Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177.139
Filter
1.
Biomaterials ; 312: 122760, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39163825

ABSTRACT

Inflammation-resident cells within arthritic sites undergo a metabolic shift towards glycolysis, which greatly aggravates rheumatoid arthritis (RA). Reprogramming glucose metabolism can suppress abnormal proliferation and activation of inflammation-related cells without affecting normal cells, holding potential for RA therapy. Single 2-deoxy-d-glucose (2-DG, glycolysis inhibitor) treatment often cause elevated ROS, which is detrimental to RA remission. The rational combination of glycolysis inhibition with anti-inflammatory intervention might cooperatively achieve favorable RA therapy. To improve drug bioavailability and exert synergetic effect, stable co-encapsulation of drugs in long circulation and timely drug release in inflamed milieu is highly desirable. Herein, we designed a stimulus-responsive hyaluronic acid-triglycerol monostearate polymersomes (HTDD) co-delivering 2-DG and dexamethasone (Dex) to arthritic sites. After intravenous injection, HTDD polymersomes facilitated prolonged circulation and preferential distribution in inflamed sites, where overexpressed matrix metalloproteinases and acidic pH triggered drug release. Results indicated 2-DG can inhibit the excessive cell proliferation and activation, and improve Dex bioavailability by reducing Dex efflux. Dex can suppress inflammatory signaling and prevent 2-DG-induced oxidative stress. Thus, the combinational strategy ultimately mitigated RA by inhibiting glycolysis and hindering inflammatory signaling. Our study demonstrated the great potential in RA therapy by reprogramming glucose metabolism in arthritic sites.


Subject(s)
Arthritis, Rheumatoid , Deoxyglucose , Dexamethasone , Glucose , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Animals , Glucose/metabolism , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Mice , Deoxyglucose/pharmacology , Inflammation/drug therapy , Glycolysis/drug effects , Polymers/chemistry , Hyaluronic Acid/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Male , Humans , Cell Proliferation/drug effects
2.
Cell Death Dis ; 15(10): 728, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39368995

ABSTRACT

Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. Abnormal cellular energy metabolism is a hallmark of LUAD. Glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) is a member of the γ-glutamylcyclotransferase family and an unfolded protein response pathway regulatory gene. Its biological function and molecular regulatory mechanism, especially regarding energy metabolism underlying LUAD, remain unclear. By utilizing tissue microarray and data from The Cancer Genome Atlas and Gene Expression Omnibus, we found that CHAC1 expression was markedly higher in LUAD tissues than in non-tumor tissues, and was positively correlated with poor prognosis. Phenotypically, CHAC1 overexpression enhanced the proliferation, migration, invasion, tumor sphere formation, and glycolysis ability of LUAD cells, resulting in tumor growth both in vitro and in vivo. Mechanistically, through a shotgun mass spectrometry-based proteomic approach and high-throughput RNA sequencing, we found that CHAC1 acted as a bridge connecting UBA2 and PKM2, enhancing the SUMOylation of PKM2. The SUMOylated PKM2 then transferred from the cytoplasm to the nucleus, activating the expression of glycolysis-related genes and enhancing the Warburg effect. Lastly, E2F Transcription Factor 1 potently activated CHAC1 transcription by directly binding to the CHAC1 promoter in LUAD cells. The results of this study implied that CHAC1 regulates energy metabolism and promotes glycolysis in LUAD progression.


Subject(s)
Adenocarcinoma of Lung , Carrier Proteins , Glucose , Lung Neoplasms , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Thyroid Hormones/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Glucose/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Animals , Disease Progression , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Mice , Cell Line, Tumor , Cell Proliferation , Mice, Nude , Cell Nucleus/metabolism , Male , Gene Expression Regulation, Neoplastic , Glycolysis , Female , Cell Movement , Mice, Inbred BALB C
3.
Metabolomics ; 20(5): 112, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369160

ABSTRACT

BACKGROUND: Cancer cells exhibit remarkable metabolic plasticity, enabling them to adapt to fluctuating nutrient conditions. This study investigates the impact of a combination of low glucose levels and inhibition of stearoyl-CoA desaturase 1 (SCD1) using A939572 on cancer metabolic plasticity and growth. METHODS: A comprehensive metabolomic and lipidomic analysis was conducted to unravel the intricate changes in cellular metabolites and lipids. MCF-7 cells were subjected to low glucose conditions, and SCD1 was inhibited using A939572. The resulting alterations in metabolic pathways and lipid profiles were explored to elucidate the synergistic effects on cancer cell physiology. RESULTS: The combination of low glucose and A939572-induced SCD1 inhibition significantly impaired cancer cell metabolic plasticity. Metabolomic analysis highlighted shifts in key glycolytic and amino acid pathways, indicating the cells' struggle to adapt to restricted glucose availability. Lipidomic profiling revealed alterations in lipid composition, implying disruptions in membrane integrity and signaling cascades. CONCLUSION: Our findings underscore the critical roles of glucose availability and SCD1 activity in sustaining cancer metabolic plasticity and growth. Simultaneously targeting these pathways emerges as a promising strategy to impede cancer progression. The comprehensive metabolomic and lipidomic analysis provides a detailed roadmap of molecular alterations induced by this combination treatment, that may help identify potential therapeutic targets.


Subject(s)
Glucose , Lipidomics , Metabolomics , Stearoyl-CoA Desaturase , Humans , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Glucose/metabolism , MCF-7 Cells , Lipidomics/methods , Metabolomics/methods , Lipid Metabolism/drug effects , Female , Cell Proliferation/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Metabolome/drug effects
4.
Nat Commun ; 15(1): 8658, 2024 Oct 06.
Article in English | MEDLINE | ID: mdl-39370422

ABSTRACT

The intensive nutrient requirements needed to sustain T cell activation and proliferation, combined with competition for nutrients within the tumor microenvironment, raise the prospect that glucose availability may limit CAR-T cell function. Here, we seek to test the hypothesis that stable overexpression (OE) of the glucose transporter GLUT1 in primary human CAR-T cells would improve their function and antitumor potency. We observe that GLUT1OE in CAR-T cells increases glucose consumption, glycolysis, glycolytic reserve, and oxidative phosphorylation, and these effects are associated with decreased T cell exhaustion and increased Th17 differentiation. GLUT1OE also induces broad metabolic reprogramming associated with increased glutathione-mediated resistance to reactive oxygen species, and increased inosine accumulation. When challenged with tumors, GLUT1OE CAR-T cells secrete more proinflammatory cytokines and show enhanced cytotoxicity in vitro, and demonstrate superior tumor control and persistence in mouse models. Our collective findings support a paradigm wherein glucose availability is rate limiting for effector CAR-T cell function and demonstrate that enhancing glucose availability via GLUT1OE could augment antitumor immune function.


Subject(s)
Glucose Transporter Type 1 , Glucose , Glycolysis , T-Lymphocytes , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Humans , Animals , Mice , Glucose/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Cell Differentiation , Cell Line, Tumor , Lymphocyte Activation/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Cytokines/metabolism , Cellular Reprogramming/genetics , Metabolic Reprogramming
5.
Front Endocrinol (Lausanne) ; 15: 1394252, 2024.
Article in English | MEDLINE | ID: mdl-39351534

ABSTRACT

Background: Stress urinary incontinence (SUI) is a common condition characterized by urethral sphincter failure and urine leakage. Its prevalence in women is higher than in men, and estimates of crude prevalence rates vary widely due to factors such as research methodologies, study populations, and underreporting by patients. This variability hinders research and impacts patient diagnosis, treatment, and quality of life. The complex etiology of SUI is not fully understood, and previous studies have primarily focused on non-invasive indicators. While emerging observational research suggests a correlation between SUI in women and abnormalities in lipid and blood metabolism, the underlying biological mechanisms and causal relationships require further investigation. This study aims to explore the causalities between SUI in women and lipid and blood metabolism. Methods: Using bidirectional univariate Mendelian randomization (MR), we investigated the causal association between SUI liability in women (case/control = 5,924/399,509) from UK Biobank and lipid and glucose metabolism, indicated by total cholesterol (TC, N = 61,166), low-density lipoproteins (LDL, N = 58,381), high-density lipoproteins (HDL, N = 60,812), triglycerides (TG, N = 60,027), fasting glucose (FG, N = 19,745), and fasting insulin (FI, N = 38,238) from ENGAGE consortium. To account for potential confounding effects, multivariable MR (MVMR) analyses were performed, adjusting for body mass index (BMI) and separately among lipid and glucose metabolism. Results: We found that increased genetically proxied TC, LDL, and HDL levels were associated with an elevated risk of SUI in women (OR: 1.090-1.117, all P < 0.05), These associations were further supported by MVMR analyses with adjustment for BMI (OR: 1.087-1.114, all P < 0.05). Conversely, increased FG and FI were associated with reduced SUI reliability in women (OR: 0.731-0.815, all P < 0.05). When adjusting among lipid and glucose metabolism, only HDL and FI demonstrated causal effects. Reverse MR analyses provided no genetic evidence supporting the causal effect of SUI in women on lipid and blood metabolism (all P > 0.05). Conclusions: Our results reported that increased TC, LDL, and HDL are linked to higher SUI susceptibility in women, while higher FG and FI levels have a protective effect. In overweight/obese women with metabolic abnormalities, the positive associations between TC, LDL, and HDL levels and SUI indicate a higher risk.


Subject(s)
Lipid Metabolism , Mendelian Randomization Analysis , Urinary Incontinence, Stress , Humans , Female , Urinary Incontinence, Stress/genetics , Urinary Incontinence, Stress/epidemiology , Urinary Incontinence, Stress/etiology , Middle Aged , Lipid Metabolism/genetics , Blood Glucose/metabolism , Case-Control Studies , Aged , Adult , Lipids/blood , Polymorphism, Single Nucleotide , Glucose/metabolism
6.
Endocr Regul ; 58(1): 206-214, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-39352777

ABSTRACT

Objective. Carboxypeptidase E (CPE) plays an important role in the biosynthesis of neurotransmitters and peptide hormones including insulin. It also promotes cell proliferation, survival, and invasion of tumor cells. The endoplasmic reticulum stress, hypoxia, and nutrient supply are significant factors of malignant tumor growth including glioblastoma. There are data indicating that the knockdown of the endoplasmic reticulum to nucleus signaling 1 (ERN1) suppressed glioblastoma cell proliferation and increased invasiveness of these cells. The present study aims to investigate the regulation of the CPE gene in U87MG glioblastoma cells by ERN1 knockdown, hypoxia, and glucose or glutamine deprivations with the intent to reveal the role of ERN1 signaling in the regulation of this gene expression and function in tumorigenesis. Methods. Human glioblastoma cells U87MG (transfected by an empty vector; control) and ERN1 knockdown cells with inhibited ERN1 endoribonuclease and protein kinase (dnERN1) or only ERN1 endoribonuclease (dnrERN1) were used. Hypoxia was introduced by dimethyloxalylglycine; for glucose and glutamine deprivations, the cells were cultured in DMEM medium without glucose or glutamine for 16 h, respectively. The expression level of the CPE gene was studied by quantitative RT-PCR and normalized to ACTB. Results. It was found that inhibition of endoribonuclease and protein kinase activities of ERN1 led to a strong up-regulation of CPE gene expression in glioblastoma cells. The expression of this gene also increased in glioblastoma cells after silencing ERN1. At the same time, the expression of this gene did not significantly change in cells with inhibited ERN1 endoribonuclease only. The expression of the CPE gene was resistant to hypoxia in control U87MG cells, but increased in cells with ERN1 knockdown. The expression of this gene was up-regulated under glutamine deprivation in control glioblastoma cells, but decreased upon ERN1 knockdown. However, glucose deprivation decreased the expression of CPE gene in both types of used cells, but ERN1 inhibition enhanced this effect. Conclusion. The results of the present study demonstrate that inhibition of ERN1 strongly up-regulated the expression of pro-oncogenic CPE gene through protein kinase activity of ERN1 and that increased CPE gene expression possibly participates in ERN1 knockdown-mediated invasiveness of glioblastoma cells.


Subject(s)
Carboxypeptidase H , Endoplasmic Reticulum Stress , Endoribonucleases , Gene Expression Regulation, Neoplastic , Glioblastoma , Protein Serine-Threonine Kinases , Humans , Glioblastoma/metabolism , Glioblastoma/genetics , Glioblastoma/pathology , Carboxypeptidase H/metabolism , Carboxypeptidase H/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/metabolism , Endoribonucleases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Glucose/metabolism , Gene Knockdown Techniques , Cell Hypoxia/physiology , Signal Transduction/physiology
7.
BMC Plant Biol ; 24(1): 931, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39375603

ABSTRACT

BACKGROUND: Cold is an important environmental limiting factor affecting plant yield and quality. Capsicum (chili pepper), a tropical and subtropical vegetable crop, is extremely sensitive to cold. Although H2S is an important signaling regulator in the responses of plant growth and development to abiotic stress, few studies have examined its effects on cold-sensitive capsicum varieties. Through biotechnology methods to enhance the cold resistance of peppers, to provide some reference for pepper breeding, investigated molecular regulation by H2S of responses to cold stress in cold-sensitive capsicum plants, via physiological and transcriptomic analyses. RESULTS: In capsicum seedlings, exogenous H2S enhanced relative electrical conductivity (REC) and levels of malondialdehyde (MDA) under cold stress, maintained membrane integrity, increased the activity of enzymatic and non-enzymatic antioxidants, balanced reactive oxygen species levels (O2·- and H2O2), and improved photosynthesis, mitigating the damage caused by cold. In addition, 416 differentially expressed genes (DEGs) were involved in the response to cold stress after H2S treatment. These DEGs were mainly enriched in the ascorbate-glutathione and starch-sucrose metabolic pathways and plant hormone signal-transduction pathways. Exogenous H2S altered the expression of key enzyme-encoding genes such as GST, APX, and MDHAR in the ascorbate-glutathione metabolism pathway, as well as that of regulatory genes for stimulatory hormones (auxin, cytokinins, and gibberellins) and inhibitory hormones (including jasmonate and salicylic acid) in the plant hormone signal-transduction pathway, helping to maintain the energy supply and intracellular metabolic stability under cold stress. CONCLUSIONS: These findings reveal that exogenous H2S improves cold tolerance in cold-sensitive capsicum plants, elucidating the molecular mechanisms underlying its responses to cold stress. This study provides a theoretical basis for exploring and improving cold tolerance in capsicum plants.


Subject(s)
Antioxidants , Capsicum , Gene Expression Regulation, Plant , Glucose , Hydrogen Sulfide , Capsicum/genetics , Capsicum/physiology , Capsicum/metabolism , Antioxidants/metabolism , Hydrogen Sulfide/metabolism , Glucose/metabolism , Cold-Shock Response/genetics , Cold Temperature , Seedlings/genetics , Seedlings/metabolism , Seedlings/physiology , Seedlings/growth & development , Malondialdehyde/metabolism , Reactive Oxygen Species/metabolism
8.
Front Endocrinol (Lausanne) ; 15: 1427058, 2024.
Article in English | MEDLINE | ID: mdl-39377070

ABSTRACT

Background: In addition to conventional treatment and modifications in physical activity and diet, alternative strategies have been investigated to manage, prevent, or delay diabetes in humans. In this regard, one strategy has relied on the immunomodulatory properties of mycobacteria, whereby Bacillus Calmette-Guerin, an attenuated live strain of Mycobacterium bovis, has been shown to improve glycemic control in patients with diabetes and to alleviate hyperglycemia in selected murine models of diabetes. A novel heat-killed (HK) whole-cell preparation of Mycobacterium aurum (M. aurum) is currently under development as a potential food supplement; nevertheless, its potential bioactivity remains largely unknown. Thus, the present study investigated the potential prophylactic anti-diabetic effects of HK M. aurum in streptozotocin (STZ)-induced diabetic mice. Methods: Mice were divided into three groups: the STZ-induced diabetic group was injected with a single intraperitoneal high dose of STZ, the HK M. aurum-treated diabetic group was prophylactically treated with three doses of HK M. aurum 6 weeks before STZ injection, and the control non-diabetic group was given three intradermal injections of borate-buffered saline and an intraperitoneal injection of citrate buffer. Liver lactate dehydrogenase (LDH), uncoupling protein 2 (UCP2), and glucose transporter 2 (GLUT2) and skeletal muscle LDH, UCP3, and GLUT4 protein expression levels in different mouse groups were determined by Western blot. Results: Our results indicated that HK M. aurum did not cause any significant changes in glycemic levels of normal non-diabetic mice. Prophylactic administration of three doses of HK M. aurum to diabetic mice resulted in a significant reduction in their blood glucose levels when compared to those in control diabetic mice. Prophylactic treatment of diabetic mice with HK M. aurum significantly restored their disturbed protein expression levels of liver UCP2 and LDH as well as of skeletal muscle UCP3. On the other hand, prophylactic treatment of diabetic mice with HK M. aurum had no significant effect on their liver GLUT2 and skeletal muscle GLUT4 and LDH protein expression levels. Conclusions: Our findings provide the first evidence that HK M. aurum possesses a hyperglycemia-lowering capacity and might support its future use as a food supplement for the amelioration of diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Liver , Muscle, Skeletal , Oxidative Stress , Animals , Mice , Diabetes Mellitus, Experimental/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Liver/metabolism , Liver/drug effects , Hyperglycemia/prevention & control , Hyperglycemia/metabolism , Oxidative Stress/drug effects , Male , Blood Glucose/metabolism , Streptozocin , Mycobacterium , Hot Temperature , Glucose/metabolism
9.
Pol Merkur Lekarski ; 52(4): 427-432, 2024.
Article in English | MEDLINE | ID: mdl-39360723

ABSTRACT

OBJECTIVE: Aim: The aim of the study was to investigate the activity of bioenergetic processes in rats under conditions of simultaneous exposure to malathion and carbon tetrachloride and after the use of enterosgel. PATIENTS AND METHODS: Materials and Methods: Experiments were conducted on rats. The rats were divided into nine groups.Malathion was administered daily (for 30 days) at a dose of 20 mg / kg body weight of the animal. Tetrachloromethane was administered twice (every other day) as a 50% oil solution at a dose of 1.0 ml / kg body weight. The intensity of energy supply processes was assessed by the activity of succinate dehydrogenase and cytochrome oxidase, impaired carbohydrate metabolism in terms of glucose and glycogen. RESULTS: Results: It was noted that succinate dehydrogenase activity in the liver decreased 2 times, in the myocardium - 1.6 times. On the thirty and seventh day of administration of toxicants after enterosorbent use, succinate dehydrogenase activity increased in the liver by 20%, cytochrome oxidase by 27%, in the myocardium - by 31% and 23%, respectively. The content of glucose in the serum after exposure to toxicants increased maximally (2.4 times) at the end of the study. In contrast, the glycogen content in the liver decreased by 48%, in the myocardium by 13%. The use of enterosgel resulted in a decrease in serum glucose. CONCLUSION: Conclusions: The use of enterosgel leads to the restoration of energy processes in the body of affected rats, which is confirmed by increased activity of mitochondrial enzymes, lowering glucose and increasing glycogen in the studied organs.


Subject(s)
Carbon Tetrachloride , Energy Metabolism , Liver , Malathion , Succinate Dehydrogenase , Animals , Rats , Energy Metabolism/drug effects , Succinate Dehydrogenase/metabolism , Liver/drug effects , Liver/metabolism , Liver/enzymology , Male , Myocardium/metabolism , Rats, Wistar , Electron Transport Complex IV/metabolism , Glucose/metabolism , Glycogen/metabolism , Insecticides
10.
Nat Commun ; 15(1): 8588, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39362888

ABSTRACT

Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11ß-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11ß-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11ß-HSD1 by using bone-targeted 11ß-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11ß-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Diet, High-Fat , Mice, Inbred C57BL , Mice, Knockout , Obesity , Osteoblasts , Animals , Diet, High-Fat/adverse effects , Osteoblasts/metabolism , Osteoblasts/drug effects , Male , Obesity/metabolism , Obesity/etiology , Obesity/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Mice , Osteogenesis/drug effects , Glucose/metabolism , Glucocorticoids/metabolism , Signal Transduction , Bone Resorption/metabolism , Bone Resorption/prevention & control
11.
Sci Rep ; 14(1): 23031, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39363015

ABSTRACT

Tumor cells promote malignant behaviors such as proliferation, invasion, and metastasis of cancer cells through glucose metabolic reprogramming, but the role of the H-dependent sugar cotransporter SLC45A4 in regulating metabolic reprogramming in ovarian cancer (OC) remains largely unknown. This study aimed to investigate the effects of SLC45A4 silencing on the transcriptome spectrum of ovarian cancer cells (OCC), glucose uptake, lactic acid production, intracellular ATP levels, and the expression and activity of HIF-α glycolysis signaling pathway. The results showed that SLC45A4 is overexpressed in OC and its elevated expression correlates with adverse clinical outcomes in OC patients. Silencing of SLC45A4 significantly inhibited the proliferation, invasion, and metastasis of OCC by suppressing glucose uptake and glycolysis, and it also reduced the expression of HIF-α glycolysis signaling pathway in OC tissues. In vivo experiments using shRNA to knock down SLC45A4 in xenograft models in nude mice demonstrated a significant inhibition of tumor growth. These findings suggest that SLC45A4 silencing can restrain the malignant progression of OC by inhibiting glucose uptake in OCC and affecting the reprogramming of glycolytic energy metabolism, indicating that SLC45A4 may serve as a potential therapeutic target for OC intervention.


Subject(s)
Cell Proliferation , Glycolysis , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Animals , Cell Line, Tumor , Mice , Mice, Nude , Disease Progression , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Signal Transduction , Metabolic Reprogramming
12.
Elife ; 132024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365738

ABSTRACT

Artificially sweetened beverages containing noncaloric monosaccharides were suggested as healthier alternatives to sugar-sweetened beverages. Nevertheless, the potential detrimental effects of these noncaloric monosaccharides on blood vessel function remain inadequately understood. We have established a zebrafish model that exhibits significant excessive angiogenesis induced by high glucose, resembling the hyperangiogenic characteristics observed in proliferative diabetic retinopathy (PDR). Utilizing this model, we observed that glucose and noncaloric monosaccharides could induce excessive formation of blood vessels, especially intersegmental vessels (ISVs). The excessively branched vessels were observed to be formed by ectopic activation of quiescent endothelial cells (ECs) into tip cells. Single-cell transcriptomic sequencing analysis of the ECs in the embryos exposed to high glucose revealed an augmented ratio of capillary ECs, proliferating ECs, and a series of upregulated proangiogenic genes. Further analysis and experiments validated that reduced foxo1a mediated the excessive angiogenesis induced by monosaccharides via upregulating the expression of marcksl1a. This study has provided new evidence showing the negative effects of noncaloric monosaccharides on the vascular system and the underlying mechanisms.


Consuming too much sugar can damage blood vessels and contribute to diseases like diabetes and heart disease. Artificial sweeteners have been suggested as a healthier alternative, and are now included in many products like sodas and baked goods. However, some studies have suggested that people who consume large amounts of artificial sweeteners also have an increased risk of cardiovascular disease. Others suggest individuals may also experience spikes in blood sugar levels similar to those observed in people with diabetes. Yet few studies have examined how artificial sweeteners affect the network of vessels that transport blood and other substances around the body. To investigate this question, Wang, Zhao, Xu, et al. studied zebrafish embryos which had been exposed to sugar and a type of artificial sweetener known as non-caloric monosaccharides. Various imaging tools revealed that high levels of sugar caused the embryos to produce more new blood vessels via a process called angiogenesis. This excessive growth of blood vessels has previously been linked to diabetic complications, including cardiovascular disease. Wang, Zhao, Xu, et al. found that zebrafish embryos exposed to several different non-caloric monosaccharides developed similar blood vessel problems. All the sweeteners tested caused immature cells lining the blood vessels to develop into active tip cells that promote angiogenesis. This led to more new blood vessels forming that branch off already existing veins and arteries. These findings suggest that artificial sweeteners may cause the same kind of damage to blood vessels as sugar. This may explain why people who consume a lot of artificial sweeteners are at risk of developing heart disease and high blood sugar levels. Future studies could help scientists learn more about how genetics or other factors affect the health impact of sugars and artificial sweeteners. This may lead to a greater understanding of the long-term health effects of artificially sweetened foods.


Subject(s)
Forkhead Box Protein O1 , Monosaccharides , Neovascularization, Physiologic , Zebrafish , Animals , Neovascularization, Physiologic/drug effects , Monosaccharides/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Glucose/metabolism , Glucose/pharmacology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Signal Transduction , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Angiogenesis
13.
Sci Adv ; 10(40): eadq7305, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39356752

ABSTRACT

Solid tumors are characterized by dysfunctional vasculature that limits perfusion and delivery of nutrients to the tumor microenvironment. Limited perfusion coupled with the high metabolic demand of growing tumors has led to the hypothesis that many tumors experience metabolic stress driven by limited availability of nutrients such as glucose, oxygen, and amino acids in the tumor. Such metabolic stress has important implications for the biology of cells in the microenvironment, affecting both disease progression and response to therapies. Recently, techniques have been developed to identify limiting nutrients and resulting metabolic stresses in solid tumors. These techniques have greatly expanded our understanding of the metabolic limitations in tumors. This review will discuss these experimental tools and the emerging picture of metabolic limitations in tumors arising from recent studies using these approaches.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Neoplasms/metabolism , Neoplasms/pathology , Animals , Glucose/metabolism
14.
Sci Rep ; 14(1): 22875, 2024 10 02.
Article in English | MEDLINE | ID: mdl-39358483

ABSTRACT

Achieving high-gravity fermentation in the industrial production of fuel ethanol, and enhancing the fermentation efficiency of high-salt raw materials, such as waste molasses, can significantly reduce wastewater output and process costs. Therefore, the development of hyperosmotic-tolerant industrial Saccharomyces cerevisiae strains, capable of resisting high-salt stress, offers both environmental and economic benefits. Our previous study highlighted the potential of CRZ1 overexpression as a strategy to improve the yeast strain's resistance to high-salt stress, however, the underlying molecular mechanisms remain unexplored. The fermentation capabilities of the CRZ1-overexpressing strain, KCR3, and its parental strain, KF7, were evaluated under condition of 1.25 M NaCl at 35 °C. Compared to KF7, KCR3 showed an 81% increase in glucose consumption (129.25 ± 0.83 g/L) and a 105% increase in ethanol production (47.59 ± 0.93 g/L), with a yield of 0.37 g/g. Comparative transcriptomic analysis showed that under high-salt stress, KCR3 exhibited significantly upregulated expression of genes associated with ion transport, stress response, gluconeogenesis, and the utilization of alternative carbon sources, while genes related to glycolysis and the biosynthesis of ribosomes, amino acids, and fatty acids were notably downregulated compared to KF7. Crz1 likely expands its influence by regulating the expression of numerous transcription factors, thereby impacting genes involved in multiple aspects of cellular function. The study revealed the regulatory mechanism of Crz1 under high-salt stress, thereby providing guidance for the construction of salt-tolerant strains.


Subject(s)
Ethanol , Fermentation , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Salt Tolerance , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Salt Tolerance/genetics , Ethanol/metabolism , Glucose/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Profiling
15.
Acta Crystallogr D Struct Biol ; 80(Pt 10): 733-743, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39361356

ABSTRACT

ß-Glucosidase from the thermophilic bacterium Caldicellulosiruptor saccharolyticus (Bgl1) has been denoted as having an attractive catalytic profile for various industrial applications. Bgl1 catalyses the final step of in the decomposition of cellulose, an unbranched glucose polymer that has attracted the attention of researchers in recent years as it is the most abundant renewable source of reduced carbon in the biosphere. With the aim of enhancing the thermostability of Bgl1 for a broad spectrum of biotechnological processes, it has been subjected to structural studies. Crystal structures of Bgl1 and its complex with glucose were determined at 1.47 and 1.95 Šresolution, respectively. Bgl1 is a member of glycosyl hydrolase family 1 (GH1 superfamily, EC 3.2.1.21) and the results showed that the 3D structure of Bgl1 follows the overall architecture of the GH1 family, with a classical (ß/α)8 TIM-barrel fold. Comparisons of Bgl1 with sequence or structural homologues of ß-glucosidase reveal quite similar structures but also unique structural features in Bgl1 with plausible functional roles.


Subject(s)
beta-Glucosidase , beta-Glucosidase/chemistry , beta-Glucosidase/metabolism , Crystallography, X-Ray , Caldicellulosiruptor/enzymology , Models, Molecular , Protein Conformation , Bacterial Proteins/chemistry , Catalytic Domain , Glucose/metabolism , Amino Acid Sequence
16.
Proc Natl Acad Sci U S A ; 121(41): e2403033121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39365816

ABSTRACT

Mammalian cell culture processes are widely utilized for biotherapeutics production, disease diagnostics, and biosensors, and hence, should be optimized to support robust cell growth and viability. However, toxic by-products accumulate in cultures due to inefficiencies in metabolic activities and nutrient utilization. In this study, we applied comprehensive 13C stable-isotope tracing of amino acids and glucose to two Immunoglobulin G (IgG) producing Chinese Hamster Ovary (CHO) cell lines to identify secreted by-products and trace their origins. CHO cells were cultured in media formulations missing a single amino acid or glucose supplemented with a 13C-tracer of the missing substrate, followed by gas chromatography-mass spectrometry (GC-MS) analysis to track labeled carbon flows and identify by-products. We tracked the sources of all secreted by-products and verified the identity of 45 by-products, majority of which were derived from glucose, leucine, isoleucine, valine, tyrosine, tryptophan, methionine, and phenylalanine. In addition to by-products identified previously, we identified several metabolites including 2-hydroxyisovaleric acid, 2-aminobutyric acid, L-alloisoleucine, ketoisoleucine, 2-hydroxy-3-methylvaleric acid, desmeninol, and 2-aminobutyric acid. When added to CHO cell cultures at different concentrations, certain metabolites inhibited cell growth while others including 2-hydroxy acids, surprisingly, reduced lactate accumulation. In vitro enzymatic analysis indicated that 2-hydroxy acids were metabolized by lactate dehydrogenase suggesting a possible mechanism for lowered lactate accumulation, e.g., competitive substrate inhibition. The 13C-labeling assisted metabolomics pipeline developed and the metabolites identified will serve as a springboard to reduce undesirable by-products accumulation and alleviate inefficient substrate utilization in mammalian cultures used for biomanufacturing and other applications through altered media formulations and pathway engineering strategies.


Subject(s)
Amino Acids , Carbon Isotopes , Cricetulus , Glucose , Animals , CHO Cells , Glucose/metabolism , Amino Acids/metabolism , Carbon Isotopes/metabolism , Gas Chromatography-Mass Spectrometry/methods , Cell Culture Techniques/methods , Cricetinae , Immunoglobulin G/metabolism , Isotope Labeling/methods
17.
Mol Med Rep ; 30(6)2024 Dec.
Article in English | MEDLINE | ID: mdl-39370816

ABSTRACT

Solute carrier family 12 member 5 (SLC12A5) is an oncogene in numerous types of cancer, however its function in breast cancer (BC) remains elusive. ETS translocation variant 4 (ETV4) promotes BC. Therefore, the present study aimed to elucidate the role of SLC12A5 in ferroptosis and glucose metabolism in BC cells as well as to understand the underlying mechanism. Analysis of data from the UALCAN database demonstrated expression levels of SLC12A5 in BC and its association with prognosis. Reverse transcription­quantitative PCR and western blotting were conducted to evaluate the expression levels of SLC12A5 and ETV4 in BC cells. The abilities of BC cells to proliferate, migrate and invade were assessed using Cell Counting Kit­8, colony formation, wound healing and Transwell assays. Thiobarbituric acid reactive substances assay and a C11 BODIPY 581/591 probe were used to evaluate lipid peroxidation. Ferroptosis resistance was evaluated by the measurement of Fe2+ and ferroptosis­related solute carrier family 7a member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), acyl­CoA synthetase long­chain family member 4 (ACSL4) and transferrin receptor 1 (TFR1) protein levels. Glycolysis was assessed via evaluation of extracellular acidification rate, oxygen consumption rate, lactate production and glucose consumption. Finally, luciferase reporter and chromatin immunoprecipitation assay were used to verify the interaction between ETV4 and the SLC12A5 promoter. UALCAN database analysis indicated that SLC12A5 was upregulated in BC tissues and cells and that SLC12A5 elevation indicated a poor prognosis of patients with BC. SLC12A5 knockdown suppressed the BC cell proliferative, migratory and invasive capabilities. Moreover, SLC12A5 knockdown decreased BC cell ferroptosis resistance and glucose metabolism reprogramming. The transcription factor ETV4 was demonstrated to bind to the SLC12A5 promoter and upregulate its transcription. Furthermore, ETV4 overexpression counteracted the suppressive effect of SLC12A5 knockdown on the BC cell proliferative, migratory and invasive abilities, as well as on ferroptosis resistance and glucose metabolism reprogramming. Transcriptional activation of SLC12A5 by ETV4 modulated the migration, invasion, ferroptosis resistance and glucose metabolism reprogramming of BC cells.


Subject(s)
Breast Neoplasms , Ferroptosis , Gene Expression Regulation, Neoplastic , Glucose , Transcriptional Activation , Humans , Ferroptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Glucose/metabolism , Female , Cell Line, Tumor , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Adenovirus E1A Proteins/metabolism , Adenovirus E1A Proteins/genetics , Cell Proliferation , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-ets/genetics , Prognosis , MCF-7 Cells , Cell Movement/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Metabolic Reprogramming
18.
Arch Microbiol ; 206(10): 422, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352521

ABSTRACT

D-xylose, one of the most abundant sugars in lignocellulosic biomass, is not widely used to produce bioproducts with added value, in part due to the absence of industrial microorganisms able to metabolize it efficiently. Herbaspirillum seropedicae Z69 is a ß-proteobacterium able to accumulate poly-3-hydroxybutyrate, a biodegradable thermoplastic biopolymer, with contents higher than 50%. It metabolizes D-xylose by non-phosphorylative pathways. In the genome of Z69, we found the genes xylFGH (ABC D-xylose transporter), xylB, xylD, and xylC (superior non-phosphorylative pathway), and the transcriptional regulator xylR, forming the xyl cluster. We constructed the knock-out mutant Z69ΔxylR that has a reduced growth in D-xylose and in D-glucose, compared with Z69. In addition, we analyzed the expression of xyl genes by RT-qPCR and promoter fusion. These results suggest that XylR activates the expression of genes at the xyl cluster in the presence of D-xylose. On the other hand, XylR does not regulate the expression of xylA, mhpD (lower non-phosphorylative pathways) and araB (L-arabinose dehydrogenase) genes. The participation of D-glucose in the regulation mechanism of these genes must still be elucidated. These results contribute to the development of new strains adapted to consume lignocellulosic sugars for the production of value-added bioproducts.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Herbaspirillum , Multigene Family , Xylose , Xylose/metabolism , Herbaspirillum/genetics , Herbaspirillum/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Polyesters/metabolism , Hydroxybutyrates/metabolism , Glucose/metabolism , Promoter Regions, Genetic , Polyhydroxybutyrates
19.
Nat Commun ; 15(1): 8632, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39366937

ABSTRACT

Rapid glucose supply is crucial for animal survival during stress response. How the timescale of stress-induced glucose release precisely controlled by hypothalamic corticotropin-releasing hormone (CRH) neurons remains unclear. Here, we show that stress-induced hyperglycemia can be divided into at least two stages in male mice: the first fast stage is mediated by hypothalamus (paraventricular to ventromedial hypothalamus)-sympathetic (raphe pallidus nucleus to intermediolateral nucleus)-liver (HSL) axis activity; the second delayed stage is mediated by adrenal activity. Blocking the activity of HSL axis impairs predatory evoked flight responses, indicating that the HSL pathway activity is necessary for stress coping. We further reveal the intracellular signal cascade for CRH signal in the hypothalamus, which is mediated by GABAA receptor ß3 subunit phosphorylation at S408/409, results in prevention of GABAA receptor membrane recruitment. Thus, we uncovered the precise timescale of glucose supply during stress which is mediated by adrenal independent HSL and adrenal dependent pathway respectively.


Subject(s)
Corticotropin-Releasing Hormone , Hyperglycemia , Hypothalamus , Liver , Receptors, GABA-A , Animals , Male , Hyperglycemia/metabolism , Liver/metabolism , Corticotropin-Releasing Hormone/metabolism , Mice , Hypothalamus/metabolism , Receptors, GABA-A/metabolism , Stress, Physiological , Glucose/metabolism , Signal Transduction , Mice, Inbred C57BL , Phosphorylation , Neurons/metabolism , Adrenal Glands/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
20.
Sci Rep ; 14(1): 23147, 2024 10 04.
Article in English | MEDLINE | ID: mdl-39366987

ABSTRACT

Reprogrammed glucose metabolism is considered as the hallmark of cancer with therapeutic implications. Phytocompounds have potential to inhibit cancer metabolism. Here, we tested the ability of Withaferin A (WA), a withanolide derived from Withania somnifera, in modulating cancer metabolism. The assessed effect of WA on aerobic glycolysis in breast cancer cell lines showed that WA decreases the glucose uptake, lactate production and ATP generation by inhibiting the expression of key glycolytic enzymes i.e., GLUT1, HK2 and PKM2. We also identified that WA induced inhibition of cancer glycolysis by targeting c-myc as validated by silencing experiments followed by metabolic readouts. Decreased glycolysis resulted in reduced cell viability, biomass and colony forming ability of breast cancer cells. To further validate our in vitro findings in breast cancer patients, we analyzed 90 metabolic pathways in ~ 2000 breast tumors and observed that glycolysis is the most deregulated pathway in breast tumors. Deregulated glycolysis also predicted poor prognosis in breast cancer patients. In addition, patient data showed correlation between c-myc expression and glycolytic deregulation in breast cancer. Taken together, our results highlight the role of WA in inhibiting breast cancer metabolism via c-myc/glycolysis axis.


Subject(s)
Breast Neoplasms , Glycolysis , Proto-Oncogene Proteins c-myc , Withanolides , Withanolides/pharmacology , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Glycolysis/drug effects , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Glucose/metabolism , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL