1
|
Xu W, Borges K. Case for supporting astrocyte energetics in glucose transporter 1 deficiency syndrome. Epilepsia 2024; 65:2213-2226. [PMID: 38767952 DOI: 10.1111/epi.18013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
In glucose transporter 1 deficiency syndrome (Glut1DS), glucose transport into brain is reduced due to impaired Glut1 function in endothelial cells at the blood-brain barrier. This can lead to shortages of glucose in brain and is thought to contribute to seizures. Ketogenic diets are the first-line treatment and, among many beneficial effects, provide auxiliary fuel in the form of ketone bodies that are largely metabolized by neurons. However, Glut1 is also the main glucose transporter in astrocytes. Here, we review data indicating that glucose shortage may also impact astrocytes in addition to neurons and discuss the expected negative biochemical consequences of compromised astrocytic glucose transport for neurons. Based on these effects, auxiliary fuels are needed for both cell types and adding medium chain triglycerides (MCTs) to ketogenic diets is a biochemically superior treatment for Glut1DS compared to classical ketogenic diets. MCTs provide medium chain fatty acids (MCFAs), which are largely metabolized by astrocytes and not neurons. MCFAs supply energy and contribute carbons for glutamine and γ-aminobutyric acid synthesis, and decanoic acid can also block α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors. MCTs do not compete with metabolism of ketone bodies mostly occurring in neurons. Triheptanoin, an anaplerotic but also gluconeogenic uneven MCT, may be another potential addition to ketogenic diets, although maintenance of "ketosis" can be difficult. Gene therapy has also targeted both endothelial cells and astrocytes. Other approaches to increase fuel delivery to the brain currently investigated include exchange of Glut1DS erythrocytes with healthy cells, infusion of lactate, and pharmacological improvement of glucose transport. In conclusion, although it remains difficult to assess impaired astrocytic energy metabolism in vivo, astrocytic energy needs are most likely not met by ketogenic diets in Glut1DS. Thus, we propose prospective studies including monitoring of blood MCFA levels to find optimal doses for add-on MCT to ketogenic diets and assessing of short- and long-term outcomes.
Collapse
Affiliation(s)
- Weizhi Xu
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Karin Borges
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
2
|
Drzewicka K, Zasłona Z. Metabolism-driven glycosylation represents therapeutic opportunities in interstitial lung diseases. Front Immunol 2024; 15:1328781. [PMID: 38550597 PMCID: PMC10973144 DOI: 10.3389/fimmu.2024.1328781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
Metabolic changes are coupled with alteration in protein glycosylation. In this review, we will focus on macrophages that are pivotal in the pathogenesis of pulmonary fibrosis and sarcoidosis and thanks to their adaptable metabolism are an attractive therapeutic target. Examples presented in this review demonstrate that protein glycosylation regulates metabolism-driven immune responses in macrophages, with implications for fibrotic processes and granuloma formation. Targeting proteins that regulate glycosylation, such as fucosyltransferases, neuraminidase 1 and chitinase 1 could effectively block immunometabolic changes driving inflammation and fibrosis, providing novel avenues for therapeutic interventions.
Collapse
|
3
|
Olszańska J, Pietraszek-Gremplewicz K, Domagalski M, Nowak D. Mutual impact of adipocytes and colorectal cancer cells growing in co-culture conditions. Cell Commun Signal 2023; 21:130. [PMID: 37316878 DOI: 10.1186/s12964-023-01155-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/29/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common malignancy worldwide. CRC cells are situated in an adipocyte-rich microenvironment, which leads to interactions between adipocytes and CRC cells. Upon exposure to cancer cells, adipocytes transform into cancer-associated adipocytes (CAAs), and as a result, they gain features that promote tumor progression. The aim of this research was to shed more light on the detailed role of interactions between adipocytes and CRC cells associated with cancer progression in the context of these alterations. METHODS To implement adipocyte-CRC cell interaction, a co-culture model was applied. The analyses mainly focused on the metabolic modifications within CAAs and CRC cells, as well as the proliferation and migration potential of CRC cells. The impact of CRC on adipocytes was investigated by qRT-PCR analysis and Oil Red O staining. Proliferation and migration of CRC cells upon co-culture were tested with videomicroscopy, XTT, and a wound healing assay. Metabolic changes within CAAs and CRC cells were investigated based on lipid droplet formation, cell cycle analysis, gene and protein expression by qRT-PCR, and western blotting techniques. RESULTS CRC cells induced reprogramming of adipocytes into CAAs, which was connected with downregulation of lipid droplet formation in CAAs and alteration in adipocyte features. CAAs showed decreased metabolism-related gene expression, phosphorylation of Akt, ERK kinases, STAT3, and lactate secretion in comparison to the control. CAAs also promoted the migration, proliferation, and lipid droplet accumulation of CRC cells. After co-culturing with adipocytes, there was a shift to the G2/M phase of the cell cycle according to the differences in cyclin expression. CONCLUSION There are complex bidirectional interactions between adipocytes and CRC cells that may be connected with the induction of CRC cell progression. Video Abstract.
Collapse
Affiliation(s)
- Joanna Olszańska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | | | - Mikołaj Domagalski
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| |
Collapse
|
4
|
Phukhum P, Phetcharaburanin J, Chaleekarn K, Kittirat Y, Kulthawatsiri T, Namwat N, Loilome W, Khuntikeo N, Titapun A, Wangwiwatsin A, Khampitak T, Suksawat M, Klanrit P. The impact of hypoxia and oxidative stress on proteo-metabolomic alterations of 3D cholangiocarcinoma models. Sci Rep 2023; 13:3072. [PMID: 36810897 PMCID: PMC9944917 DOI: 10.1038/s41598-023-30204-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
The three-dimensional multicellular spheroid (3D MCS) model has been employed in cholangiocarcinoma research as it generates 3D architecture and includes more physiological relevance with the multicellular arrangement. However, it is also essential to explain the molecular signature in this microenvironment and its structural complexity. The results indicated that poorly differentiated CCA cell lines were unable to form 3D MCS due to the lack of cell adhesion molecules with more mesenchymal marker expression. The well-differentiated CCA and cholangiocyte cell lines were able to develop 3D MCSs with round shapes, smooth perimeter, and cell adhesion molecules that led to the hypoxic and oxidative microenvironment detected. For MMNK-1, KKU-213C, and KKU-213A MCSs, the proteo-metabolomic analysis showed proteins and metabolic products altered compared to 2D cultures, including cell-cell adhesion molecules, energy metabolism-related enzymes and metabolites, and oxidative-related metabolites. Therefore, the 3D MCSs provide different physiological states with different phenotypic signatures compared to 2D cultures. Considering the 3D model mimics more physiological relevance, it might lead to an alternate biochemical pathway, targeting to improve drug sensitivity for CCA treatment.
Collapse
Affiliation(s)
- Pimpawadee Phukhum
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Jutarop Phetcharaburanin
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Kwuanjira Chaleekarn
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Yingpinyapat Kittirat
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Thanaporn Kulthawatsiri
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Nisana Namwat
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Watcharin Loilome
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Narong Khuntikeo
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Attapol Titapun
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Arporn Wangwiwatsin
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Tueanjit Khampitak
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Manida Suksawat
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Poramate Klanrit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Khon Kaen University Phenome Centre, Khon Kaen, 40002, Thailand. .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
5
|
Huang G, Zhao D, Lan C, Wu B, Li X, Lou S, Zheng Y, Huang Y, Hu Z, Jia B. Glucose-assisted trophic conversion of Chlamydomonas reinhardtii by expression of glucose transporter GLUT1. ALGAL RES 2022. [DOI: 10.1016/j.algal.2021.102626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
6
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
7
|
Silva JVV, Ganesan S, Wickramasinghe HKJP, Stepanchenko N, Kaya CA, Beitz DC, Appuhamy JADRN. Effects of branched-chain amino acids on glucose uptake and lactose synthesis rates in bovine mammary epithelial cells and lactating mammary tissue slices. J Dairy Sci 2021; 105:1717-1730. [PMID: 34802743 DOI: 10.3168/jds.2021-20950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/15/2021] [Indexed: 11/19/2022]
Abstract
Even though supplementations of essential AA (EAA) are often related to increased lactose yields in dairy cows, underlying mechanisms connecting EAA availability to the mammary glands and lactose synthesis are poorly understood. The objective of this study was to examine the effects of branched-chain AA (BCAA) including Leu, Ile, and Val on (1) glucose transporter (GLUT1) abundance and glucose uptake, (2) the abundance of proteins regulating lactose synthesis pathway, and (3) fractional synthesis rates of lactose (FSR) using bovine mammary epithelial cells (BMEC) and mammary tissues slices (MTS). The BMEC (n = 4) were allocated randomly to regular Dulbecco's Modified Eagle Medium with Ham's F12 (DMEM/F12) media (+EAA) or +EAA deficient (by 90%) in all EAA (-EAA), all BCAA (-BCAA), only Leu (-Leu), only Ile (-Ile) or only Val (-Val). Western immunoblotting analyses, depletion of glucose in media, and a proteomic analysis were performed to determine the abundance of GLUT1 in the cell membrane, net glucose uptake, and the abundance of enzymes involved in lactose synthesis pathway in BMEC, respectively. The MTS (n = 6) were allocated randomly to DMEM/F12 media having all EAA and 13C-glucose at concentrations similar to plasma concentrations of cows (+EAAp), and +EAAp deprived of all BCAA (-BCAAp) or only Leu (-Leup) for 3 h. The 13C enrichments of free glucose pool in MTS (EGlu-free) and the enrichments of glucose incorporated into lactose in MTS and media [ELactose-bound (T&M)] were determined and used in calculating FSR. In BMEC, -BCAA increased the fraction of total GLUT1 translocated to the cell membrane and the fraction that was potentially glycosylated compared with +EAA. Among individual BCAA, only -Leu was associated with a 63% increase in GLUT1 translocated to the cell membrane and a 40% increase in glucose uptake of BMEC. The -BCAA tended to be related to a 75% increase in the abundance of hexokinase in BMEC. Deprivation of Leu tended to increase glucose uptake of MTS but did not affect EGlu-free, ELactose-bound (T&M), or FSR relative to +EAAp. On the other hand, -BCAAp did not affect glucose uptake of MTS but was related to lower ELactose-bound (T&M), or FSR relative to +EAAp. Considering together, decreasing Leu supply to mammary tissues enhances GLUT1 and thus glucose uptake, which, however, does not affect lactose synthesis rates. Moreover, the deficiency of other BCAA, Ile, and Val alone or together with the deficiency of Leu seemed to decrease lactose synthesis rates without affecting glucose uptake. The data also emphasize the importance of addressing the effect of the supply of other nutrients to the mammary glands than the precursor supply in describing the synthesis of a milk component.
Collapse
Affiliation(s)
- J V V Silva
- Department of Animal Science, Iowa State University, Ames 50011
| | - S Ganesan
- Department of Animal Science, Iowa State University, Ames 50011
| | | | - N Stepanchenko
- Department of Animal Science, Iowa State University, Ames 50011
| | - C A Kaya
- Department of Livestock and Crop Production, Dicle University, Diyarbakir, 21280, Turkey
| | - D C Beitz
- Department of Animal Science, Iowa State University, Ames 50011
| | | |
Collapse
|
8
|
Cazzato G, Colagrande A, Cimmino A, Abbatepaolo C, Bellitti E, Romita P, Lospalluti L, Foti C, Arezzo F, Loizzi V, Lettini T, Sablone S, Resta L, Cormio G, Ingravallo G, Rossi R. GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives. Cells 2021; 10:cells10113090. [PMID: 34831313 PMCID: PMC8624914 DOI: 10.3390/cells10113090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/03/2021] [Accepted: 11/07/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Malignant melanoma is the most aggressive of skin cancers and the 19th most common cancer worldwide, with an estimated age-standardized incidence rate of 2.8-3.1 per 100,000; although there have been clear advances in therapeutic treatment, the prognosis of MM patients with Breslow thickness greater than 1 mm is still quite poor today. The study of how melanoma cells manage to survive and proliferate by consuming glucose has been partially addressed in the literature, but some rather interesting results are starting to be present. METHODS A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and a search of PubMed and Web of Sciences (WoS) databases was performed until 27 September 2021 using the terms: glucose transporter 1 and 3 and GLUT1/3 in combination with each of the following: melanoma, neoplasm and immunohistochemistry. RESULTS In total, 46 records were initially identified in the literature search, of which six were duplicates. After screening for eligibility and inclusion criteria, 16 publications were ultimately included. CONCLUSIONS the results discussed regarding the role and expression of GLUT are still far from definitive, but further steps toward understanding and stopping this mechanism have, at least in part, been taken. New studies and new discoveries should lead to further clarification of some aspects since the various mechanisms of glucose uptake by neoplastic cells are not limited to the transporters of the GLUT family alone.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
- Correspondence: ; Tel.: +39-3405203641
| | - Anna Colagrande
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Antonietta Cimmino
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Caterina Abbatepaolo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Emilio Bellitti
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Paolo Romita
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Lucia Lospalluti
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Caterina Foti
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Francesca Arezzo
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Vera Loizzi
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Teresa Lettini
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Sara Sablone
- Section of Legal Medicine, Department of Interdisciplinary Medicine, Bari Policlinico Hospital, University of Bari, “Aldo Moro”, 70124 Bari, Italy;
| | - Leonardo Resta
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Gennaro Cormio
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Roberta Rossi
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| |
Collapse
|
9
|
Tanioka T, Maeda K, Takahashi R, Iwamoto S. The Ang III/AT2R Pathway Enhances Glucose Uptake by Improving GLUT1 Expression in 3T3-L1 Adipocytes. Biol Pharm Bull 2021; 44:1014-1018. [PMID: 34193683 DOI: 10.1248/bpb.b20-00946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiotensin III (Ang III) is a heptapeptide derived from Ang II that has been confirmed as the preferred agonist of angiotensin II type 2 receptor (AT2R). Recent studies have revealed AT2R mainly exerts anti-inflammation effects. However, the effects of the Ang III/AT2R pathway on adipocytes remain unknown. Here, the effects of Ang III on glucose uptake were examined. The results showed that AT2R expression was upregulated during adipogenesis in 3T3-L1 preadipocytes, whereas AT1R expression was diminished. Also, Ang III (10 nM) significantly increased glucose uptake by 3T3-L1 adipocytes, which was blocked by PD123319, an AT2R blocker, but not by irbesartan, an AT1R blocker. Ang III also induced the expression of glucose transporter type 1 (GLUT1). These stimulatory effects were inhibited by pretreatment with PD123319, but not with irbesartan. Together, these results indicate that Ang III enhances glucose uptake by upregulating GLUT1 expression via AT2R.
Collapse
Affiliation(s)
- Toshihiro Tanioka
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| | - Kohei Maeda
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| | - Rei Takahashi
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| | - Sanju Iwamoto
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University
| |
Collapse
|
10
|
Matsuo S, Hiasa M, Omote H. Functional characterization and tissue localization of the facilitative glucose transporter GLUT12. J Biochem 2020; 168:611-620. [PMID: 32761185 DOI: 10.1093/jb/mvaa090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/28/2020] [Indexed: 11/13/2022] Open
Abstract
Facilitative glucose transporters (GLUTs) play crucial roles in glucose utilization and homeostasis. GLUT12 was initially isolated as a novel GLUT4-like transporter involved in insulin-dependent glucose transport. However, tissue distribution and biochemical properties of GLUT12 are not well understood. In this study, we investigated the basic kinetic properties and tissue distribution of GLUT12. Human GLUT12 and GLUT1 were overexpressed and purified using Ni-NTA column chromatography. Reconstituted proteoliposomes showed time-dependent d-glucose transport activity, which was inhibited by phloretin and dehydroascorbate. Dose dependence of glucose transport revealed a KM and Vmax values of 6.4 mM and 1.2 μmol/mg/min, respectively, indicating that GLUT12 is a high-affinity type GLUT. Glucose transport by GLUT12 was inhibited by ATP and glucose-1-phosphate, glucose-6-phosphate and disaccharides (properties similar to those of GLUT1). Indirect immunohistochemistry revealed the distribution of mouse GLUT12 in the apical region of distal tubules and collecting ducts in the kidney and epithelial cells of the jejunum. In addition to these cells, GLUT12 was present in chromaffin cells in the adrenal medulla, the anterior pituitary lobe, as well as the thyroid and pyloric glands. These tissue distributions suggest a unique function of GLUT12, besides that of an insulin-dependent glucose transport.
Collapse
Affiliation(s)
- Shunsuke Matsuo
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
11
|
Platelets and Defective N-Glycosylation. Int J Mol Sci 2020; 21:ijms21165630. [PMID: 32781578 PMCID: PMC7460655 DOI: 10.3390/ijms21165630] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
N-glycans are covalently linked to an asparagine residue in a simple acceptor sequence of proteins, called a sequon. This modification is important for protein folding, enhancing thermodynamic stability, and decreasing abnormal protein aggregation within the endoplasmic reticulum (ER), for the lifetime and for the subcellular localization of proteins besides other functions. Hypoglycosylation is the hallmark of a group of rare genetic diseases called congenital disorders of glycosylation (CDG). These diseases are due to defects in glycan synthesis, processing, and attachment to proteins and lipids, thereby modifying signaling functions and metabolic pathways. Defects in N-glycosylation and O-glycosylation constitute the largest CDG groups. Clotting and anticlotting factor defects as well as a tendency to thrombosis or bleeding have been described in CDG patients. However, N-glycosylation of platelet proteins has been poorly investigated in CDG. In this review, we highlight normal and deficient N-glycosylation of platelet-derived molecules and discuss the involvement of platelets in the congenital disorders of N-glycosylation.
Collapse
|
12
|
Pifferi F, Cunnane SC, Guesnet P. Evidence of the Role of Omega-3 Polyunsaturated Fatty Acids in Brain Glucose Metabolism. Nutrients 2020; 12:nu12051382. [PMID: 32408634 PMCID: PMC7285025 DOI: 10.3390/nu12051382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 11/30/2022] Open
Abstract
In mammals, brain function, particularly neuronal activity, has high energy needs. When glucose is supplemented by alternative oxidative substrates under different physiological conditions, these fuels do not fully replace the functions fulfilled by glucose. Thus, it is of major importance that the brain is almost continuously supplied with glucose from the circulation. Numerous studies describe the decrease in brain glucose metabolism during healthy or pathological ageing, but little is known about the mechanisms that cause such impairment. Although it appears difficult to determine the exact role of brain glucose hypometabolism during healthy ageing or during age-related neurodegenerative diseases such as Alzheimer’s disease, uninterrupted glucose supply to the brain is still of major importance for proper brain function. Interestingly, a body of evidence suggests that dietary n-3 polyunsaturated fatty acids (PUFAs) might play significant roles in brain glucose regulation. Thus, the goal of the present review is to summarize this evidence and address the role of n-3 PUFAs in brain energy metabolism. Taken together, these data suggest that ensuring an adequate dietary supply of n-3 PUFAs could constitute an essential aspect of a promising strategy to promote optimal brain function during both healthy and pathological ageing.
Collapse
Affiliation(s)
- Fabien Pifferi
- Unité Mixte de Recherche (UMR), Centre Nationnal de la Recherche Scientifique (CNRS), Museum National d’Histoire Naturelle (MNHN) 7179, Mécanismes Adaptatifs et Evolution (MECADEV), 1 Avenue du Petit Château, 91800 Brunoy, France
- Correspondence:
| | - Stephen C. Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Research Center on Aging, Sherbrooke, QC J1H 4C4, Canada
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | | |
Collapse
|
13
|
Pharmacologic inhibition of N-linked glycan trimming with kifunensine disrupts GLUT1 trafficking and glucose uptake. Biochimie 2020; 174:18-29. [PMID: 32298759 DOI: 10.1016/j.biochi.2020.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/26/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
The facilitative glucose transport GLUT1 (SLC2A1) is a constitutively expressed membrane protein involved in basal uptake of blood glucose. GLUT1 modification by N-linked glycosylation at a single asparagine residue (N45) appears to play multiple roles in the trafficking, stability and transport activity of this protein. Here we examine the role of complex N-glycosylation on GLUT1 function in renal epithelial cells by arresting this modification at the high-mannose stage with the mannosidase I inhibitor kifunensine. Consistent with prior work in which GLUT1 glycosylation was completely inhibited, we find that kifunensine treatment results in a time-dependent decrease of up to 40% in cellular glucose uptake. We further demonstrate that this effect is primarily a result of deficient GLUT1 trafficking to the cell membrane due to quality control mechanisms that instead direct GLUT1 to the ER-associated degradation (ERAD) pathway. Unlike tunicamycin, which inhibits the first step in N-glycosyl transfer and causes dramatic cell cycle arrest, kifunensine causes only a modest decrease in GLUT1 levels and cell cycle progression in both normal and transformed renal cells. The effect of kifunensine on the cell cycle appears to be independent of its effect on GLUT1, since all renal cell types in this study displayed decreased proliferation regardless of their dependence on glucose uptake for growth and survival. Together these results indicate that proper N-glycan processing plays an important role in directing GLUT1 to the cell surface and that disruption of mannosidase activity results in aberrant degradation of GLUT1 by the ERAD pathway.
Collapse
|
14
|
Kuzyk CL, Anderson CC, Roede JR. Simvastatin Induces Delayed Apoptosis Through Disruption of Glycolysis and Mitochondrial Impairment in Neuroblastoma Cells. Clin Transl Sci 2020; 13:563-572. [PMID: 31917509 PMCID: PMC7214657 DOI: 10.1111/cts.12740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Simvastatin, a commonly used cholesterol-lowering drug, inhibits the mevalonate pathway involved in the synthesis of the mitochondrial electron carrier coenzyme Q10 (CoQ10), as well as other bioenergetics substrates. The purpose of this study was to investigate simvastatin exposure on mitochondrial respiration, metabolic fuel preferences, and glucose utilization. We hypothesized that simvastatin at a noncytotoxic dose will impair energy metabolism in human neuroblastoma cells. SK-N-AS cells were exposed at acute and chronic time points and evaluated in a Seahorse XF analyzer, revealing decreased mitochondrial and glycolytic parameters. Flow cytometry showed a significant induction of apoptosis in simvastatin-treated cells at 48 hours. Finally, multiple techniques were used to show that simvastatin-mediated impairment of bioenergetics is more complex than CoQ10 depletion or hampered glucose uptake. Therefore, the data reported here represent a biphasic hit to mitochondria followed by reduction in glucose and glutamine metabolism in neuroblastoma; adding mechanism to potential pleotropic effects of statins.
Collapse
Affiliation(s)
- Crystal L Kuzyk
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
15
|
Tin A, Marten J, Halperin Kuhns VL, Li Y, Wuttke M, Kirsten H, Sieber KB, Qiu C, Gorski M, Yu Z, Giri A, Sveinbjornsson G, Li M, Chu AY, Hoppmann A, O'Connor LJ, Prins B, Nutile T, Noce D, Akiyama M, Cocca M, Ghasemi S, van der Most PJ, Horn K, Xu Y, Fuchsberger C, Sedaghat S, Afaq S, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boerwinkle E, Bottinger EP, Boutin TS, Brumat M, Burkhardt R, Campana E, Campbell A, Campbell H, Carroll RJ, Catamo E, Chambers JC, Ciullo M, Concas MP, Coresh J, Corre T, Cusi D, Felicita SC, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Delgado G, Demirkan A, Devuyst O, Dittrich K, Eckardt KU, Ehret G, Endlich K, Evans MK, Gansevoort RT, Gasparini P, Giedraitis V, Gieger C, Girotto G, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hayward C, Hicks AA, Hofer E, Holm H, Huang W, Hutri-Kähönen N, Hwang SJ, Ikram MA, Lewis RM, Ingelsson E, Jakobsdottir J, Jonsdottir I, Jonsson H, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kanai M, Kerr SM, Kiess W, Kleber ME, Koenig W, Kooner JS, Körner A, Kovacs P, Krämer BK, Kronenberg F, Kubo M, Kühnel B, La Bianca M, Lange LA, Lehne B, Lehtimäki T, Liu J, Loeffler M, Loos RJF, Lyytikäinen LP, Magi R, Mahajan A, Martin NG, März W, Mascalzoni D, Matsuda K, Meisinger C, Meitinger T, Metspalu A, Milaneschi Y, O'Donnell CJ, Wilson OD, Gaziano JM, Mishra PP, Mohlke KL, Mononen N, Montgomery GW, Mook-Kanamori DO, Müller-Nurasyid M, Nadkarni GN, Nalls MA, Nauck M, Nikus K, Ning B, Nolte IM, Noordam R, O'Connell JR, Olafsson I, Padmanabhan S, Penninx BWJH, Perls T, Peters A, Pirastu M, Pirastu N, Pistis G, Polasek O, Ponte B, Porteous DJ, Poulain T, Preuss MH, Rabelink TJ, Raffield LM, Raitakari OT, Rettig R, Rheinberger M, Rice KM, Rizzi F, Robino A, Rudan I, Krajcoviechova A, Cifkova R, Rueedi R, Ruggiero D, Ryan KA, Saba Y, Salvi E, Schmidt H, Schmidt R, Shaffer CM, Smith AV, Smith BH, Spracklen CN, Strauch K, Stumvoll M, Sulem P, Tajuddin SM, Teren A, Thiery J, Thio CHL, Thorsteinsdottir U, Toniolo D, Tönjes A, Tremblay J, Uitterlinden AG, Vaccargiu S, van der Harst P, van Duijn CM, Verweij N, Völker U, Vollenweider P, Waeber G, Waldenberger M, Whitfield JB, Wild SH, Wilson JF, Yang Q, Zhang W, Zonderman AB, Bochud M, Wilson JG, Pendergrass SA, Ho K, Parsa A, Pramstaller PP, Psaty BM, Böger CA, Snieder H, Butterworth AS, Okada Y, Edwards TL, Stefansson K, Susztak K, Scholz M, Heid IM, Hung AM, Teumer A, Pattaro C, Woodward OM, Vitart V, Köttgen A. Target genes, variants, tissues and transcriptional pathways influencing human serum urate levels. Nat Genet 2019; 51:1459-1474. [PMID: 31578528 PMCID: PMC6858555 DOI: 10.1038/s41588-019-0504-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022]
Abstract
Elevated serum urate levels cause gout and correlate with cardiometabolic diseases via poorly understood mechanisms. We performed a trans-ancestry genome-wide association study of serum urate in 457,690 individuals, identifying 183 loci (147 previously unknown) that improve the prediction of gout in an independent cohort of 334,880 individuals. Serum urate showed significant genetic correlations with many cardiometabolic traits, with genetic causality analyses supporting a substantial role for pleiotropy. Enrichment analysis, fine-mapping of urate-associated loci and colocalization with gene expression in 47 tissues implicated the kidney and liver as the main target organs and prioritized potentially causal genes and variants, including the transcriptional master regulators in the liver and kidney, HNF1A and HNF4A. Experimental validation showed that HNF4A transactivated the promoter of ABCG2, encoding a major urate transporter, in kidney cells, and that HNF4A p.Thr139Ile is a functional variant. Transcriptional coregulation within and across organs may be a general mechanism underlying the observed pleiotropy between urate and cardiometabolic traits.
Collapse
Affiliation(s)
- Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Welch Centre for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA.
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Yong Li
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Karsten B Sieber
- Target Sciences-Genetics, GlaxoSmithKline, Collegeville, PA, USA
| | - Chengxiang Qiu
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Zhi Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayush Giri
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Man Li
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Anselm Hoppmann
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Luke J O'Connor
- Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bram Prins
- Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - Teresa Nutile
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
| | - Damia Noce
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Massimiliano Cocca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Sahar Ghasemi
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Yizhe Xu
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Saima Afaq
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Institute of Public Health & Social Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nisha Bansal
- Division of Nephrology, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
| | | | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Eric Boerwinkle
- Human Genetics Centre, University of Texas Health Science Centre, Houston, TX, USA
| | - Erwin P Bottinger
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Marco Brumat
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Ralph Burkhardt
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Eric Campana
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eulalia Catamo
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Marina Ciullo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tanguy Corre
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Daniele Cusi
- Institute of Biomedical Technologies, Italy National Research Council, Milano, Italy
- Bio4Dreams, Milano, Italy
| | | | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Aiko P J de Vries
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Graciela Delgado
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Katalin Dittrich
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georg Ehret
- Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Karlhans Endlich
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Paolo Gasparini
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Giorgia Girotto
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pavel Hamet
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Medpharmgene, Montreal, Quebec, Canada
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Andrew A Hicks
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Hilma Holm
- deCODE Genetics, Amgen Inc., Reykjavik, Iceland
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Centre, Shanghai, China
- Shanghai Industrial Technology Institute, Shanghai, China
| | - Nina Hutri-Kähönen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- The Centre for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raychel M Lewis
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Johanna Jakobsdottir
- Icelandic Heart Association, Kópavogur, Iceland
- The Centre of Public Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Helgi Jonsson
- Landspitalinn University Hospital, Reykjavík, Iceland
- University of Iceland, Reykjavík, Iceland
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Navya Shilpa Josyula
- Geisinger Research, Biomedical and Translational Informatics Institute, Rockville, MD, USA
| | - Bettina Jung
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shona M Kerr
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wieland Kiess
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Biostatistics, University of Ulm, Ulm, Germany
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, 323 School of Public Health, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Antje Körner
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Centre Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michiaki Kubo
- RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
| | - Martina La Bianca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jun Liu
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Reedik Magi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Winfried März
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | | | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Christa Meisinger
- Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-Universität München at UNIKA-T Augsburg, Augsburg, Germany
| | - Thomas Meitinger
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christopher J O'Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Otis D Wilson
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
| | - J Michael Gaziano
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Centre, Leiden, the Netherlands
| | - Martina Müller-Nurasyid
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Boting Ning
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali University Hospital, Reykjavik, Iceland
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Thomas Perls
- Department of Medicine, Geriatrics Section, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Mario Pirastu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Giorgio Pistis
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
- Gen-info Ltd, Zagreb, Croatia
| | - Belen Ponte
- Nephrology Service, Department of Specialties in Internal Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Tanja Poulain
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ton J Rabelink
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
- Einthoven Laboratory of Experimental Vascular Research, Leiden University Medical Centre, Leiden, the Netherlands
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Rainer Rettig
- Institute of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Myriam Rheinberger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Federica Rizzi
- Department of Health Sciences, University of Milan, Milano, Italy
- ePhood Scientific Unit, ePhood SRL, Milano, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Alena Krajcoviechova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Thomayer Hospital, Prague, Czech Republic
| | - Renata Cifkova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Medicine II, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Kathleen A Ryan
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yasaman Saba
- Molecular Biology and Biochemistry, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milano, Italy
- Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Christian M Shaffer
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Albert V Smith
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Stumvoll
- Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
| | | | - Salman M Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Andrej Teren
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Heart Centre Leipzig, Leipzig, Germany
| | - Joachim Thiery
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Chris H L Thio
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | - Anke Tönjes
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Johanne Tremblay
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Simona Vaccargiu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Centre for Cardiovascular Research, the Netherlands Heart Institute, Utrecht, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Genomics plc, Oxford, UK
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerard Waeber
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - John B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sarah H Wild
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - James F Wilson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London, UK
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Centre, Jackson, MS, USA
| | - Sarah A Pendergrass
- Geisinger Research, Biomedical and Translational Informatics Institute, Danville, PA, USA
| | - Kevin Ho
- Kidney Health Research Institute, Geisinger, Danville, PA, USA
- Department of Nephrology, Geisinger, Danville, PA, USA
| | - Afshin Parsa
- Division of Kidney, Urologic and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, Department of Epidemiology, Department of Health Service, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology and Rheumatology, Kliniken Südostbayern AG, Traunstein, Germany
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adam S Butterworth
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Centre, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Katalin Susztak
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Adriana M Hung
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | | | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
16
|
Li Y, Liu Y, Zhu H, Chen X, Tian M, Wei Y, Gong Y, Jiang J. N-acetylglucosaminyltransferase I promotes glioma cell proliferation and migration through increasing the stability of the glucose transporter GLUT1. FEBS Lett 2019; 594:358-366. [PMID: 31494931 DOI: 10.1002/1873-3468.13596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/28/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
Abstract
Abnormal alteration of N-glycosylation structure contributes to glioma progression. N-acetylglucosaminyltransferase I (MGAT1) plays an essential role in the conversion of processed high-mannose cores into complex or hybrid N-linked oligosaccharide structures. The function of MGAT1 in glioma development remains largely unknown. Here, we found that the expression of MGAT1 is higher in glioblastoma compared to normal brain tissues. Inhibition of EGFR signalling pathway or serum starvation reduces MGAT1 expression. Knockdown of MGAT1 inhibits glioma cell proliferation and migration. Furthermore, MGAT1 promotes complex N-glycosylation of glucose transporter 1 (Glut1) and increases Glut1 protein levels. In summary, our findings indicate that MGAT1 is highly expressed in glioblastoma and promotes glioma cells at least partly through upregulation of Glut1 protein.
Collapse
Affiliation(s)
- Yinan Li
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingchao Liu
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hongda Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoning Chen
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mi Tian
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyan Wei
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianhai Jiang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Ushiyama A, Priyadarshana C, Setiawan R, Miyazaki H, Ishikawa N, Tajima A, Asano A. Membrane raft-mediated regulation of glucose signaling pathway leading to acrosome reaction in chicken sperm†. Biol Reprod 2019; 100:1482-1491. [DOI: 10.1093/biolre/ioz015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/21/2018] [Accepted: 02/01/2019] [Indexed: 01/13/2023] Open
Affiliation(s)
- Ai Ushiyama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Chathura Priyadarshana
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Rangga Setiawan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Hitoshi Miyazaki
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Naoto Ishikawa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Atsushi Tajima
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| | - Atsushi Asano
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki—, Japan
| |
Collapse
|
18
|
Large-scale whole-exome sequencing association studies identify rare functional variants influencing serum urate levels. Nat Commun 2018; 9:4228. [PMID: 30315176 PMCID: PMC6185909 DOI: 10.1038/s41467-018-06620-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/13/2018] [Indexed: 02/08/2023] Open
Abstract
Elevated serum urate levels can cause gout, an excruciating disease with suboptimal treatment. Previous GWAS identified common variants with modest effects on serum urate. Here we report large-scale whole-exome sequencing association studies of serum urate and kidney function among ≤19,517 European ancestry and African-American individuals. We identify aggregate associations of low-frequency damaging variants in the urate transporters SLC22A12 (URAT1; p = 1.3 × 10−56) and SLC2A9 (p = 4.5 × 10−7). Gout risk in rare SLC22A12 variant carriers is halved (OR = 0.5, p = 4.9 × 10−3). Selected rare variants in SLC22A12 are validated in transport studies, confirming three as loss-of-function (R325W, R405C, and T467M) and illustrating the therapeutic potential of the new URAT1-blocker lesinurad. In SLC2A9, mapping of rare variants of large effects onto the predicted protein structure reveals new residues that may affect urate binding. These findings provide new insights into the genetic architecture of serum urate, and highlight molecular targets in SLC22A12 and SLC2A9 for lowering serum urate and preventing gout. Elevated serum urate levels are a risk factor for gout. Here, Tin et al. perform whole-exome sequencing in 19,517 individuals and detect low-frequency genetic variants in urate transporter genes, SLC22A12 and SLC2A9, associated with serum urate levels and confirm their damaging nature in vitro and in silico.
Collapse
|
19
|
Sandoval KE, Wooten JS, Harris MP, Schaller ML, Umbaugh DS, Witt KA. Mfsd2a and Glut1 Brain Nutrient Transporters Expression Increase with 32-Week Low and High Lard Compared with Fish-Oil Dietary Treatment in C57Bl/6 Mice. Curr Dev Nutr 2018; 2:nzy065. [PMID: 30338310 PMCID: PMC6186908 DOI: 10.1093/cdn/nzy065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/27/2018] [Accepted: 07/26/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Diet-mediated alterations of critical brain nutrient transporters, major facilitator super family domain-containing 2a (Mfsd2a) and glucose transporter 1 (Glut1), have wide reaching implications in brain health and disease. OBJECTIVE The aim of the study was to examine the impact of long-term low- and high-fat diets with lard or fish oil on critical brain nutrient transporters, Mfsd2a and Glut1. METHODS Eight-week-old male C57BL/6 mice were fed 1 of the following 4 diets for 32 wk: 10% of kcal from lard, 10% of kcal from fish oil, 41% of kcal from lard, or 41% of kcal from fish oil. Body weight and blood chemistries delineated dietary effects. Cortical and subcortical Mfsd2a and Glut1 mRNA and protein expression were evaluated, with other supportive nutrient-sensitive targets also assessed for mRNA expression changes. RESULTS Fish-oil diets increased cortical Mfsd2a mRNA expression compared with lard diets. Subcortical Mfsd2a mRNA expression decreased as the percentage of fat in the diet increased. There was an interaction between the type and percentage of fat with cortical and subcortical Mfsd2a and cortical Glut1 protein expression. In the lard diet groups, protein expression of cortical and subcortical Mfsd2a and cortical Glut1 significantly increased as fat percentage increased. As the fat percentage increased in the fish-oil diet groups, protein expression of cortical and subcortical Mfsd2a and cortical Glut1 did not change. When comparing the fish-oil groups with 10% lard, cortical Mfsd2a protein expression was significantly higher in the 10% and 41% fish-oil groups, whereas cortical Glut1 protein expression was significantly higher in only the 10% fish-oil group. A positive correlation between cortical peroxisome proliferator-activated receptor γ mRNA expression and Mfsd2a protein expression was shown. CONCLUSION Corresponding to chronic dietary treatment, an interaction between the type of fat and the percentage of fat exists respective to changes in brain expression of the key nutrient transporters Mfsd2a and Glut1.
Collapse
Affiliation(s)
| | - Joshua S Wooten
- Applied Health, School of Education, Health, and Human Behavior, Southern Illinois University Edwardsville, Edwardsville, IL
| | - Mathew P Harris
- Applied Health, School of Education, Health, and Human Behavior, Southern Illinois University Edwardsville, Edwardsville, IL
| | - Megan L Schaller
- Applied Health, School of Education, Health, and Human Behavior, Southern Illinois University Edwardsville, Edwardsville, IL
| | - David S Umbaugh
- Pharmaceutical Sciences, School of Pharmacy, Edwardsville, IL
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Edwardsville, IL
| |
Collapse
|
20
|
The Nutrient-Sensing Hexosamine Biosynthetic Pathway as the Hub of Cancer Metabolic Rewiring. Cells 2018; 7:cells7060053. [PMID: 29865240 PMCID: PMC6025041 DOI: 10.3390/cells7060053] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Alterations in glucose and glutamine utilizing pathways and in fatty acid metabolism are currently considered the most significant and prevalent metabolic changes observed in almost all types of tumors. Glucose, glutamine and fatty acids are the substrates for the hexosamine biosynthetic pathway (HBP). This metabolic pathway generates the “sensing molecule” UDP-N-Acetylglucosamine (UDP-GlcNAc). UDP-GlcNAc is the substrate for the enzymes involved in protein N- and O-glycosylation, two important post-translational modifications (PTMs) identified in several proteins localized in the extracellular space, on the cell membrane and in the cytoplasm, nucleus and mitochondria. Since protein glycosylation controls several key aspects of cell physiology, aberrant protein glycosylation has been associated with different human diseases, including cancer. Here we review recent evidence indicating the tight association between the HBP flux and cell metabolism, with particular emphasis on the post-transcriptional and transcriptional mechanisms regulated by the HBP that may cause the metabolic rewiring observed in cancer. We describe the implications of both protein O- and N-glycosylation in cancer cell metabolism and bioenergetics; focusing our attention on the effect of these PTMs on nutrient transport and on the transcriptional regulation and function of cancer-specific metabolic pathways.
Collapse
|
21
|
O-GlcNAcylation: key regulator of glycolytic pathways. J Bioenerg Biomembr 2018; 50:189-198. [DOI: 10.1007/s10863-018-9742-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
|
22
|
Gonzalez-Menendez P, Hevia D, Mayo JC, Sainz RM. The dark side of glucose transporters in prostate cancer: Are they a new feature to characterize carcinomas? Int J Cancer 2017; 142:2414-2424. [DOI: 10.1002/ijc.31165] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/01/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Pedro Gonzalez-Menendez
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - David Hevia
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - Juan C. Mayo
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - Rosa M. Sainz
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| |
Collapse
|
23
|
Ahmed A, Arshad M, Malik A, Parveen S, Alsenaidy AM. Camelus dromedarius glucose transporter 4: in silico analysis, cloning, expression, purification and characterisation in E. coli. Arch Physiol Biochem 2017; 123:254-264. [PMID: 28440667 DOI: 10.1080/13813455.2017.1312460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Camels have exceptional carbohydrate metabolism as their plasma glucose level is high and have low whole body insulin sensitivity, similar to that observed in type 2 diabetes patients. We aimed at studing an important component of insulin signalling pathway, the GLUT4, in camel. Camelus dromedarius GLUT4 (CdGLUT4) CDS is 1530 nucleotide in length that encodes for a 55KDa protein. CdGLUT4 has 23 amino acid substitutions and 3N-glycosylation sites, compared to 2 in Human GLUT4. 3 D structures of CdGLUT4 and HsGLUT4 generated by homology modelling revealed conservation of characteristic signature motifs. CdGLUT4 was cloned and expressed optimally in C43(DE3)pLysS strain and maximum detergent solubility was observed in n-Dodecyl-β-d-maltopyranoside. These preliminary data provide information on residual differences between CdGLUT4 and HsGLUT4 that may be responsible for camel's unique glucose metabolism. These differences are postulated to assist in designing and development of efficacious GLUT4 that might help in management of diabetic patients.
Collapse
Affiliation(s)
- Anwar Ahmed
- a Protein Research Chair, Department of Biochemistry, College of Science , King Saud University , Riyadh , Saudi Arabia
- b Centre of Excellence in Biotechnology Research, Department of Biochemistry, College of Science , King Saud University , Riyadh , Saudi Arabia
| | - Mohammed Arshad
- a Protein Research Chair, Department of Biochemistry, College of Science , King Saud University , Riyadh , Saudi Arabia
| | - Ajamaluddin Malik
- a Protein Research Chair, Department of Biochemistry, College of Science , King Saud University , Riyadh , Saudi Arabia
| | - Shama Parveen
- c Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi , India
| | - Abdulrahman M Alsenaidy
- a Protein Research Chair, Department of Biochemistry, College of Science , King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|
24
|
Tee SS, Park JM, Hurd RE, Brimacombe KR, Boxer MB, Massoud TF, Rutt BK, Spielman DM. PKM2 activation sensitizes cancer cells to growth inhibition by 2-deoxy-D-glucose. Oncotarget 2017; 8:90959-90968. [PMID: 29207616 PMCID: PMC5710897 DOI: 10.18632/oncotarget.19630] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 07/06/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer metabolism has emerged as an increasingly attractive target for interfering with tumor growth. Small molecule activators of pyruvate kinase isozyme M2 (PKM2) suppress tumor formation but have an unknown effect on established tumors. We demonstrate that TEPP-46, a PKM2 activator, results in increased glucose consumption, providing the rationale for combining PKM2 activators with the toxic glucose analog, 2-deoxy-D-glucose (2-DG). Combination treatment resulted in reduced viability of a range of cell lines in standard cell culture conditions at concentrations of drugs that had no effect when used alone. This effect was replicated in vivo on established subcutaneous tumors. We further demonstrated the ability to detect acute metabolic differences in combination treatment using hyperpolarized magnetic resonance spectroscopy (MRS). Combination treated tumors displayed a higher pyruvate to lactate 13C-label exchange 2 hr post-treatment. This ability to assess the effect of drugs non-invasively may accelerate the implementation and clinical translation of drugs that target cancer metabolism.
Collapse
Affiliation(s)
- Sui Seng Tee
- Department of Radiology, Stanford University, Stanford, CA, USA.,Current/Present address: Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae Mo Park
- Department of Radiology, Stanford University, Stanford, CA, USA.,Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralph E Hurd
- Applied Sciences Laboratory, GE Healthcare, Menlo Park, CA, USA
| | - Kyle R Brimacombe
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA.,NIH Chemical Genomics Center, Bethesda, MD, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Tarik F Massoud
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Brian K Rutt
- Department of Radiology, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
25
|
Clarke JD, Novak P, Lake AD, Hardwick RN, Cherrington NJ. Impaired N-linked glycosylation of uptake and efflux transporters in human non-alcoholic fatty liver disease. Liver Int 2017; 37:1074-1081. [PMID: 28097795 PMCID: PMC5479731 DOI: 10.1111/liv.13362] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/30/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS N-linked glycosylation of proteins is critical for proper protein folding and trafficking to the plasma membrane. Drug transporters are one class of proteins that have reduced function when glycosylation is impaired. N-linked glycosylation of plasma proteins has also been investigated as a biomarker for several liver diseases, including non-alcoholic fatty liver disease (NAFLD). The purpose of this study was to assess the transcriptomic expression of genes involved in protein processing and glycosylation, and to determine the glycosylation status of key drug transporters during human NAFLD progression. METHODS Human liver samples diagnosed as healthy, steatosis, and non-alcoholic steatohepatitis (NASH) were analysed for gene expression of glycosylation-related genes and for protein glycosylation using immunoblot. RESULTS Genes involved in protein processing in the ER and biosynthesis of N-glycans were significantly enriched for down-regulation in NAFLD progression. Included in the down regulated N-glycan biosynthesis category were genes involved in the oligosaccharyltransferase complex, N-glycan quality control, N-glycan precursor biosynthesis, N-glycan trimming to the core, and N-glycan extension from the core. N-glycan degradation genes were unaltered in the progression to NASH. Immunoblot analysis of the uptake transporters organic anion transporting polypeptide-1B1 (OATP1B1), OATP1B3, OATP2B1, and Sodium/Taurocholate Co-transporting Polypeptide (NTCP) and the efflux transporter multidrug resistance-associated protein 2 (MRP2) demonstrated a significant loss of glycosylation following the progression to NASH. CONCLUSIONS These data suggest that the loss of glycosylation of key uptake and efflux transporters in humans NASH may influence transporter function and contribute to altered drug disposition observed in NASH.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Petr Novak
- Biology Centre ASCR, Institute of Plant Molecular Biology, Ceske Budejovice, Czech Republic
| | - April D Lake
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Rhiannon N Hardwick
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
26
|
Shtraizent N, DeRossi C, Nayar S, Sachidanandam R, Katz LS, Prince A, Koh AP, Vincek A, Hadas Y, Hoshida Y, Scott DK, Eliyahu E, Freeze HH, Sadler KC, Chu J. MPI depletion enhances O-GlcNAcylation of p53 and suppresses the Warburg effect. eLife 2017. [PMID: 28644127 PMCID: PMC5495572 DOI: 10.7554/elife.22477] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Rapid cellular proliferation in early development and cancer depends on glucose metabolism to fuel macromolecule biosynthesis. Metabolic enzymes are presumed regulators of this glycolysis-driven metabolic program, known as the Warburg effect; however, few have been identified. We uncover a previously unappreciated role for Mannose phosphate isomerase (MPI) as a metabolic enzyme required to maintain Warburg metabolism in zebrafish embryos and in both primary and malignant mammalian cells. The functional consequences of MPI loss are striking: glycolysis is blocked and cells die. These phenotypes are caused by induction of p53 and accumulation of the glycolytic intermediate fructose 6-phosphate, leading to engagement of the hexosamine biosynthetic pathway (HBP), increased O-GlcNAcylation, and p53 stabilization. Inhibiting the HBP through genetic and chemical methods reverses p53 stabilization and rescues the Mpi-deficient phenotype. This work provides mechanistic evidence by which MPI loss induces p53, and identifies MPI as a novel regulator of p53 and Warburg metabolism. DOI:http://dx.doi.org/10.7554/eLife.22477.001
Collapse
Affiliation(s)
- Nataly Shtraizent
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Charles DeRossi
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Shikha Nayar
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Ravi Sachidanandam
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Liora S Katz
- Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Adam Prince
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Anna P Koh
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Adam Vincek
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Yoav Hadas
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Yujin Hoshida
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Donald K Scott
- Department of Medicine, Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Hudson H Freeze
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Kirsten C Sadler
- Biology Program, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Jaime Chu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
27
|
Narayan DS, Chidlow G, Wood JP, Casson RJ. Glucose metabolism in mammalian photoreceptor inner and outer segments. Clin Exp Ophthalmol 2017; 45:730-741. [PMID: 28334493 DOI: 10.1111/ceo.12952] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 02/25/2017] [Accepted: 03/20/2017] [Indexed: 12/22/2022]
Abstract
Photoreceptors are the first-order neurons of the visual pathway, converting light into electrical signals. Rods and cones are the two main types of photoreceptors in the mammalian retina. Rods are specialized for sensitivity at the expense of resolution and are responsible for vision in dimly lit conditions. Cones are responsible for high acuity central vision and colour vision. Many human retinal diseases are characterized by a progressive loss of photoreceptors. Photoreceptors consist of four primary regions: outer segments, inner segments, cell bodies and synaptic terminals. Photoreceptors consume large amounts of energy, and therefore, energy metabolism may be a critical juncture that links photoreceptor function and survival. Cones require more energy than rods, and cone degeneration is the main cause of clinically significant vision loss in retinal diseases. Photoreceptor segments are capable of utilizing various energy substrates, including glucose, to meet their large energy demands. The pathways by which photoreceptor segments meet their energy demands remain incompletely understood. Improvements in the understanding of glucose metabolism in photoreceptor segments may provide insight into the reasons why photoreceptors degenerate due to energy failure. This may, in turn, assist in developing bio-energetic therapies aimed at protecting photoreceptors.
Collapse
Affiliation(s)
- Daniel S Narayan
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - John Pm Wood
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - Robert J Casson
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| |
Collapse
|
28
|
Uemura T, Ito S, Ohta Y, Tachikawa M, Wada T, Terasaki T, Ohtsuki S. Abnormal N-Glycosylation of a Novel Missense Creatine Transporter Mutant, G561R, Associated with Cerebral Creatine Deficiency Syndromes Alters Transporter Activity and Localization. Biol Pharm Bull 2017; 40:49-55. [PMID: 28049948 DOI: 10.1248/bpb.b16-00582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cerebral creatine deficiency syndromes (CCDSs) are caused by loss-of-function mutations in creatine transporter (CRT, SLC6A8), which transports creatine at the blood-brain barrier and into neurons of the central nervous system (CNS). This results in low cerebral creatine levels, and patients exhibit mental retardation, poor language skills and epilepsy. We identified a novel human CRT gene missense mutation (c.1681 G>C, G561R) in Japanese CCDSs patients. The purpose of the present study was to evaluate the reduction of creatine transport in G561R-mutant CRT-expressing 293 cells, and to clarify the mechanism of its functional attenuation. G561R-mutant CRT exhibited greatly reduced creatine transport activity compared to wild-type CRT (WT-CRT) when expressed in 293 cells. Also, the mutant protein is localized mainly in intracellular membrane fraction, while WT-CRT is localized in plasma membrane. Western blot analysis revealed a 68 kDa band of WT-CRT protein in plasma membrane fraction, while G561R-mutant CRT protein predominantly showed bands at 55, 110 and 165 kDa in crude membrane fraction. The bands of both WT-CRT and G561R-mutant CRT were shifted to 50 kDa by N-glycosidase treatment. Our results suggest that the functional impairment of G561R-mutant CRT was probably caused by incomplete N-linked glycosylation due to misfolding during protein maturation, leading to oligomer formation and changes of cellular localization.
Collapse
Affiliation(s)
- Tatsuki Uemura
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | | | | | | | | | | | | |
Collapse
|
29
|
Tumova S, Kerimi A, Porter KE, Williamson G. Transendothelial glucose transport is not restricted by extracellular hyperglycaemia. Vascul Pharmacol 2016; 87:219-229. [DOI: 10.1016/j.vph.2016.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/29/2016] [Accepted: 11/02/2016] [Indexed: 01/18/2023]
|
30
|
Ganti K, Massimi P, Manzo-Merino J, Tomaić V, Pim D, Playford MP, Lizano M, Roberts S, Kranjec C, Doorbar J, Banks L. Interaction of the Human Papillomavirus E6 Oncoprotein with Sorting Nexin 27 Modulates Endocytic Cargo Transport Pathways. PLoS Pathog 2016; 12:e1005854. [PMID: 27649450 PMCID: PMC5029876 DOI: 10.1371/journal.ppat.1005854] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 08/09/2016] [Indexed: 01/05/2023] Open
Abstract
A subset of high-risk Human Papillomaviruses (HPVs) are the causative agents of a large number of human cancers, of which cervical is the most common. Two viral oncoproteins, E6 and E7, contribute directly towards the development and maintenance of malignancy. A characteristic feature of the E6 oncoproteins from cancer-causing HPV types is the presence of a PDZ binding motif (PBM) at its C-terminus, which confers interaction with cellular proteins harbouring PDZ domains. Here we show that this motif allows E6 interaction with Sorting Nexin 27 (SNX27), an essential component of endosomal recycling pathways. This interaction is highly conserved across E6 proteins from multiple high-risk HPV types and is mediated by a classical PBM-PDZ interaction but unlike many E6 targets, SNX27 is not targeted for degradation by E6. Rather, in HPV-18 positive cell lines the association of SNX27 with components of the retromer complex and the endocytic transport machinery is altered in an E6 PBM-dependent manner. Analysis of a SNX27 cargo, the glucose transporter GLUT1, reveals an E6-dependent maintenance of GLUT1 expression and alteration in its association with components of the endocytic transport machinery. Furthermore, knockdown of E6 in HPV-18 positive cervical cancer cells phenocopies the loss of SNX27, both in terms of GLUT1 expression levels and its vesicular localization, with a concomitant marked reduction in glucose uptake, whilst loss of SNX27 results in slower cell proliferation in low nutrient conditions. These results demonstrate that E6 interaction with SNX27 can alter the recycling of cargo molecules, one consequence of which is modulation of nutrient availability in HPV transformed tumour cells.
Collapse
Affiliation(s)
- Ketaki Ganti
- International Centre for Genetic Engineering and Biotechnology, Padriciano, Trieste, Italy
| | - Paola Massimi
- International Centre for Genetic Engineering and Biotechnology, Padriciano, Trieste, Italy
| | - Joaquin Manzo-Merino
- CONACyT Research Fellow, Instituto Nacional de Cancerologia, Mexico/Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico. Col. Seccion XVI, Tlalpan, Mexico
| | - Vjekoslav Tomaić
- International Centre for Genetic Engineering and Biotechnology, Padriciano, Trieste, Italy
- Division of Molecular Medicine, Ruđjer Bošković Institute, Zagreb, Croatia
| | - David Pim
- International Centre for Genetic Engineering and Biotechnology, Padriciano, Trieste, Italy
| | - Martin P. Playford
- National Heart, Blood and Lung Institute, NIH, Bethesda, Maryland, United States of America
| | - Marcela Lizano
- Instituto Nacional de Cancerologia, Mexico/Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico. Col. Seccion XVI, Tlalpan, Mexico
| | - Sally Roberts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christian Kranjec
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Padriciano, Trieste, Italy
- * E-mail:
| |
Collapse
|
31
|
Gu M, Chen A, Sun S, Xu G. Complex Regulation of Plant Phosphate Transporters and the Gap between Molecular Mechanisms and Practical Application: What Is Missing? MOLECULAR PLANT 2016; 9:396-416. [PMID: 26714050 DOI: 10.1016/j.molp.2015.12.012] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/18/2015] [Accepted: 12/11/2015] [Indexed: 05/18/2023]
Abstract
It has been almost 25 years since the first report of the gene encoding a high-affinity phosphate transporter (PT), PHO84, in yeast. Since then, an increasing number of yeast PHO84 homologs as well as other genes encoding proteins with phosphate (Pi) transport activities have been identified and functionally characterized in diverse plant species. Great progress has been made also in deciphering the molecular mechanism underlying the regulation of the abundance and/or activity of these genes and their products. The regulatory genes affect plant Pi homeostasis commonly through direct or indirect regulation of the abundance of PTs at different levels. However, little has been achieved in the use of PTs for developing genetically modified crops with high phosphorus use efficiency (PUE). This might be a consequence of overemphasizing Pi uptake from the rhizosphere and lack of knowledge about the roles of PTs in Pi transport and recycling within the plant that are required to optimize PUE. Here, we mainly focused on the genes encoding proteins with Pi transport activities and the emerging understanding of their regulation at the transcriptional, post-transcriptional, translational, and post-translational levels. In addition, we propose potential strategies for effective use of PTs in improving plant growth and development.
Collapse
Affiliation(s)
- Mian Gu
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China; MOA Key Laboratory of Plant Nutrition and Fertilization in Lower-Middle Reaches of the Yangtze River, Nanjing 210095, China
| | - Aiqun Chen
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China; MOA Key Laboratory of Plant Nutrition and Fertilization in Lower-Middle Reaches of the Yangtze River, Nanjing 210095, China
| | - Shubin Sun
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China; MOA Key Laboratory of Plant Nutrition and Fertilization in Lower-Middle Reaches of the Yangtze River, Nanjing 210095, China
| | - Guohua Xu
- State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China; MOA Key Laboratory of Plant Nutrition and Fertilization in Lower-Middle Reaches of the Yangtze River, Nanjing 210095, China.
| |
Collapse
|
32
|
Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol Neurobiol 2016; 54:1046-1077. [PMID: 26801191 DOI: 10.1007/s12035-015-9672-6] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Glucose transporters (GLUTs) at the blood-brain barrier maintain the continuous high glucose and energy demands of the brain. They also act as therapeutic targets and provide routes of entry for drug delivery to the brain and central nervous system for treatment of neurological and neurovascular conditions and brain tumours. This article first describes the distribution, function and regulation of glucose transporters at the blood-brain barrier, the major ones being the sodium-independent facilitative transporters GLUT1 and GLUT3. Other GLUTs and sodium-dependent transporters (SGLTs) have also been identified at lower levels and under various physiological conditions. It then considers the effects on glucose transporter expression and distribution of hypoglycemia and hyperglycemia associated with diabetes and oxygen/glucose deprivation associated with cerebral ischemia. A reduction in glucose transporters at the blood-brain barrier that occurs before the onset of the main pathophysiological changes and symptoms of Alzheimer's disease is a potential causative effect in the vascular hypothesis of the disease. Mutations in glucose transporters, notably those identified in GLUT1 deficiency syndrome, and some recreational drug compounds also alter the expression and/or activity of glucose transporters at the blood-brain barrier. Approaches for drug delivery across the blood-brain barrier include the pro-drug strategy whereby drug molecules are conjugated to glucose transporter substrates or encapsulated in nano-enabled delivery systems (e.g. liposomes, micelles, nanoparticles) that are functionalised to target glucose transporters. Finally, the continuous development of blood-brain barrier in vitro models is important for studying glucose transporter function, effects of disease conditions and interactions with drugs and xenobiotics.
Collapse
|
33
|
Navale AM, Paranjape AN. Glucose transporters: physiological and pathological roles. Biophys Rev 2016; 8:5-9. [PMID: 28510148 DOI: 10.1007/s12551-015-0186-2] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 12/01/2015] [Indexed: 12/17/2022] Open
Abstract
Glucose is a primary energy source for most cells and an important substrate for many biochemical reactions. As glucose is a need of each and every cell of the body, so are the glucose transporters. Consequently, all cells express these important proteins on their surface. In recent years developments in genetics have shed new light on the types and physiology of various glucose transporters, of which there are two main types-sodium-glucose linked transporters (SGLTs) and facilitated diffusion glucose transporters (GLUT)-which can be divided into many more subclasses. Transporters differ in terms of their substrate specificity, distribution and regulatory mechanisms. Glucose transporters have also received much attention as therapeutic targets for various diseases. In this review, we attempt to present a simplified view of this complex topic which may be of interest to researchers involved in biochemical and pharmacological research.
Collapse
Affiliation(s)
- Archana M Navale
- Department of Pharmacology, Faculty of Pharmacy, Parul University, P.O. Limda, Waghodia Taluka, Vadodara District, 391760, Gujarat, India.
| | | |
Collapse
|
34
|
Stelzl T, Baranov T, Geillinger KE, Kottra G, Daniel H. Effect of N-glycosylation on the transport activity of the peptide transporter PEPT1. Am J Physiol Gastrointest Liver Physiol 2016; 310:G128-41. [PMID: 26585416 DOI: 10.1152/ajpgi.00350.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/10/2015] [Indexed: 01/31/2023]
Abstract
The intestinal peptide transporter PEPT1 provides bulk quantities of amino acids to epithelial cells. PEPT1 is a high-capacity and low-affinity solute carrier of the SLC15 family found in apical membranes of enterocytes in small intestine and distal colon. Surprisingly, murine PEPT1 (mPEPT1) has an apparent molecular mass of ∼95 kDa in the small intestine but ∼105 kDa in the large intestine. Here we describe studies on mPEPT1 protein glycosylation and how glycans affect transport function. Putative N-glycosylation sites of mPEPT1 were altered by site-directed mutagenesis followed by expression in Xenopus laevis oocytes. Replacement of six asparagine residues (N) at positions N50, N406, N439, N510, N515, and N532 by glutamine (Q) resulted in a decrease of the mPEPT1 mass by around 35 kDa. Electrophysiology revealed all glycosylation-deficient transporters to be functional with comparable expression levels in oocyte membranes. Strikingly, the mutant protein with N50Q exhibited a twofold decreased affinity for Gly-Sar but a 2.5-fold rise in the maximal inward currents compared with the wild-type protein. Elevated maximal transport currents were also recorded for cefadroxil and tri-l-alanine. Tracer flux studies performed with [(14)C]-Gly-Sar confirmed the reduction in substrate affinity and showed twofold increased maximal transport rates for the N50Q transporter. Elimination of individual N-glycosylation sites did not alter membrane expression in oocytes or overall transport characteristics except for the mutant protein N50Q. Because transporter surface density was not altered in N50Q, removal of the glycan at this location appears to accelerate the substrate turnover rate.
Collapse
Affiliation(s)
- Tamara Stelzl
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Tatjana Baranov
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Kerstin E Geillinger
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Gabor Kottra
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Hannelore Daniel
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| |
Collapse
|
35
|
Martínez-Quintana JA, Kikuta S, Felix-Portillo M, Peregrino-Uriarte AB, Yepiz-Plascencia G. A novel functional glucose transporter in the white shrimp Litopenaeus vannamei -LvGLUT2- is up-regulated during hypoxia in hepatopancreas. MARINE ENVIRONMENTAL RESEARCH 2015; 112:61-67. [PMID: 26415036 DOI: 10.1016/j.marenvres.2015.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/11/2015] [Accepted: 09/13/2015] [Indexed: 06/05/2023]
Abstract
In hypoxia conditions, the white shrimp Litopenaeus vannamei shifts its energetic metabolism from aerobic to anaerobic, requiring more glucose uptake into the cells by GLUT proteins. We here report a novel glucose transporter in shrimp. The Lvglut2 cDNA is 2473 bp-long containing an ORF of 1458 bp encoding 486 amino acid residues. The deduced protein has the features of a facilitative sugar transporter. The Lvglut2 gene product tagged with GFP was expressed in the cell membrane of Xenopus oocytes. In the same expression system, untagged LvGLUT2 resulted to be a bidirectional glucose transporter that functions moving glucose down its concentration gradient in and out of the cell. Lvglut2 mRNA is expressed in hepatopancreas while in muscle and gills it was not detected. Hypoxia up-regulates the expression of Lvglut2 transcripts in hepatopancreas. These results provide a better understanding of facilitative glucose transporters and gene regulation during hypoxia in crustaceans.
Collapse
Affiliation(s)
| | - Shingo Kikuta
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | |
Collapse
|
36
|
Chien MW, Lin MH, Huang SH, Fu SH, Hsu CY, Yen BLJ, Chen JT, Chang DM, Sytwu HK. Glucosamine Modulates T Cell Differentiation through Down-regulating N-Linked Glycosylation of CD25. J Biol Chem 2015; 290:29329-44. [PMID: 26468284 DOI: 10.1074/jbc.m115.674671] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Indexed: 01/16/2023] Open
Abstract
Glucosamine has immunomodulatory effects on autoimmune diseases. However, the mechanism(s) through which glucosamine modulates different T cell subsets and diseases remain unclear. We demonstrate that glucosamine impedes Th1, Th2, and iTreg but promotes Th17 differentiation through down-regulating N-linked glycosylation of CD25 and subsequently inhibiting its downstream Stat5 signaling in a dose-dependent manner. The effect of glucosamine on T helper cell differentiation was similar to that induced by anti-IL-2 treatment, further supporting an IL-2 signaling-dependent modulation. Interestingly, excess glucose rescued this glucosamine-mediated regulation, suggesting a functional competition between glucose and glucosamine. High-dose glucosamine significantly decreased Glut1 N-glycosylation in Th1-polarized cells. This finding suggests that both down-regulated IL-2 signaling and Glut1-dependent glycolytic metabolism contribute to the inhibition of Th1 differentiation by glucosamine. Finally, glucosamine treatment inhibited Th1 cells in vivo, prolonged the survival of islet grafts in diabetic recipients, and exacerbated the severity of EAE. Taken together, our results indicate that glucosamine interferes with N-glycosylation of CD25, and thereby attenuates IL-2 downstream signaling. These effects suggest that glucosamine may be an important modulator of T cell differentiation and immune homeostasis.
Collapse
Affiliation(s)
- Ming-Wei Chien
- From the Graduate Institute of Life Sciences, Department and Graduate Institute of Microbiology and Immunology
| | - Ming-Hong Lin
- Department and Graduate Institute of Microbiology and Immunology
| | | | - Shin-Huei Fu
- Department and Graduate Institute of Microbiology and Immunology
| | - Chao-Yuan Hsu
- From the Graduate Institute of Life Sciences, Department and Graduate Institute of Microbiology and Immunology
| | - B Lin-Ju Yen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, 35053 Taiwan
| | | | - Deh-Ming Chang
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490 Taiwan and
| | - Huey-Kang Sytwu
- From the Graduate Institute of Life Sciences, Department and Graduate Institute of Microbiology and Immunology,
| |
Collapse
|
37
|
Liemburg-Apers DC, Willems PHGM, Koopman WJH, Grefte S. Interactions between mitochondrial reactive oxygen species and cellular glucose metabolism. Arch Toxicol 2015; 89:1209-26. [PMID: 26047665 PMCID: PMC4508370 DOI: 10.1007/s00204-015-1520-y] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
Mitochondrial reactive oxygen species (ROS) production and detoxification are tightly balanced. Shifting this balance enables ROS to activate intracellular signaling and/or induce cellular damage and cell death. Increased mitochondrial ROS production is observed in a number of pathological conditions characterized by mitochondrial dysfunction. One important hallmark of these diseases is enhanced glycolytic activity and low or impaired oxidative phosphorylation. This suggests that ROS is involved in glycolysis (dys)regulation and vice versa. Here we focus on the bidirectional link between ROS and the regulation of glucose metabolism. To this end, we provide a basic introduction into mitochondrial energy metabolism, ROS generation and redox homeostasis. Next, we discuss the interactions between cellular glucose metabolism and ROS. ROS-stimulated cellular glucose uptake can stimulate both ROS production and scavenging. When glucose-stimulated ROS production, leading to further glucose uptake, is not adequately counterbalanced by (glucose-stimulated) ROS scavenging systems, a toxic cycle is triggered, ultimately leading to cell death. Here we inventoried the various cellular regulatory mechanisms and negative feedback loops that prevent this cycle from occurring. It is concluded that more insight in these processes is required to understand why they are (un)able to prevent excessive ROS production during various pathological conditions in humans.
Collapse
Affiliation(s)
- Dania C. Liemburg-Apers
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter H. G. M. Willems
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Werner J. H. Koopman
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Sander Grefte
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
- />Department of Human and Animal Physiology, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands
| |
Collapse
|
38
|
Park MS. Molecular Dynamics Simulations of the Human Glucose Transporter GLUT1. PLoS One 2015; 10:e0125361. [PMID: 25919356 PMCID: PMC4412407 DOI: 10.1371/journal.pone.0125361] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/12/2015] [Indexed: 01/31/2023] Open
Abstract
Glucose transporters (GLUTs) provide a pathway for glucose transport across membranes. Human GLUTs are implicated in devastating diseases such as heart disease, hyper- and hypo-glycemia, type 2 diabetes and cancer. The human GLUT1 has been recently crystalized in the inward-facing open conformation. However, there is no other structural information for other conformations. The X-ray structures of E. coli Xylose permease (XylE), a glucose transporter homolog, are available in multiple conformations with and without the substrates D-xylose and D-glucose. XylE has high sequence homology to human GLUT1 and key residues in the sugar-binding pocket are conserved. Here we construct a homology model for human GLUT1 based on the available XylE crystal structure in the partially occluded outward-facing conformation. A long unbiased all atom molecular dynamics simulation starting from the model can capture a new fully opened outward-facing conformation. Our investigation of molecular interactions at the interface between the transmembrane (TM) domains and the intracellular helices (ICH) domain in the outward- and inward-facing conformation supports that the ICH domain likely stabilizes the outward-facing conformation in GLUT1. Furthermore, inducing a conformational transition, our simulations manifest a global asymmetric rocker switch motion and detailed molecular interactions between the substrate and residues through the water-filled selective pore along a pathway from the extracellular to the intracellular side. The results presented here are consistent with previously published biochemical, mutagenesis and functional studies. Together, this study shed light on the structure and functional relationships of GLUT1 in multiple conformational states.
Collapse
Affiliation(s)
- Min-Sun Park
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
- * E-mail:
| |
Collapse
|
39
|
Console L, Scalise M, Tarmakova Z, Coe IR, Indiveri C. N-linked glycosylation of human SLC1A5 (ASCT2) transporter is critical for trafficking to membrane. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1636-45. [PMID: 25862406 DOI: 10.1016/j.bbamcr.2015.03.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/11/2022]
Abstract
The human amino acid transporter SLC1A5 (ASCT2) contains two N-glycosylation sites (N163 and N212) located in the large extracellular loop. In the homology structural model of ASCT2 these Asn residues are extracellularly exposed. Mutants of the two Asn exhibited altered electrophoretic mobility. N163Q and N212Q displayed multiple bands with apparent molecular masses from 80kDa to 50kDa. N163/212Q displayed a single band of 50kDa corresponding to the unglycosylated protein. The presence in membrane of WT and mutants was evaluated by protein biotinylation assay followed by immunoblotting. The double mutation significantly impaired the presence of the protein in membrane, without impairment in protein synthesis. [(3)H]glutamine transport was measured in cells transiently transfected with the WT or mutants. N163/212Q exhibited a strongly reduced transport activity correlating with reduced surface expression. The same proteins extracted from cells and reconstituted in liposomes showed comparable transport activities demonstrating that the intrinsic transport function of the mutants was not affected. The rate of endocytosis of ASCT2 was assayed by a reversible biotinylation strategy. N212Q and N163/212Q showed strongly increased rates of endocytosis respect to WT. ASCT2 stability was determined using cycloheximide. N163Q or N163/212Q showed a slightly or significantly lower stability with respect to WT. To assess trafficking to the membrane, a brefeldin-based assay, which caused retention of proteins in ER, was performed. One hour after brefeldin removal WT protein was localized to the plasma membrane while the double mutant was localized in the cytosol. The results demonstrate that N-glycosylation is critical for trafficking.
Collapse
Affiliation(s)
- Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; Department of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto M5B 2K3, Canada
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Zlatina Tarmakova
- Department of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto M5B 2K3, Canada
| | - Imogen R Coe
- Department of Chemistry and Biology, Ryerson University, 350 Victoria St., Toronto M5B 2K3, Canada
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| |
Collapse
|
40
|
Rojas R, Segovia C, Trombert AN, Santander J, Manque P. The effect of tunicamycin on the glucose uptake, growth, and cellular adhesion in the protozoan parasite Crithidia fasciculata. Curr Microbiol 2014; 69:541-8. [PMID: 24894907 DOI: 10.1007/s00284-014-0620-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/08/2014] [Indexed: 01/17/2023]
Abstract
Crithidia fasciculata represents a very interesting model organism to study biochemical, cellular, and genetic processes unique to members of the family of the Trypanosomatidae. Thus, C. fasciculata parasitizes several species of insects and has been widely used to test new therapeutic strategies against parasitic infections. By using tunicamycin, a potent inhibitor of glycosylation in asparaginyl residues of glycoproteins (N-glycosylation), we demonstrate that N-glycosylation in C. fasciculata cells is involved in modulating glucose uptake, dramatically impacting growth, and cell adhesion. C. fasciculata treated with tunicamycin was severely affected in their ability to replicate and to adhere to polystyrene substrates and losing their ability to aggregate into small and large groups. Moreover, under tunicamycin treatment, the parasites were considerably shorter and rounder and displayed alterations in cytoplasmic vesicles formation. Furthermore, glucose uptake was significantly impaired in a tunicamycin dose-dependent manner; however, no cytotoxic effect was observed. Interestingly, this effect was reversible. Thus, when tunicamycin was removed from the culture media, the parasites recovered its growth rate, cell adhesion properties, and glucose uptake. Collectively, these results suggest that changes in the tunicamycin-dependent glycosylation levels can influence glucose uptake, cell growth, and adhesion in the protozoan parasite C. fasciculata.
Collapse
Affiliation(s)
- Robert Rojas
- Nucleus for Microbiology and Immunity, Center for Genomics and Bioinformatics, Faculty of Sciences, Universidad Mayor, Campus Huechuraba, Camino La Pirámide 5750, Huechuraba, Santiago, Chile,
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Palmer CS, Ostrowski M, Gouillou M, Tsai L, Yu D, Zhou J, Henstridge DC, Maisa A, Hearps AC, Lewin SR, Landay A, Jaworowski A, McCune JM, Crowe SM. Increased glucose metabolic activity is associated with CD4+ T-cell activation and depletion during chronic HIV infection. AIDS 2014; 28:297-309. [PMID: 24335483 PMCID: PMC4293200 DOI: 10.1097/qad.0000000000000128] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Glucose metabolism plays a fundamental role in supporting the growth, proliferation and effector functions of T cells. We investigated the impact of HIV infection on key processes that regulate glucose uptake and metabolism in primary CD4 and CD8 T cells. DESIGN AND METHODS Thirty-eight HIV-infected treatment-naive, 35 HIV+/combination antiretroviral therapy, seven HIV+ long-term nonprogressors and 25 HIV control individuals were studied. Basal markers of glycolysis [e.g. glucose transporter-1 (Glut1) expression, glucose uptake, intracellular glucose-6-phosphate, and L-lactate] were measured in T cells. The cellular markers of immune activation, CD38 and HLA-DR, were measured by flow cytometry. RESULTS The surface expression of the Glut1 is up-regulated in CD4 T cells in HIV-infected patients compared with uninfected controls. The percentage of circulating CD4Glut1 T cells was significantly increased in HIV-infected patients and was not restored to normal levels following combination antiretroviral therapy. Basal markers of glycolysis were significantly higher in CD4Glut1 T cells compared to CD4Glut1 T cells. The proportion of CD4Glut1 T cells correlated positively with the expression of the cellular activation marker, HLA-DR, on total CD4 T cells, but inversely with the absolute CD4 T-cell count irrespective of HIV treatment status. CONCLUSION Our data suggest that Glut1 is a potentially novel and functional marker of CD4 T-cell activation during HIV infection. In addition, Glut1 expression on CD4 T cells may be exploited as a prognostic marker for CD4 T-cell loss during HIV disease progression.
Collapse
Affiliation(s)
- Clovis S Palmer
- aCentre for Biomedical Research, Burnet Institute, Melbourne, Australia bInstituto de Investigaciones Biomédicas en Retrovirus y SIDA. Facultad de Medicina, Buenos Aires, Argentina cCentre for Population Health, Burnet Institute, Melbourne dLaboratory of Molecular Immunomodulation, School of Biomedical Sciences, Monash University, Clayton eCellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute fDepartment of Infectious Diseases, Monash University gInfectious Diseases Department, The Alfred hospital, Melbourne, Australia hDepartment of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois iDivision of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Talukder JR, Griffin A, Jaima A, Boyd B, Wright J. Lactoferrin ameliorates prostaglandin E2-mediated inhibition of Na+-glucose cotransport in enterocytes. Can J Physiol Pharmacol 2014; 92:9-20. [DOI: 10.1139/cjpp-2013-0211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Various immunoinflammatory cytokines are produced during chronic intestinal inflammation, which inhibits Na+-glucose cotransport (SGLT1) in villus cells. Lactoferrin (Lf), abundantly present in colostrum, is a multifunctional glycoprotein that is absorbed by receptor-mediated transcytosis in humans and animals and has been shown to exert anti-inflammatory effects. Therefore, this study aimed to examine whether Lf would prevent PGE2 effect on SGLT1 for glucose absorption in enterocytes. Intestinal epithelial cells (IEC-6) were grown on transwell plates, treated with phlorizin, PGE2, AH6809, and Lf, and 3-O-methyl d-glucopyranose (OMG) uptake was measured in 10 days postconfluent. Na+-dependent OMG uptake, phlorizin, and immunoblotting studies established the activity and apical membrane localization of SGLT1 in IEC-6 cells. PGE2 inhibited SGLT1 in a concentration- and time-dependent manner with an inhibitory constant (Ki) of 50.0 nmol/L and that was antagonized by prostanoid receptor inhibitor, AH6809. PGE2 did not alter Na+/K+-ATPase activity. In contrast, quantitative real-time polymerase chain reaction and Western blot analyses revealed that SGLT1-specific transcripts and protein expression level were decreased 3-fold by PGE2. Furthermore, PGE2 treatment increased intracellular cyclic adenosine monophosphate (cAMP) and Ca2+ concentrations and decreased SGLT1 expression on the apical membrane, and these effects were ameliorated by Lf. Therefore, we conclude that Lf ameliorates the PGE2 inhibition of SGLT1 most likely via the Ca2+- and cAMP-signaling pathways.
Collapse
Affiliation(s)
- Jamilur R. Talukder
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Ashley Griffin
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Antara Jaima
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Brittney Boyd
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Jaleesa Wright
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| |
Collapse
|
44
|
Abstract
GLUT4 is regulated by its intracellular localization. In the absence of insulin, GLUT4 is efficiently retained intracellularly within storage compartments in muscle and fat cells. Upon insulin stimulation (and contraction in muscle), GLUT4 translocates from these compartments to the cell surface where it transports glucose from the extracellular milieu into the cell. Its implication in insulin-regulated glucose uptake makes GLUT4 not only a key player in normal glucose homeostasis but also an important element in insulin resistance and type 2 diabetes. Nevertheless, how GLUT4 is retained intracellularly and how insulin acts on this retention mechanism is largely unclear. In this review, the current knowledge regarding the various molecular processes that govern GLUT4 physiology is discussed as well as the questions that remain.
Collapse
|
45
|
Cura AJ, Carruthers A. Role of monosaccharide transport proteins in carbohydrate assimilation, distribution, metabolism, and homeostasis. Compr Physiol 2013; 2:863-914. [PMID: 22943001 DOI: 10.1002/cphy.c110024] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The facilitated diffusion of glucose, galactose, fructose, urate, myoinositol, and dehydroascorbicacid in mammals is catalyzed by a family of 14 monosaccharide transport proteins called GLUTs. These transporters may be divided into three classes according to sequence similarity and function/substrate specificity. GLUT1 appears to be highly expressed in glycolytically active cells and has been coopted in vitamin C auxotrophs to maintain the redox state of the blood through transport of dehydroascorbate. Several GLUTs are definitive glucose/galactose transporters, GLUT2 and GLUT5 are physiologically important fructose transporters, GLUT9 appears to be a urate transporter while GLUT13 is a proton/myoinositol cotransporter. The physiologic substrates of some GLUTs remain to be established. The GLUTs are expressed in a tissue specific manner where affinity, specificity, and capacity for substrate transport are paramount for tissue function. Although great strides have been made in characterizing GLUT-catalyzed monosaccharide transport and mapping GLUT membrane topography and determinants of substrate specificity, a unifying model for GLUT structure and function remains elusive. The GLUTs play a major role in carbohydrate homeostasis and the redistribution of sugar-derived carbons among the various organ systems. This is accomplished through a multiplicity of GLUT-dependent glucose sensing and effector mechanisms that regulate monosaccharide ingestion, absorption,distribution, cellular transport and metabolism, and recovery/retention. Glucose transport and metabolism have coevolved in mammals to support cerebral glucose utilization.
Collapse
Affiliation(s)
- Anthony J Cura
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
46
|
Liao ND, Chiang TA, Lee WY. Glucose transporter 1 protein detected by enzyme-linked immunosorbent assay and immunocytochemistry. Cancer Cytopathol 2013; 121:695-702. [DOI: 10.1002/cncy.21324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 01/27/2023]
Affiliation(s)
- Nai-Ding Liao
- Department of Cytopathology; Chi Mei Medical Center; Tainan Taiwan
| | - Tai-An Chiang
- Department of Medical Laboratory Science and Biotechnology; Chung Hwa University of Medical Technology; Tainan Taiwan
| | - Wen-Ying Lee
- Department of Cytopathology; Chi Mei Medical Center; Tainan Taiwan
- Department of Medical Laboratory Science and Biotechnology; Chung Hwa University of Medical Technology; Tainan Taiwan
- Department of Pathology, College of Medicine; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
47
|
Chip S, Nitsch C, Wellmann S, Kapfhammer JP. Subfield-specific neurovascular remodeling in the entorhino-hippocampal-organotypic slice culture as a response to oxygen-glucose deprivation and excitotoxic cell death. J Cereb Blood Flow Metab 2013; 33:508-18. [PMID: 23232944 PMCID: PMC3618384 DOI: 10.1038/jcbfm.2012.190] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Transient ischemia causes delayed neurodegeneration in selective brain areas, particularly in the CA1 field of the hippocampus. This is accompanied by neurovascular impairment. It is unknown whether neurodegeneration is the cause or consequence of vascular changes. In an entorhino-hippocampal-organotypic slice culture system with well-preserved blood vessels, we studied the interplay between neurodegeneration and neurovasculature. Short-term oxygen and glucose deprivation (OGD) resulted in upregulation of hypoxic markers and with a delay of 24 to 48 hours in selective nerve cell death in CA1. In parallel, local vessel density decreased as detected by markers of endothelial cells and of the extracellular matrix. Claudin-5, a tight junction protein and marker of the blood-brain barrier was reduced. Preventing neuronal death with tetrodotoxin or 6-cyano-7-nitroquinoxaline-2,3-dione rescued blood vessels, suggesting that vessel loss is not due to OGD per se but a consequence of neuronal death. Induction of excitotoxic neuronal death with AMPA caused widespread neurodegeneration, but vessel reduction was confined to CA1. In dentate gyrus without neuronal loss, vessel density increased. We propose that neuronal stress and death influence maintenance, loss and remodeling of the neurovasculature and that the type of vascular response is in addition determined by local factors within the hippocampus.
Collapse
Affiliation(s)
- Sophorn Chip
- Department of Biomedicine, Section of Functional Neuroanatomy, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
48
|
Pyla R, Poulose N, Jun JY, Segar L. Expression of conventional and novel glucose transporters, GLUT1, -9, -10, and -12, in vascular smooth muscle cells. Am J Physiol Cell Physiol 2013; 304:C574-89. [PMID: 23302780 DOI: 10.1152/ajpcell.00275.2012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intimal hyperplasia is characterized by exaggerated proliferation of vascular smooth muscle cells (VSMCs). Enhanced VSMC growth is dependent on increased glucose uptake and metabolism. Facilitative glucose transporters (GLUTs) are comprised of conventional GLUT isoforms (GLUT1-5) and novel GLUT isoforms (GLUT6-14). Previous studies demonstrate that GLUT1 overexpression or GLUT10 downregulation contribute to phenotypic changes in VSMCs. To date, the expression profile of all 14 GLUT isoforms has not been fully examined in VSMCs. Using the proliferative and differentiated phenotypes of human aortic VSMCs, the present study has determined the relative abundance of GLUT1-14 mRNAs by quantitative real-time PCR analysis. Twelve GLUT mRNAs excluding GLUT7 and GLUT14 were detectable in VSMCs. In the proliferative phenotype, the relative abundance of key GLUT mRNAs was GLUT1 (∼43%)>GLUT10 (∼26%)>GLUT9 (∼13%)>GLUT12 (∼4%), whereas in the differentiated phenotype the relative abundance was GLUT10 (∼28%)>GLUT1 (∼25%)>GLUT12 (∼20%)>GLUT9 (∼14%), together constituting 86-87% of total GLUT transcripts. To confirm the expression of key GLUT proteins, immunoblot and immunocytochemical analyses were performed using GLUT isoform-specific primary antibodies. The protein bands characteristic of GLUT1, -9, -10, and -12 were detected in VSMCs in parallel with respective positive controls. In particular, GLUT1 protein expression showed different molecular forms representative of altered glycosylation. While GLUT1 protein displayed a predominant distribution in the plasma membrane, GLUT9, -10, and -12 proteins were mostly distributed in the intracellular compartments. The present study provides the first direct evidence for GLUT9 and GLUT12 expression in VSMCs in conjunction with the previously identified GLUT1 and GLUT10.
Collapse
Affiliation(s)
- Rajkumar Pyla
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912-2450, USA
| | | | | | | |
Collapse
|
49
|
Luni C, Marth JD, Doyle FJ. Computational modeling of glucose transport in pancreatic β-cells identifies metabolic thresholds and therapeutic targets in diabetes. PLoS One 2012; 7:e53130. [PMID: 23300881 PMCID: PMC3531366 DOI: 10.1371/journal.pone.0053130] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/23/2012] [Indexed: 11/18/2022] Open
Abstract
Pancreatic β-cell dysfunction is a diagnostic criterion of Type 2 diabetes and includes defects in glucose transport and insulin secretion. In healthy individuals, β-cells maintain plasma glucose concentrations within a narrow range in concert with insulin action among multiple tissues. Postprandial elevations in blood glucose facilitate glucose uptake into β-cells by diffusion through glucose transporters residing at the plasma membrane. Glucose transport is essential for glycolysis and glucose-stimulated insulin secretion. In human Type 2 diabetes and in the mouse model of obesity-associated diabetes, a marked deficiency of β-cell glucose transporters and glucose uptake occurs with the loss of glucose-stimulated insulin secretion. Recent studies have shown that the preservation of glucose transport in β-cells maintains normal insulin secretion and blocks the development of obesity-associated diabetes. To further elucidate the underlying mechanisms, we have constructed a computational model of human β-cell glucose transport in health and in Type 2 diabetes, and present a systems analysis based on experimental results from human and animal studies. Our findings identify a metabolic threshold or "tipping point" whereby diminished glucose transport across the plasma membrane of β-cells limits intracellular glucose-6-phosphate production by glucokinase. This metabolic threshold is crossed in Type 2 diabetes and results in β-cell dysfunction including the loss of glucose stimulated insulin secretion. Our model further discriminates among molecular control points in this pathway wherein maximal therapeutic intervention is achieved.
Collapse
Affiliation(s)
- Camilla Luni
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
- Sansum Diabetes Research Institute, Santa Barbara, California, United States of America
| | - Jamey D. Marth
- Center for Nanomedicine, Sanford-Burnham Medical Research Institute and the Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Francis J. Doyle
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
- Sansum Diabetes Research Institute, Santa Barbara, California, United States of America
- * E-mail:
| |
Collapse
|
50
|
Bentley PA, Shao Y, Misra Y, Morielli AD, Zhao FQ. Characterization of bovine glucose transporter 1 kinetics and substrate specificities in Xenopus oocytes. J Dairy Sci 2012; 95:1188-97. [PMID: 22365203 DOI: 10.3168/jds.2011-4430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 11/03/2011] [Indexed: 11/19/2022]
Abstract
Glucose is an essential substrate for lactose synthesis and an important energy source in milk production. Glucose uptake in the mammary gland, therefore, plays a critical role in milk synthesis. Facilitative glucose transporters (GLUT) mediate glucose uptake in the mammary gland. Glucose transporter 1 (GLUT1) is the major facilitative glucose transporter expressed in the bovine mammary gland and has been shown to localize to the basolateral membrane of mammary epithelial cells. Glucose transporter 1 is, therefore, thought to play a major role in glucose uptake during lactation. The objective of this study was to determine the transport kinetic properties and substrate specificity of bovine GLUT1 using the Xenopus oocyte model. Bovine GLUT1 (bGLUT1) was expressed in Xenopus oocytes by microinjection of in vitro transcribed cRNA and was found to be localized to the plasma membrane, which resulted in increased glucose uptake. This bGLUT1-mediated glucose uptake was dramatically inhibited by specific facilitative glucose transport inhibitors, cytochalasin B, and phloretin. Kinetic analysis of bovine and human GLUT1 was conducted under zero-trans conditions using radio-labeled 2-deoxy-D-glucose and the principles of Michaelis-Menten kinetics. Bovine GLUT1 exhibited a Michaelis constant (K(m)) of 9.8 ± 3.0mM for 2-deoxy-d-glucose, similar to 11.7 ± 3.7 mM for human GLUT1. Transport by bGLUT1 was inhibited by mannose and galactose, but not fructose, indicating that bGLUT1 may also be able to transport mannose and galactose. Our data provides functional insight into the transport properties of bGLUT1 in taking up glucose across mammary epithelial cells for milk synthesis.
Collapse
Affiliation(s)
- P A Bentley
- Laboratory of Lactation Physiology, Department of Animal Science, University of Vermont, Burlington 05405, USA
| | | | | | | | | |
Collapse
|