral nerves. Finally, emerging evidence supports an important role for connexins and possibly pannexins in the supply and regulation of metabolic energy. In this review, we will first define these critical metabolic transporter subtypes and then examine their localization in the peripheral nervous system. We will subsequently discuss the evidence, which comes both from experiments in animal models and observations from human diseases, supporting critical roles played by these metabolic transporters in the peripheral nervous system. Despite progress made in understanding the function of these transporters, many questions and some discrepancies remain, and these will also be addressed throughout this review. Peripheral nerve metabolism is fundamentally important and renewed interest in these pathways should help to answer many of these questions and potentially provide new treatments for neurologic diseases that are partly, or completely, caused by disruption of metabolism.
Collapse
Affiliation(s)
- Atul Rawat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brett M Morrison
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
17
|
Annandale M, Daniels LJ, Li X, Neale JPH, Chau AHL, Ambalawanar HA, James SL, Koutsifeli P, Delbridge LMD, Mellor KM. Fructose Metabolism and Cardiac Metabolic Stress. Front Pharmacol 2021; 12:695486. [PMID: 34267663 PMCID: PMC8277231 DOI: 10.3389/fphar.2021.695486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease is one of the leading causes of mortality in diabetes. High fructose consumption has been linked with the development of diabetes and cardiovascular disease. Serum and cardiac tissue fructose levels are elevated in diabetic patients, and cardiac production of fructose via the intracellular polyol pathway is upregulated. The question of whether direct myocardial fructose exposure and upregulated fructose metabolism have potential to induce cardiac fructose toxicity in metabolic stress settings arises. Unlike tightly-regulated glucose metabolism, fructose bypasses the rate-limiting glycolytic enzyme, phosphofructokinase, and proceeds through glycolysis in an unregulated manner. In vivo rodent studies have shown that high dietary fructose induces cardiac metabolic stress and functional disturbance. In vitro, studies have demonstrated that cardiomyocytes cultured in high fructose exhibit lipid accumulation, inflammation, hypertrophy and low viability. Intracellular fructose mediates post-translational modification of proteins, and this activity provides an important mechanistic pathway for fructose-related cardiomyocyte signaling and functional effect. Additionally, fructose has been shown to provide a fuel source for the stressed myocardium. Elucidating the mechanisms of fructose toxicity in the heart may have important implications for understanding cardiac pathology in metabolic stress settings.
Collapse
Affiliation(s)
- M Annandale
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L J Daniels
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - X Li
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J P H Neale
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - A H L Chau
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - H A Ambalawanar
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - S L James
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - P Koutsifeli
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L M D Delbridge
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - K M Mellor
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Staubach P, Koch AK, Langhorst J, Schreiber S, Röcken C, Helwig U. Expression of the fructose transporter GLUT5 in patients with fructose malabsorption. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2021; 59:531-539. [PMID: 34130330 DOI: 10.1055/a-1156-4386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Patients with abdominal symptoms are frequently diagnosed with fructose malabsorption (FM). Fructose is absorbed by monosaccharide transporters located in the brush border of the human small intestine. The aim of this study was to investigate the histoanatomical distribution of the main fructose transporter GLUT5. MATERIALS AND METHODS We studied 223 patients diagnosed with FM by a hydrogen breath test and grouped according to their response to a fructose-free diet. The control group were 42 healthy individuals and 29 patients with celiac disease (CD). The fructose breath test was done with 50 g fructose. The expression of Glut5 in duodenal biopsy specimens was studied by immunohistochemistry. The Kruskal-Wallis-test and Mann-Whitney U-test were used to carry out the statistical analysis. RESULTS The histoanatomical expression pattern of GLUT5 did not differ significantly between those patients with FM who responded completely to a fructose-free diet (n = 183) and healthy individuals (n = 42); nor did it correlate to H2 production measured in fructose breath testing. In patients with FM, the GLUT5 expression pattern did not differ between those individuals responding to a fructose-free diet and those who did not. However, GLUT5 expression pattern was significantly different in patients with CD (n = 29) compared to patients with FM and to healthy individuals (p = 0.009). CONCLUSION GLUT5 expression patterns are not be related to adult patients with FM. However, in secondary malabsorption, a decreased GLUT5 expression was found. Further investigation is needed to understand the essential factors in FM and the influence on functional gastrointestinal disorders.
Collapse
Affiliation(s)
- Pia Staubach
- Department of Pathology, Christian-Albrechts-University Kiel, Germany
| | - Anna Katharina Koch
- Department of Internal and Integrative Medicine, Kliniken Essen-Mitte, University of Duisburg-Essen, Germany
| | - Jost Langhorst
- Department of Internal and Integrative Medicine, Kliniken Essen-Mitte, University of Duisburg-Essen, Germany
| | - Stefan Schreiber
- Department of Internal Medicine, Christian-Albrechts-University Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University Kiel, Germany
| | - Ulf Helwig
- Specialist Practice for Internal Medicine, Oldenburg, Oldenburg Germany.,Department of Internal Medicine, Christian-Albrechts-University Kiel, Germany
| |
Collapse
|
19
|
Drew D, North RA, Nagarathinam K, Tanabe M. Structures and General Transport Mechanisms by the Major Facilitator Superfamily (MFS). Chem Rev 2021; 121:5289-5335. [PMID: 33886296 PMCID: PMC8154325 DOI: 10.1021/acs.chemrev.0c00983] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/12/2022]
Abstract
The major facilitator superfamily (MFS) is the largest known superfamily of secondary active transporters. MFS transporters are responsible for transporting a broad spectrum of substrates, either down their concentration gradient or uphill using the energy stored in the electrochemical gradients. Over the last 10 years, more than a hundred different MFS transporter structures covering close to 40 members have provided an atomic framework for piecing together the molecular basis of their transport cycles. Here, we summarize the remarkable promiscuity of MFS members in terms of substrate recognition and proton coupling as well as the intricate gating mechanisms undergone in achieving substrate translocation. We outline studies that show how residues far from the substrate binding site can be just as important for fine-tuning substrate recognition and specificity as those residues directly coordinating the substrate, and how a number of MFS transporters have evolved to form unique complexes with chaperone and signaling functions. Through a deeper mechanistic description of glucose (GLUT) transporters and multidrug resistance (MDR) antiporters, we outline novel refinements to the rocker-switch alternating-access model, such as a latch mechanism for proton-coupled monosaccharide transport. We emphasize that a full understanding of transport requires an elucidation of MFS transporter dynamics, energy landscapes, and the determination of how rate transitions are modulated by lipids.
Collapse
Affiliation(s)
- David Drew
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Rachel A. North
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Kumar Nagarathinam
- Center
of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, D-23538, Lübeck, Germany
| | - Mikio Tanabe
- Structural
Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Oho 1-1, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|
20
|
Zhou L, Lu R, Liu Q, Xiao B, Hai L, Guo L, Wu Y, Zheng Y. Two branched fructose modification improves tumor targeting delivery of liposomes to breast cancer in intro and in vivo. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Gonçalves AS, Andrade N, Martel F. Intestinal fructose absorption: Modulation and relation to human diseases. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Elferink H, Bruekers JPJ, Veeneman GH, Boltje TJ. A comprehensive overview of substrate specificity of glycoside hydrolases and transporters in the small intestine : "A gut feeling". Cell Mol Life Sci 2020; 77:4799-4826. [PMID: 32506169 PMCID: PMC7658089 DOI: 10.1007/s00018-020-03564-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023]
Abstract
The human body is able to process and transport a complex variety of carbohydrates, unlocking their nutritional value as energy source or as important building block. The endogenous glycosyl hydrolases (glycosidases) and glycosyl transporter proteins located in the enterocytes of the small intestine play a crucial role in this process and digest and/or transport nutritional sugars based on their structural features. It is for these reasons that glycosidases and glycosyl transporters are interesting therapeutic targets to combat sugar related diseases (such as diabetes) or to improve drug delivery. In this review we provide a detailed overview focused on the molecular structure of the substrates involved as a solid base to start from and to fuel research in the area of therapeutics and diagnostics.
Collapse
Affiliation(s)
- Hidde Elferink
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525, Nijmegen, The Netherlands
| | - Jeroen P J Bruekers
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525, Nijmegen, The Netherlands
| | | | - Thomas J Boltje
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525, Nijmegen, The Netherlands.
| |
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
|
Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 2020; 472:1207-1248. [PMID: 32829466 PMCID: PMC7462918 DOI: 10.1007/s00424-020-02439-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 12/23/2022]
Abstract
Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstr 6, 97070, Würzburg, Germany.
| |
Collapse
|
26
|
Holman GD. Structure, function and regulation of mammalian glucose transporters of the SLC2 family. Pflugers Arch 2020; 472:1155-1175. [PMID: 32591905 PMCID: PMC7462842 DOI: 10.1007/s00424-020-02411-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
The SLC2 genes code for a family of GLUT proteins that are part of the major facilitator superfamily (MFS) of membrane transporters. Crystal structures have recently revealed how the unique protein fold of these proteins enables the catalysis of transport. The proteins have 12 transmembrane spans built from a replicated trimer substructure. This enables 4 trimer substructures to move relative to each other, and thereby alternately opening and closing a cleft to either the internal or the external side of the membrane. The physiological substrate for the GLUTs is usually a hexose but substrates for GLUTs can include urate, dehydro-ascorbate and myo-inositol. The GLUT proteins have varied physiological functions that are related to their principal substrates, the cell type in which the GLUTs are expressed and the extent to which the proteins are associated with subcellular compartments. Some of the GLUT proteins translocate between subcellular compartments and this facilitates the control of their function over long- and short-time scales. The control of GLUT function is necessary for a regulated supply of metabolites (mainly glucose) to tissues. Pathophysiological abnormalities in GLUT proteins are responsible for, or associated with, clinical problems including type 2 diabetes and cancer and a range of tissue disorders, related to tissue-specific GLUT protein profiles. The availability of GLUT crystal structures has facilitated the search for inhibitors and substrates and that are specific for each GLUT and that can be used therapeutically. Recent studies are starting to unravel the drug targetable properties of each of the GLUT proteins.
Collapse
Affiliation(s)
- Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| |
Collapse
|
27
|
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
|
28
|
Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease. Pflugers Arch 2020; 472:1273-1298. [PMID: 32591906 PMCID: PMC7462924 DOI: 10.1007/s00424-020-02417-x] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
A family of facilitative glucose transporters (GLUTs) is involved in regulating tissue-specific glucose uptake and metabolism in the liver, skeletal muscle, and adipose tissue to ensure homeostatic control of blood glucose levels. Reduced glucose transport activity results in aberrant use of energy substrates and is associated with insulin resistance and type 2 diabetes. It is well established that GLUT2, the main regulator of hepatic hexose flux, and GLUT4, the workhorse in insulin- and contraction-stimulated glucose uptake in skeletal muscle, are critical contributors in the control of whole-body glycemia. However, the molecular mechanism how insulin controls glucose transport across membranes and its relation to impaired glycemic control in type 2 diabetes remains not sufficiently understood. An array of circulating metabolites and hormone-like molecules and potential supplementary glucose transporters play roles in fine-tuning glucose flux between the different organs in response to an altered energy demand.
Collapse
|
29
|
Finessi M, Bisi G, Deandreis D. Hyperglycemia and 18F-FDG PET/CT, issues and problem solving: a literature review. Acta Diabetol 2020; 57:253-262. [PMID: 31304560 DOI: 10.1007/s00592-019-01385-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/24/2019] [Indexed: 12/26/2022]
Abstract
Positron emission tomography/computed tomography (PET/CT) is a standard procedure for imaging cancer commonly used in the clinical practice for several diseases, in particular for cancer staging, restaging, treatment monitoring and radiation therapy planning. Despite the availability of many radiotracers, 18F-fluoro-2-deoxy-2-D-glucose ([18F]FDG) is the most used. International PET/CT guidelines propose protocols for patients' correct preparation before [18F]FDG injection, in particular with the regard of diabetic patients and therapy management. Hyperglycemic conditions and oral or insulin medication showed advantages and disadvantages on PET/CT scan accuracy: A correct knowledge of effects of these conditions on glucose metabolism assumes a fundamental role on patients management before [18F]FDG PET/CT scan.
Collapse
Affiliation(s)
- Monica Finessi
- Division of Nuclear Medicine, Department of Medical Sciences, University of Turin, AOU Città della Salute e della Scienza, Turin, Italy.
| | - Gianni Bisi
- Division of Nuclear Medicine, Department of Medical Sciences, University of Turin, AOU Città della Salute e della Scienza, Turin, Italy
| | - Désirée Deandreis
- Division of Nuclear Medicine, Department of Medical Sciences, University of Turin, AOU Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
30
|
Pu Y, Zhang H, Peng Y, Fu Q, Yue Q, Zhao Y, Guo L, Wu Y. Dual-targeting liposomes with active recognition of GLUT5 and αvβ3 for triple-negative breast cancer. Eur J Med Chem 2019; 183:111720. [DOI: 10.1016/j.ejmech.2019.111720] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/24/2019] [Accepted: 09/18/2019] [Indexed: 01/01/2023]
|
31
|
Liu B, Wang Y, Zhang Y, Yan B. Mechanisms of Protective Effects of SGLT2 Inhibitors in Cardiovascular Disease and Renal Dysfunction. Curr Top Med Chem 2019; 19:1818-1849. [PMID: 31456521 DOI: 10.2174/1568026619666190828161409] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is one of the most common forms of the disease worldwide. Hyperglycemia and insulin resistance play key roles in type 2 diabetes mellitus. Renal glucose reabsorption is an essential feature in glycaemic control. Kidneys filter 160 g of glucose daily in healthy subjects under euglycaemic conditions. The expanding epidemic of diabetes leads to a prevalence of diabetes-related cardiovascular disorders, in particular, heart failure and renal dysfunction. Cellular glucose uptake is a fundamental process for homeostasis, growth, and metabolism. In humans, three families of glucose transporters have been identified, including the glucose facilitators GLUTs, the sodium-glucose cotransporter SGLTs, and the recently identified SWEETs. Structures of the major isoforms of all three families were studied. Sodium-glucose cotransporter (SGLT2) provides most of the capacity for renal glucose reabsorption in the early proximal tubule. A number of cardiovascular outcome trials in patients with type 2 diabetes have been studied with SGLT2 inhibitors reducing cardiovascular morbidity and mortality. The current review article summarises these aspects and discusses possible mechanisms with SGLT2 inhibitors in protecting heart failure and renal dysfunction in diabetic patients. Through glucosuria, SGLT2 inhibitors reduce body weight and body fat, and shift substrate utilisation from carbohydrates to lipids and, possibly, ketone bodies. These pleiotropic effects of SGLT2 inhibitors are likely to have contributed to the results of the EMPA-REG OUTCOME trial in which the SGLT2 inhibitor, empagliflozin, slowed down the progression of chronic kidney disease and reduced major adverse cardiovascular events in high-risk individuals with type 2 diabetes. This review discusses the role of SGLT2 in the physiology and pathophysiology of renal glucose reabsorption and outlines the unexpected logic of inhibiting SGLT2 in the diabetic kidney.
Collapse
Affiliation(s)
- Ban Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuliang Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Biao Yan
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Yin F, Lan R, Wu Z, Wang Z, Wu H, Li Z, Yu H, Zhao Z, Li H. Yupingfeng polysaccharides enhances growth performance in Qingyuan partridge chicken by up-regulating the mRNA expression of SGLT1, GLUT2 and GLUT5. Vet Med Sci 2019; 5:451-461. [PMID: 30973212 PMCID: PMC6682804 DOI: 10.1002/vms3.167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The ban on the use of antibiotic in feed encouraged nutritionists to using alternatives to maintain growth performance and intestinal function of broilers. This study was conducted to evaluate the effects of Yupingfeng polysaccharides (YP) supplementation on growth performance and expression of SGLT1, GLUT2 and GLUT5 in Qingyuan partridge chicken. Experiment 1: a total of 540 chickens were randomly allocated to five groups with six replication. Dietary treatments were: (1) CON (control group), basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP; (4) T3, CON + 2 g kg-1 YP; (5) T4, CON + 4 g kg-1 YP. Experiment 2, a total of 162 were randomly allocated to three groups with three replication. Dietary treatments were: (1) CON, basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP. From days 1 to 14 and overall, chicken fed T1 diet had higher ADG. On day 42, there was increased villus height of jejunum in T1 group. On days 14 and 28, there was decreased villus height of duodenum and jejunum in T2 group. In duodenum, the expression of SGLT1 (days 21, 35 and 42), GLUT2 (days 7, 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21 and 28) was increased with YP supplementation. In jejunum, the expression of SGLT1 (days 7, 14, 21, 28 and 35), GLUT2 (days 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. In ileum, the expression of SGLT1 (days 7, 21, 35 and 42), GLUT2 (days 7, 14, 21 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. Dietary YP supplementation improves growth performance and expression of SGLT1, GLUT2 and GLUT5 in intestine.
Collapse
Affiliation(s)
- Fuquan Yin
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Ruixia Lan
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Zhengmin Wu
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Zhijing Wang
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Haohao Wu
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Zhiming Li
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Hui Yu
- School of Life Science and EngineeringFoshan UniversityFoshanGuangdongP.R. China
| | - Zhihui Zhao
- Department of Animal ScienceCollege of AgricultureGuangdong Ocean UniversityZhanjiangGuangdongP.R. China
| | - Hua Li
- School of Life Science and EngineeringFoshan UniversityFoshanGuangdongP.R. China
| |
Collapse
|
33
|
Pereira MT, Malik M, Nostro JA, Mahler GJ, Musselman LP. Effect of dietary additives on intestinal permeability in both Drosophila and a human cell co-culture. Dis Model Mech 2018; 11:dmm034520. [PMID: 30504122 PMCID: PMC6307910 DOI: 10.1242/dmm.034520] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/06/2018] [Indexed: 12/13/2022] Open
Abstract
Increased intestinal barrier permeability has been correlated with aging and disease, including type 2 diabetes, Crohn's disease, celiac disease, multiple sclerosis and irritable bowel syndrome. The prevalence of these ailments has risen together with an increase in industrial food processing and food additive consumption. Additives, including sugar, metal oxide nanoparticles, surfactants and sodium chloride, have all been suggested to increase intestinal permeability. We used two complementary model systems to examine the effects of food additives on gut barrier function: a Drosophila in vivo model and an in vitro human cell co-culture model. Of the additives tested, intestinal permeability was increased most dramatically by high sugar. High sugar also increased feeding but reduced gut and overall animal size. We also examined how food additives affected the activity of a gut mucosal defense factor, intestinal alkaline phosphatase (IAP), which fluctuates with bacterial load and affects intestinal permeability. We found that high sugar reduced IAP activity in both models. Artificial manipulation of the microbiome influenced gut permeability in both models, revealing a complex relationship between the two. This study extends previous work in flies and humans showing that diet can play a role in the health of the gut barrier. Moreover, simple models can be used to study mechanisms underlying the effects of diet on gut permeability and function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Matthew T Pereira
- Department of Biological Sciences, Binghamton University, Binghamton, New York 13902, USA
| | - Mridu Malik
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
| | - Jillian A Nostro
- Department of Biological Sciences, Binghamton University, Binghamton, New York 13902, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
| | | |
Collapse
|
34
|
Carreño D, Corro N, Torres-Estay V, Véliz LP, Jaimovich R, Cisternas P, San Francisco IF, Sotomayor PC, Tanasova M, Inestrosa NC, Godoy AS. Fructose and prostate cancer: toward an integrated view of cancer cell metabolism. Prostate Cancer Prostatic Dis 2018; 22:49-58. [PMID: 30104655 DOI: 10.1038/s41391-018-0072-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/13/2018] [Accepted: 06/29/2018] [Indexed: 01/07/2023]
Abstract
Activation of glucose transporter-1 (Glut-1) gene expression is a molecular feature of cancer cells that increases glucose uptake and metabolism. Increased glucose uptake is the basis for the clinical localization of primary tumors using positron emission tomography (PET) and 2-deoxy-2-[18F]-fluoro-D-glucose (FDG) as a radiotracer. However, previous studies have demonstrated that a considerable number of cancers, which include prostate cancer (CaP), express low to undetectable levels of Glut-1 and that FDG-PET has limited clinical applicability in CaP. This observation could be explained by a low metabolic activity of CaP cells that may be overcome using different hexoses, such as fructose, as the preferred energy source. However, these hypotheses have not been examined critically in CaP. This review article summarizes what is currently known about transport and metabolism of hexoses, and more specifically fructose, in CaP and provides experimental evidences indicating that CaP cells may have increased capacity to transport and metabolize fructose in vitro and in vivo. Moreover, this review highlights recent findings that allow better understanding of how metabolism of fructose may regulate cancer cell proliferation and how fructose uptake and metabolism, through the de novo lipogenesis pathway, may provide new opportunities for CaP early diagnosis, staging, and treatment.
Collapse
Affiliation(s)
- Daniela Carreño
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Néstor Corro
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Loreto P Véliz
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE), Department of Cell Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Paula C Sotomayor
- Center for Integrative Medicine and Innovative Science, Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Marina Tanasova
- Department of Chemistry, Michigan Technological University, Houghton, MI, 49931, USA
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Department of Cell Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro S Godoy
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
35
|
Wang Y, Wu S, Huang C, Li Y, Zhao H, Kasim V. Yin Yang 1 promotes the Warburg effect and tumorigenesis via glucose transporter GLUT3. Cancer Sci 2018; 109:2423-2434. [PMID: 29869834 PMCID: PMC6113438 DOI: 10.1111/cas.13662] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/28/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer cells typically shift their metabolism to aerobic glycolysis to fulfill the demand of energy and macromolecules to support their proliferation. Glucose transporter (GLUT) family‐mediated glucose transport is the pacesetter of aerobic glycolysis and, thus, is critical for tumor cell metabolism. Yin Yang 1 (YY1) is an oncogene crucial for tumorigenesis; however, its role in tumor cell glucose metabolism remains unclear. Here, we revealed that YY1 activates GLUT3 transcription by directly binding to its promoter and, concomitantly, enhances tumor cell aerobic glycolysis. This regulatory effect of YY1 on glucose entry into the cells is critical for YY1‐induced tumor cell proliferation and tumorigenesis. Intriguingly, YY1 regulation of GLUT3 expression, and, subsequently, of tumor cell aerobic glycolysis and tumorigenesis, occurs p53‐independently. Our results also showed that clinical drug oxaliplatin suppresses colon carcinoma cell proliferation by inhibiting the YY1/GLUT3 axis. Together, these results link YY1's tumorigenic potential with the critical first step of aerobic glycolysis. Thus, our novel findings not only provide new insights into the complex role of YY1 in tumorigenesis but also indicate the potential of YY1 as a target for cancer therapy irrespective of the p53 status.
Collapse
Affiliation(s)
- Yali Wang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shourong Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing, China
| | - Can Huang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yanjun Li
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hezhao Zhao
- Cancer Hospital and Chongqing Cancer Institute, Chongqing University, Chongqing, China
| | - Vivi Kasim
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing, China
| |
Collapse
|
36
|
Crouse MS, McLean KJ, Greseth NP, Crosswhite MR, Pereira NN, Ward AK, Reynolds LP, Dahlen CR, Neville BW, Borowicz PP, Caton JS. Maternal nutrition and stage of early pregnancy in beef heifers: Impacts on expression of glucose, fructose, and cationic amino acid transporters in utero-placental tissues. J Anim Sci 2018; 95:5563-5572. [PMID: 29293768 DOI: 10.2527/jas2017.1983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We hypothesized that maternal nutrition and day of gestation would impact utero-placental mRNA expression of the nutrient transporters , , , , and in beef heifers. Crossbred Angus heifers (n = 49) were estrous synchronized, bred via AI, assigned to nutritional treatment (CON = 100% of NRC requirements for 0.45 kg/d gain and RES = 60% of CON) and ovariohysterectomized on d 16, 34, or 50 of gestation (n = 6 to 9/d); Non-bred, non-pregnant (NB-NP) controls were fed the CON diet, not bred, and were ovariohysterectomized on d 16 of the synchronized estrous cycle = 6). The resulting arrangement of treatments was a 2 × 3 factorial + 1 (CON vs. RES × d 16, 34, or 50 + NB-NP controls). Caruncle (CAR), intercaruncular endometrium (ICAR), and fetal membranes (FM [chorioallantois]), were obtained from the pregnant uterine horn (the uterine horn containing the conceptus) immediately after ovariohysterectomy. On d 50 cotyledons (COT), intercotyledonary placenta (ICOT) and amnion (AMN) were also collected. Relative expression of nutrient transporters was determined for each tissue utilizing NB-NP-CAR and NB-NP-ICAR tissues as the baseline. For FM, NB-NP endometrium served as the baseline. There was no interaction of day × treatment ( ≥ 0.20) for any genes in CAR. However, CAR expression of was greater ( < 0.01) on d 16 compared with d 34 and 50, and , , and were greater ( ≤ 0.05) on d 34 compared with d 16 and 50. In ICAR, was the only gene to be influenced by the day × treatment interaction ( = 0.01), being greater in d 50 CON compared with d 34 CON and d 16 and 50 RES. In ICAR, expression of was greater ( < 0.01) on d 16 compared with d 34, and expression of was greater ( < 0.01) on d 34 and 50 compared with d 16. In FM, expression of was greater ( = 0.04) on d 16 compared with d 50 of gestation, and expression of was greater ( < 0.01) on d 34 and 50 compared with d 16. On d 50, expression of , , and expression were all greater ( < 0.05) in AMN compared with COT and ICOT, and expression of was greater ( < 0.01) in ICOT compared with COT and AMN. These data indicate that day was a more influential factor for mRNA expression of utero-placental glucose and cationic AA transporters than maternal nutritional status in heifers during early pregnancy.
Collapse
|
37
|
Shroff GS, Sabloff BS, Truong MT, Carter BW, Viswanathan C. PET/CT Interpretative Pitfalls in Thoracic Malignancies. Semin Ultrasound CT MR 2018; 39:282-288. [DOI: 10.1053/j.sult.2018.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
38
|
Schmidl S, Iancu CV, Choe JY, Oreb M. Ligand Screening Systems for Human Glucose Transporters as Tools in Drug Discovery. Front Chem 2018; 6:183. [PMID: 29888221 PMCID: PMC5980966 DOI: 10.3389/fchem.2018.00183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022] Open
Abstract
Hexoses are the major source of energy and carbon skeletons for biosynthetic processes in all kingdoms of life. Their cellular uptake is mediated by specialized transporters, including glucose transporters (GLUT, SLC2 gene family). Malfunction or altered expression pattern of GLUTs in humans is associated with several widespread diseases including cancer, diabetes and severe metabolic disorders. Their high relevance in the medical area makes these transporters valuable drug targets and potential biomarkers. Nevertheless, the lack of a suitable high-throughput screening system has impeded the determination of compounds that would enable specific manipulation of GLUTs so far. Availability of structural data on several GLUTs enabled in silico ligand screening, though limited by the fact that only two major conformations of the transporters can be tested. Recently, convenient high-throughput microbial and cell-free screening systems have been developed. These remarkable achievements set the foundation for further and detailed elucidation of the molecular mechanisms of glucose transport and will also lead to great progress in the discovery of GLUT effectors as therapeutic agents. In this mini-review, we focus on recent efforts to identify potential GLUT-targeting drugs, based on a combination of structural biology and different assay systems.
Collapse
Affiliation(s)
- Sina Schmidl
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
39
|
Valenzuela-Melgarejo FJ, Caro-Díaz C, Cabello-Guzmán G. Potential Crosstalk between Fructose and Melatonin: A New Role of Melatonin-Inhibiting the Metabolic Effects of Fructose. Int J Endocrinol 2018; 2018:7515767. [PMID: 30154843 PMCID: PMC6092995 DOI: 10.1155/2018/7515767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Increased consumption of energy-dense foods such as fructose-rich syrups represents one of the significant, growing concerns related to the alarming trend of overweight, obesity, and metabolic disorders worldwide. Metabolic pathways affected by fructose involve genes related to lipogenesis/lipolysis, beta-oxidation, mitochondrial biogenesis, gluconeogenesis, oxidative phosphorylation pathways, or altering of circadian production of insulin and leptin. Moreover, fructose can be a risk factor during pregnancy elevating the risk of preterm delivery, hypertension, and metabolic impairment of the mother and fetus. Melatonin is a chronobiotic and homeostatic hormone that can modulate the harmful effects of fructose via clock gene expression and metabolic pathways, modulating the expression of PPARγ, SREBF-1 (SREBP-1), hormone-sensitive lipase, C/EBP-α genes, NRF-1, PGC1α, and uncoupling protein-1. Moreover, this hormone has the capacity in the rat of reverting the harmful effects of fructose, increasing the body weight and weight ratio of the liver, and increasing the body weight and restoring the glycemia from mothers exposed to fructose. The aim of this review is to show the potential crosstalk between fructose and melatonin and their potential role during pregnancy.
Collapse
Affiliation(s)
| | - Claudia Caro-Díaz
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| | - Gerardo Cabello-Guzmán
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| |
Collapse
|
40
|
Transporter Gene Expression and Transference of Fructose in Broiler Chick Intestine. J Poult Sci 2017; 55:137-141. [PMID: 32055166 PMCID: PMC6756492 DOI: 10.2141/jpsa.0170095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022] Open
Abstract
Recent studies have suggested that a high-fructose diet leads to the development of metabolic syndrome in mammals. However, relatively little information is available regarding the absorption of fructose in the chicken intestine. We therefore investigated fructose absorption and its transporters in the chicken small intestine. The gene expression of three transporters (glucose transporter protein member 2 and 5 and sodium-dependent glucose transporter protein 1) in the jejunum of fasted chicks were lower than those in chicks fed ad libitum. The everted intestinal sacs (in vitro method for investigating intestinal absorption) showed that the concentration of fructose uptake rapidly increased within 15 min after incubation, and then gradually increased until 60 min. After 15 min of incubation, fructose uptake in the ad libitum chick intestine was approximately 2-fold that in the fasted intestine and was less than half of the glucose uptake in the ad libitum chick intestine. Our results suggest that fructose is absorbed in the small intestine of chicks and that uptake is decreased by fasting treatment with decreases in the mRNA expression of related transporters.
Collapse
|
41
|
Lau CTS, Chan C, Zhang KY, Roy VAL, Lo KKW. Photophysical, Cellular-Uptake, and Bioimaging Studies of Luminescent Ruthenium(II)-Polypyridine Complexes Containing a d
-Fructose Pendant. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201701038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Chris Tsan-Shing Lau
- Department of Chemistry; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Christina Chan
- Department of Chemistry; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Kenneth Yin Zhang
- Department of Chemistry; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Vellaisamy A. L. Roy
- Department of Material Science and Engineering; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
- State Key Laboratory of Millimeter Waves; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
- Center for Functional Photonics; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
- State Key Laboratory of Millimeter Waves; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
- Center for Functional Photonics; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong P. R. China
| |
Collapse
|
42
|
Geidl-Flueck B, Gerber PA. Insights into the Hexose Liver Metabolism-Glucose versus Fructose. Nutrients 2017; 9:E1026. [PMID: 28926951 PMCID: PMC5622786 DOI: 10.3390/nu9091026] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 12/15/2022] Open
Abstract
High-fructose intake in healthy men is associated with characteristics of metabolic syndrome. Extensive knowledge exists about the differences between hepatic fructose and glucose metabolism and fructose-specific mechanisms favoring the development of metabolic disturbances. Nevertheless, the causal relationship between fructose consumption and metabolic alterations is still debated. Multiple effects of fructose on hepatic metabolism are attributed to the fact that the liver represents the major sink of fructose. Fructose, as a lipogenic substrate and potent inducer of lipogenic enzyme expression, enhances fatty acid synthesis. Consequently, increased hepatic diacylglycerols (DAG) are thought to directly interfere with insulin signaling. However, independently of this effect, fructose may also counteract insulin-mediated effects on liver metabolism by a range of mechanisms. It may drive gluconeogenesis not only as a gluconeogenic substrate, but also as a potent inducer of carbohydrate responsive element binding protein (ChREBP), which induces the expression of lipogenic enzymes as well as gluconeogenic enzymes. It remains a challenge to determine the relative contributions of the impact of fructose on hepatic transcriptome, proteome and allosterome changes and consequently on the regulation of plasma glucose metabolism/homeostasis. Mathematical models exist modeling hepatic glucose metabolism. Future models should not only consider the hepatic adjustments of enzyme abundances and activities in response to changing plasma glucose and insulin/glucagon concentrations, but also to varying fructose concentrations for defining the role of fructose in the hepatic control of plasma glucose homeostasis.
Collapse
Affiliation(s)
- Bettina Geidl-Flueck
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091 Zurich, Switzerland.
| | - Philipp A Gerber
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
43
|
Szablewski L. Distribution of glucose transporters in renal diseases. J Biomed Sci 2017; 24:64. [PMID: 28854935 PMCID: PMC5577680 DOI: 10.1186/s12929-017-0371-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 08/23/2017] [Indexed: 02/06/2023] Open
Abstract
Kidneys play an important role in glucose homeostasis. Renal gluconeogenesis prevents hypoglycemia by releasing glucose into the blood stream. Glucose homeostasis is also due, in part, to reabsorption and excretion of hexose in the kidney.Lipid bilayer of plasma membrane is impermeable for glucose, which is hydrophilic and soluble in water. Therefore, transport of glucose across the plasma membrane depends on carrier proteins expressed in the plasma membrane. In humans, there are three families of glucose transporters: GLUT proteins, sodium-dependent glucose transporters (SGLTs) and SWEET. In kidney, only GLUTs and SGLTs protein are expressed. Mutations within genes that code these proteins lead to different renal disorders and diseases. However, diseases, not only renal, such as diabetes, may damage expression and function of renal glucose transporters.
Collapse
Affiliation(s)
- Leszek Szablewski
- Medical University of Warsaw, Chair & Department of General Biology & Parasitology, Center for Biostructure Research, 5 Chalubinskiego Str., 02-004, Warsaw, Poland.
| |
Collapse
|
44
|
Tripp J, Essl C, Iancu CV, Boles E, Choe JY, Oreb M. Establishing a yeast-based screening system for discovery of human GLUT5 inhibitors and activators. Sci Rep 2017; 7:6197. [PMID: 28740135 PMCID: PMC5524692 DOI: 10.1038/s41598-017-06262-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/05/2017] [Indexed: 01/08/2023] Open
Abstract
Human GLUT5 is a fructose-specific transporter in the glucose transporter family (GLUT, SLC2 gene family). Its substrate-specificity and tissue-specific expression make it a promising target for treatment of diabetes, metabolic syndrome and cancer, but few GLUT5 inhibitors are known. To identify and characterize potential GLUT5 ligands, we developed a whole-cell system based on a yeast strain deficient in fructose uptake, in which GLUT5 transport activity is associated with cell growth in fructose-based media or assayed by fructose uptake in whole cells. The former method is convenient for high-throughput screening of potential GLUT5 inhibitors and activators, while the latter enables detailed kinetic characterization of identified GLUT5 ligands. We show that functional expression of GLUT5 in yeast requires mutations at specific positions of the transporter sequence. The mutated proteins exhibit kinetic properties similar to the wild-type transporter and are inhibited by established GLUT5 inhibitors N-[4-(methylsulfonyl)-2-nitrophenyl]-1,3-benzodioxol-5-amine (MSNBA) and (−)-epicatechin-gallate (ECG). Thus, this system has the potential to greatly accelerate the discovery of compounds that modulate the fructose transport activity of GLUT5.
Collapse
Affiliation(s)
- Joanna Tripp
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Christine Essl
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Eckhard Boles
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
45
|
Gu TT, Song L, Chen TY, Wang X, Zhao XJ, Ding XQ, Yang YZ, Pan Y, Zhang DM, Kong LD. Fructose downregulates miR-330 to induce renal inflammatory response and insulin signaling impairment: Attenuation by morin. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600760] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/03/2017] [Accepted: 02/07/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Ting-Ting Gu
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Tian-Yu Chen
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Xing Wang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Xiao-Juan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Xiao-Qin Ding
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Yan-Zi Yang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Ying Pan
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Dong-Mei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology; School of Life Science; Nanjing University; Nanjing P. R. China
| |
Collapse
|
46
|
Specific regions of the brain are capable of fructose metabolism. Brain Res 2016; 1657:312-322. [PMID: 28034722 DOI: 10.1016/j.brainres.2016.12.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 12/21/2022]
Abstract
High fructose consumption in the Western diet correlates with disease states such as obesity and metabolic syndrome complications, including type II diabetes, chronic kidney disease, and non-alcoholic fatty acid liver disease. Liver and kidneys are responsible for metabolism of 40-60% of ingested fructose, while the physiological fate of the remaining fructose remains poorly understood. The primary metabolic pathway for fructose includes the fructose-transporting solute-like carrier transport proteins 2a (SLC2a or GLUT), including GLUT5 and GLUT9, ketohexokinase (KHK), and aldolase. Bioinformatic analysis of gene expression encoding these proteins (glut5, glut9, khk, and aldoC, respectively) identifies other organs capable of this fructose metabolism. This analysis predicts brain, lymphoreticular tissue, placenta, and reproductive tissues as possible additional organs for fructose metabolism. While expression of these genes is highest in liver, the brain is predicted to have expression levels of these genes similar to kidney. RNA in situ hybridization of coronal slices of adult mouse brains validate the in silico expression of glut5, glut9, khk, and aldoC, and show expression across many regions of the brain, with the most notable expression in the cerebellum, hippocampus, cortex, and olfactory bulb. Dissected samples of these brain regions show KHK and aldolase enzyme activity 5-10 times the concentration of that in liver. Furthermore, rates of fructose oxidation in these brain regions are 15-150 times that of liver slices, confirming the bioinformatics prediction and in situ hybridization data. This suggests that previously unappreciated regions across the brain can use fructose, in addition to glucose, for energy production.
Collapse
|
47
|
Increased Expression of the Glucose Transporter Type 1 Gene Is Associated With Worse Overall Survival in Resected Pancreatic Adenocarcinoma. Pancreas 2016; 45:974-9. [PMID: 26692443 PMCID: PMC4912950 DOI: 10.1097/mpa.0000000000000580] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES There is currently no reliable method to predict the risk of relapse after curative resection of early-stage pancreatic adenocarcinoma. Increased glucose metabolism observed on F-fluorodeoxyglucose positron emission tomography (PET) by malignant cells, the Warburg effect, is a well-known characteristic of the malignant phenotype. We investigated the role of glucose transporter type 1 (GLUT-1) gene expression, a glucose cell plasma membrane transporter, in early-stage pancreatic cancer. METHODS Associations between GLUT-1 gene expression with PET maximum standardized uptake values and histologic grade were investigated in early-stage pancreatic adenocarcinoma patients. Multivariate analysis was conducted to determine predictors of prognosis. Cox proportional hazard model was used for survival analysis. RESULTS Sixty-three patients had GLUT-1 gene analysis performed, and 50 patients had both GLUT-1 analysis and PET scan. Patients with high GLUT-1 gene expression had a decreased overall survival by univariate analysis using Cox proportional hazard model (hazard ratio, 2.82; P = 0.001) and remained significant on multivariate analysis (hazard ratio, 2.54; P = 0.03). There was no correlation of GLUT-1 gene expression with histologic grade or PET maximum standardized uptake values. CONCLUSIONS Increased GLUT-1 gene expression was associated with a decreased overall survival in pancreatic adenocarcinoma. This supports increased GLUT-1 gene expression as a potential prognostic marker in resected pancreatic adenocarcinoma.
Collapse
|
48
|
Kojo A, Yamada K, Yamamoto T. Glucose transporter 5 (GLUT5)-like immunoreactivity is localized in subsets of neurons and glia in the rat brain. J Chem Neuroanat 2016; 74:55-70. [PMID: 27036089 DOI: 10.1016/j.jchemneu.2016.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/24/2016] [Accepted: 03/24/2016] [Indexed: 12/13/2022]
Abstract
This study aimed at examining the distribution of glucose transporter 5 (GLUT5), which preferentially transports fructose, in the rat brain by immunohistochemistry and Western blotting. Small immunoreactive puncta (less than 0.7μm) were sparsely distributed all over the brain, some of which appeared to be associated with microglial processes detected by an anti-ionized calcium-binding adapter molecule 1 (Iba-1) monoclonal antibody. In addition, some of these immunoreactive puncta seemed to be associated with tanycyte processes that were labeled with anti-glial fibrillary acidic protein (GFAP) monoclonal antibody. Ependymal cells were also found to be immunopositive for GLUT5. Furthermore, several noticeable GLUT5 immunoreactive profiles were observed. GLUT5 immunoreactive neurons, confirmed by double staining with neuronal nuclei (NeuN), were seen in the entopeduncular nucleus and lateral hypothalamus. Cerebellar Purkinje cells were immunopositve for GLUT5. Dense accumulation of immunoreactive puncta, some of which were neuronal elements (confirmed by immunoelectron microscopy), were observed in the optic tract and their terminal fields, namely, superior colliculus, pretectum, nucleus of the optic tract, and medial terminal nucleus of the optic tract. In addition to the associated areas of the visual system, the vestibular and cochlear nuclei also contained dense GLUT5 immunoreactive puncta. Western blot analysis of the cerebellum indicated that the antibody used recognized the 33.5 and 37.0kDa bands that were also contained in jejunum and kidney extracts. Thus, these results suggest that GLUT5 may transport fructose in subsets of the glia and neurons for an energy source of these cells.
Collapse
Affiliation(s)
- Akiko Kojo
- Division of Medical Nutrition, Faculty of Healthcare, Tokyo Healthcare University, Setagaya-ku, Tokyo 154-8568, Japan
| | - Kentaro Yamada
- Department of Oral Science, Division of Neuroscience and Brain Functions, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Toshiharu Yamamoto
- Department of Oral Science, Division of Neuroscience and Brain Functions, Kanagawa Dental University, Yokosuka 238-8580, Japan.
| |
Collapse
|
49
|
Abstract
The heart is adapted to utilize all classes of substrates to meet the high-energy demand, and it tightly regulates its substrate utilization in response to environmental changes. Although fatty acids are known as the predominant fuel for the adult heart at resting stage, the heart switches its substrate preference toward glucose during stress conditions such as ischemia and pathological hypertrophy. Notably, increasing evidence suggests that the loss of metabolic flexibility associated with increased reliance on glucose utilization contribute to the development of cardiac dysfunction. The changes in glucose metabolism in hypertrophied hearts include altered glucose transport and increased glycolysis. Despite the role of glucose as an energy source, changes in other nonenergy producing pathways related to glucose metabolism, such as hexosamine biosynthetic pathway and pentose phosphate pathway, are also observed in the diseased hearts. This article summarizes the current knowledge regarding the regulation of glucose transporter expression and translocation in the heart during physiological and pathological conditions. It also discusses the signaling mechanisms governing glucose uptake in cardiomyocytes, as well as the changes of cardiac glucose metabolism under disease conditions.
Collapse
Affiliation(s)
- Dan Shao
- Mitochondria and Metabolism Center, University of Washington, Seattle, Washington, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle, Washington, USA
| |
Collapse
|
50
|
Nomura N, Verdon G, Kang HJ, Shimamura T, Nomura Y, Sonoda Y, Hussien SA, Qureshi AA, Coincon M, Sato Y, Abe H, Nakada-Nakura Y, Hino T, Arakawa T, Kusano-Arai O, Iwanari H, Murata T, Kobayashi T, Hamakubo T, Kasahara M, Iwata S, Drew D. Structure and mechanism of the mammalian fructose transporter GLUT5. Nature 2015; 526:397-401. [PMID: 26416735 PMCID: PMC4618315 DOI: 10.1038/nature14909] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/14/2015] [Indexed: 02/02/2023]
Abstract
The altered activity of the fructose transporter GLUT5, an isoform of the facilitated-diffusion glucose transporter family, has been linked to disorders such as type 2 diabetes and obesity. GLUT5 is also overexpressed in certain tumor cells and inhibitors are potential drugs for these conditions. Here, we describe the crystal structure of GLUT5 from Rattus norvegicus and Bos taurus in open outward- and open inward-facing conformations, respectively. GLUT5 has a major facilitator superfamily fold like other homologous monosaccharide transporters. Based on a comparison of the inward-facing structures of GLUT5 and human GLUT1, a ubiquitous glucose transporter, we show that a single point mutation is enough to switch the substrate binding preference of GLUT5 from fructose to glucose. A comparison of the substrate-free structures of GLUT5 with occluded substrate-bound structures of XylE suggests that, besides global rocker-switch like re-orientation of the bundles, local asymmetric rearrangements of C-terminal bundle helices TMs 7 and 10 underlie a “gated-pore” transport mechanism in such monosaccharide transporters.
Collapse
Affiliation(s)
- Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Grégory Verdon
- Division of Molecular Biosciences, Imperial College London, London, SW7 2AZ, U.K.,Membrane Protein Laboratory, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Chilton, Oxfordshire, OX11 0DE, U.K.,Research Complex at Harwell Rutherford, Appleton Laboratory, Harwell, Oxford, Didcot, Oxfordshire, OX11 0FA, U.K
| | - Hae Joo Kang
- Division of Molecular Biosciences, Imperial College London, London, SW7 2AZ, U.K.,Membrane Protein Laboratory, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Chilton, Oxfordshire, OX11 0DE, U.K.,Research Complex at Harwell Rutherford, Appleton Laboratory, Harwell, Oxford, Didcot, Oxfordshire, OX11 0FA, U.K
| | - Tatsuro Shimamura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yayoi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yo Sonoda
- Division of Molecular Biosciences, Imperial College London, London, SW7 2AZ, U.K
| | - Saba Abdul Hussien
- Centre for Biomembrane Research, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Aziz Abdul Qureshi
- Centre for Biomembrane Research, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Mathieu Coincon
- Centre for Biomembrane Research, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Yumi Sato
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hitomi Abe
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshiko Nakada-Nakura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tomoya Hino
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takatoshi Arakawa
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Osamu Kusano-Arai
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Takeshi Murata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Systems and Structural Biology Center, RIKEN, 1-7-22 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Takuya Kobayashi
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Michihiro Kasahara
- Laboratory of Biophysics, School of Medicine, Teikyo University, Hachioji, Tokyo 192-0395, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, ERATO, Iwata Human Receptor Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Japan Science and Technology Agency, Research Acceleration Program, Membrane Protein Crystallography Project, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Division of Molecular Biosciences, Imperial College London, London, SW7 2AZ, U.K.,Membrane Protein Laboratory, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Chilton, Oxfordshire, OX11 0DE, U.K.,Research Complex at Harwell Rutherford, Appleton Laboratory, Harwell, Oxford, Didcot, Oxfordshire, OX11 0FA, U.K.,Systems and Structural Biology Center, RIKEN, 1-7-22 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - David Drew
- Division of Molecular Biosciences, Imperial College London, London, SW7 2AZ, U.K.,Centre for Biomembrane Research, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|