Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.426
Filter
1.
BMC Cardiovasc Disord ; 24(1): 349, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987688

ABSTRACT

PURPOSE: Glycolysis and immune metabolism play important roles in acute myocardial infarction (AMI). Therefore, this study aimed to identify and experimentally validate the glycolysis-related hub genes in AMI as diagnostic biomarkers, and further explore the association between hub genes and immune infiltration. METHODS: Differentially expressed genes (DEGs) from AMI peripheral blood mononuclear cells (PBMCs) were analyzed using R software. Glycolysis-related DEGs (GRDEGs) were identified and analyzed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) for functional enrichment. A protein-protein interaction network was constructed using the STRING database and visualized using Cytoscape software. Immune infiltration analysis between patients with AMI and stable coronary artery disease (SCAD) controls was performed using CIBERSORT, and correlation analysis between GRDEGs and immune cell infiltration was performed. We also plotted nomograms and receiver operating characteristic (ROC) curves to assess the predictive accuracy of GRDEGs for AMI occurrence. Finally, key genes were experimentally validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting using PBMCs. RESULTS: A total of 132 GRDEGs and 56 GRDEGs were identified on the first day and 4-6 days after AMI, respectively. Enrichment analysis indicated that these GRDEGs were mainly clustered in the glycolysis/gluconeogenesis and metabolic pathways. Five hub genes (HK2, PFKL, PKM, G6PD, and ALDOA) were selected using the cytoHubba plugin. The link between immune cells and hub genes indicated that HK2, PFKL, PKM, and ALDOA were significantly positively correlated with monocytes and neutrophils, whereas G6PD was significantly positively correlated with neutrophils. The calibration curve, decision curve analysis, and ROC curves indicated that the five hub GRDEGs exhibited high predictive value for AMI. Furthermore, the five hub GRDEGs were validated by RT-qPCR and western blotting. CONCLUSION: We concluded that HK2, PFKL, PKM, G6PD, and ALDOA are hub GRDEGs in AMI and play important roles in AMI progression. This study provides a novel potential immunotherapeutic method for the treatment of AMI.


Subject(s)
Computational Biology , Gene Regulatory Networks , Glycolysis , Myocardial Infarction , Protein Interaction Maps , Humans , Glycolysis/genetics , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/diagnosis , Gene Expression Profiling , Databases, Genetic , Transcriptome , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Predictive Value of Tests , Male , Middle Aged , Hexokinase/genetics , Female , Case-Control Studies , Nomograms , Reproducibility of Results
2.
Cells ; 13(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38995012

ABSTRACT

Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are aggressive sarcomas that can arise both sporadically and in patients with the genetic syndrome Neurofibromatosis type 1 (NF1). Prognosis is dismal, as large dimensions, risk of relapse, and anatomical localization make surgery poorly effective, and no therapy is known. Hence, the identification of MPNST molecular features that could be hit in an efficient and selective way is mandatory to envision treatment options. Here, we find that MPNSTs express high levels of the glycolytic enzyme Hexokinase 2 (HK2), which is known to shield cancer cells from noxious stimuli when it localizes at MAMs (mitochondria-associated membranes), contact sites between mitochondria and endoplasmic reticulum. A HK2-targeting peptide that dislodges HK2 from MAMs rapidly induces a massive death of MPNST cells. After identifying different matrix metalloproteases (MMPs) expressed in the MPNST microenvironment, we have designed HK2-targeting peptide variants that harbor cleavage sites for these MMPs, making such peptides activatable in the proximity of cancer cells. We find that the peptide carrying the MMP2/9 cleavage site is the most effective, both in inhibiting the in vitro tumorigenicity of MPNST cells and in hampering their growth in mice. Our data indicate that detaching HK2 from MAMs could pave the way for a novel anti-MPNST therapeutic strategy, which could be flexibly adapted to the protease expression features of the tumor microenvironment.


Subject(s)
Hexokinase , Peptides , Hexokinase/metabolism , Hexokinase/genetics , Humans , Animals , Cell Line, Tumor , Peptides/metabolism , Peptides/pharmacology , Peptides/chemistry , Mice , Nerve Sheath Neoplasms/pathology , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Matrix Metalloproteinase 2/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Tumor Microenvironment
3.
Cell Death Dis ; 15(7): 502, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003255

ABSTRACT

Dysfunction of the ubiquitin-proteasome system (UPS) is involved in the pathogenesis of various malignancies including colorectal cancer (CRC). Ubiquitin domain containing 1 (UBTD1), a ubiquitin-like protein, regulates UPS-mediated protein degradation and tumor progression in some cancer types. However, the biological function and mechanism of UBTD1 are far from being well elucidated, and its role in CRC has not been explored yet. In our study, we analyzed CRC patients' clinical information and UBTD1 expression data, and found that the expression of UBTD1 in cancer tissue was significantly higher than that in adjacent normal tissue. Higher UBTD1 expression was significantly associated with poorer survival and more lymph node metastasis. Overexpression of UBTD1 could facilitate, while knockdown could inhibit CRC cell proliferation and migration, respectively. RNA-seq and proteomics indicated that c-Myc is an important downstream target of UBTD1. Metabolomics showed the products of the glycolysis pathway were significantly increased in UBTD1 overexpression cells. In vitro, we verified UBTD1 upregulating c-Myc protein and promoting CRC cell proliferation and migration via regulating c-Myc. UBTD1 promoted CRC cells' glycolysis, evidenced by the increased lactate production and glucose uptake following UBTD1 overexpression. Mechanistically, UBTD1 prolonged the half-life of the c-Myc protein by binding to E3 ligase ß-transducin repeat-containing protein (ß-TrCP), thereby upregulated the expression of glycolysis rate-limiting enzyme hexokinase II (HK2), and enhanced glycolysis and promoted CRC progression. In conclusion, our study revealed that UBTD1 promotes CRC progression by upregulating glycolysis via the ß-TrCP/c-Myc/HK2 pathway, suggesting its potential as a prognostic biomarker and therapeutic target in CRC.


Subject(s)
Cell Proliferation , Colorectal Neoplasms , Disease Progression , Glycolysis , Proto-Oncogene Proteins c-myc , Up-Regulation , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Ubiquitins/metabolism , Ubiquitins/genetics , Male , Female , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement , Middle Aged , Mice, Nude , Animals , Hexokinase/metabolism , Hexokinase/genetics , Mice , Protein Stability
4.
Theranostics ; 14(9): 3509-3525, 2024.
Article in English | MEDLINE | ID: mdl-38948065

ABSTRACT

Rationale: Current treatments for ocular angiogenesis primarily focus on blocking the activity of vascular endothelial growth factor (VEGF), but unfavorable side effects and unsatisfactory efficacy remain issues. The identification of novel targets for anti-angiogenic treatment is still needed. Methods: We investigated the role of tsRNA-1599 in ocular angiogenesis using endothelial cells, a streptozotocin (STZ)-induced diabetic model, a laser-induced choroidal neovascularization model, and an oxygen-induced retinopathy model. CCK-8 assays, EdU assays, transwell assays, and matrigel assays were performed to assess the role of tsRNA-1599 in endothelial cells. Retinal digestion assays, Isolectin B4 (IB4) staining, and choroidal sprouting assays were conducted to evaluate the role of tsRNA-1599 in ocular angiogenesis. Transcriptomic analysis, metabolic analysis, RNA pull-down assays, and mass spectrometry were utilized to elucidate the mechanism underlying angiogenic effects mediated by tsRNA-1599. Results: tsRNA-1599 expression was up-regulated in experimental ocular angiogenesis models and endothelial cells in response to angiogenic stress. Silencing of tsRNA-1599 suppressed angiogenic effects in endothelial cells in vitro and inhibited pathological ocular angiogenesis in vivo. Mechanistically, tsRNA-1599 exhibited little effect on VEGF signaling but could cause reduced glycolysis and NAD+/NADH production in endothelial cells by regulating the expression of HK2 gene through interacting with YBX1, thus affecting endothelial effects. Conclusions: Targeting glycolytic reprogramming of endothelial cells by a tRNA-derived small RNA represents an exploitable therapeutic approach for ocular neovascular diseases.


Subject(s)
Choroidal Neovascularization , Endothelial Cells , Glycolysis , Animals , Glycolysis/drug effects , Mice , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Humans , Y-Box-Binding Protein 1/metabolism , Y-Box-Binding Protein 1/genetics , Angiogenesis Inhibitors/pharmacology , Hexokinase/metabolism , Hexokinase/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL , Male , Disease Models, Animal , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/genetics , Human Umbilical Vein Endothelial Cells , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism
5.
Nat Commun ; 15(1): 6046, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025848

ABSTRACT

Energy status and nutrients regulate photosynthetic protein expression. The unicellular green alga Chromochloris zofingiensis switches off photosynthesis in the presence of exogenous glucose (+Glc) in a process that depends on hexokinase (HXK1). Here, we show that this response requires that cells lack sufficient iron (-Fe). Cells grown in -Fe+Glc accumulate triacylglycerol (TAG) while losing photosynthesis and thylakoid membranes. However, cells with an iron supplement (+Fe+Glc) maintain photosynthesis and thylakoids while still accumulating TAG. Proteomic analysis shows that known photosynthetic proteins are most depleted in heterotrophy, alongside hundreds of uncharacterized, conserved proteins. Photosynthesis repression is associated with enzyme and transporter regulation that redirects iron resources to (a) respiratory instead of photosynthetic complexes and (b) a ferredoxin-dependent desaturase pathway supporting TAG accumulation rather than thylakoid lipid synthesis. Combining insights from diverse organisms from green algae to vascular plants, we show how iron and trophic constraints on metabolism aid gene discovery for photosynthesis and biofuel production.


Subject(s)
Chlorophyta , Glucose , Iron , Lipid Metabolism , Photosynthesis , Triglycerides , Iron/metabolism , Glucose/metabolism , Triglycerides/metabolism , Chlorophyta/metabolism , Chlorophyta/genetics , Thylakoids/metabolism , Proteomics , Hexokinase/metabolism , Hexokinase/genetics , Chlorophyceae/metabolism , Chlorophyceae/genetics
6.
Nat Commun ; 15(1): 5410, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926365

ABSTRACT

METTL3 is the catalytic subunit of the methyltransferase complex, which mediates m6A modification to regulate gene expression. In addition, METTL3 regulates transcription in an enzymatic activity-independent manner by driving changes in high-order chromatin structure. However, how these functions of the methyltransferase complex are coordinated remains unknown. Here we show that the methyltransferase complex coordinates its enzymatic activity-dependent and independent functions to regulate cellular senescence, a state of stable cell growth arrest. Specifically, METTL3-mediated chromatin loops induce Hexokinase 2 expression through the three-dimensional chromatin organization during senescence. Elevated Hexokinase 2 expression subsequently promotes liquid-liquid phase separation, manifesting as stress granule phase separation, by driving metabolic reprogramming. This correlates with an impairment of translation of cell-cycle related mRNAs harboring polymethylated m6A sites. In summary, our results report a coordination of m6A-dependent and -independent function of the methyltransferase complex in regulating senescence through phase separation driven by metabolic reprogramming.


Subject(s)
Cellular Senescence , Chromatin , Methyltransferases , Stress Granules , Methyltransferases/metabolism , Methyltransferases/genetics , Chromatin/metabolism , Humans , Stress Granules/metabolism , Stress Granules/genetics , Hexokinase/metabolism , Hexokinase/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Adenosine/metabolism , Adenosine/analogs & derivatives , HEK293 Cells , Metabolic Reprogramming , Phase Separation
7.
Signal Transduct Target Ther ; 9(1): 158, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862461

ABSTRACT

Cell membrane-camouflaged nanoparticles possess inherent advantages derived from their membrane structure and surface antigens, including prolonged circulation in the bloodstream, specific cell recognition and targeting capabilities, and potential for immunotherapy. Herein, we introduce a cell membrane biomimetic nanodrug platform termed MPB-3BP@CM NPs. Comprising microporous Prussian blue nanoparticles (MPB NPs) serving as both a photothermal sensitizer and carrier for 3-bromopyruvate (3BP), these nanoparticles are cloaked in a genetically programmable cell membrane displaying variants of signal regulatory protein α (SIRPα) with enhanced affinity to CD47. As a result, MPB-3BP@CM NPs inherit the characteristics of the original cell membrane, exhibiting an extended circulation time in the bloodstream and effectively targeting CD47 on the cytomembrane of colorectal cancer (CRC) cells. Notably, blocking CD47 with MPB-3BP@CM NPs enhances the phagocytosis of CRC cells by macrophages. Additionally, 3BP, an inhibitor of hexokinase II (HK2), suppresses glycolysis, leading to a reduction in adenosine triphosphate (ATP) levels and lactate production. Besides, it promotes the polarization of tumor-associated macrophages (TAMs) towards an anti-tumor M1 phenotype. Furthermore, integration with MPB NPs-mediated photothermal therapy (PTT) enhances the therapeutic efficacy against tumors. These advantages make MPB-3BP@CM NPs an attractive platform for the future development of innovative therapeutic approaches for CRC. Concurrently, it introduces a universal approach for engineering disease-tailored cell membranes for tumor therapy.


Subject(s)
CD47 Antigen , Cell Membrane , Colorectal Neoplasms , Nanoparticles , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Nanoparticles/chemistry , Humans , CD47 Antigen/genetics , Mice , Cell Membrane/metabolism , Cell Membrane/genetics , Animals , Pyruvates/chemistry , Pyruvates/pharmacology , Hexokinase/genetics , Cell Line, Tumor , Macrophages/metabolism , Macrophages/drug effects , Ferrocyanides
8.
Am J Chin Med ; 52(3): 865-884, 2024.
Article in English | MEDLINE | ID: mdl-38790085

ABSTRACT

Ovarian cancer is a common, highly lethal tumor. Herein, we reported that S-phase kinase-associated protein 2 (Skp2) is essential for the growth and aerobic glycolysis of ovarian cancer cells. Skp2 was upregulated in ovarian cancer tissues and associated with poor clinical outcomes. Using a customized natural product library screening, we found that xanthohumol inhibited aerobic glycolysis and cell viability of ovarian cancer cells. Xanthohumol facilitated the interaction between E3 ligase Cdh1 and Skp2 and promoted the Ub-K48-linked polyubiquitination of Skp2 and degradation. Cdh1 depletion reversed xanthohumol-induced Skp2 downregulation, enhancing HK2 expression and glycolysis in ovarian cancer cells. Finally, a xenograft tumor model was employed to examine the antitumor efficacy of xanthohumol in vivo. Collectively, we discovered that xanthohumol promotes the binding between Skp2 and Cdh1 to suppress the Skp2/AKT/HK2 signal pathway and exhibits potential antitumor activity for ovarian cancer cells.


Subject(s)
Flavonoids , Glycolysis , Ovarian Neoplasms , Propiophenones , S-Phase Kinase-Associated Proteins , Ubiquitination , Propiophenones/pharmacology , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , Flavonoids/pharmacology , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Glycolysis/drug effects , Animals , Signal Transduction/drug effects , Cadherins/metabolism , Carcinogenesis/drug effects , Antigens, CD/metabolism , Hexokinase/metabolism , Cell Line, Tumor , Proto-Oncogene Proteins c-akt/metabolism , Mice , Phytotherapy , Mice, Nude , Antineoplastic Agents, Phytogenic/pharmacology
9.
Diabetes Res Clin Pract ; 212: 111721, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821414

ABSTRACT

AIMS: Autoantibodies against hexokinase 1 (HK1) were recently proposed to be associated with diabetic macular edema (DME). We hypothesized that anti-HK1 autoantibodies can be used as DME markers and to predict DME onset. MATERIALS AND METHODS: Serum from patients with 1) DME, 2) diabetes mellitus (DM), 3) allergies or autoimmunities, and 4) control subjects was tested for anti-HK1 and anti-hexokinase 2 (HK2) autoantibodies by immunoblotting. Patients with DM were prospectively followed for up to nine years, and the association of anti-HK1 antibodies with new-onset DME was evaluated. The vitreous humor was also tested for autoantibodies. RESULTS: Among patients with DME, 32 % were positive for anti-HK1 autoantibodies (42 % of those with underlying type 1 DM and 31 % of those with underlying type 2 DM), and 12 % were positive for anti-HK2 autoantibodies, with only partial overlap of these two groups of patients. Anti-HK1 positive were also 7 % of patients with DM, 6 % of patients with allergies and autoimmunities, and 3 % of control subjects. The latter three groups were anti-HK2 negative. Only one of seven patients with DM who were initially anti-HK1 positive developed DME. CONCLUSIONS: Anti-HK1 autoantibodies can be used as DME markers but fail to predict DME onset.


Subject(s)
Autoantibodies , Diabetic Retinopathy , Hexokinase , Macular Edema , Humans , Hexokinase/immunology , Autoantibodies/blood , Autoantibodies/immunology , Diabetic Retinopathy/immunology , Diabetic Retinopathy/blood , Male , Female , Middle Aged , Macular Edema/immunology , Macular Edema/blood , Aged , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/blood , Prospective Studies , Adult , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/blood , Biomarkers/blood
10.
Chem Biol Interact ; 397: 111062, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38763349

ABSTRACT

Acute lung injury is the leading cause of paraquat (PQ) poisoning-related mortality. The mechanism by which macrophages are involved in PQ-induced acute lung injury remains unclear. In recent years, the role of metabolic reprogramming in macrophage functional transformation has received significant attention. The current study aimed to identify the role of altered macrophage glucose metabolism and molecular mechanisms in PQ poisoning-induced acute lung injury. We established a model of acute lung injury in PQ-intoxicated mice via the intraperitoneal injection of PQ. PQ exposure induces macrophage M1 polarization and promotes the release of inflammatory factors, which causes the development of acute lung injury in mice. In vitro analysis revealed that PQ altered glucose metabolism, which could be reversed by siRNA transfection to silence the expression of HK1, a key enzyme in glucose metabolism. RNA sequencing revealed that the ERK/MAPK pathway was the crucial molecular mechanism of PQ pathogenesis. Further, U0126, an ERK inhibitor, could inhibit PQ-induced HK1 activation and macrophage M1 polarization. These findings provide novel insights into the previously unrecognized mechanism of ERK/MAPK-HK1 activation in PQ poisoning.


Subject(s)
Acute Lung Injury , Glucose , Hexokinase , MAP Kinase Signaling System , Macrophages , Mice, Inbred C57BL , Paraquat , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Paraquat/toxicity , Mice , Glucose/metabolism , Macrophages/metabolism , Macrophages/drug effects , Hexokinase/metabolism , MAP Kinase Signaling System/drug effects , Male , Signal Transduction/drug effects , RAW 264.7 Cells
11.
Cytokine ; 180: 156663, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38815522

ABSTRACT

BACKGROUND: Glioma represents the predominant malignant brain tumor. This investigation endeavors to elucidate the impact and prospective mechanisms of glycolysis-related lncARSR on glioma. METHODS: LncARSR level was assessed in normal glial cells and glioma cells. Cell proliferation, migration, and invasion measurements were conducted through CCK-8, wound healing, and transwell assay. Flow cytometry was utilized to measure cell apoptosis and cell cycle. Biochemical assay kits and immunoblotting were employed to measure the content of glycolysis-related indicators and protein expression, respectively. We analyzed the impact of both lncARSR knockdown and overexpression of the Signal Transducer and Activator of Transcription 3 (STAT3) on Hexokinase 2 (HK2) using dual luciferase reporter assays and Chromatin Immunoprecipitation (ChIP) experiments. Further assessment of the impact of lncARSR on glioma progression was conducted through animal experiments. RESULTS: LncARSR was expressed at elevated levels in glioma cells compared to normal glial cells. Overexpressing lncARSR enhanced proliferation, migration, invasion, and G2/M phase arrest in glioma cells and also increased glucose, lactate, ATP production, as well as the expression of HK2, PFK1, PKM2, GLUT1, and LDHA. STAT3 binding to the HK2 gene promoter was weakened following the knockdown of lncARSR. Upregulation of STAT3 reversed the suppressed functions of knocking down lncARSR on cell proliferation, migration, invasion, G2/M phase arrest, and glycolysis and counteracted its promotional effect on cell apoptosis. In vivo, knocking down lncARSR inhibits glioma growth and ki67 and PCNA expression. CONCLUSION: LncARSR promotes the development of glioma by enhancing glycolysis through the STAT3-HK2 axis.


Subject(s)
Cell Movement , Cell Proliferation , Glioma , Glycolysis , Hexokinase , RNA, Long Noncoding , STAT3 Transcription Factor , STAT3 Transcription Factor/metabolism , Glioma/metabolism , Glioma/pathology , Glioma/genetics , Hexokinase/metabolism , Hexokinase/genetics , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Animals , Cell Movement/genetics , Mice , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Apoptosis , Signal Transduction
12.
Ecotoxicol Environ Saf ; 279: 116500, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38795416

ABSTRACT

Hexavalent chromium [Cr(VI)] is one of the most common environmental contaminants due to its tremendous industrial applications, but its effects and mechanism remain to be investigated. Our previous studies showed that Cr(VI) exposure caused malignant transformation and tumorigenesis. This study showed that glycolytic proteins HK2 and LDHA levels were statistically significant changed in blood samples of Cr(VI)-exposed workers and in Cr-T cells compared to the control subjects and parental cells. HK2 and LDHA knockdown inhibited cell proliferation and angiogenesis, and higher HK2 and LDHA expression levels are associated with advanced stages and poor prognosis of lung cancer. We found that miR-218 levels were significantly decreased and miR-218 directly targeted HK2 and LDHA for inhibiting their expression. Overexpression of miR-218 inhibited glucose consumption and lactate production in Cr-T cells. Further study found that miR-218 inhibited tumor growth and angiogenesis by decreasing HK2 and LDHA expression in vivo. MiR-218 levels were negatively correlated with HK2 and LDHA expression levels and cancer development in human lung and other cancers. These results demonstrated that miR-218/HK2/LDHA pathway is vital for regulating Cr(VI)-induced carcinogenesis and human cancer development.


Subject(s)
Carcinogenesis , Chromium , Hexokinase , Lung Neoplasms , MicroRNAs , Up-Regulation , MicroRNAs/genetics , Humans , Chromium/toxicity , Hexokinase/genetics , Hexokinase/metabolism , Carcinogenesis/chemically induced , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Prognosis , Animals , Cell Proliferation/drug effects , L-Lactate Dehydrogenase/metabolism , Occupational Exposure/adverse effects , Mice , Isoenzymes
13.
PLoS One ; 19(5): e0303060, 2024.
Article in English | MEDLINE | ID: mdl-38723008

ABSTRACT

In the current study we investigated the impact of combination of rutin and vitamin A on glycated products, the glyoxalase system, oxidative markers, and inflammation in animals fed a high-fat high-fructose (HFFD) diet. Thirty rats were randomly divided into six groups (n = 5). The treatments, metformin (120 mg/kg), rutin (100 mg/kg), vitamin A (43 IU/kg), and a combination of rutin (100 mg/kg) and vitamin A (43 IU/kg) were given to relevant groups of rats along with high-fructose high-fat diet for 42 days. HbA1c, D-lactate, Glyoxylase-1, Hexokinase 2, malondialdehyde (MDA), glutathione peroxidase (GPx), catalase (CAT), nuclear transcription factor-B (NF-κB), interleukin-6 (IL-6), interleukin-8 (IL-8) and histological examinations were performed after 42 days. The docking simulations were conducted using Auto Dock package. The combined effects of rutin and vitamin A in treated rats significantly (p < 0.001) reduced HbA1c, hexokinase 2, and D-lactate levels while preventing cellular damage. The combination dramatically (p < 0.001) decreased MDA, CAT, and GPx in treated rats and decreased the expression of inflammatory cytokines such as IL-6 andIL-8, as well as the transcription factor NF-κB. The molecular docking investigations revealed that rutin had a strong affinity for several important biomolecules, including as NF-κB, Catalase, MDA, IL-6, hexokinase 2, and GPx. The results propose beneficial impact of rutin and vitamin A as a convincing treatment strategy to treat AGE-related disorders, such as diabetes, autism, alzheimer's, atherosclerosis.


Subject(s)
Diet, High-Fat , Fructose , Hyperglycemia , Inflammation , Oxidative Stress , Rutin , Vitamin A , Animals , Rutin/pharmacology , Oxidative Stress/drug effects , Fructose/adverse effects , Rats , Diet, High-Fat/adverse effects , Vitamin A/pharmacology , Vitamin A/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Male , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Molecular Docking Simulation , Rats, Wistar , Disease Models, Animal , Glycosylation/drug effects , Metformin/pharmacology , Glycated Hemoglobin/metabolism , NF-kappa B/metabolism , Hexokinase/metabolism , Catalase/metabolism
14.
J Transl Med ; 22(1): 460, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750462

ABSTRACT

BACKGROUND: Chaperonin Containing TCP1 Subunit 6 A (CCT6A) is a prominent protein involved in the folding and stabilization of newly synthesized proteins. However, its roles and underlying mechanisms in lung adenocarcinoma (LUAD), one of the most aggressive cancers, remain elusive. METHODS: Our study utilized in vitro cell phenotype experiments to assess CCT6A's impact on the proliferation and invasion capabilities of LUAD cell lines. To delve into CCT6A's intrinsic mechanisms affecting glycolysis and proliferation in lung adenocarcinoma, we employed transcriptomic sequencing and liquid chromatography-mass spectrometry analysis. Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (CHIP) assays were also conducted to substantiate the mechanism. RESULTS: CCT6A was found to be significantly overexpressed in LUAD and associated with a poorer prognosis. The silencing of CCT6A inhibited the proliferation and migration of LUAD cells and elevated apoptosis rates. Mechanistically, CCT6A interacted with STAT1 protein, forming a complex that enhances the stability of STAT1 by protecting it from ubiquitin-mediated degradation. This, in turn, facilitated the transcription of hexokinase 2 (HK2), a critical enzyme in aerobic glycolysis, thereby stimulating LUAD's aerobic glycolysis and progression. CONCLUSION: Our findings reveal that the CCT6A/STAT1/HK2 axis orchestrated a reprogramming of glucose metabolism and thus promoted LUAD progression. These insights position CCT6A as a promising candidate for therapeutic intervention in LUAD treatment.


Subject(s)
Adenocarcinoma of Lung , Cell Proliferation , Chaperonin Containing TCP-1 , Disease Progression , Glycolysis , Hexokinase , Lung Neoplasms , STAT1 Transcription Factor , Humans , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Hexokinase/metabolism , STAT1 Transcription Factor/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Chaperonin Containing TCP-1/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Apoptosis , Signal Transduction , Neoplasm Invasiveness
15.
Sci Rep ; 14(1): 12143, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802677

ABSTRACT

Microglia are natural immune cells in the central nervous system, and the activation of microglia is accompanied by a reprogramming of glucose metabolism. In our study, we investigated the role of long non-coding RNA taurine-upregulated gene 1 (TUG1) in regulating microglial glucose metabolism reprogramming and activation. BV2 cells were treated with Lipopolysaccharides (LPS)/Interferon-γ (IFN-γ) to establish a microglial activation model. The glycolysis inhibitor 2-Deoxy-D-glucose (2-DG) was used as a control. The expression levels of TUG1 mRNA and proinflammatory cytokines such as Interleukin-1ß (IL-1ß), Interleukin -6, and Tumor Necrosis Factor-α mRNA and anti-inflammatory cytokines such as IL-4, Arginase 1(Arg1), CD206, and Ym1 were detected by RT-qPCR. TUG1 was silenced using TUG1 siRNA and knocked out using CRISPR/Cas9. The mRNA and protein expression levels of key enzymes involved in glucose metabolism, such as Hexokinase2, Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), Lactate dehydrogenase, Glucose 6 phosphate dehydrogenase, and Pyruvate dehydrogenase (PDH), were determined by RT-qPCR and Western blotting. The glycolytic rate of microglial cells was measured using Seahorse. Differential metabolites were determined by metabolomics, and pathway enrichment was performed using these differential metabolites. Our findings revealed that the expression of TUG1 was elevated in proinflammatory-activated microglia and positively correlated with the levels of inflammatory factors. The expression of anti-inflammatory cytokines such as IL-4, Arg1, CD206, and Ym1 were decreased when induced with LPS/IFN-γ. However, this decrease was reversed by the treatment with 2-DG. Silencing of GAPDH led to an increase in the expression of TUG1 and inflammatory factors. TUG1 knockout (TUG1KO) inhibited the expression of glycolytic key enzymes and promoted the expression of oxidative phosphorylation key enzymes, shifting the metabolic profile of activated microglia from glycolysis to oxidative phosphorylation. Additionally, TUG1KO reduced the accumulation of metabolites, facilitating the restoration of the tricarboxylic acid cycle and enhancing oxidative phosphorylation in microglia. Furthermore, the downregulation of TUG1 was found to reduce the expression of both proinflammatory and anti-inflammatory cytokines under normal conditions. Interestingly, when induced with LPS/IFN-γ, TUG1 downregulation showed a potentially beneficial effect on microglia in terms of inflammation. Downregulation of TUG1 expression inhibits glycolysis and facilitates the shift of microglial glucose metabolism from glycolysis to oxidative phosphorylation, promoting their transformation towards an anti-inflammatory phenotype and exerting anti-inflammatory effects in BV2.


Subject(s)
Glucose , Glycolysis , Lipopolysaccharides , Microglia , RNA, Long Noncoding , Microglia/metabolism , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Glucose/metabolism , Mice , Lipopolysaccharides/pharmacology , Cytokines/metabolism , Inflammation/metabolism , Inflammation/genetics , Interferon-gamma/metabolism , beta-N-Acetylhexosaminidases/metabolism , beta-N-Acetylhexosaminidases/genetics , Cell Line , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mannose-Binding Lectins/genetics , Deoxyglucose/pharmacology , Interleukin-4/metabolism , Interleukin-1beta/metabolism , Metabolic Reprogramming , Arginase , Hexokinase , Lectins
16.
Cancer Immunol Immunother ; 73(7): 122, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714539

ABSTRACT

Neuroblastoma (NB) is the most common and deadliest extracranial solid tumor in children. Targeting tumor-associated macrophages (TAMs) is a strategy for attenuating tumor-promoting states. The crosstalk between cancer cells and TAMs plays a pivotal role in mediating tumor progression in NB. The overexpression of Hexokinase-3 (HK3), a pivotal enzyme in glucose metabolism, has been associated with poor prognosis in NB patients. Furthermore, it correlates with the infiltration of M2-like macrophages within NB tumors, indicating its significant involvement in tumor progression. Therefore, HK3 not only directly regulates the malignant biological behaviors of tumor cells, such as proliferation, migration, and invasion, but also recruits and polarizes M2-like macrophages through the PI3K/AKT-CXCL14 axis in neuroblastoma. The secretion of lactate and histone lactylation alterations within tumor cells accompanies this interaction. Additionally, elevated expression of HK3 in M2-TAMs was found at the same time. Modulating HK3 within M2-TAMs alters the biological behavior of tumor cells, as demonstrated by our in vitro studies. This study highlights the pivotal role of HK3 in the progression of NB malignancy and its intricate regulatory network with M2-TAMs. It establishes HK3 as a promising dual-functional biomarker and therapeutic target in combating neuroblastoma.


Subject(s)
Hexokinase , Neuroblastoma , Tumor-Associated Macrophages , Neuroblastoma/metabolism , Neuroblastoma/pathology , Humans , Hexokinase/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cell Proliferation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Cell Movement , Chemokines, CXC/metabolism , Animals , Tumor Microenvironment/immunology
17.
Cell Signal ; 119: 111183, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636768

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide, with Hepatitis B virus (HBV) infection being the leading cause. This study aims to investigate the role of HBV in HCC pathogenesis involving glucose metabolism. Long non-coding RNA (lncRNA) OIP5-AS1 was significantly downregulated in HBV-positive HCC patients, and its low expression indicated a poor prognosis. This lncRNA was primarily localized in the cytoplasm, acting as a tumor suppressor. HBV protein X (HBx) repressed OIP5-AS1 expression by inhibiting a ligand-activated transcriptional factor peroxisome proliferator-activated receptor α (PPARα). Furthermore, mechanistic studies revealed that OIP5-AS1 inhibited tumor growth by suppressing Hexokinase domain component 1 (HKDC1)-mediated glycolysis. The expression of HKDC1 could be enhanced by transcriptional factor sterol regulatory element-binding protein 1 (SREBP1). OIP5-AS1 facilitated the ubiquitination and degradation of SREBP1 to suppress HKDC1 transcription, which inhibited glycolysis. The results suggest that lncRNA OIP5-AS1 plays an anti-oncogenic role in HBV-positive HCC via the HBx/OIP5-AS1/HKDC1 axis, providing a promising diagnostic marker and therapeutic target for HBV-positive HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Gene Expression Regulation, Neoplastic , Glycolysis , Hexokinase , Liver Neoplasms , RNA, Long Noncoding , Trans-Activators , Viral Regulatory and Accessory Proteins , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Glycolysis/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Hexokinase/metabolism , Hexokinase/genetics , Animals , Hepatitis B virus , Male , Cell Line, Tumor , Down-Regulation , Mice , Mice, Nude , Female , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Mice, Inbred BALB C , PPAR alpha/metabolism , PPAR alpha/genetics
18.
Acta Biomater ; 181: 308-316, 2024 06.
Article in English | MEDLINE | ID: mdl-38570107

ABSTRACT

Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.


Subject(s)
Acute Lung Injury , CRISPR-Cas Systems , Hexokinase , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Animals , Mice , Hexokinase/genetics , Hexokinase/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/drug effects , Administration, Inhalation , Liposomes/chemistry , RAW 264.7 Cells , Male , Cellular Reprogramming/drug effects , Gene Editing , Glycolysis/drug effects
19.
Metab Brain Dis ; 39(5): 719-729, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38687460

ABSTRACT

BACKGROUND: Glioma is the main subtype of primary central nervous system (CNS) tumor with high malignancy and poor prognosis under current therapeutic approaches. Glycolysis and suppressive tumor microenvironment (TME) are key markers of glioma with great importance for aggressive features of glioma and inferior clinical outcomes. Hexokinase 3 (HK3) is an important rate-limiting enzyme in glycolysis, but its function in glioma remains unknown. METHODS: This study comprehensively assessed the expression distribution and immunological effect of HK3 via pan-cancer analysis based on datasets from Genotype Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), and The Cancer Genome Atlas (TCGA). Furthermore, it explored the malignant phenotype and genomic landscape between low-HK3 and high-HK3 expression groups in gliomas from Chinese Glioma Genome Atlas (CGGA) and TCGA. Moreover, data from the TIMER website predicted the relationship between macrophage infiltration and HK3 expression. Also, single-cell sequencing data were used to validate the relationship. RESULTS: For pan-cancer patients, HK3 was expressed in various cancers. The results showed that HK3 was highly expressed in gliomas and positively correlated with tumor-infiltrating immune cells (TIICs), immune checkpoints, immunomodulators, and chemokines. Meanwhile, HK3 expression was highest in normal immune cells and tissues. In gliomas, the expression of HK3 was found to be closely correlated with the malignant clinical characteristics and the infiltration of macrophages. Also, HK3 was proven to be positively associated with macrophage through single-cell sequencing data and immunohistochemistry techniques. Finally, it is predicted that samples with high HK3 expression are often malignant entities and also significant genomic aberrations of driver oncogenes. CONCLUSIONS: This is the first comprehensive research to figure out the relationship between HK3 and TME characteristics in gliomas. HK3 is positively associated with macrophage infiltration and can induce the immunosuppressive TME and malignant phenotype of gliomas.


Subject(s)
Brain Neoplasms , Glioma , Hexokinase , Tumor Microenvironment , Humans , Glioma/pathology , Glioma/genetics , Glioma/immunology , Glioma/enzymology , Hexokinase/metabolism , Hexokinase/genetics , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Tumor Microenvironment/immunology , Macrophages/metabolism , Macrophages/pathology , Gene Expression Regulation, Neoplastic
20.
Cell Cycle ; 23(4): 435-447, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38626328

ABSTRACT

The precise mechanisms underlying the inhibitory effects of SIRT3, a mitochondrial sirtuin protein, on hepatocellular carcinoma (HCC) development, as well as its impact on mitochondrial respiration, remain poorly understood. We assessed sirtuins 3 (SIRT3) levels in HCC tissues and Huh7 cells cultured under hypoxic condition. We investigated the effects of SIRT3 on cell proliferation, glycolytic metabolism, mitochondrial respiration, mitophagy, and mitochondrial biogenesis in Huh7 cells. Besides, we explored the potential mechanisms regulating SIRT3 expression in hypoxically cultured Huh7 cells. Gradual reduction in SIRT3 expressions were observed in both adjacent tumor tissues and tumor tissues. Similarly, SIRT3 expressions were diminished in Huh7 cells cultured under hypoxic condition. Forced expression of SIRT3 attenuated the growth of hypoxically cultured Huh7 cells. SIRT3 overexpression led to a decrease in extracellular acidification rate while increasing oxygen consumption rate. SIRT3 downregulated the levels of hexokinase 2 and pyruvate kinase M2. Moreover, SIRT3 enhanced mitophagy signaling, as indicated by mtKeima, and upregulated key proteins involved in various mitophagic pathways while reducing intracellular reactive oxygen species levels. Furthermore, SIRT3 increased proxisome proliferator-activated receptor-gamma coactivator 1α levels and the amount of mitochondrial DNA in Huh7 cells. Notably, ß-catenin expressions were elevated in Huh7 cells cultured under hypoxic condition. Antagonists and agonists of ß-catenin respectively upregulated and downregulated SIRT3 expressions in hypoxically cultured Huh7 cells. The modulationsof glycolysis and mitochondrial respiration represent the primary mechanism through which SIRT3, suppressed by ß-catenin, inhibits HCC cell proliferation.


Subject(s)
Carcinoma, Hepatocellular , Cell Hypoxia , Glycolysis , Liver Neoplasms , Sirtuin 3 , beta Catenin , Humans , beta Catenin/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hexokinase/metabolism , Hexokinase/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Mitochondria/metabolism , Mitophagy/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 3/metabolism , Sirtuin 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL