Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.336
Filter
1.
J Mol Neurosci ; 74(2): 52, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38724832

ABSTRACT

Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism-based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease.


Subject(s)
Artemisinins , Brain Neoplasms , Glioblastoma , Glutamine , Glioblastoma/metabolism , Glioblastoma/drug therapy , Humans , Glutamine/metabolism , Cell Line, Tumor , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Artemisinins/therapeutic use , Artemisinins/pharmacology , Reactive Oxygen Species/metabolism , Glutaminase/metabolism , Glutaminase/antagonists & inhibitors , Tumor Microenvironment , Apoptosis , Thiadiazoles/pharmacology , Thiadiazoles/therapeutic use , Cell Movement , Benzeneacetamides/pharmacology , Benzeneacetamides/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology
2.
Mil Med Res ; 11(1): 28, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711073

ABSTRACT

BACKGROUND: Intervertebral disc degeneration (IVDD) is a multifaceted condition characterized by heterogeneity, wherein the balance between catabolism and anabolism in the extracellular matrix of nucleus pulposus (NP) cells plays a central role. Presently, the available treatments primarily focus on relieving symptoms associated with IVDD without offering an effective cure targeting its underlying pathophysiological processes. D-mannose (referred to as mannose) has demonstrated anti-catabolic properties in various diseases. Nevertheless, its therapeutic potential in IVDD has yet to be explored. METHODS: The study began with optimizing the mannose concentration for restoring NP cells. Transcriptomic analyses were employed to identify the mediators influenced by mannose, with the thioredoxin-interacting protein (Txnip) gene showing the most significant differences. Subsequently, small interfering RNA (siRNA) technology was used to demonstrate that Txnip is the key gene through which mannose exerts its effects. Techniques such as colocalization analysis, molecular docking, and overexpression assays further confirmed the direct regulatory relationship between mannose and TXNIP. To elucidate the mechanism of action of mannose, metabolomics techniques were employed to pinpoint glutamine as a core metabolite affected by mannose. Next, various methods, including integrated omics data and the Gene Expression Omnibus (GEO) database, were used to validate the one-way pathway through which TXNIP regulates glutamine. Finally, the therapeutic effect of mannose on IVDD was validated, elucidating the mechanistic role of TXNIP in glutamine metabolism in both intradiscal and orally treated rats. RESULTS: In both in vivo and in vitro experiments, it was discovered that mannose has potent efficacy in alleviating IVDD by inhibiting catabolism. From a mechanistic standpoint, it was shown that mannose exerts its anti-catabolic effects by directly targeting the transcription factor max-like protein X-interacting protein (MondoA), resulting in the upregulation of TXNIP. This upregulation, in turn, inhibits glutamine metabolism, ultimately accomplishing its anti-catabolic effects by suppressing the mitogen-activated protein kinase (MAPK) pathway. More importantly, in vivo experiments have further demonstrated that compared with intradiscal injections, oral administration of mannose at safe concentrations can achieve effective therapeutic outcomes. CONCLUSIONS: In summary, through integrated multiomics analysis, including both in vivo and in vitro experiments, this study demonstrated that mannose primarily exerts its anti-catabolic effects on IVDD through the TXNIP-glutamine axis. These findings provide strong evidence supporting the potential of the use of mannose in clinical applications for alleviating IVDD. Compared to existing clinically invasive or pain-relieving therapies for IVDD, the oral administration of mannose has characteristics that are more advantageous for clinical IVDD treatment.


Subject(s)
Cell Cycle Proteins , Glutamine , Intervertebral Disc Degeneration , Mannose , Intervertebral Disc Degeneration/drug therapy , Mannose/pharmacology , Mannose/therapeutic use , Animals , Rats , Glutamine/pharmacology , Glutamine/metabolism , Male , Rats, Sprague-Dawley , Humans , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism
3.
PLoS One ; 19(5): e0303471, 2024.
Article in English | MEDLINE | ID: mdl-38718074

ABSTRACT

OBJECTIVE: Preeclampsia (PE) is a severe complication of unclear pathogenesis associated with pregnancy. This research aimed to elucidate the properties of immune cell infiltration and potential biomarkers of PE based on bioinformatics analysis. MATERIALS AND METHODS: Two PE datasets were imported from the Gene ExpressioOmnibus (GEO) and screened to identify differentially expressed genes (DEGs). Significant module genes were identified by weighted gene co-expression network analysis (WGCNA). DEGs that interacted with key module genes (GLu-DEGs) were analyzed further by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. The diagnostic value of the genes was assessed using receiver operating characteristic (ROC) curves and protein-protein interaction (PPI) networks were constructed using GeneMANIA, and GSVA analysis was performed using the MSigDB database. Immune cell infiltration was analyzed using the TISIDB database, and StarBase and Cytoscape were used to construct an RBP-mRNA network. The identified hub genes were validated in two independent datasets. For further confirmation, placental tissue from healthy pregnant women and women with PE were collected and analyzed using both RT-qPCR and immunohistochemistry. RESULTS: A total of seven GLu-DEGs were obtained and were found to be involved in pathways associated with the transport of sulfur compounds, PPAR signaling, and energy metabolism, shown by GO and KEGG analyses. GSVA indicated significant increases in adipocytokine signaling. Furthermore, single-sample Gene Set Enrichment Analysis (ssGSEA) indicated that the levels of activated B cells and T follicular helper cells were significantly increased in the PE group and were negatively correlated with GLu-DEGs, suggesting their potential importance. CONCLUSION: In summary, the results showed a correlation between glutamine metabolism and immune cells, providing new insights into the understandingPE pathogenesis and furnishing evidence for future advances in the treatment of this disease.


Subject(s)
Gene Regulatory Networks , Glutamine , Pre-Eclampsia , Protein Interaction Maps , Humans , Pre-Eclampsia/genetics , Pre-Eclampsia/immunology , Female , Pregnancy , Protein Interaction Maps/genetics , Glutamine/metabolism , Computational Biology/methods , Gene Ontology , Gene Expression Profiling , Adult , Placenta/metabolism , Placenta/immunology
4.
Braz J Med Biol Res ; 57: e13468, 2024.
Article in English | MEDLINE | ID: mdl-38808890

ABSTRACT

In this double-blind placebo-controlled randomized investigation, we assessed the tolerability of glutamine in older adults recruited from three daycare centers. The relevance of studying glutamine supplementation in elderly patients lies in its potential to provide a well-tolerated intervention. Glutamine, a crucial amino acid, plays a vital role in various physiological processes, including immune function and protein synthesis. Understanding its impact on older adults is essential, given the potential implications for their health and well-being. Participants received a daily dose of 12.4 g of oral effervescent glutamine (EGln group) or maltodextrin (placebo group) for 60 days. Fifteen patients from each group completed the study. The mean ages were 77.0±9.1 and 79.0±6.9 years for the EGln and placebo groups, respectively. We evaluated body mass index, aminogram, hemogram, plasma levels of glucose, prealbumin, albumin, urea, creatinine, uric acid, C-reactive protein, vitamin D, calcium, sodium, potassium, and the plasma activities of aspartate aminotransferase and alanine aminotransferase. Notably, we quantified a broad array of inflammatory markers and growth factors providing a holistic understanding of the potential effects of glutamine supplementation. The results demonstrated that oral glutamine did not induce significant changes in any evaluated parameters, and no adverse effects were reported. This finding suggested that the dosage of glutamine used in this study was well-tolerated and safe. This information contributes to the broader understanding of glutamine supplementation, emphasizing its safety and supporting its potential as a viable intervention for maintaining health in aging individuals.


Subject(s)
Dietary Supplements , Glutamine , Humans , Glutamine/administration & dosage , Double-Blind Method , Aged , Male , Female , Aged, 80 and over , Biomarkers/blood
5.
Mol Cell Biol ; 44(4): 149-163, 2024.
Article in English | MEDLINE | ID: mdl-38725392

ABSTRACT

Phenylacetylglutamine (PAGln), a gut metabolite is substantially elevated in heart failure (HF). The increase of PAGln in plasma is associated with atrial fibrillation (AF), and contributes to AF pathogenesis. However, the role of PAGln in AF with HF remains uncertain. Therefore, this study aimed to determine the effect of PAGln on AF after HF. Thoracic aortic coarctation (TAC) created overpressure-induced HF mice for 4 weeks. Histopathology, biochemical, echocardiographic for assessment of cardiac function, and electrophysiological examination of several electrophysiological indexes (ERP, SNRT, and the occurrence rate of AF) were performed at the end of the HF mice model. We found that plasma PAGln levels were significantly elevated in PAGln-treated HF mice and that PAGln aggravated maladaptive structural remodeling and electrical remodeling, which aggravated the vulnerability of AF, shortened the ERP duration, prolonged the SNRT, increased the occurrence rate of AF in HF mice. Mechanistically, PAGln exacerbated ROS accumulation and increased the levels of phosphorylated PLB and CAMK II. Overall, PAGln played a vital role in promoting the occurrence of AF in HF mice by activating the CAMK II signaling pathway.


Subject(s)
Atrial Fibrillation , Heart Failure , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/etiology , Mice , Heart Failure/etiology , Heart Failure/metabolism , Male , Mice, Inbred C57BL , Disease Models, Animal , Glutamine/metabolism , Glutamine/analogs & derivatives , Glutamine/pharmacology , Signal Transduction/drug effects , Reactive Oxygen Species/metabolism
6.
Gac Med Mex ; 160(1): 1-8, 2024.
Article in English | MEDLINE | ID: mdl-38753562

ABSTRACT

BACKGROUND: Protein interactions participate in many molecular mechanisms involved in cellular processes. The human TATA box binding protein (hTBP) interacts with Antennapedia (Antp) through its N-terminal region, specifically via its glutamine homopeptides. This PolyQ region acts as a binding site for other transcription factors under normal conditions, but when it expands, it generates spinocerebellar ataxia 17 (SCA17), whose protein aggregates in the brain prevent its correct functioning. OBJECTIVE: To determine whether the hTBP glutamine-rich region is involved in its interaction with homeoproteins and the role it plays in the formation of protein aggregates in SCA17. MATERIAL AND METHODS: We characterized hTBP interaction with other homeoproteins using BiFC, and modeled SCA17 in Drosophila melanogaster by targeting hTBPQ80 to the fly brain using UAS/GAL4. RESULTS: There was hTBP interaction with homeoproteins through its glutamine-rich region, and hTBP protein aggregates with expanded glutamines were found to affect the locomotor capacity of flies. CONCLUSIONS: The study of hTBP interactions opens the possibility for the search for new therapeutic strategies in neurodegenerative pathologies such as SCA17.


ANTECEDENTES: Las interacciones proteicas participan en una gran cantidad de mecanismos moleculares que rigen los procesos celulares. La proteína de unión a la caja TATA humana (hTBP) interacciona con Antennapedia (Antp) a través de su extremo N-terminal, específicamente a través de sus homopéptidos de glutaminas. Esta región PolyQ sirve como sitio de unión a factores de transcripción en condiciones normales, pero cuando se expande genera la ataxia espinal cerebelosa 17 (SCA17), cuyos agregados proteicos en el cerebro impiden su funcionamiento correcto. OBJETIVO: Determinar si la región rica en glutaminas de hTBP interviene en su interacción con homeoproteínas y el papel que tiene en la formación de agregados proteicos en SCA17. MATERIAL Y MÉTODOS: Se caracterizó la interacción de hTBP con otras homeoproteínas usando BiFC y se modeló SCA17 en Drosophila melanogaster dirigiendo hTBPQ80 al cerebro de las moscas usando UAS/GAL4. RESULTADOS: Existió interacción de hTBP con homeoproteínas a través de su región rica en glutaminas. Los agregados proteicos de hTBP con las glutaminas expandidas afectaron la capacidad locomotriz de las moscas. CONCLUSIONES: El estudio de las interacciones de hTBP abre la posibilidad para la búsqueda de nuevas estrategias terapéuticas en patologías neurodegenerativas como SCA17.


Subject(s)
Disease Models, Animal , Drosophila melanogaster , Spinocerebellar Ataxias , TATA-Box Binding Protein , Animals , Drosophila melanogaster/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/genetics , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/genetics , Humans , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Glutamine/metabolism , Protein Aggregates/physiology , Peptides/metabolism , Brain/metabolism
7.
Mol Med ; 30(1): 64, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760723

ABSTRACT

BACKGROUND: Insulin like growth factor II mRNA binding protein 3 (IGF2BP3) has been implicated in numerous inflammatory and cancerous conditions. However, its precise molecular mechanisms in endometriosis (EMs) remains unclear. The aim of this study is to examine the influence of IGF2BP3 on the occurrence and progression of EMs and to elucidate its underlying molecular mechanism. METHODS: Efects of IGF2BP3 on endometriosis were confrmed in vitro and in vivo. Based on bioinformatics analysis, RNA immunoprecipitation (RIP), RNA pull-down assays and Fluorescent in situ hybridization (FISH) were used to show the association between IGF2BP3 and UCA1. Single-cell spatial transcriptomics analysis shows the expression distribution of glutaminase 1 (GLS1) mRNA in EMs. Study the effect on glutamine metabolism after ectopic endometriotic stromal cells (eESCs) were transfected with Sh-IGF2BP3 and Sh-UCA1 lentivirus. RESULTS: Immunohistochemical staining have revealed that IGF2BP3 was upregulated in ectopic endometriotic lesions (EC) compared to normal endometrial tissues (EN). The proliferation and migration ability of eESCs were greatly reduced by downregulating IGF2BP3. Additionally, IGF2BP3 has been observed to interact with urothelial carcinoma associated 1 (UCA1), leading to increased stability of GLS1 mRNA and subsequently enhancing glutamine metabolism. Results also demonstrated that IGF2BP3 directly interacts with the 3' UTR region of GLS1 mRNA, influencing its expression and stability. Furthermore, UCA1 was able to bind with c-MYC protein, stabilizing c-MYC mRNA and consequently enhancing GLS1 expression through transcriptional promotion. CONCLUSION: These discoveries underscored the critical involvement of IGF2BP3 in the elevation and stability of GLS1 mRNA in the context of glutamine metabolism by interacting with UCA1 in EMs. The implications of our study extended to the identification of possible therapeutic targets for individuals with EMs.


Subject(s)
Endometriosis , Glutaminase , Glutamine , RNA Stability , RNA, Long Noncoding , RNA-Binding Proteins , Female , Humans , Glutaminase/metabolism , Glutaminase/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Endometriosis/metabolism , Endometriosis/genetics , Endometriosis/pathology , Glutamine/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Proliferation , Adult , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression Regulation , Protein Binding
8.
Sensors (Basel) ; 24(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38793927

ABSTRACT

Specialized cancer treatments have the potential to exploit glutamine dependence to increase patient survival rates. Glutamine diagnostics capable of tracking a patient's response to treatment would enable a personalized treatment dosage to optimize the tradeoff between treatment success and dangerous side effects. Current clinical glutamine testing requires sophisticated and expensive lab-based tests, which are not broadly available on a frequent, individualized basis. To address the need for a low-cost, portable glutamine diagnostic, this work engineers a cell-free glutamine biosensor to overcome assay background and signal-to-noise limitations evident in previously reported studies. The findings from this work culminate in the development of a shelf-stable, paper-based, colorimetric glutamine test with a high signal strength and a high signal-to-background ratio for dramatically improved signal resolution. While the engineered glutamine test is important progress towards improving the management of cancer and other health conditions, this work also expands the assay development field of the promising cell-free biosensing platform, which can facilitate the low-cost detection of a broad variety of target molecules with high clinical value.


Subject(s)
Biosensing Techniques , Glutamine , Metabolic Engineering , Biosensing Techniques/methods , Glutamine/metabolism , Metabolic Engineering/methods , Humans , Genetic Engineering/methods , Paper , Colorimetry/methods , Cell-Free System
9.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791327

ABSTRACT

Cancer cells adeptly manipulate their metabolic processes to evade immune detection, a phenomenon intensifying the complexity of cancer progression and therapy. This review delves into the critical role of cancer cell metabolism in the immune-editing landscape, highlighting how metabolic reprogramming facilitates tumor cells to thrive despite immune surveillance pressures. We explore the dynamic interactions within the tumor microenvironment (TME), where cancer cells not only accelerate their glucose and amino acid metabolism but also induce an immunosuppressive state that hampers effective immune response. Recent findings underscore the metabolic competition between tumor and immune cells, particularly focusing on how this interaction influences the efficacy of emerging immunotherapies. By integrating cutting-edge research on the metabolic pathways of cancer cells, such as the Warburg effect and glutamine addiction, we shed light on potential therapeutic targets. The review proposes that disrupting these metabolic pathways could enhance the response to immunotherapy, offering a dual-pronged strategy to combat tumor growth and immune evasion.


Subject(s)
Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Neoplasms/pathology , Tumor Microenvironment/immunology , Immunotherapy/methods , Animals , Warburg Effect, Oncologic , Glutamine/metabolism , Tumor Escape , Metabolic Networks and Pathways
10.
Medicina (Kaunas) ; 60(5)2024 May 03.
Article in English | MEDLINE | ID: mdl-38792944

ABSTRACT

Acute kidney injury represents a significant threat in cardiac surgery regarding complications and costs. Novel preventive approaches are needed, as the therapeutic modalities are still limited. As experimental studies have demonstrated, glutamine, a conditionally essential amino acid, might have a protective role in this setting. Moreover, the levels of glutamine after the cardiopulmonary bypass are significantly lower. In clinical practice, various trials have investigated the effects of glutamine supplementation on cardiac surgery with encouraging results. However, these studies are heterogeneous regarding the selection criteria, timing, dose, outcomes studied, and way of glutamine administration. This narrative review aims to present the potential role of glutamine in cardiac surgery-associated acute kidney injury prevention, starting from the experimental studies and guidelines to the clinical practice and future directions.


Subject(s)
Acute Kidney Injury , Cardiac Surgical Procedures , Dietary Supplements , Glutamine , Humans , Acute Kidney Injury/prevention & control , Acute Kidney Injury/etiology , Cardiac Surgical Procedures/adverse effects , Glutamine/administration & dosage , Postoperative Complications/prevention & control , Animals , Disease Models, Animal
11.
Front Immunol ; 15: 1369289, 2024.
Article in English | MEDLINE | ID: mdl-38756785

ABSTRACT

Background: This study aims to identify precise biomarkers for breast cancer to improve patient outcomes, addressing the limitations of traditional staging in predicting treatment responses. Methods: Our analysis encompassed data from over 7,000 breast cancer patients across 14 datasets, which included in-house clinical data and single-cell data from 8 patients (totaling 43,766 cells). We utilized an integrative approach, applying 10 machine learning algorithms in 54 unique combinations to analyze 100 existing breast cancer signatures. Immunohistochemistry assays were performed for empirical validation. The study also investigated potential immunotherapies and chemotherapies. Results: Our research identified five consistent glutamine metabolic reprogramming (GMR)-related genes from multi-center cohorts, forming the foundation of a novel GMR-model. This model demonstrated superior accuracy in predicting recurrence and mortality risks compared to existing clinical and molecular features. Patients classified as high-risk by the model exhibited poorer outcomes. IHC validation in 30 patients reinforced these findings, suggesting the model's broad applicability. Intriguingly, the model indicates a differential therapeutic response: low-risk patients may benefit more from immunotherapy, whereas high-risk patients showed sensitivity to specific chemotherapies like BI-2536 and ispinesib. Conclusions: The GMR-model marks a significant leap forward in breast cancer prognosis and the personalization of treatment strategies, offering vital insights for the effective management of diverse breast cancer patient populations.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Glutamine , Machine Learning , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Glutamine/metabolism , Biomarkers, Tumor/metabolism , Prognosis , Gene Expression Regulation, Neoplastic , Middle Aged , Transcriptome , Metabolic Reprogramming
12.
Commun Biol ; 7(1): 608, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769385

ABSTRACT

Diverse tumor metabolic phenotypes are influenced by the environment and genetic lesions. Whether these phenotypes extend to rhabdomyosarcoma (RMS) and how they might be leveraged to design new therapeutic approaches remains an open question. Thus, we utilized a Pax7Cre-ER-T2/+; NrasLSL-G12D/+; p53fl/fl (P7NP) murine model of sarcoma with mutations that most frequently occur in human embryonal RMS. To study metabolism, we infuse 13C-labeled glucose or glutamine into mice with sarcomas and show that sarcomas consume more glucose and glutamine than healthy muscle tissue. However, we reveal a marked shift from glucose consumption to glutamine metabolism after radiation therapy (RT). In addition, we show that inhibiting glutamine, either through genetic deletion of glutaminase (Gls1) or through pharmacological inhibition of glutaminase, leads to significant radiosensitization in vivo. This causes a significant increase in overall survival for mice with Gls1-deficient compared to Gls1-proficient sarcomas. Finally, Gls1-deficient sarcomas post-RT elevate levels of proteins involved in natural killer cell and interferon alpha/gamma responses, suggesting a possible role of innate immunity in the radiosensitization of Gls1-deficient sarcomas. Thus, our results indicate that glutamine contributes to radiation response in a mouse model of RMS.


Subject(s)
Glutaminase , Glutamine , Sarcoma , Animals , Glutamine/metabolism , Mice , Glutaminase/metabolism , Glutaminase/genetics , Glutaminase/antagonists & inhibitors , Sarcoma/metabolism , Sarcoma/radiotherapy , Sarcoma/genetics , Glucose/metabolism , Disease Models, Animal , Radiation Tolerance
13.
Discov Med ; 36(183): 836-841, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38665031

ABSTRACT

BACKGROUND: Over 80% of lung cancer cases constitute non-small cell lung cancer (NSCLC), making it the most prevalent type of lung cancer globally and the leading cause of cancer-related deaths. The treatment of NSCLC patients with gefitinib has demonstrated promising initial efficacy. However, the underlying mechanism remains unclear. This study aims to investigate how gefitinib affects the mitogen-activated protein kinase kinase (MEK)/extracellular regulated protein kinases (ERK) signaling pathway-mediated growth and death of NSCLC cells. METHODS: In this study, the NSCLC cell line A549 was cultured in vitro and divided into a control group and a gefitinib group. The viability of the A549 cells was assessed using the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. Flow cytometry was employed to detect apoptosis in A549 cells, and the expression of glutamate dehydrogenase (GDH1) mRNA in these cells was determined using real-time quantitative PCR (RT-PCR). Western blotting was utilized to evaluate the protein expression levels of key components in the MEK/ERK signaling pathway, including phospho-MEK1/2, MEK1/2, phospho-ERK1/2, and ERK1/2. Additionally, intracellular glutamine content in A549 cells was measured using a colorimetric method. RESULTS: In contrast to the control group, the proliferation of A549 cells, the transcription level of glutamate dehydrogenase (GDH1), the intracellular glutamine content, and the protein expression levels of phospho-MEK1/2 and phospho-ERK1/2 were significantly lower in the gefitinib group. Moreover, apoptosis markedly increased. CONCLUSIONS: Gefitinib expedites apoptosis and diminishes proliferation in the NSCLC cell line A549 by downregulating the epidermal growth factor receptor (EGFR)/MEK/ERK signaling pathway. This effect is accomplished by fostering the expression of GDH1 to augment glutaminolysis in A549 cells.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung , Gefitinib , Glutamine , Lung Neoplasms , MAP Kinase Signaling System , Humans , Gefitinib/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , A549 Cells , Glutamine/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Glutamate Dehydrogenase/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Line, Tumor
14.
Exp Mol Med ; 56(4): 1013-1026, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38684915

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent because it kills cancer cells while sparing normal cells. However, many cancers, including pancreatic ductal adenocarcinoma (PDAC), exhibit intrinsic or acquired resistance to TRAIL, and the molecular mechanisms underlying TRAIL resistance in cancers, particularly in PDAC, remain unclear. In this study, we demonstrated that glutamine (Gln) endows PDAC cells with resistance to TRAIL through KDM4C-mediated epigenetic regulation of cFLIP. Inhibition of glutaminolysis significantly reduced the cFLIP level, leading to TRAIL-mediated formation of death-inducing signaling complexes. Overexpression of cFLIP dramatically rescued PDAC cells from TRAIL/Gln deprivation-induced apoptosis. Alpha-Ketoglutarate (aKG) supplementation significantly reversed the decrease in the cFLIP level induced by glutaminolysis inhibition and rescued PDAC cells from TRAIL/Gln deprivation-induced apoptosis. Knockdown of glutamic-oxaloacetic transaminase 2, which facilitates the conversion of oxaloacetate and glutamate into aspartate and aKG, decreased aKG production and the cFLIP level and activated TRAIL-induced apoptosis. AKG-mediated epigenetic regulation was necessary for maintaining a high level of cFLIP. Glutaminolysis inhibition increased the abundance of H3K9me3 in the cFLIP promoter, indicating that Gln-derived aKG production is important for Jumonji-domain histone demethylase (JHDM)-mediated cFLIP regulation. The JHDM KDM4C regulated cFLIP expression by binding to its promoter, and KDM4C knockdown sensitized PDAC cells to TRAIL-induced apoptosis. The present findings suggest that Gln-derived aKG production is required for KDM4C-mediated epigenetic regulation of cFLIP, which leads to resistance to TRAIL.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , Drug Resistance, Neoplasm , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glutamine , Jumonji Domain-Containing Histone Demethylases , Pancreatic Neoplasms , TNF-Related Apoptosis-Inducing Ligand , Humans , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Glutamine/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Apoptosis/drug effects , Ketoglutaric Acids/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Aspartate Aminotransferase, Cytoplasmic/metabolism , Aspartate Aminotransferase, Cytoplasmic/genetics , Animals , Promoter Regions, Genetic
15.
Endocr Regul ; 58(1): 91-100, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38656254

ABSTRACT

Objective. Glucose and glutamine supply as well as serine synthesis and endoplasmic reticulum (ER) stress are important factors of glioblastoma growth. Previous studies showed that the knockdown of ERN1 (ER to nucleus signaling 1) suppressed glioblastoma cell proliferation and modified the sensitivity of numerous gene expressions to nutrient deprivations. The present study is aimed to investigate the impact of glucose and glutamine deprivations on the expression of serine synthesis genes in U87MG glioblastoma cells in relation to ERN1 knockdown with the intent to reveal the role of ERN1 signaling pathway on the ER stress-dependent regulation of these gene expressions. Clarification of the regulatory mechanisms of serine synthesis is a great significance for glioblastoma therapy. Methods. The control U87MG glioblastoma cells (transfected by empty vector) and ERN1 knockdown cells (transfected by dominant-negative ERN1) were exposed under glucose and glutamine deprivation conditions for 16 h. RNA was extracted from cells and reverse transcribed. The expression level of PHGDH (phosphoglycerate dehydrogenase), PSAT1 (phosphoserine amino-transferase 1), PSPH (phosphoserine phosphatase), ATF4 (activating transcription factor 4), and SHMT1 (serine hydroxymethyltransferase 1) genes was studied by real-time qPCR and normalized to ACTB. Results. It was found that the expression level of genes responsible for serine synthesis such as PHGDH, PSAT1, PSPH, and transcription factor ATF4 was up-regulated in U87MG glioblastoma cells under glucose and glutamine deprivations. Furthermore, inhibition of ERN1 significantly enhances the impact of glucose and especially glutamine deprivations on these gene expressions. At the same time, the expression of the SHMT1 gene, which is responsible for serine conversion to glycine, was down-regulated in both nutrient deprivation conditions with more significant changes in ERN1 knockdown glioblastoma cells. Conclusion. Taken together, the results of present study indicate that the expression of genes responsible for serine synthesis is sensitive to glucose and glutamine deprivations in gene-specific manner and that suppression of ERN1 signaling significantly modifies the impact of both glucose and glutamine deprivations on PHGDH, PSAT1, PSPH, ATF4, and SHMT1 gene expressions and reflects the ERN1-mediated genome reprograming introduced by nutrient deprivation condition.


Subject(s)
Endoribonucleases , Gene Expression Regulation, Neoplastic , Glioblastoma , Glucose , Glutamine , Phosphoglycerate Dehydrogenase , Phosphoric Monoester Hydrolases , Protein Serine-Threonine Kinases , Serine , Transaminases , Humans , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/metabolism , Glucose/metabolism , Glutamine/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Serine/biosynthesis , Signal Transduction
16.
Nutrients ; 16(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38674821

ABSTRACT

Understanding the nutritional content of protein supplements is crucial for optimal nutritional planning among athletes and other people. Distribution of macronutrients and aminograms in the main products available in the national Chilean market remains unknown. A descriptive cross-sectional study was conducted to identify the main protein supplements available in the Chilean market. Information on macronutrients and aminograms from the nutritional labels of each product was extracted. The analysis considered the content per portion and per 100 g. Cluster analysis models and graphical representations were explored. Eighty protein shakes were assessed in the Santiago de Chile market. The median protein dosage was 32 g (range from 25 to 52), and the median energy value stood at 390 kcal (range from 312 to 514). The median protein content per 100 g of product was found to be 75 g (range from 42.5 to 97.2). The combined median concentration of amino acids was 4749.75 mg. Among these, the essential amino acid L-Tryptophan exhibited the lowest concentration at 1591.50 mg, while the conditional amino acid L-Glutamine had the highest median concentration at 17,336 mg. There was a significant prevalence of animal-derived products, placing specific emphasis on protein supplements that feature elevated levels of the amino acids L-Glutamine and L-Leucine.


Subject(s)
Dietary Proteins , Dietary Supplements , Nutritive Value , Chile , Cross-Sectional Studies , Dietary Proteins/analysis , Humans , Amino Acids/analysis , Food Labeling , Tryptophan/analysis , Nutrients/analysis , Leucine/analysis , Energy Intake , Glutamine/analysis
17.
Nat Commun ; 15(1): 3534, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670989

ABSTRACT

Glutamine synthetase (GS) is vital in maintaining ammonia and glutamate (Glu) homeostasis in living organisms. However, the natural enzyme relies on adenosine triphosphate (ATP) to activate Glu, resulting in impaired GS function during ATP-deficient neurotoxic events. To date, no reports demonstrate using artificial nanostructures to mimic GS function. In this study, we synthesize aggregation-induced emission active polyP-Mn nanosheets (STPE-PMNSs) based on end-labeled polyphosphate (polyP), exhibiting remarkable GS-like activity independent of ATP presence. Further investigation reveals polyP in STPE-PMNSs serves as phosphate source to activate Glu at low ATP levels. This self-feeding mechanism offers a significant advantage in regulating Glu homeostasis at reduced ATP levels in nerve cells during excitotoxic conditions. STPE-PMNSs can effectively promote the conversion of Glu to glutamine (Gln) in excitatory neurotoxic human neuroblastoma cells (SH-SY5Y) and alleviate Glu-induced neurotoxicity. Additionally, the fluorescence signal of nanosheets enables precise monitoring of the subcellular distribution of STPE-PMNSs. More importantly, the intracellular fluorescence signal is enhanced in a conversion-responsive manner, allowing real-time tracking of reaction progression. This study presents a self-sustaining strategy to address GS functional impairment caused by ATP deficiency in nerve cells during neurotoxic events. Furthermore, it offers a fresh perspective on the potential biological applications of polyP-based nanostructures.


Subject(s)
Adenosine Triphosphate , Glutamate-Ammonia Ligase , Glutamic Acid , Glutamine , Manganese , Nanostructures , Neurons , Polyphosphates , Glutamate-Ammonia Ligase/metabolism , Humans , Polyphosphates/chemistry , Polyphosphates/metabolism , Polyphosphates/pharmacology , Nanostructures/chemistry , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Neurons/metabolism , Neurons/drug effects , Glutamine/metabolism , Manganese/metabolism , Manganese/chemistry , Biocompatible Materials/chemistry
18.
Mol Nutr Food Res ; 68(9): e2300704, 2024 May.
Article in English | MEDLINE | ID: mdl-38656560

ABSTRACT

SCOPE: This study investigates the potential of glutamine to mitigate intestinal mucositis and dysbiosis caused by the chemotherapeutic agent 5-fluorouracil (5-FU). METHODS AND RESULTS: Over twelve days, Institute of Cancer Research (ICR) mice are given low (0.5 mg kg-1) or high (2 mg kg-1) doses of L-Glutamine daily, with 5-FU (50 mg kg-1) administered between days six and nine. Mice receiving only 5-FU exhibited weight loss, diarrhea, abnormal cell growth, and colonic inflammation, correlated with decreased mucin proteins, increased endotoxins, reduced fecal short-chain fatty acids, and altered gut microbiota. Glutamine supplementation counteracted these effects by inhibiting the Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) pathway, modulating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) oxidative stress proteins, and increasing mammalian target of rapamycin (mTOR) levels, thereby enhancing microbial diversity and protecting intestinal mucosa. CONCLUSIONS: These findings underscore glutamine's potential in preventing 5-FU-induced mucositis by modulating gut microbiota and inflammation pathways.


Subject(s)
Fluorouracil , Gastrointestinal Microbiome , Glutamine , Intestinal Mucosa , Mucositis , Animals , Gastrointestinal Microbiome/drug effects , Fluorouracil/adverse effects , Glutamine/pharmacology , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice, Inbred ICR , Male , Toll-Like Receptor 4/metabolism , NF-E2-Related Factor 2/metabolism , Dysbiosis/chemically induced , Dysbiosis/drug therapy , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , TOR Serine-Threonine Kinases/metabolism , Antimetabolites, Antineoplastic/adverse effects , Heme Oxygenase-1/metabolism
19.
J Cancer Res Clin Oncol ; 150(4): 211, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662258

ABSTRACT

BACKGROUND: Circular ribose nucleic acids (circRNAs), an abundant type of noncoding RNAs, are widely expressed in eukaryotic cells and exert a significant impact on the initiation and progression of various disorders, including different types of cancer. However, the specific role of various circRNAs in colorectal cancer (CRC) pathology is still not fully understood. METHODS: The initial step involved the use of quantitative reverse transcription polymerase chain reaction (RT-qPCR) to assess the expression levels of circRNAs and messenger RNA (mRNA) in CRC cell lines and tissues. Subsequently, functional analyses of circCOL1A1 knockdown were conducted in vitro and in vivo through cell counting kit (CCK)-8, colony formation and transwell assays, as well as xenograft mouse model of tumor formation. Molecular expression and interactions were investigated using luciferase reporter assays, Western blot analysis, RNA immunoprecipitation (RIP), and immunohistochemical staining. RESULTS: The RT-qPCR results revealed elevated levels of circCOL1A1 expressions in CRC tissues and cell lines as compared to the normal counterparts. In addition, circCOL1A1 expression level was found to be correlated with TNM stage, lymph node metastases, distant metastases, and invasion. Knockdown of circCOL1A1 resulted in impaired invasion, migration, and proliferation of CRC cells, and suppressed tumor generation in the animal model. We further demonstrated that circCOL1A1 could act as a sponge for miR-214-3p, suppressing miR-214-3p activity and leading to the upregulation of GLS1 protein to promote glutamine metabolism. CONCLUSION: These findings suggest that circCOL1A1 functions as an oncogenic molecule to promote CRC progression via miR-214-3p/GLS1 axis, hinting on the potential of circCOL1A1 as a therapeutic target for CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colorectal Neoplasms , Glutaminase , Glutamine , MicroRNAs , Neoplasm Invasiveness , RNA, Circular , Up-Regulation , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , RNA, Circular/genetics , Xenograft Model Antitumor Assays
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167152, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582012

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a kind of hepatobiliary tumor that is increasing in incidence and mortality. The gut microbiota plays a role in the onset and progression of cancer, however, the specific mechanism by which the gut microbiota acts on ICC remains unclear. In this study, feces and plasma from healthy controls and ICC patients were collected for 16S rRNA sequencing or metabolomics analysis. Gut microbiota analysis showed that gut microbiota abundance and biodiversity were altered in ICC patients compared with controls. Plasma metabolism analysis showed that the metabolite glutamine content of the ICC patient was significantly higher than that of the controls. KEGG pathway analysis showed that glutamine plays a vital role in ICC. In addition, the use of antibiotics in ICC animals further confirmed that changes in gut microbiota affect changes in glutamine. Further experiments showed that supplementation with glutamine inhibited ferroptosis and downregulated ALK5 and NOX1 expression in HuCCT1 cells. ALK5 overexpression or NOX1 overexpression increased NOX1, p53, PTGS2, ACSL4, LPCAT3, ROS, MDA and Fe2+ and decreased FTH1, SLC7A11 and GSH. Knockdown of NOX1 suppressed FIN56-induced ferroptosis. In vivo, supplementation with glutamine promoted tumor growth. Overexpression of ALK5 repressed tumor growth and induced ferroptosis in nude mice, which could be reversed by the addition of glutamine. Our results suggested that the gut microbiota altered glutamine metabolism to inhibit ferroptosis in ICC by regulating the ALK5/NOX1 axis.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Ferroptosis , Gastrointestinal Microbiome , Glutamine , NADPH Oxidase 1 , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/microbiology , Cholangiocarcinoma/drug therapy , Ferroptosis/drug effects , Humans , Glutamine/metabolism , NADPH Oxidase 1/metabolism , NADPH Oxidase 1/genetics , Animals , Gastrointestinal Microbiome/drug effects , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/microbiology , Mice , Male , Cell Line, Tumor , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/genetics , Mice, Nude , Female , Middle Aged , Gene Expression Regulation, Neoplastic/drug effects , Receptor, Transforming Growth Factor-beta Type I
SELECTION OF CITATIONS
SEARCH DETAIL
...