To see the other types of publications on this topic, follow the link: GLUT12.

Journal articles on the topic 'GLUT12'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'GLUT12.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Stuart, Charles A., Deling Yin, Mary E. A. Howell, Rhesa J. Dykes, John J. Laffan, and Arny A. Ferrando. "Hexose transporter mRNAs for GLUT4, GLUT5, and GLUT12 predominate in human muscle." American Journal of Physiology-Endocrinology and Metabolism 291, no. 5 (2006): E1067—E1073. http://dx.doi.org/10.1152/ajpendo.00250.2006.

Full text
Abstract:
In the past few years, 8 additional members of the facilitative hexose transporter family have been identified, giving a total of 14 members of the SLC2A family of membrane-bound hexose transporters. To determine which of the new hexose transporters were expressed in muscle, mRNA concentrations of 11 glucose transporters (GLUTs) were quantified and compared. RNA from muscle from 10 normal volunteers was subjected to RT-PCR. Primers were designed that amplified 78- to 241-base fragments, and cDNA standards were cloned for GLUT1, GLUT2, GLUT3, GLUT4, GLUT5, GLUT6, GLUT8, GLUT9, GLUT10, GLUT11, GLUT12, and GAPDH. Seven of these eleven hexose transporters were detectable in normal human muscle. The rank order was GLUT4, GLUT5, GLUT12, GLUT8, GLUT11, GLUT3, and GLUT1, with corresponding concentrations of 404 ± 49, 131 ± 14, 33 ± 4, 5.5 ± 0.5, 4.1 ± 0.4, 1.2 ± .0.1, and 0.9 ± 0.2 copies/ng RNA (means ± SE), respectively, for the 10 subjects. Concentrations of mRNA for GLUT4, GLUT5, and GLUT12 were much higher than those for the remainder of the GLUTs and together accounted for 98% of the total GLUT isoform mRNA. Immunoblots of muscle homogenates verified that the respective proteins for GLUT4, GLUT5, and GLUT12 were present in normal human muscle. Immunofluorescent studies demonstrated that GLUT4 and GLUT12 were predominantly expressed in type I oxidative fibers; however, GLUT5 was expressed predominantly in type II (white) fibers.
APA, Harvard, Vancouver, ISO, and other styles
2

Pyla, Rajkumar, Ninu Poulose, John Y. Jun, and Lakshman Segar. "Expression of conventional and novel glucose transporters, GLUT1, -9, -10, and -12, in vascular smooth muscle cells." American Journal of Physiology-Cell Physiology 304, no. 6 (2013): C574—C589. http://dx.doi.org/10.1152/ajpcell.00275.2012.

Full text
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.
APA, Harvard, Vancouver, ISO, and other styles
3

Schaan, Beatriz D’Agord, and Ubiratan Fabres Machado. "Glucose transporters in animal models of diabetes and hypertension." American Journal of Physiology-Renal Physiology 291, no. 3 (2006): F702—F703. http://dx.doi.org/10.1152/ajprenal.00065.2006.

Full text
Abstract:
Renal tubular glucose reabsorption is mediated by facilitative glucose transporter (GLUT) proteins and energy-dependent sodium glucose luminal transporters. Glucose transport in the diabetic kidney is upregulated and has been implicated in the pathogenesis of progressive diabetic nephropathy. Hyperglycemia, hypertension, and activation of the renin-angiotensin system are believed important in the development of the disease. The present study examines the renal expression of the facilitative glucose transporters GLUT1 and GLUT12 in rat models of diabetic nephropathy. Sprague-Dawley and transgenic (mRen-2)27 rats received either streptozotocin-induced diabetes or vehicle. GLUT12 expression and localization were determined by immunohistochemistry, immunoblotting, in situ hybridization, and confocal immunofluorescence. GLUT1 immunolabeling was detected on the basolateral membrane throughout the nephron. GLUT12 was localized to the distal tubules and collecting ducts. A significant increase in GLUT12 immunolabeling was measured in Ren-2 controls and Ren-2 diabetic animals compared with Sprague-Dawley controls. GLUT12 expression was higher in Ren-2 diabetic compared with Sprague-Dawley diabetic rats. Long-term diabetes resulted in significant increases in GLUT1 levels in the renal proximal tubules and expression was higher in Ren-2 diabetic than Sprague-Dawley diabetic rats. GLUT12 protein was localized to the cytoplasm and to the apical membrane of human and rat distal tubules and collecting ducts. The apical localization of GLUT12 in the distal tubules and collecting ducts suggests that it could contribute to additional glucose reabsorption in the late nephron. Levels of both GLUT1 and GLUT12 are elevated in animal models of hypertension and diabetic nephropathy.
APA, Harvard, Vancouver, ISO, and other styles
4

Linden, Kelly C., Carrie L. DeHaan, Yuan Zhang, et al. "Renal expression and localization of the facilitative glucose transporters GLUT1 and GLUT12 in animal models of hypertension and diabetic nephropathy." American Journal of Physiology-Renal Physiology 290, no. 1 (2006): F205—F213. http://dx.doi.org/10.1152/ajprenal.00237.2004.

Full text
Abstract:
Renal tubular glucose reabsorption is mediated by facilitative glucose transporter (GLUT) proteins and energy-dependent sodium glucose luminal transporters. Glucose transport in the diabetic kidney is upregulated and has been implicated in the pathogenesis of progressive diabetic nephropathy. Hyperglycemia, hypertension, and activation of the renin-angiotensin system are believed important in the development of the disease. The present study examines the renal expression of the facilitative glucose transporters GLUT1 and GLUT12 in rat models of diabetic nephropathy. Sprague-Dawley and transgenic (mRen-2)27 rats received either streptozotocin-induced diabetes or vehicle. GLUT12 expression and localization were determined by immunohistochemistry, immunoblotting, in situ hybridization, and confocal immunofluorescence. GLUT1 immunolabeling was detected on the basolateral membrane throughout the nephron. GLUT12 was localized to the distal tubules and collecting ducts. A significant increase in GLUT12 immunolabeling was measured in Ren-2 controls and Ren-2 diabetic animals compared with Sprague-Dawley controls. GLUT12 expression was higher in Ren-2 diabetic compared with Sprague-Dawley diabetic rats. Long-term diabetes resulted in significant increases in GLUT1 levels in the renal proximal tubules and expression was higher in Ren-2 diabetic than Sprague-Dawley diabetic rats. GLUT12 protein was localized to the cytoplasm and to the apical membrane of human and rat distal tubules and collecting ducts. The apical localization of GLUT12 in the distal tubules and collecting ducts suggests that it could contribute to additional glucose reabsorption in the late nephron. Levels of both GLUT1 and GLUT12 are elevated in animal models of hypertension and diabetic nephropathy.
APA, Harvard, Vancouver, ISO, and other styles
5

Matsuo, Shunsuke, Miki Hiasa, and Hiroshi Omote. "Functional characterization and tissue localization of the facilitative glucose transporter GLUT12." Journal of Biochemistry 168, no. 6 (2020): 611–20. http://dx.doi.org/10.1093/jb/mvaa090.

Full text
Abstract:
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.
APA, Harvard, Vancouver, ISO, and other styles
6

Stuart, Charles A., Mary E. A. Howell, Yi Zhang, and Deling Yin. "Insulin-Stimulated Translocation of Glucose Transporter (GLUT) 12 Parallels That of GLUT4 in Normal Muscle." Journal of Clinical Endocrinology & Metabolism 94, no. 9 (2009): 3535–42. http://dx.doi.org/10.1210/jc.2009-0162.

Full text
Abstract:
Context: GLUT4 is the predominant glucose transporter isoform expressed in fat and muscle. In GLUT4 null mice, insulin-stimulated glucose uptake into muscle was diminished but not eliminated, suggesting that another insulin-sensitive system was present. Objective: This study was intended to determine whether insulin caused GLUT12 translocation in muscle. Design: Six normal volunteers had muscle biopsies before and after euglycemic insulin infusions. Setting: Infusions and biopsies were performed in an outpatient clinic. Participants: Subjects were nonobese, young adults with no family history of diabetes. Main Outcome Measures: GLUT12, GLUT4, and GLUT1 proteins were quantified in muscle biopsy fractions. Cultured myoblasts were used to determine whether GLUT12 translocation was phosphatidyl inositol-3 kinase (PI3-K)-dependent. Intervention. Insulin was infused at 40 mU/m2 · min for 3 h. Results: In human muscle, insulin caused a shift of a portion of GLUT12 from intracellular low-density microsomes to the plasma membrane (PM) fraction (17% in PM at baseline, 38% in PM after insulin). Insulin increased GLUT4 in PM from 13 to 42%. GLUT1 was predominantly in the PM fractions at baseline and did not change significantly after insulin. L6 myoblasts in culture also expressed and translocated GLUT12 in response to insulin, but inhibiting PI3-K prevented the translocation of GLUT12 and GLUT4. Conclusions: Insulin causes GLUT12 to translocate from an intracellular location to the plasma membrane in normal human skeletal muscle. Translocation of GLUT12 in cultured myoblasts was dependent on activation of PI3-K. GLUT12 may have evolutionarily preceded GLUT4 and now provides redundancy to the dominant GLUT4 system in muscle. Insulin induces sequestered GLUT12 to move to the muscle cell surface.
APA, Harvard, Vancouver, ISO, and other styles
7

Pujol-Giménez, Jonai, Alejandra Pérez, Alejandro M. Reyes, Donald D. F. Loo, and Maria Pilar Lostao. "Functional characterization of the human facilitative glucose transporter 12 (GLUT12) by electrophysiological methods." American Journal of Physiology-Cell Physiology 308, no. 12 (2015): C1008—C1022. http://dx.doi.org/10.1152/ajpcell.00343.2014.

Full text
Abstract:
GLUT12 is a member of the facilitative family of glucose transporters. The goal of this study was to characterize the functional properties of GLUT12, expressed in Xenopus laevis oocytes, using radiotracer and electrophysiological methods. Our results showed that GLUT12 is a facilitative sugar transporter with substrate selectivity: d-glucose ≥ α-methyl-d-glucopyranoside (α-MG) > 2-deoxy-d-glucose(2-DOG) > d-fructose = d-galactose. α-MG is a characteristic substrate of the Na+/glucose (SGLT) family and has not been shown to be a substrate of any of the GLUTs. In the absence of sugar, 22Na+ was transported through GLUT12 at a higher rate (40%) than noninjected oocytes, indicating that there is a Na+ leak through GLUT12. Genistein, an inhibitor of GLUT1, also inhibited sugar uptake by GLUT12. Glucose uptake was increased by the PKA activator 8-bromoadenosine 3′,5′-cyclic monophosphate (8-Br-cAMP) but not by the PKC activator phorbol-12-myristate-13-acetate (PMA). In high K+ concentrations, glucose uptake was blocked. Addition of glucose to the external solution induced an inward current with a reversal potential of approximately −15 mV and was blocked by Cl− channel blockers, indicating the current was carried by Cl− ions. The sugar-activated Cl− currents were unaffected by genistein. In high external K+ concentrations, sugar-activated Cl− currents were also blocked, indicating that GLUT12 activity is voltage dependent. Furthermore, glucose-induced current was increased by the PKA activator 8-Br-cAMP but not by the PKC activator PMA. These new features of GLUT12 are very different from those described for other GLUTs, indicating that GLUT12 must have a specific physiological role within glucose homeostasis, still to be discovered.
APA, Harvard, Vancouver, ISO, and other styles
8

Wood, I. Stuart, and Paul Trayhurn. "Glucose transporters (GLUT and SGLT): expanded families of sugar transport proteins." British Journal of Nutrition 89, no. 1 (2003): 3–9. http://dx.doi.org/10.1079/bjn2002763.

Full text
Abstract:
The number of known glucose transporters has expanded considerably over the past 2 years. At least three, and up to six, Na+-dependent glucose transporters (SGLT1–SGLT6; gene name SLC5A) have been identified. Similarly, thirteen members of the family of facilitative sugar transporters (GLUT1–GLUT12 and HMIT; gene name SLC2A) are now recognised. These various transporters exhibit different substrate specificities, kinetic properties and tissue expression profiles. The number of distinct gene products, together with the presence of several different transporters in certain tissues and cells (for example, GLUT1, GLUT4, GLUT5, GLUT8, GLUT12 and HMIT in white adipose tissue), indicates that glucose delivery into cells is a process of considerable complexity.
APA, Harvard, Vancouver, ISO, and other styles
9

Barrett, Montana Renae, and Michael Scott Davis. "Conditioning-induced expression of novel glucose transporters in canine skeletal muscle homogenate." PLOS ONE 18, no. 5 (2023): e0285424. http://dx.doi.org/10.1371/journal.pone.0285424.

Full text
Abstract:
Athletic conditioning can increase the capacity for insulin-stimulated skeletal muscle glucose uptake through increased sarcolemmal expression of GLUT4 and potentially additional novel glucose transporters. We used a canine model that has previously demonstrated conditioning-induced increases in basal, insulin- and contraction-stimulated glucose uptake to identify whether expression of glucose transporters other than GLUT4 was upregulated by athletic conditioning. Skeletal muscle biopsies were obtained from 12 adult Alaskan Husky racing sled dogs before and after a full season of conditioning and racing, and homogenates from those biopsies were assayed for expression of GLUT1, GLUT3, GLUT4, GLUT6, GLUT8, and GLUT12 using western blots. Athletic conditioning resulted in a 1.31 ± 0.70 fold increase in GLUT1 (p <0.0001), 1.80 ± 1.99 fold increase in GLUT4 (p = 0.005), and 2.46 ± 2.39 fold increase in GLUT12 (p = 0.002). The increased expression of GLUT1 helps explain the previous findings of conditioning-induced increases in basal glucose clearance in this model, and the increase in GLUT12 provides an alternative mechanism for insulin- and contraction-mediated glucose uptake and likely contributes to the substantial conditioning-induced increases in insulin sensitivity in highly trained athletic dogs. Furthermore, these results suggest that athletic dogs can serve as a valuable resource for the study of alternative glucose transport mechanisms in higher mammals.
APA, Harvard, Vancouver, ISO, and other styles
10

Pouliot, Frederic, Salma Meizou, Cassandra Ringuette Goulet, et al. "GLUT1 expression in high-risk prostate cancer: Correlation with 18F-FDG-PET/CT and clinical outcome." Journal of Clinical Oncology 38, no. 6_suppl (2020): 291. http://dx.doi.org/10.1200/jco.2020.38.6_suppl.291.

Full text
Abstract:
291 Background: Tumour FDG-uptake is of prognostic value in high-risk and metastatic prostate cancer (PCa). The aim of this study is to investigate the underlying glucose metabolism mechanisms of 18F-FDG-uptake on PET/CT imaging in PCa. Methods: Retrospective analysis was conducted for 94 patients diagnosed with a Gleason sum ≥8 at biopsy who underwent 18F-FDG-PET/CT imaging before radical prostatectomy. GLUT1, GLUT12 and HK2 expression were blindly scored after immunohistochemistry on radical prostatectomy specimens by 3 pathologists. 18F-FDG-uptake in primary lesion was measured by a blinded reader using maximum standardized uptake value (SUVmax). Correlations between GLUT1, GLUT12 and HK2 and SUVmax were assessed using Spearman’s rank correlation test. Survival probabilities were based on the Kaplan-Meier method. Results: With a median follow-up of 4.5 years, 56% (n=53) of patients had biochemical recurrence, 7% (n=7) progressed to castration-resistant PCa (CRPC) disease, 13% (n=12) developed metastasis and 6% (n=6) died. Correlation was found between GLUT1 expression and SUVmax level (r=0.2512, p=0.0182). In addition, SUVmax was significantly higher in tumours with high GLUT1 expression (n=17, 5.74±1.67) than tumours with low GLUT1 expression (n=71, 2.68±0.31, P=0.0037). Also, contrary to GLUT12 and HEX2 expression, a significant association was found between GLUT-1 expression levels and SUVmax index (p=0.004), lymph node status (p=0.046), volume of cancer (P=0.013), CRPC-free survival (p=0.02) and metastasis-free survival (p=0.04). Conclusions: GLUT1 expression in PCa tumours correlates with 18F-FDG-uptake and poor prognostic factors. These results suggest that this transporter is involved in the molecular mechanism of 18F-FDG-uptake in high-risk PCa and raise interest in targeting metabolic dependencies of PCa cells as a selective anticancer strategy.
APA, Harvard, Vancouver, ISO, and other styles
11

Gil-Iturbe, Eva, Elisa Félix-Soriano, Neira Sáinz, et al. "Effect of aging and obesity on GLUT12 expression in small intestine, adipose tissue, muscle, and kidney and its regulation by docosahexaenoic acid and exercise in mice." Applied Physiology, Nutrition, and Metabolism 45, no. 9 (2020): 957–67. http://dx.doi.org/10.1139/apnm-2019-0721.

Full text
Abstract:
Obesity is characterized by excessive fat accumulation and inflammation. Aging has also been characterized as an inflammatory condition, frequently accompanied by accumulation of visceral fat. Beneficial effects of exercise and n-3 long-chain polyunsaturated fatty acids in metabolic disorders have been described. Glucose transporter 12 (GLUT12) is one of the less investigated members of the GLUT family. Glucose, insulin, and tumor necrosis factor alpha (TNF-α) induce GLUT12 translocation to the membrane in muscle, adipose tissue, and intestine. We aimed to investigate GLUT12 expression in obesity and aging, and under diet supplementation with docosahexaenoic acid (DHA) alone or in combination with physical exercise in mice. Aging increased GLUT12 expression in intestine, kidney, and adipose tissue, whereas obesity reduced it. No changes on the transporter occurred in skeletal muscle. In obese 18-month-old mice, DHA further decreased GLUT12 in the 4 organs. Aerobic exercise alone did not modify GLUT12, but the changes triggered by exercise were able to prevent the DHA-diminishing effect, and almost restored GLUT12 basal levels. In conclusion, the downregulation of metabolism in aging would be a stimulus to upregulate GLUT12 expression. Contrary, obesity, an excessive energy condition, would induce GLUT12 downregulation. The combination of exercise and DHA would contribute to restore basal function of GLUT12. Novelty In small intestine, kidney and adipose tissue aging increases GLUT12 protein expression whereas obesity reduces it. Dietary DHA decreases GLUT12 in small intestine, kidney, adipose tissue and skeletal muscle. Exercise alone does not modify GLUT12 expression, nevertheless exercise prevents the DHA-diminishing effect on GLUT12.
APA, Harvard, Vancouver, ISO, and other styles
12

Wilson-O’Brien, Amy L., Carrie L. DeHaan, and Suzanne Rogers. "Mitogen-Stimulated and Rapamycin-Sensitive Glucose Transporter 12 Targeting and Functional Glucose Transport in Renal Epithelial Cells." Endocrinology 149, no. 3 (2007): 917–24. http://dx.doi.org/10.1210/en.2007-0985.

Full text
Abstract:
We hypothesized that glucose transporter 12 (GLUT12) is involved in regulation of glucose flux in distal renal tubules in response to elevated glucose. We used the Madin-Darby canine kidney polarized epithelial cell model and neutralizing antibodies to analyze GLUT12 targeting and directional GLUT12-mediated glucose transport. At physiological glucose concentrations, GLUT12 was localized to a perinuclear position. High glucose and serum treatment resulted in GLUT12 localization to the apical membrane. This mitogen-stimulated targeting of GLUT12 was inhibited by rapamycin, the specific inhibitor of mammalian target of rapamycin (mTOR). The functional role of GLUT12 was also examined. We constructed a GLUT12 cDNA containing a c-Myc epitope tag in the fifth exofacial loop. Assays of glucose transport at the apical membrane were performed using Transwell filters. By comparing transport assays in the presence of neutralizing anti-c-Myc monoclonal antibody, we specifically measured GLUT12-mediated glucose transport at the apical surface. GLUT12-mediated glucose transport was mitogen dependent and rapamycin sensitive. Our results implicate mTOR signaling in a novel pathway of glucose transporter protein targeting and glucose transport. Activity of the mTOR pathway has been associated with diabetic kidney disease. Our results provide evidence for a link between GLUT12 protein trafficking, glucose transport and signaling molecules central to the control of metabolic disease processes.
APA, Harvard, Vancouver, ISO, and other styles
13

Kyrtata, Natalia, Ben Dickie, Hedley Emsley, and Laura Parkes. "Glucose transporters in Alzheimer's disease." BJPsych Open 7, S1 (2021): S265—S266. http://dx.doi.org/10.1192/bjo.2021.707.

Full text
Abstract:
BackgroundPhysiological brain function depends on tight glucose regulation, including transport and phosphorylation, the first step in its metabolism. Impaired glucose regulation is increasingly implicated in the pathophysiology of Alzheimer's disease (AD). Glucose hypometabolism in AD may be at least partly due to impaired glucose transport at the blood-brain barrier (BBB). Glucose transporters (GLUTs) are an integral component of the BBB. There is evidence of a significant reduction in vascular and non-vascular forms of GLUT1 and GLUT3 in AD brains compared to age-matched controls. Glucose transport, as well as phosphorylation, appears to be a rate limiting step for glucose metabolism in the brain. We have reviewed the literature on glucose transport abnormalities in AD and the effect such abnormalities have on the brain.MethodPublished literature between 1st January 1946 and 1st November 2019 was identified using EMBASE and MEDLINE databases and titles and abstracts were scanned. Human studies (autopsy and imaging) and data from animal models were included while reviews, letters and cellular or molecular studies were excluded from the search.ResultAutopsy studies in AD patients show significant reductions in GLUT3 in areas of the brain closely associated with AD pathology. Patients with AD and diabetes showed greater reductions of GLUT1 and GLUT3. A longitudinal study showed significant reductions in GLUT3 levels which correlated with greater amyloid-β (Aβ) and neurofibrillary tangle pathological burden in participants with AD pathology at post-mortem but without evidence of cognitive dysfunction in their lifetime. Some studies showed increased GLUT1, with others showing reduced GLUT1, levels in AD brain. A newly recognised GLUT12 appears to be increased in AD. Animal studies showed similar results with GLUT1 and GLUT3 knockout animal models exhibiting AD pathology, while overexpression of GLUT1 or treatment with metformin decreased Aβ toxicity in a Drosophila model of AD. GLUT2 levels were increased in both human AD brain and in an animal model of AD. Imaging studies using fluorodeoxyglucose [18F]FDG with positron emission tomography (FDG-PET) in AD subjects show reductions in glucose transport and glucose metabolism in areas most affected in AD. A small randomised control trial showed anti-diabetic medications improved the glucose transport in AD subjects.ConclusionGLUTs play a significant role in AD pathology with evidence suggesting that GLUT3 reductions may precede the onset of clinical symptoms, while GLUT2 and GLUT12 may have a compensatory role. Repurposing anti-diabetic drugs shows promising results in both animal and human studies of AD.
APA, Harvard, Vancouver, ISO, and other styles
14

Chandler, Jenalle D., Elizabeth D. Williams, John L. Slavin, James D. Best, and Suzanne Rogers. "Expression and localization of GLUT1 and GLUT12 in prostate carcinoma." Cancer 97, no. 8 (2003): 2035–42. http://dx.doi.org/10.1002/cncr.11293.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

White, Mark A., Efrosini Tsouko, Chenchu Lin, et al. "GLUT12 promotes prostate cancer cell growth and is regulated by androgens and CaMKK2 signaling." Endocrine-Related Cancer 25, no. 4 (2018): 453–69. http://dx.doi.org/10.1530/erc-17-0051.

Full text
Abstract:
Despite altered metabolism being an accepted hallmark of cancer, it is still not completely understood which signaling pathways regulate these processes. Given the central role of androgen receptor (AR) signaling in prostate cancer, we hypothesized that AR could promote prostate cancer cell growth in part through increasing glucose uptake via the expression of distinct glucose transporters. Here, we determined that AR directly increased the expression ofSLC2A12, the gene that encodes the glucose transporter GLUT12. In support of these findings, gene signatures of AR activity correlated withSLC2A12expression in multiple clinical cohorts. Functionally, GLUT12 was required for maximal androgen-mediated glucose uptake and cell growth in LNCaP and VCaP cells. Knockdown of GLUT12 also decreased the growth of C4-2, 22Rv1 and AR-negative PC-3 cells. This latter observation corresponded with a significant reduction in glucose uptake, indicating that additional signaling mechanisms could augment GLUT12 function in an AR-independent manner. Interestingly, GLUT12 trafficking to the plasma membrane was modulated by calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-5′-AMP-activated protein kinase (AMPK) signaling, a pathway we previously demonstrated to be a downstream effector of AR. Inhibition of CaMKK2-AMPK signaling decreased GLUT12 translocation to the plasma membrane by inhibiting the phosphorylation of TBC1D4, a known regulator of glucose transport. Further, AR increasedTBC1D4expression. Correspondingly, expression ofTBC1D4correlated with AR activity in prostate cancer patient samples. Taken together, these data demonstrate that prostate cancer cells can increase the functional levels of GLUT12 through multiple mechanisms to promote glucose uptake and subsequent cell growth.
APA, Harvard, Vancouver, ISO, and other styles
16

Ware, Bruce, Marie Bevier, Yoshinori Nishijima, Suzanne Rogers, Cynthia A. Carnes, and Véronique A. Lacombe. "Chronic heart failure selectively induces regional heterogeneity of insulin-responsive glucose transporters." American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 301, no. 5 (2011): R1300—R1306. http://dx.doi.org/10.1152/ajpregu.00822.2010.

Full text
Abstract:
Glucose uptake across the sarcolemma is regulated by a family of membrane proteins called glucose transporters (GLUTs), which includes GLUT4 (the major cardiac isoform) and GLUT12 (a novel, second insulin-sensitive isoform). Potential regional patterns in glucose transport across the cardiac chambers have not been examined; thus, we hypothesized that insulin-responsive GLUT4 and -12 protein and gene expression would be chamber specific in healthy subjects and during chronic heart failure (HF). Using a canine model of tachypacing-induced, progressive, chronic HF, total GLUT protein and messenger RNA in both ventricles and atria (free wall and appendage) were investigated by immunoblotting and real-time PCR. In controls, GLUT4, but not GLUT12, protein content was significantly higher in the atria compared with the ventricles, with the highest content in the right atrium (RA; P < 0.001). GLUT4 and GLUT12 mRNA levels were similar across the cardiac chambers. During chronic HF, GLUT4 and GLUT12 protein content was highest in the left ventricle (LV; by 2.5- and 4.2-fold, respectively, P < 0.01), with a concomitant increase in GLUT4 and GLUT12 mRNA ( P < 0.001). GLUT4, but not GLUT12, protein content was decreased in RA during chronic HF ( P = 0.001). In conclusion, GLUT4 protein was differentially expressed across the chambers in the healthy heart, and this regional pattern was reversed during HF. Our data suggest that LV was the primary site dependent on both GLUT4 and GLUT12 during chronic HF. In addition, the paradoxical decrease in GLUT4 content in RA may induce perturbations in atrial energy production during chronic HF.
APA, Harvard, Vancouver, ISO, and other styles
17

Toyoda, Yu, Tappei Takada, Hiroshi Miyata, et al. "Identification of GLUT12/SLC2A12 as a urate transporter that regulates the blood urate level in hyperuricemia model mice." Proceedings of the National Academy of Sciences 117, no. 31 (2020): 18175–77. http://dx.doi.org/10.1073/pnas.2006958117.

Full text
Abstract:
Recent genome-wide association studies have revealed some genetic loci associated with serum uric acid levels and susceptibility to gout/hyperuricemia which contain potential candidates of physiologically important urate transporters. One of these novel loci is located upstream ofSGK1andSLC2A12, suggesting that variations in these genes increase the risks of hyperuricemia and gout. We herein focused onSLC2A12encoding a transporter, GLUT12, the physiological function of which remains unclear. As GLUT12 belongs to the same protein family as a well-recognized urate transporter GLUT9, we hypothesized that GLUT12 mediates membrane transport of urate. Therefore, we conducted functional assays and analyzedGlut12knockout hyperuricemia model mice, generated using the CRISPR-Cas9 system. Our results revealed that GLUT12 acts as a physiological urate transporter and its dysfunction elevates the blood urate concentration. This study provides insights into the deeper understanding of the urate regulatory system in the body, which is also important for pathophysiology of gout/hyperuricemia.
APA, Harvard, Vancouver, ISO, and other styles
18

Yu, Qinghua, Liqi Zhu, Jian Lin, et al. "Functional Analyse of GLUT1 and GLUT12 in Glucose Uptake in Goat Mammary Gland Epithelial Cells." PLoS ONE 8, no. 5 (2013): e65013. http://dx.doi.org/10.1371/journal.pone.0065013.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Purcell, S. H., L. B. Aerni-Flessner, A. R. Willcockson, K. A. Diggs-Andrews, S. J. Fisher, and K. H. Moley. "Improved Insulin Sensitivity by GLUT12 Overexpression in Mice." Diabetes 60, no. 5 (2011): 1478–82. http://dx.doi.org/10.2337/db11-0033.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Miller, Peter J., Kiera A. Finucane, Megan Hughes, and Feng-Qi Zhao. "Cloning and Expression of Bovine Glucose Transporter GLUT12." Mammalian Genome 16, no. 11 (2005): 873–83. http://dx.doi.org/10.1007/s00335-005-0080-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Macheda, Maria, Elizabeth Williams, James Best, Mary Wlodek, and Suzanne Rogers. "Expression and localisation of GLUT1 and GLUT12 glucose transporters in the pregnant and lactating rat mammary gland." Cell and Tissue Research 311, no. 1 (2003): 91–97. http://dx.doi.org/10.1007/s00441-002-0661-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Gaster, M., A. Handberg, A. Sch�rmann, H. G. Joost, H. Beck-Nielsen, and H. D. Schr�der. "GLUT11, but not GLUT8 or GLUT12, is expressed in human skeletal muscle in a fibre type-specific pattern." Pfl�gers Archiv European Journal of Physiology 448, no. 1 (2004): 105–13. http://dx.doi.org/10.1007/s00424-003-1219-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Rogers, Suzanne, Jenalle D. Chandler, Alison L. Clarke, Steven Petrou, and James D. Best. "Glucose transporter GLUT12-functional characterization in Xenopus laevis oocytes." Biochemical and Biophysical Research Communications 308, no. 3 (2003): 422–26. http://dx.doi.org/10.1016/s0006-291x(03)01417-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Chen, Bei, Yunfeng Wang, Manying Geng, Xi Lin, and Wenxue Tang. "Localization of Glucose Transporter 10 to Hair Cells’ Cuticular Plate in the Mouse Inner Ear." BioMed Research International 2018 (June 14, 2018): 1–7. http://dx.doi.org/10.1155/2018/7817453.

Full text
Abstract:
This study aimed to investigate the localization pattern of glucose transporters (Gluts) in mouse cochlea. Genome-wide gene expression analysis using CodeLink™ bioarrays indicated that Glut1 and Glut10 were highly expressed (~10-fold) in mouse cochlea compared with the other members of glucose transporters (Glut2-6, Glut8, and Glut9). Semiquantitative RT-PCR and western blotting confirmed that Glut10 expression in mouse cochlea was high throughout the embryogenesis and postnatal development. Immunofluorescent staining showed that Glut10 protein was localized in the cuticular plate of the outer and inner cochlear hair cells and in the ampullary crest of the vestibular system. Based on these results, it was supposed that Glut10 may contribute to glucose transport from the endolymph to the hair cells across the cuticular plate.
APA, Harvard, Vancouver, ISO, and other styles
25

Pehlivanoglu, Suray, Ozge Burcu Sahan, Sebnem Pehlivanoglu, and Kadriye Aktas Kont. "Epithelial mesenchymal transition regulator TWIST1 transcription factor stimulates glucose uptake through upregulation of GLUT1, GLUT3, and GLUT12 in vitro." In Vitro Cellular & Developmental Biology - Animal 57, no. 10 (2021): 933–43. http://dx.doi.org/10.1007/s11626-021-00635-w.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Pujol-Gimenez, Jonai, Eva Martisova, Alberto Perez-Mediavilla, María Pilar Lostao, and Maria J. Ramirez. "Expression of the Glucose Transporter GLUT12 in Alzheimer's Disease Patients." Journal of Alzheimer's Disease 42, no. 1 (2014): 97–101. http://dx.doi.org/10.3233/jad-132498.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Gude, N. M., J. L. Stevenson, S. Rogers, et al. "GLUT12 Expression in Human Placenta in First Trimester and Term." Placenta 24, no. 5 (2003): 566–70. http://dx.doi.org/10.1053/plac.2002.0925.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Gil-Iturbe, Eva, Maite Solas, Mar Cuadrado-Tejedo, et al. "GLUT12 Expression in Brain of Mouse Models of Alzheimer’s Disease." Molecular Neurobiology 57, no. 2 (2019): 798–805. http://dx.doi.org/10.1007/s12035-019-01743-1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Zhou, Y., P. L. Kaye, and M. Pantaleon. "48. Cloning and characterisation of mouse GLUT12 in preimplantation embryos." Reproduction, Fertility and Development 15, no. 9 (2003): 48. http://dx.doi.org/10.1071/srb03ab48.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

DeBosch, Brian J., Maggie Chi, and Kelle H. Moley. "Glucose Transporter 8 (GLUT8) Regulates Enterocyte Fructose Transport and Global Mammalian Fructose Utilization." Endocrinology 153, no. 9 (2012): 4181–91. http://dx.doi.org/10.1210/en.2012-1541.

Full text
Abstract:
Enterocyte fructose absorption is a tightly regulated process that precedes the deleterious effects of excess dietary fructose in mammals. Glucose transporter (GLUT)8 is a glucose/fructose transporter previously shown to be expressed in murine intestine. The in vivo function of GLUT8, however, remains unclear. Here, we demonstrate enhanced fructose-induced fructose transport in both in vitro and in vivo models of enterocyte GLUT8 deficiency. Fructose exposure stimulated [14C]-fructose uptake and decreased GLUT8 protein abundance in Caco2 colonocytes, whereas direct short hairpin RNA-mediated GLUT8 knockdown also stimulated fructose uptake. To assess GLUT8 function in vivo, we generated GLUT8-deficient (GLUT8KO) mice. GLUT8KO mice exhibited significantly greater jejunal fructose uptake at baseline and after high-fructose diet (HFrD) feeding vs. wild-type mice. Strikingly, long-term HFrD feeding in GLUT8KO mice exacerbated fructose-induced increases in blood pressure, serum insulin, low-density lipoprotein and total cholesterol vs. wild-type controls. Enhanced fructose uptake paralleled with increased abundance of the fructose and glucose transporter, GLUT12, in HFrD-fed GLUT8KO mouse enterocytes and in Caco2 cultures exposed to high-fructose medium. We conclude that GLUT8 regulates enterocyte fructose transport by regulating GLUT12, and that disrupted GLUT8 function has deleterious long-term metabolic sequelae. GLUT8 may thus represent a modifiable target in the prevention and treatment of malnutrition or the metabolic syndrome.
APA, Harvard, Vancouver, ISO, and other styles
31

Rogers, Suzanne, Susan E. Docherty, John L. Slavin, Michael A. Henderson, and James D. Best. "Differential expression of GLUT12 in breast cancer and normal breast tissue." Cancer Letters 193, no. 2 (2003): 225–33. http://dx.doi.org/10.1016/s0304-3835(03)00010-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Gude, N. M., J. L. Stevenson, P. Murthi, et al. "Expression of GLUT12 in the fetal membranes of the human placenta." Placenta 26, no. 1 (2005): 67–72. http://dx.doi.org/10.1016/j.placenta.2004.04.006.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Jiménez-Amilburu, Vanesa, Susanne Jong-Raadsen, Jeroen Bakkers, Herman P. Spaink, and Rubén Marín-Juez. "GLUT12 deficiency during early development results in heart failure and a diabetic phenotype in zebrafish." Journal of Endocrinology 224, no. 1 (2014): 1–15. http://dx.doi.org/10.1530/joe-14-0539.

Full text
Abstract:
Cardiomyopathies-associated metabolic pathologies (e.g., type 2 diabetes and insulin resistance) are a leading cause of mortality. It is known that the association between these pathologies works in both directions, for which heart failure can lead to metabolic derangements such as insulin resistance. This intricate crosstalk exemplifies the importance of a fine coordination between one of the most energy-demanding organs and an equilibrated carbohydrate metabolism. In this light, to assist in the understanding of the role of insulin-regulated glucose transporters (GLUTs) and the development of cardiomyopathies, we have developed a model forglut12deficiency in zebrafish. GLUT12 is a novel insulin-regulated GLUT expressed in the main insulin-sensitive tissues, such as cardiac muscle, skeletal muscle, and adipose tissue. In this study, we show thatglut12knockdown impacts the development of the embryonic heart resulting in abnormal valve formation. Moreover,glut12-deficient embryos also exhibited poor glycemic control. Glucose measurements showed that these larvae were hyperglycemic and resistant to insulin administration. Transcriptome analysis demonstrated that a number of genes known to be important in cardiac development and function as well as metabolic mediators were dysregulated in these larvae. These results indicate thatglut12is an essential GLUT in the heart where the reduction in glucose uptake due toglut12deficiency leads to heart failure presumably due to the lack of glucose as energy substrate. In addition, the diabetic phenotype displayed by these larvae afterglut12abrogation highlights the importance of this GLUT during early developmental stages.
APA, Harvard, Vancouver, ISO, and other styles
34

Pujol-Gimenez, Jonai, Fátima Pérez de Heredia, Miguel Angel Idoate, Rachel Airley, María Pilar Lostao, and Andrew Robert Evans. "Could GLUT12 be a Potential Therapeutic Target in Cancer Treatment? A Preliminary Report." Journal of Cancer 6, no. 2 (2015): 139–43. http://dx.doi.org/10.7150/jca.10429.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Zhou, Yuchan, Peter L. Kaye, and Marie Pantaleon. "Identification of the facilitative glucose transporter 12 gene Glut12 in mouse preimplantation embryos." Gene Expression Patterns 4, no. 6 (2004): 621–31. http://dx.doi.org/10.1016/j.modgep.2004.04.010.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Waller, Amanda P., Michael George, Anuradha Kalyanasundaram, et al. "GLUT12 functions as a basal and insulin-independent glucose transporter in the heart." Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1832, no. 1 (2013): 121–27. http://dx.doi.org/10.1016/j.bbadis.2012.09.013.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Gil-Iturbe, Eva, Rosa Castilla-Madrigal, Jaione Barrenetxe, Ana Cristina Villaro, and María Pilar Lostao. "GLUT12 expression and regulation in murine small intestine and human Caco-2 cells." Journal of Cellular Physiology 234, no. 4 (2018): 4396–408. http://dx.doi.org/10.1002/jcp.27231.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Fam, Barbara C., Laura J. Rose, Rebecca Sgambellone, Zheng Ruan, Joseph Proietto, and Sofianos Andrikopoulos. "Normal muscle glucose uptake in mice deficient in muscle GLUT4." Journal of Endocrinology 214, no. 3 (2012): 313–27. http://dx.doi.org/10.1530/joe-12-0032.

Full text
Abstract:
Skeletal muscle insulin resistance is a major characteristic underpinning type 2 diabetes. Impairments in the insulin responsiveness of the glucose transporter,Glut4 (Slc2a4), have been suggested to be a contributing factor to this disturbance. We have produced muscle-specificGlut4knockout (KO) mice using Cre/LoxP technology on a C57BL6/J background and shown undetectable levels of GLUT4 in both skeletal muscle and heart. Our aim was to determine whether complete deletion of muscle GLUT4 does in fact lead to perturbations in glucose homoeostasis. Glucose tolerance, glucose turnover and 2-deoxyglucose uptake into muscle and fat under basal and insulin-stimulated conditions were assessed in 12-week-old KO and control mice using the oral glucose tolerance test (OGTT) and hyperinsulinaemic/euglycaemic clamp respectively. KO mice weighed ∼17% less and had significantly heavier hearts compared with control mice. Basally, plasma glucose and plasma insulin were significantly lower in the KO compared with control mice, which conferred normal glucose tolerance. Despite the lack of GLUT4 in the KO mouse muscle, glucose uptake was not impaired in skeletal muscle but was reduced in heart under insulin-stimulated conditions. Neither GLUT1 nor GLUT12 protein levels were altered in the skeletal muscle or heart tissue of our KO mice. High-fat feeding did not alter glucose tolerance in the KO mice but led to elevated plasma insulin levels during the glucose tolerance test. Our study demonstrates that deletion of muscle GLUT4 does not adversely affect glucose disposal and glucose tolerance and that compensation from other transporters may contribute to this unaltered homoeostasis of glucose.
APA, Harvard, Vancouver, ISO, and other styles
39

Houbrechts, Anne M., An Beckers, Pieter Vancamp, et al. "Age-Dependent Changes in Glucose Homeostasis in Male Deiodinase Type 2 Knockout Zebrafish." Endocrinology 160, no. 11 (2019): 2759–72. http://dx.doi.org/10.1210/en.2019-00445.

Full text
Abstract:
Abstract Thyroid hormones (THs) are crucial regulators of glucose metabolism and insulin sensitivity. Moreover, inactivating mutations in type 2 deiodinase (DIO2), the major TH-activating enzyme, have been associated with type 2 diabetes mellitus in both humans and mice. We studied the link between Dio2 deficiency and glucose homeostasis in fasted males of two different Dio2 knockout (KO) zebrafish lines. Young adult Dio2KO zebrafish (6 to 9 months) were hyperglycemic. Both insulin and glucagon expression were increased, whereas β and α cell numbers in the main pancreatic islet were similar to those in wild-types. Insulin receptor expression in skeletal muscle was decreased at 6 months, accompanied by a strong downregulation of hexokinase and pyruvate kinase expression. Blood glucose levels in Dio2KO zebrafish, however, normalized around 1 year of age. Older mutants (18 to 24 months) were normoglycemic, and increased insulin and glucagon expression was accompanied by a prominent increase in pancreatic islet size and β and α cell numbers. Older Dio2KO zebrafish also showed strongly decreased expression of glucagon receptors in the gastrointestinal system as well as decreased expression of glucose transporters GLUT2 and GLUT12, glucose-6-phosphatase, and glycogen synthase 2. This study shows that Dio2KO zebrafish suffer from transient hyperglycemia, which is counteracted with increasing age by a prominent hyperplasia of the endocrine pancreas together with decreases in hepatic glucagon sensitivity and intestinal glucose uptake. Further research on the mechanisms allowing compensation in older Dio2KO zebrafish may help to identify new therapeutic targets for (TH deficiency–related) hyperglycemia.
APA, Harvard, Vancouver, ISO, and other styles
40

McMillin, Shawna L., Parker L. Evans, William M. Taylor, et al. "Muscle-Specific Ablation of Glucose Transporter 1 (GLUT1) Does Not Impair Basal or Overload-Stimulated Skeletal Muscle Glucose Uptake." Biomolecules 12, no. 12 (2022): 1734. http://dx.doi.org/10.3390/biom12121734.

Full text
Abstract:
Glucose transporter 1 (GLUT1) is believed to solely mediate basal (insulin-independent) glucose uptake in skeletal muscle; yet recent work has demonstrated that mechanical overload, a model of resistance exercise training, increases muscle GLUT1 levels. The primary objective of this study was to determine if GLUT1 is necessary for basal or overload-stimulated muscle glucose uptake. Muscle-specific GLUT1 knockout (mGLUT1KO) mice were generated and examined for changes in body weight, body composition, metabolism, systemic glucose regulation, muscle glucose transporters, and muscle [3H]-2-deoxyglucose uptake ± the GLUT1 inhibitor BAY-876. [3H]-hexose uptake ± BAY-876 was also examined in HEK293 cells-expressing GLUT1-6 or GLUT10. mGLUT1KO mice exhibited no impairments in body weight, lean mass, whole body metabolism, glucose tolerance, basal or overload-stimulated muscle glucose uptake. There was no compensation by the insulin-responsive GLUT4. In mGLUT1KO mouse muscles, overload stimulated higher expression of mechanosensitive GLUT6, but not GLUT3 or GLUT10. In control and mGLUT1KO mouse muscles, 0.05 µM BAY-876 impaired overload-stimulated, but not basal glucose uptake. In the GLUT-HEK293 cells, BAY-876 inhibited glucose uptake via GLUT1, GLUT3, GLUT4, GLUT6, and GLUT10. Collectively, these findings demonstrate that GLUT1 does not mediate basal muscle glucose uptake and suggest that a novel glucose transport mechanism mediates overload-stimulated glucose uptake.
APA, Harvard, Vancouver, ISO, and other styles
41

Maaßen, Tjorge, Siranush Vardanyan, Anton Brosig, et al. "Monosomy-3 Alters the Expression Profile of the Glucose Transporters GLUT1-3 in Uveal Melanoma." International Journal of Molecular Sciences 21, no. 24 (2020): 9345. http://dx.doi.org/10.3390/ijms21249345.

Full text
Abstract:
Monosomy-3 in uveal melanoma (UM) cells increases the risk of fatal metastases. The gene encoding the low-affinity glucose transporter GLUT2 resides on chromosome 3q26.2. Here, we analyzed the expression of the glucose transporters GLUT1, GLUT2, and GLUT3 with regard to the histological and clinical factors by performing immunohistochemistry on the primary tumors of n = 33 UM patients. UMs with monosomy-3 exhibited a 57% lower immunoreactivity for GLUT2 and a 1.8×-fold higher ratio of GLUT1 to total GLUT1-3. The combined levels of GLUT1-3 proteins were reduced in the irradiated but not the non-irradiated tumors with monosomy-3. GLUT3 expression was stronger in the irradiated samples with disomy-3 versus monosomy-3, but the ratio of the GLUT3 isoform to total GLUT1-3 did not differ with regard to the monosomy-3 status in the irradiated or non-irradiated subgroups. Systemic metastases were associated with the presence of monosomy-3 in the primary and circulating tumor cells as well as a higher GLUT1 ratio. Upregulation of the high-affinity glucose transporter GLUT1 possibly as a compensation for the low-affinity isoform GLUT2 may be enhancing the basal glucose uptake in the UM cells with monosomy-3. Prevention of hyperglycemia might, therefore, be a valuable approach to delay the lethal UM metastases.
APA, Harvard, Vancouver, ISO, and other styles
42

Aghayan, M., L. V. Rao, R. M. Smith, et al. "Developmental expression and cellular localization of glucose transporter molecules during mouse preimplantation development." Development 115, no. 1 (1992): 305–12. http://dx.doi.org/10.1242/dev.115.1.305.

Full text
Abstract:
Two general mechanisms mediate glucose transport, one is a sodium-coupled glucose transporter found in the apical border of intestinal and kidney epithelia, while the other is a sodium-independent transport system. Of the latter, several facilitated transporters have been identified, including GLUT1 (erythrocyte/brain), GLUT2 (liver) and GLUT4 (adipose/muscle) isoforms. In this study, we used Western-blot analysis and high resolution immunoelectron microscopy (IEM) to investigate the stage-related expression and cellular localization of GLUT1, 2 and 4. The Western blot results demonstrate that GLUT1 is detectable in the oocyte and throughout preimplantation development. GLUT2 isoforms were not detectable until the blastocyst stage, while the GLUT4 isoform was undetectable in the oocyte through blastocyst stages. The present findings confirm previous studies at the molecular level which demonstrated that mRNAs encoding the same GLUT isoforms are detectable at corresponding developmental stages. GLUT1 and GLUT2 display different cellular distributions at the blastocyst stage as shown by IEM studies. GLUT1 has a widespread distribution in both trophectoderm and inner cell mass cells, while GLUT2 is located on trophectoderm membranes facing the blastocyst cavity. This observation suggests a different functional significance for these isoforms during mouse preimplantation development.
APA, Harvard, Vancouver, ISO, and other styles
43

Mohani, Chandra Irwanadi, Achmad Rudijanto, Aulanni’am ., and Setyawati Soeharto. "DLBS3233 reduces inflammatory marker on kidney by increasing expression GLUT1 and GLUT2 in diabetic rats." F1000Research 11 (August 23, 2022): 976. http://dx.doi.org/10.12688/f1000research.123091.1.

Full text
Abstract:
Introduction: Diabetic kidney disease (DKD), as a diabetes mellitus type 2 (DMT2) complications, is getting more prevalent nowadays. Inflammation is one of the renal injury mechanisms evaluated through the surge in in TNF-α and NF-κβ expression. Impaired expression of gluten transporter 1 (GLUT1) and GLUT2 reduces glucose uptake. DBLS3233 is a novel anti-diabetes agent and Indonesian herbal product responsible for glucose control and upregulation of insulin signal transduction. We performed an experiment on DLBS3233 to examine the response of TNF-α and NF-κβ and the expression of GLUT 1 and GLUT2. Methods: A total of 30 adult male Wistar rats were randomly divided into six groups (n=5 per group): nondiabetic rats in the control group (group 1); untreated diabetic rats (group 2); diabetic rats treated with DLBS3233 4,5mg/kgBW (group 3); 9mg/kgBW (group 4); 18mg/kgBW (group 5), and diabetic rats treated with pioglitazone (group 6). Immunohistochemistry was performed to examine the expression of GLUT1 and GLUT2 in the pancreas and expression of TNF-α and NF-κβ in the kidney. The data was then analyzed by ANOVA. Results: In the DBLS3233 group, reduced expression of both TNF-α and NF-κβ was seen through immunohistochemistry, whereas GLUT1 and GLUT2 were intensified compared to untreated groups. From statistical analysis, we obtained significantly lower expression of TNF-α and NF-κβ, as well as enhanced GLUT1 and GLUT2 expression compared to untreated groups (p<0.05). Conclusions: DBLS3233 significantly reduces the inflammatory process and enhances the expression of GLUT1 and GLUT2 diabetic rats.
APA, Harvard, Vancouver, ISO, and other styles
44

Coudert, Edouard, Géraldine Pascal, Joëlle Dupont, et al. "Phylogenesis and Biological Characterization of a New Glucose Transporter in the Chicken (Gallus gallus), GLUT12." PLOS ONE 10, no. 10 (2015): e0139517. http://dx.doi.org/10.1371/journal.pone.0139517.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Flessner, Lauren B., and Kelle H. Moley. "Similar [DE]XXXL[LI] Motifs Differentially Target GLUT8 and GLUT12 in Chinese Hamster Ovary Cells." Traffic 10, no. 3 (2009): 324–33. http://dx.doi.org/10.1111/j.1600-0854.2008.00866.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Matsuzaka, Takashi, and Hitoshi Shimano. "GLUT12: a second insulin-responsive glucose transporters as an emerging target for type 2 diabetes." Journal of Diabetes Investigation 3, no. 2 (2011): 130–31. http://dx.doi.org/10.1111/j.2040-1124.2011.00177.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Dominguez, J. H., K. Camp, L. Maianu, and W. T. Garvey. "Glucose transporters of rat proximal tubule: differential expression and subcellular distribution." American Journal of Physiology-Renal Physiology 262, no. 5 (1992): F807—F812. http://dx.doi.org/10.1152/ajprenal.1992.262.5.f807.

Full text
Abstract:
In the late proximal tubule, glucose reabsorption progressively lowers the concentration of luminal glucose, and concentrative glucose influx increases to ensure complete glucose reabsorption. The change in glucose influx is effected by luminal Na(+)-dependent glucose transporters (Na(+)-GLUT), which exhibit higher Na(+)-to-glucose stoichiometric ratios in the late proximal tubule. In this work, the corresponding changes in the axial distribution of basolateral glucose efflux transporters (GLUTs) were examined. mRNAs encoding high-affinity facilitative basolateral transporter GLUT1, low-affinity GLUT2, and apical Na(+)-GLUT were identified in mixed populations of proximal convoluted and straight tubules. The organization of the cognate proteins was also appraised on Western blots. GLUT1 was present in glomeruli, proximal convoluted, and straight tubules, GLUT2 was only expressed in the proximal convoluted tubule, and Na(+)-GLUT was present in both proximal convoluted and straight segments. GLUT1 and GLUT2 were confined to the basolateral membrane, whereas Na(+)-GLUT was preferentially localized to the brush-border membrane. These data are consistent with the idea that glucose influx in early and late proximal tubule is achieved through Na(+)-GLUT, that GLUT1 and GLUT2 are responsible for glucose efflux in the early proximal tubule, and that in the late proximal tubule, where transcellular glucose flux is lower, only GLUT1 mediates glucose efflux.
APA, Harvard, Vancouver, ISO, and other styles
48

Pujol-Giménez, Jonai, Jaione Barrenetxe, Pedro González-Muniesa, and Maria Pilar Lostao. "The facilitative glucose transporter GLUT12: what do we know and what would we like to know?" Journal of Physiology and Biochemistry 69, no. 2 (2012): 325–33. http://dx.doi.org/10.1007/s13105-012-0213-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Macheda, Maria, Darren Kelly, James Best, and Suzanne Rogers. "Expression during rat fetal development of GLUT12 - a member of the class III hexose transporter family." Anatomy and Embryology 205, no. 5-6 (2002): 441–52. http://dx.doi.org/10.1007/s00429-002-0263-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

Macheda, Maria L., Darren J. Kelly, James D. Best, and Suzanne Rogers. "Expression during rat fetal development of GLUT12 – a member of the class III hexose transporter family." Anatomy and Embryology 206, no. 4 (2003): 335. http://dx.doi.org/10.1007/s00429-002-0303-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!