Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 78
1.
Intensive Care Med ; 50(1): 46-55, 2024 Jan.
Article En | MEDLINE | ID: mdl-37922010

PURPOSE: The purpose of this study was to profile genetic causal factors of acute respiratory distress syndrome (ARDS) and early predict patients at high ARDS risk. METHODS: We performed a phenome-wide Mendelian Randomization analysis through summary statistics of an ARDS genome-wide association study (1250 cases and 1583 controls of European ancestry) and 33,150 traits. Transcriptomic data from human blood and lung tissues of a preclinical mouse model were used to validate biomarkers, which were further used to construct a prediction model and nomogram. RESULTS: A total of 1736 traits, including 1223 blood RNA, 159 plasma proteins, and 354 non-gene phenotypes (classified by Biochemistry, Anthropometry, Disease, Nutrition and Habit, Immunology, and Treatment), exhibited a potentially causal relationship with ARDS development, which were accessible through a user-friendly interface platform called CARDS (Causal traits for Acute Respiratory Distress Syndrome). Regarding candidate blood RNA, four genes were validated, namely TMEM176B, SLC2A5, CDC45, and VSIG8, showing differential expression in blood of ARDS patients compared to controls, as well as dynamic expression in mouse lung tissues. Importantly, the addition of four blood genes and five immune cell proportions significantly improved the prediction performance of ARDS development, with 0.791 of the area under the curve from receiver-operator characteristic, compared to 0.725 for the basic model consisting of Acute Physiology and Chronic Health Evaluation (APACHE) III Score, sex, body mass index, bacteremia, and sepsis. A model-based nomogram was also developed for the clinical practice. CONCLUSION: This study identifies a wide range of ARDS relevant factors and develops a promising prediction model, enhancing early clinical management and intervention for ARDS development.


Genome-Wide Association Study , Respiratory Distress Syndrome , Humans , Animals , Mice , Genetic Profile , Biomarkers , Respiratory Distress Syndrome/etiology , RNA , Glucose Transporter Type 5/genetics
2.
Aging (Albany NY) ; 15(22): 12966-12981, 2023 11 16.
Article En | MEDLINE | ID: mdl-37980162

BACKGROUND: Prostate cancer (PC) is a common urinary system malignancy, and advanced PC patients had a poor prognosis due to recurrence or distant metastasis. Therefore, it's imperative to reveal more details in tumorigenesis and prognosis of PC patients. METHODS: The miRNA and mRNA expression profile data of 485 PC patients were obtained from The Cancer Genome Atlas database. The univariate Cox regression was applied to screen miRNAs relating to prognosis of PC. Then miRTarBase was used to predict target mRNAs of miRNAs. The hsa-mir-503/hsa-mir-1247 knockdown in 22RV1 cells was established to evaluate the effect of these two miRNAs on tumor cell migration and invasion ability. Flow cytometry was used to detect the effect of hsa-mir-503/hsa-mir-1247 knockdown on 22RV1 apoptosis rate. RESULTS: Univariate Cox regression analysis identified hsa-mir-503 as a poor and hsa-mir-1247 as a favorable prognostic marker. Totally 649 target mRNAs were screened, among which DUSP19, FGF2, and SLC2A5 had a negative correlation with hsa-mir-503, while FGF2 and VSTM4 had a positive correlation with hsa-mir-1247. In 22RV1 cells, hsa-mir-503 was up-regulated, and hsa-mir-1247 was down-regulated. hsa-mir-503 knockdown attenuated the migration and invasion of 22RV1 cells, while hsa-mir-1247 knockdown exhibited the opposite effect. In addition, hsa-mir-503 knockdown promoted 22RV1 cell apoptosis. hsa-mir-1247 overexpression significantly inhibited the tumor growth of PC in vivo. CONCLUSIONS: Herein, we demonstrated that hsa-mir-503 and hsa-mir-1247 could serve as new prognostic markers of PC, and hsa-mir-1247 had great potential to inhibit PC progression by suppressing the migration and invasion ability in vitro and in vivo.


MicroRNAs , Prostatic Neoplasms , Male , Humans , Fibroblast Growth Factor 2/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , RNA, Messenger , Gene Expression Regulation, Neoplastic , Dual-Specificity Phosphatases/genetics , Glucose Transporter Type 5/genetics
3.
Genes Chromosomes Cancer ; 62(7): 412-422, 2023 07.
Article En | MEDLINE | ID: mdl-37102302

Pediatric acute myeloid leukemia (AML) is a poor prognostic subtype of pediatric leukemia. However, the detailed characteristics of many genetic abnormalities are yet to be established in this disease. Although TP53 and RB1 are established as representative tumor suppressor genes in various cancers, alterations of these two genes, especially RB1, have not been characterized in pediatric AML. We performed next-generation sequencing in 328 pediatric AML patients from the Japanese AML-05 trial to ascertain TP53 and RB1 alterations, and their prognostic implications. We identified seven patients with TP53 alterations (2.1%) and six patients with RB1 alterations (1.8%). These alterations were found in only patients without RUNX1::RUNX1T1, CBFB::MYH11, or KMT2A rearrangements. TP53 and RB1 were frequently co-deleted with their neighboring genes PRPF8 and ELF1, respectively. Patients with TP53 alterations had significantly lower 5-year overall survival (OS; 14.3% vs. 71.4%, p < 0.001) and lower 5-year event-free survival (EFS; 0% vs. 56.3%, p < 0.001); similarly, patients with RB1 had significantly lower 5-year OS (0% vs. 71.8%, p < 0.001) and lower 5-year EFS (0% vs. 56.0%, p < 0.001) when compared to patients without these alterations. In gene expression analyses, oxidative phosphorylation, glycolysis, and protein secretion were upregulated in patients with TP53 and/or RB1 alterations. Additionally, Kaplan-Meier analysis revealed that high expressions of SLC2A5, KCNAB2, and CD300LF were related to poor OS of non-core-binding factor AML patients (p < 0.001, p = 0.001, and p = 0.021, respectively). This study will contribute to the development of risk-stratified therapy and precision medicine in pediatric AML.


Leukemia, Myeloid, Acute , Humans , Child , Mutation , Leukemia, Myeloid, Acute/pathology , Prognosis , Kaplan-Meier Estimate , Tumor Suppressor Protein p53/genetics , Glucose Transporter Type 5/genetics , Ubiquitin-Protein Ligases/genetics , Retinoblastoma Binding Proteins/genetics
4.
Br J Dermatol ; 186(6): 997-1009, 2022 06.
Article En | MEDLINE | ID: mdl-35441365

BACKGROUND: Dermatofibrosarcoma protuberans (DFSP) is a rare and marginal cutaneous sarcoma of intermediate-grade malignancy, for which the genomic landscape remains unclear. Understanding the landscape of DFSP will help to further classify the genomic pathway of malignant development in soft tissue. OBJECTIVES: To identify the comprehensive molecular pathogenesis of DFSP. METHODS: In this study, the comprehensive genomic features, with 53 tumour-normal pairs of DFSP, were revealed by whole-genome sequencing. RESULTS: The mutational signature 1 (C > T mutation at CpG dinucleotides) is featured in DFSP, resulting in higher mutations in DNA replication. Interestingly, the recurrence of DFSP is correlated with low tumour mutation burden. Novel mutation genes in DFSP were identified, including MUC4/6, KMT2C and BRCA1, and subsequently, three molecular subtypes of DFSP were classified on the basis of MUC4 and MUC6 mutations. Various structural aberrations including genomic rearrangements were identified in DSFPs, particularly in 17q and 22q, which cause oncogene amplification (AKT1, SPHK1, COL1A1, PDGFß) or tumour suppressor deletion (CDKN2A/B). In addition to gene fusion of COL1A1-PDGFß [t(17;22)], we identified gene fusion of SLC2A5-BTBD7 [t(1;14)] in DFSP through whole-genome sequencing, and verified it experimentally. Enrichment analysis of altered molecules revealed that DNA repair, cell cycle, phosphoinositide 3-kinase and Janus kinase pathways were primarily involved in DFSP. CONCLUSIONS: This is the first large-scale whole-genome sequencing for DFSP, and our findings describe the comprehensive genomic landscape, highlighting the molecular complexity and genomic aberrations of DFSP. Our findings also provide novel potential diagnostic and therapeutic targets for this disease. What is already known about this topic? Chromosomal translocation between chromosome 17 and chromosome 22 is the main feature in the pathogenesis of dermatofibrosarcoma protuberans (DFSP). What does this study add? We describe the comprehensive genomic landscape of DFSP, highlighting the molecular complexity and genomic aberrations. Our findings provide novel potential diagnostic and therapeutic targets for this disease. What is the translational message? Our study revealed novel molecular subtypes of DFSP based on genetic mutations, which benefits precision diagnosis. We also found oncogene amplification, including AKT1 and SPHK1, which provides novel potential target molecules for further DFSP treatment. In addition to gene fusion of COL1A1-PDGFß, we identified a novel gene fusion of SLC2A5-BTBD7 in DFSP, which is a novel potential diagnostic and therapeutic target for this disease.


Dermatofibrosarcoma , Skin Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Dermatofibrosarcoma/genetics , Dermatofibrosarcoma/pathology , Gene Rearrangement , Genomics , Glucose Transporter Type 5/genetics , Humans , Oncogene Proteins, Fusion/genetics , Phosphatidylinositol 3-Kinases/metabolism , Skin Neoplasms/pathology
5.
BMC Gastroenterol ; 22(1): 167, 2022 Apr 06.
Article En | MEDLINE | ID: mdl-35387598

BACKGROUND: While role of ALDOB-related gene variants for hereditary fructose intolerance is well established, contribution of gene variants for acquired fructose malabsorption (e.g. SLC2A5, GLUT5) is not well understood. METHODS: Patients referred to fructose breath test were further selected to identify those having acquired fructose malabsorption. Molecular analysis of genomic DNA included (I) exclusion of 3 main ALDOB gene variants causing hereditary fructose intolerance and (II) sequencing analysis of SLC2A5 gene comprising complete coding region, at least 20 bp of adjacent intronic regions and 700 bp of proximal promoter. RESULTS: Among 494 patients, 35 individuals with acquired fructose malabsorption were identified based on pathological fructose-breath test and normal lactose-breath test. Thirty four of them (97%) had negative tissue anti-transglutaminase and/or deamidated gliadin antibodies in their medical records. Molecular analysis of SLC2A5 gene of all 35 subjects identified 5 frequent and 5 singular gene variants mostly in noncoding regions (promoter and intron). Allele frequencies of gene variants were similar to those reported in public databases strongly implying that none of them was associated with acquired fructose malabsorption. CONCLUSIONS: Gene variants of coding exons, adjacent intronic regions and proximal promoter region of SLC2A5 gene are unlikely to contribute to genetic predisposition of acquired fructose malabsorption.


Fructose Intolerance , Breath Tests , Exons , Fructose , Fructose Intolerance/diagnosis , Fructose Intolerance/genetics , Glucose Transporter Type 5/genetics , Humans , Promoter Regions, Genetic
6.
Sci Rep ; 12(1): 1429, 2022 01 26.
Article En | MEDLINE | ID: mdl-35082341

The passive transport of glucose and related hexoses in human cells is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT3 is a high-affinity glucose transporter primarily responsible for glucose entry in neurons. Changes in its expression have been implicated in neurodegenerative diseases and cancer. GLUT3 inhibitors can provide new ways to probe the pathophysiological role of GLUT3 and tackle GLUT3-dependent cancers. Through in silico screening of an ~ 8 million compounds library against the inward- and outward-facing models of GLUT3, we selected ~ 200 ligand candidates. These were tested for in vivo inhibition of GLUT3 expressed in hexose transporter-deficient yeast cells, resulting in six new GLUT3 inhibitors. Examining their specificity for GLUT1-5 revealed that the most potent GLUT3 inhibitor (G3iA, IC50 ~ 7 µM) was most selective for GLUT3, inhibiting less strongly only GLUT2 (IC50 ~ 29 µM). None of the GLUT3 inhibitors affected GLUT5, three inhibited GLUT1 with equal or twofold lower potency, and four showed comparable or two- to fivefold better inhibition of GLUT4. G3iD was a pan-Class 1 GLUT inhibitor with the highest preference for GLUT4 (IC50 ~ 3.9 µM). Given the prevalence of GLUT1 and GLUT3 overexpression in many cancers and multiple myeloma's reliance on GLUT4, these GLUT3 inhibitors may discriminately hinder glucose entry into various cancer cells, promising novel therapeutic avenues in oncology.


Drug Discovery , Glucose Transporter Type 3/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Binding Sites , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/antagonists & inhibitors , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 5/antagonists & inhibitors , Glucose Transporter Type 5/chemistry , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry
7.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article En | MEDLINE | ID: mdl-34884473

Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body's metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) is a fructose transporter primarily expressed in microglia within the central nervous system. However, little is known about the nutritional regulation of Slc2a5 expression in microglia and its role in the regulation of metabolism. The present study aimed to address the hypothesis that nutrients affect microglial activity by altering the expression of glucose transporter genes. Murine microglial cell line SIM-A9 cells and primary microglia from mouse brain were exposed to different concentrations of glucose and levels of microglial activation markers and glucose transporter genes were measured. High concentration of glucose increased levels of the immediate-early gene product c-Fos, a marker of cell activation, Slc2a5 mRNA, and pro-inflammatory cytokine genes in microglial cells in a time-dependent manner, while fructose failed to cause these changes. Glucose-induced changes in pro-inflammatory gene expression were partially attenuated in SIM-A9 cells treated with the GLUT5 inhibitor. These findings suggest that an increase in local glucose availability leads to the activation of microglia by controlling their carbohydrate sensing mechanism through both GLUT5-dependent and -independent mechanisms.


Glucose Transporter Type 5/genetics , Glucose/pharmacology , Microglia/cytology , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Mice , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins c-fos/genetics
8.
Gut Microbes ; 13(1): 1993582, 2021.
Article En | MEDLINE | ID: mdl-34793284

Many chronic diseases are associated with decreased abundance of the gut commensal Faecalibacterium prausnitzii. This strict anaerobe can grow on dietary fibers, e.g., prebiotics, and produce high levels of butyrate, often associated to epithelial metabolism and health. However, little is known about other F. prausnitzii metabolites that may affect the colonic epithelium. Here, we analyzed prebiotic cross-feeding between F. prausnitzii and intestinal epithelial (Caco-2) cells in a "Human-oxygen Bacteria-anaerobic" coculture system. Inulin-grown F. prausnitzii enhanced Caco-2 viability and suppressed inflammation- and oxidative stress-marker expression. Inulin-grown F. prausnitzii produced excess butyrate and fructose, but only fructose efficiently promoted Caco-2 growth. Finally, fecal microbial taxonomy analysis (16S sequencing) from healthy volunteers (n = 255) showed the strongest positive correlation for F. prausnitzii abundance and stool fructose levels. We show that fructose, produced and accumulated in a fiber-rich colonic environment, supports colonic epithelium growth, while butyrate does not.


Faecalibacterium prausnitzii/metabolism , Fructose/metabolism , Intestinal Mucosa/metabolism , Inulin/metabolism , Anaerobiosis , Butyrates/analysis , Butyrates/metabolism , Caco-2 Cells , Cell Proliferation , Cell Survival , Coculture Techniques , Feces/chemistry , Feces/microbiology , Fructose/analysis , Gastrointestinal Microbiome , Glucose/analysis , Glucose/metabolism , Glucose Transporter Type 5/genetics , Humans , Inflammation/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Pectins/metabolism , Prebiotics
9.
J Immunol Res ; 2021: 9938397, 2021.
Article En | MEDLINE | ID: mdl-34604392

Lung adenocarcinoma (LUAD) is a major subtype of lung cancer with a relatively poor prognosis, requiring novel therapeutic approaches. Great advances in new immunotherapy strategies have shown encouraging results in lung cancer patients. This study is aimed at elucidating the function of SLC2A5 in the prognosis and pathogenesis of LUAD by analyzing public databases. The differential expression of SLC2A5 in various tissues from Oncomine, GEPIA, and other databases was obtained, and SLC2A5 expression at the protein level in normal and tumor tissues was detected with the use of the HPA database. Then, we used the UALCAN database to analyze the expression of SLC2A5 in different clinical feature subgroups. Notably, in both PrognoScan and Kaplan-Meier plotter databases, we found a certain association between SLC2A5 and poor OS outcomes in LUAD patients. Studies based on the TIMER database show a strong correlation between SLC2A5 expression and various immune cell infiltrates and markers. The data analysis in the UALCAN database showed that the decreased promoter methylation level of SLC2A5 in LUAD may lead to the high expression of SLC2A5. Finally, we used the LinkedOmics database to evaluate the SLC2A5-related coexpression and functional networks in LUAD and to investigate their role in tumor immunity. These findings suggest that SLC2A5 correlated with immune infiltration can be used as a candidate diagnostic and prognostic biomarker in LUAD patients.


Adenocarcinoma of Lung/genetics , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 5/genetics , Immune System/metabolism , Lung Neoplasms/genetics , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Glucose Transporter Type 5/metabolism , Humans , Immune System/cytology , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Young Adult
10.
Sci Rep ; 11(1): 13751, 2021 07 02.
Article En | MEDLINE | ID: mdl-34215797

Glucose is an essential energy source for cells. In humans, its passive diffusion through the cell membrane is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT2 transports both glucose and fructose with low affinity and plays a critical role in glucose sensing mechanisms. Alterations in the function or expression of GLUT2 are involved in the Fanconi-Bickel syndrome, diabetes, and cancer. Distinguishing GLUT2 transport in tissues where other GLUTs coexist is challenging due to the low affinity of GLUT2 for glucose and fructose and the scarcity of GLUT-specific modulators. By combining in silico ligand screening of an inward-facing conformation model of GLUT2 and glucose uptake assays in a hexose transporter-deficient yeast strain, in which the GLUT1-5 can be expressed individually, we identified eleven new GLUT2 inhibitors (IC50 ranging from 0.61 to 19.3 µM). Among them, nine were GLUT2-selective, one inhibited GLUT1-4 (pan-Class I GLUT inhibitor), and another inhibited GLUT5 only. All these inhibitors dock to the substrate cavity periphery, close to the large cytosolic loop connecting the two transporter halves, outside the substrate-binding site. The GLUT2 inhibitors described here have various applications; GLUT2-specific inhibitors can serve as tools to examine the pathophysiological role of GLUT2 relative to other GLUTs, the pan-Class I GLUT inhibitor can block glucose entry in cancer cells, and the GLUT2/GLUT5 inhibitor can reduce the intestinal absorption of fructose to combat the harmful effects of a high-fructose diet.


Drug Discovery , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 5/antagonists & inhibitors , Small Molecule Libraries/chemistry , Computer Simulation , Diabetes Mellitus/drug therapy , Fanconi Syndrome/drug therapy , Glucose/genetics , Glucose/metabolism , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/ultrastructure , Glucose Transporter Type 5/chemistry , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/ultrastructure , Humans , Ligands , Neoplasms/drug therapy , Protein Conformation/drug effects , User-Computer Interface
11.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G232-G242, 2021 08 01.
Article En | MEDLINE | ID: mdl-34133236

The Western diet has been suggested to contribute to the rising incidence of inflammatory bowel diseases. This has led to the hypothesis that fructose, a component of the Western diet, could play a role in the pathogenesis of inflammatory bowel diseases. A high-fructose diet is known to exacerbate experimental colitis. This study tested whether the expression of GLUT5, the fructose transporter, is a determinant of the severity of experimental colitis during elevated fructose consumption and whether ileal inflammation is associated with altered GLUT5 expression in Crohn's disease. Studies in genetically engineered mice showed that in comparison to Glut5+/+ mice, feeding a 15 kcal% fructose diet to Glut5-/- mice led to worse dextran sodium sulfate (DSS)-induced colitis. This effect was associated with elevated levels of colonic fructose and a shift in the fecal microbiota in Glut5-/- mice. Importantly, treatment with broad-spectrum antibiotics protected against the worsening of colitis mediated by dietary fructose in Glut5-/- mice. Gene expression analysis revealed that GLUT5 levels are reduced in the intestines of patients with ileal Crohn's disease. Moreover, levels of GLUT5 negatively correlated with expression of proinflammatory mediators in these samples. Collectively, these results demonstrate that dietary constituent (fructose)-host gene (GLUT5) interactions can shape the colonic microbiota, thereby impacting the severity of colitis.NEW & NOTEWORTHY This study provides the first evidence that reduced levels of GLUT5, the fructose transporter, worsen experimental colitis upon fructose feeding, an effect mediated by changes in the gut microbiota. Moreover, GLUT5 expression is reduced in Crohn's ileitis. Overall, these findings demonstrate the importance of interactions between dietary fructose and host GLUT5 as determinants of both the composition of colonic microbiota and severity of experimental colitis.


Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Fructose/metabolism , Glucose Transporter Type 5/metabolism , Animals , Colitis, Ulcerative/etiology , Dietary Sugars/adverse effects , Dietary Sugars/metabolism , Fructose/adverse effects , Gastrointestinal Microbiome , Glucose Transporter Type 5/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Sodium Dodecyl Sulfate/toxicity
12.
Br J Cancer ; 125(5): 734-747, 2021 08.
Article En | MEDLINE | ID: mdl-34188196

BACKGROUND: SLC2A5 is a high-affinity fructose transporter, which is frequently upregulated in multiple human malignant tumours. However, the function and molecular mechanism of SLC2A5 in colorectal cancer (CRC) remain unknown. METHODS: We detected the expression levels of SLC2A5 in CRC tissues and CRC cell lines by western blotting, qRT-PCR and immunohistochemistry. CRC cell lines with stable overexpression or knockdown of SLC2A5 were constructed to evaluate the functional roles of SLC2A5 in vitro through conventional assays. An intrasplenic inoculation model was established in mice to investigate the effect of SLC2A5 in promoting metastasis in vivo. Methylation mass spectrometry sequencing, methylation specific PCR, bisulphite sequencing PCR, ChIP-qPCR and luciferase reporter assay were performed to investigate the molecular mechanism underlying transcriptional activation of SLC2A5. RESULTS: We found that SLC2A5 was upregulated in colorectal tumour tissues. Functionally, a high level of SLC2A5 expression was associated with increased invasion and metastasis capacities of CRC cells both in vitro and in vivo. Mechanistically, we unveiled that S100P could integrate to a specific region of SLC2A5 promoter, thereby reducing its methylation levels and activating SLC2A5 transcription. CONCLUSIONS: Our results reveal a novel mechanism that S100P mediates the promoter demethylation and transcription activation of SLC2A5, thereby promoting the metastasis of CRC.


Calcium-Binding Proteins/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Neoplasm Proteins/metabolism , Up-Regulation , Animals , Caco-2 Cells , Case-Control Studies , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Promoter Regions, Genetic , Transcriptional Activation
13.
Sci Rep ; 11(1): 10885, 2021 05 25.
Article En | MEDLINE | ID: mdl-34035344

The relationship between cerebral glucose metabolism and glucose transporter expression after intracerebral hemorrhage (ICH) is unclear. Few studies have used positron emission tomography (PET) to explore cerebral glucose metabolism after ICH in rodents. In this study, we produced ICH in mice with an intrastriatal injection of collagenase to investigate whether glucose metabolic changes in 18F-fluoro-2-deoxy-D-glucose (FDG)-PET images are associated with expression of glucose transporters (GLUTs) over time. On days 1 and 3 after ICH, the ipsilateral striatum exhibited significant hypometabolism. However, by days 7 and 14, glucose metabolism was significantly higher in the ipsilateral striatum than in the contralateral striatum. The contralateral hemisphere did not show hypermetabolism at any time after ICH. Qualitative immunofluorescence and Western blotting indicated that the expression of GLUT1 in ipsilateral striatum decreased on days 1 and 3 after ICH and gradually returned to baseline by day 21. The 18F-FDG uptake after ICH was associated with expression of GLUT1 but not GLUT3 or GLUT5. Our data suggest that ipsilateral cerebral glucose metabolism decreases in the early stage after ICH and increases progressively in the late stage. Changes in 18F-FDG uptake on PET imaging are associated with the expression of GLUT1 in the ipsilateral striatum.


Cerebral Hemorrhage/diagnostic imaging , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 5/metabolism , Glucose/metabolism , Animals , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Collagenases/adverse effects , Corpus Striatum/metabolism , Disease Models, Animal , Fluorodeoxyglucose F18/administration & dosage , Gene Expression Regulation , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/genetics , Glucose Transporter Type 5/genetics , Male , Mice , Positron Emission Tomography Computed Tomography
14.
Cancer Res ; 81(11): 2824-2832, 2021 06 01.
Article En | MEDLINE | ID: mdl-33762358

Clinical localization of primary tumors and sites of metastasis by PET is based on the enhanced cellular uptake of 2-deoxy-2-[18F]-fluoro-D-glucose (FDG). In prostate cancer, however, PET-FDG imaging has shown limited clinical applicability, suggesting that prostate cancer cells may utilize hexoses other than glucose, such as fructose, as the preferred energy source. Our previous studies suggested that prostate cancer cells overexpress fructose transporters, but not glucose transporters, compared with benign cells. Here, we focused on validating the functional expression of fructose transporters and determining whether fructose can modulate the biology of prostate cancer cells in vitro and in vivo. Fructose transporters, Glut5 and Glut9, were significantly upregulated in clinical specimens of prostate cancer when compared with their benign counterparts. Fructose levels in the serum of patients with prostate cancer were significantly higher than healthy subjects. Functional expression of fructose transporters was confirmed in prostate cancer cell lines. A detailed kinetic characterization indicated that Glut5 represents the main functional contributor in mediating fructose transport in prostate cancer cells. Fructose stimulated proliferation and invasion of prostate cancer cells in vitro. In addition, dietary fructose increased the growth of prostate cancer cell line-derived xenograft tumors and promoted prostate cancer cell proliferation in patient-derived xenografts. Gene set enrichment analysis confirmed that fructose stimulation enriched for proliferation-related pathways in prostate cancer cells. These results demonstrate that fructose promotes prostate cancer cell growth and aggressiveness in vitro and in vivo and may represent an alternative energy source for prostate cancer cells. SIGNIFICANCE: This study identifies increased expression of fructose transporters in prostate cancer and demonstrates a role for fructose as a key metabolic substrate supporting prostate cancer cells, revealing potential therapeutic targets and biomarkers.


Biomarkers, Tumor/metabolism , Diet/adverse effects , Fructose/pharmacology , Gene Expression Regulation, Neoplastic , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 5/metabolism , Prostatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 5/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Insect Mol Biol ; 30(1): 1-8, 2021 02.
Article En | MEDLINE | ID: mdl-32920918

Hypoxic stress is linked to various cardiovascular disorders (e.g., stroke, myocardial infarction), mediated, at least in part, by a reduction in ATP synthesis. Fructose-driven glycolysis is proposed as an alternative pathway capable of sustaining ATP production even under anoxic conditions. Here, we tested the hypothesis that facilitating fructose-driven metabolism exerts a protective effect against anoxic stress in Drosophila. Genetically modified flies with the human fructose transporter (GluT5) and ketohexokinase (KHK) genes downstream of upstream activating sequence (UAS) were constructed. The GAL4-UAS system was confirmed to: (i) increase the expression of GluT5 and KHK in a tissue-specific and a time-dependent manner (i.e., whole flies [with Act5c-gene switch GAL4 driver], neurons [with elav-gene switch GAL4 driver]) and (ii) reduce mortality of flies when placed under anoxic stress. Taken together, these data suggest that increasing fructose metabolism may be a clinically relevant approach to minimize hypoxia-induced cellular damage.


Drosophila , Fructose/metabolism , Hypoxia , Animals , Drosophila/genetics , Drosophila/metabolism , Fructokinases/genetics , Glucose Transporter Type 5/genetics , Glycolysis , Humans , Hypoxia/metabolism , Hypoxia/prevention & control
16.
J Photochem Photobiol B ; 211: 111995, 2020 Oct.
Article En | MEDLINE | ID: mdl-32836050

A most crucial feature of biological adaptation is the maintenance of a close temporal relationship of behaviour and physiology with prevailing 24-h light-dark environment, which is rapidly changing with increasing nighttime illumination. This study investigated developmental effects of the loss of night on circadian behaviour, metabolism and gene expressions in diurnal zebra finches born and raised under LL, with controls on 12L:12D. Birds under LD were entrained, and showed normal body mass and a significant 24-h rhythm in both activity-rest pattern and mRNA expression of candidate genes that we measured. But, under LL, birds gained weight and accumulated lipid in the liver. Intriguingly, at the end of the experiment, the majority (4/5th) of birds under LL were rhythmic in activity despite arrhythmic expression in the hypothalamus of c-Fos (neuronal activity), Rhodopsin and Mel1-a genes (light perception), and clock genes (Bmal1, Per2 and Rev-erb ß). In peripheral tissues, LL induced variable clock gene expressions. Whereas 24-h mRNA rhythm was abolished for Bmal1 in both liver and gut, it persisted for Per2 and Rev-erb ß in liver, and for Per2 in gut. Further, we found under LL, the loss of 24-h rhythm in hepatic expression of Fasn and Cd36/Fat (biosynthesis and its uptake), and gut expression of Sglt1, Glut5, Cd36 and Pept1 (nutrient absorption) genes. As compared to LD, baseline mRNA levels of Fasn and Cd36 genes were attenuated under LL. Among major transporter genes, Sglt1 (glucose) and Cd36 (fat) genes were arrhythmic, while Glut5 (glucose) and Pept1 (protein) genes were rhythmic but with phase differences under LL, compared to LD. These results demonstrate dissociation of circadian behaviour from clock gene rhythms, and provide molecular insights into possible mechanisms at different levels (behaviour and physiology) that diurnal animals might employ in order to adapt to an emerging overly illuminated-night urban environment.


Circadian Clocks/physiology , Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Hypothalamus/physiology , Metabolism/physiology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Finches , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Light , Liver , Peptide Transporter 1/genetics , Peptide Transporter 1/metabolism , Photoperiod , RNA, Messenger/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Stomach
17.
Technol Cancer Res Treat ; 19: 1533033819894308, 2020.
Article En | MEDLINE | ID: mdl-31918632

AIMS: The solute carrier family 2 (SLC2) genes are comprised of 14 members which are essential for the maintenance of glucose uptake and survival of tumour cells. This study was performed to investigate the associations of SLC2 family gene expression with mortality in acute myeloid leukemia (AML). METHODS: Clinical features and SLC2 family gene expression data were obtained from The Cancer Genome Atlas and Gene Expression Omnibus database. The associations between SLC2 family gene expression and clinicopathologic features were analyzed using linear regression model. Kaplan-Meier survival, univariate, multivariate survival analyses and validation analysis were performed to analyze the associations between SLC2 family gene expression and patients' overall survival. RESULTS: Patient mortality was positively associated with age and cytogenetic risk in AML patients. Kaplan-Meier survival analysis suggested that patients with high SLC2A5 and SLC2A10 expression showed poorer survival than those with low SLC2A5 and SLC2A10 expression. In contrast, patients with high SLC2A13 expression exhibited better prognosis than those with low SLC2A13 expression (P < 0.05 for all cases, log rank test). Multivariate survival analysis and validation analysis confirmed that high expression of SLC2A5 and SLC2A10 and low expression of SLC2A13 were associated with increased mortality (P = 0.00, Odd ratio [OR]:4.05, 95% Confidence Interval [CI]: 1.73-10.22; P = 0.00, OR: 3.66, 95% CI: 1.54-9.25; and P = 0.01, OR: 0.26, 95% CI: 0.09-0.68, respectively). CONCLUSION: SLC family gene expression, such as SLC2A5, SLC2A10 and SLC2A13, was significantly associated with prognosis of AML patients, their expression levels might become useful prognostic biomarkers in AML.


Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 5/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Databases, Genetic/statistics & numerical data , Female , Gene Expression Profiling , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 5/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Prognosis , Survival Rate
18.
Vet Med Sci ; 5(3): 451-461, 2019 08.
Article En | MEDLINE | ID: mdl-30973212

The ban on the use of antibiotic in feed encouraged nutritionists to using alternatives to maintain growth performance and intestinal function of broilers. This study was conducted to evaluate the effects of Yupingfeng polysaccharides (YP) supplementation on growth performance and expression of SGLT1, GLUT2 and GLUT5 in Qingyuan partridge chicken. Experiment 1: a total of 540 chickens were randomly allocated to five groups with six replication. Dietary treatments were: (1) CON (control group), basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP; (4) T3, CON + 2 g kg-1 YP; (5) T4, CON + 4 g kg-1 YP. Experiment 2, a total of 162 were randomly allocated to three groups with three replication. Dietary treatments were: (1) CON, basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP. From days 1 to 14 and overall, chicken fed T1 diet had higher ADG. On day 42, there was increased villus height of jejunum in T1 group. On days 14 and 28, there was decreased villus height of duodenum and jejunum in T2 group. In duodenum, the expression of SGLT1 (days 21, 35 and 42), GLUT2 (days 7, 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21 and 28) was increased with YP supplementation. In jejunum, the expression of SGLT1 (days 7, 14, 21, 28 and 35), GLUT2 (days 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. In ileum, the expression of SGLT1 (days 7, 21, 35 and 42), GLUT2 (days 7, 14, 21 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. Dietary YP supplementation improves growth performance and expression of SGLT1, GLUT2 and GLUT5 in intestine.


Chickens/growth & development , Dietary Supplements/analysis , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Developmental/drug effects , Polysaccharides/pharmacology , Animal Feed/analysis , Animals , Chickens/anatomy & histology , Chickens/genetics , Diet/veterinary , Gene Expression Regulation, Developmental/genetics , Glucose Transporter Type 2/drug effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 5/drug effects , Glucose Transporter Type 5/genetics , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/drug effects , Intestine, Small/anatomy & histology , Intestine, Small/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , Random Allocation , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/genetics , Up-Regulation
19.
J Am Coll Nutr ; 38(8): 670-680, 2019.
Article En | MEDLINE | ID: mdl-31008696

Objectives: Dysfunctional metabolism of carbohydrates is a fundamental component of many dietary-related disorders. It has been hypothesized that plant extracts containing high levels of antioxidants may have the ability to stabilize carbohydrate regulation. The aim of this study was to assess the effects of a polyphenol-rich sugarcane extract on cellular pathways related to carbohydrate metabolism.Methods: We evaluated the antioxidant activity of a polyphenol-rich sugarcane extract obtained by a patented hydrophobic extract process and its therapeutic potential to regulate carbohydrate metabolism and protect against metabolic disorders such as type 2 diabetes.Results: Quantitative analytical studies support that the polyphenol-rich sugarcane extract has a high concentration of polyphenols and antioxidant compounds. The follow-up cellular studies via Caco-2 cells and dysfunctional ß-cell models suggested that the polyphenol-rich sugarcane extract may help deter glucose and fructose uptake in intestinal cells and restore insulin production in dysfunctional ß-cells-key functions in managing diabetic conditions.Conclusions: These findings suggest that sugarcane polyphenols may modulate cellular mechanism in a manner that is beneficial to health.


Antioxidants/pharmacology , Hypoglycemic Agents/pharmacology , Plant Extracts/chemistry , Polyphenols/pharmacology , Saccharum/chemistry , Animals , Antioxidants/chemistry , Cell Line , Chromatography, Liquid , Cricetinae , Gene Expression Regulation/drug effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Humans , Hypoglycemic Agents/chemistry , Insulin , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mass Spectrometry , Polyphenols/chemistry
20.
Food Chem ; 277: 604-608, 2019 Mar 30.
Article En | MEDLINE | ID: mdl-30502192

d-Allulose has been reported to have beneficial health effects. However, the transport system(s) mediating intestinal d-allulose transport has not yet been clearly identified. The aim of this study was to investigate whether intestinal d-allulose transport is mediated by glucose transporter type 5 (GLUT5). When d-allulose alone was gavaged, plasma d-allulose levels were dramatically higher in rats previously fed fructose. This suggests enhanced intestinal d-allulose absorption paralleled increases in GLUT5 expression observed only in fructose-fed rats. When d-allulose was gavaged with d-fructose, previously observed increases in plasma d-allulose levels were dampened and delayed, indicating d-fructose inhibited transepithelial d-allulose transport into plasma. Tracer D-[14C]-fructose uptake rate was reduced to 54.8% in 50 mM d-allulose and to 16.4% in 50 mM d-fructose, suggesting d-allulose competed with D-[14C]-fructose and the affinity of d-allulose for GLUT5 was lower than that of d-fructose. GLUT5 clearly mediates, likely at lower affinity relative to d-fructose, intestinal d-allulose transport.


Fructose/metabolism , Glucose Transporter Type 5/metabolism , Intestine, Small/enzymology , Animals , Biological Transport , Blood Glucose , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/metabolism , Fructose/blood , Glucose/analysis , Glucose/metabolism , Glucose Transporter Type 5/genetics , Male , Rats , Rats, Sprague-Dawley , Substrate Specificity
...