Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.556
Filtrer
1.
CNS Neurosci Ther ; 30(8): e14913, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39123294

RÉSUMÉ

BACKGROUND: Hyperglycemia-induced neuroinflammation significantly contributes to diabetic neuropathic pain (DNP), but the underlying mechanisms remain unclear. OBJECTIVE: To investigate the role of Sirt3, a mitochondrial deacetylase, in hyperglycemia-induced neuroinflammation and DNP and to explore potential therapeutic interventions. METHOD AND RESULTS: Here, we found that Sirt3 was downregulated in spinal dorsal horn (SDH) of diabetic mice by RNA-sequencing, which was further confirmed at the mRNA and protein level. Sirt3 deficiency exacerbated hyperglycemia-induced neuroinflammation and DNP by enhancing microglial aerobic glycolysis in vivo and in vitro. Overexpression of Sirt3 in microglia alleviated inflammation by reducing aerobic glycolysis. Mechanistically, high-glucose stimulation activated Akt, which phosphorylates and inactivates FoxO1. The inactivation of FoxO1 diminished the transcription of Sirt3. Besides that, we also found that hyperglycemia induced Sirt3 degradation via the mitophagy-lysosomal pathway. Blocking Akt activation by GSK69093 or metformin rescued the degradation of Sirt3 protein and transcription inhibition of Sirt3 mRNA, which substantially diminished hyperglycemia-induced inflammation. Metformin in vivo treatment alleviated neuroinflammation and diabetic neuropathic pain by rescuing hyperglycemia-induced Sirt3 downregulation. CONCLUSION: Hyperglycemia induces metabolic reprogramming and inflammatory activation in microglia through the regulation of Sirt3 transcription and degradation. This novel mechanism identifies Sirt3 as a potential drug target for treating DNP.


Sujet(s)
Diabète expérimental , Neuropathies diabétiques , Régulation négative , Glycolyse , Hyperglycémie , Souris de lignée C57BL , Microglie , Sirtuine-3 , Animaux , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Souris , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/physiologie , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/physiologie , Hyperglycémie/métabolisme , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Mâle , Diabète expérimental/métabolisme , Diabète expérimental/complications , Neuropathies diabétiques/métabolisme , Inflammation/métabolisme , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/étiologie , Metformine/pharmacologie
2.
J Biochem Mol Toxicol ; 38(8): e23799, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132768

RÉSUMÉ

It is well established that pyruvate kinase M2 (PKM2) activity contributes to metabolic reprogramming in various cancers, including colorectal cancer (CRC). Estrogen or 17ß-estradiol (E2) signaling is also known to modulate glycolysis markers in cancer cells. However, whether the inhibition of PKM2 combined with E2 treatment could adversely affect glucose metabolism in CRC cells remains to be investigated. First, we confirmed the metabolic plasticity of CRC cells under varying environmental conditions. Next, we identified glycolysis markers that were upregulated in CRC patients and assessed in vitro mRNA levels following E2 treatment. We found that PKM2 expression, which is highly upregulated in CRC clinical samples, is not altered by E2 treatment in CRC cells. In this study, glucose uptake, generation of reactive oxygen species (ROS), lactate production, cell viability, and apoptosis were evaluated in CRC cells following E2 treatment, PKM2 silencing, or a combination of both. Compared to individual treatments, combination therapy resulted in a significant reduction in cell viability and enhanced apoptosis. Glucose uptake and ROS production were markedly reduced in PKM2-silenced E2-treated cells. The data presented here suggest that E2 signaling combined with PKM2 inhibition cumulatively targets glucose metabolism in a manner that negatively impacts CRC cell growth. These findings hold promise for novel therapeutic strategies targeting altered metabolic pathways in CRC.


Sujet(s)
Tumeurs colorectales , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Hormones thyroïdiennes/métabolisme , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Oestrogènes/pharmacologie , , Oestradiol/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Glucose/métabolisme , Protéines de transport/métabolisme , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , Pyruvate kinase/génétique , Glycolyse/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Femelle
3.
Sci Rep ; 14(1): 18862, 2024 08 14.
Article de Anglais | MEDLINE | ID: mdl-39143171

RÉSUMÉ

Cell adhesion to the extracellular matrix and its natural outcome of cell spreading, along with the maintenance of barrier activity, are essential behaviors of epithelial cells, including retinal pigment epithelium (RPE). Disruptions in these characteristics can result in severe vision-threatening diseases such as diabetic macular edema and age-related macular degeneration. However, the precise mechanisms underlying how RPE cells regulate their barrier integrity and cell spreading are not fully understood. This study aims to elucidate the relative importance of upper glycolytic components in governing these cellular behaviors of RPE cells. Electric Cell-Substrate Impedance Sensing (ECIS) technology was utilized to assess in real-time the effects of targeting various upper glycolytic enzymes on RPE barrier function and cell spreading by measuring cell resistance and capacitance, respectively. Specific inhibitors used included WZB117 for Glut1 inhibition, Lonidamine for Hexokinase inhibition, PFK158 for PFKFB3/PFK axis inhibition, and TDZD-8 for Aldolase inhibition. Additionally, the viability of RPE cells was evaluated using a lactate dehydrogenase (LDH) cytotoxicity assay. The most significant decrease in electrical resistance and increase in capacitance of RPE cells were observed due to dose-dependent inhibition of Glut1 using WZB117, as well as Aldolase inhibition with TDZD-8. LDH level analysis at 24-72 h post-treatment with WZB117 (1 and 10 µM) or TDZD-8 (1 µM) showed no significant difference compared to the control, indicating that the disruption of RPE functionality was not attributed to cell death. Lastly, inhibition of other upper glycolytic components, including PFKFB3/PFK with PFK158 or Hexokinase with Lonidamine, did not significantly affect RPE cell behavior. This study provides insights into the varied roles of upper glycolytic components in regulating the functionality of RPE cells. Specifically, it highlights the critical roles of Glut1 and Aldolase in preserving barrier integrity and promoting RPE cell adhesion and spreading. Such understanding will guide the development of safe interventions to treat RPE cell dysfunction in various retinal disorders.


Sujet(s)
Glycolyse , Épithélium pigmentaire de la rétine , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/cytologie , Glycolyse/effets des médicaments et des substances chimiques , Humains , Transporteur de glucose de type 1/métabolisme , Hexokinase/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Adhérence cellulaire/effets des médicaments et des substances chimiques , Impédance électrique , Phosphofructokinase-2/métabolisme , Phosphofructokinase-2/antagonistes et inhibiteurs
4.
Sci Rep ; 14(1): 19003, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152152

RÉSUMÉ

Gastric cancer (GC) remains a global disease with a high mortality rate, the lack of effective treatments and the high toxicity of side effects are primary causes for its poor prognosis. Hence, urgent efforts are needed to find safe and effective therapeutic strategies. Gypenoside (Gyp) is a widely used natural product that regulates blood glucose to improve disease progression with few toxic side effects. Given the crucial role of abnormal glycometabolism in driving tumor malignancy, it is important to explore the association between Gyp and glycometabolism in GC and understand the mechanism of action by which Gyp influences glycometabolism. In this study, we demonstrated that Gyp suppresses GC proliferation and migration both in vitro and in vivo. We identified that Gyp suppresses the malignant progression of GC by inhibiting glycolysis using network pharmacology and metabolomics. Transcriptome analysis revealed that the Hippo pathway is a key regulator of glycolysis by Gyp in GC. Furthermore, Gyp induced upregulation of LATS1/2 proteins, leading to increased YAP phosphorylation and decreased TAZ protein expression. The YAP agonist XMU-MP-1 rescued the inhibitory effect of Gyp on GC proliferation by reversing glycolysis. These findings confirmed that Gyp inhibits GC proliferation by targeting glycolysis through the Hippo pathway. Our study examined the role of Gyp in the malignant progression of GC, explored its therapeutic prospects, elucidated a mechanism by which Gyp suppresses GC proliferation through interference with the glycolytic process, thus providing a potential novel therapeutic strategy for GC patients.


Sujet(s)
Prolifération cellulaire , Glycolyse , Gynostemma , Voie de signalisation Hippo , Protein-Serine-Threonine Kinases , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Lignée cellulaire tumorale , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Mouvement cellulaire/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
5.
Sci Rep ; 14(1): 18252, 2024 08 06.
Article de Anglais | MEDLINE | ID: mdl-39107469

RÉSUMÉ

Brown fat is a therapeutic target for the treatment of obesity-associated metabolic diseases. However, nutritional intervention strategies for increasing the mass and activity of human brown adipocytes have not yet been established. To identify vitamins required for brown adipogenesis and adipocyte browning, chemical compound-induced brown adipocytes (ciBAs) were converted from human dermal fibroblasts under serum-free and vitamin-free conditions. Choline was found to be essential for adipogenesis. Additional treatment with pantothenic acid (PA) provided choline-induced immature adipocytes with browning properties and metabolic maturation, including uncoupling protein 1 (UCP1) expression, lipolysis, and mitochondrial respiration. However, treatment with high PA concentrations attenuated these effects along with decreased glycolysis. Transcriptome analysis showed that a low PA concentration activated metabolic genes, including the futile creatine cycle-related thermogenic genes, which was reversed by a high PA concentration. Riboflavin treatment suppressed thermogenic gene expression and increased lipolysis, implying a metabolic pathway different from that of PA. Thiamine treatment slightly activated thermogenic genes along with decreased glycolysis. In summary, our results suggest that specific B vitamins and choline are uniquely involved in the regulation of adipocyte browning via cellular energy metabolism in a concentration-dependent manner.


Sujet(s)
Adipocytes bruns , Choline , Acide pantothénique , Riboflavine , Thiamine , Humains , Riboflavine/pharmacologie , Acide pantothénique/pharmacologie , Acide pantothénique/métabolisme , Adipocytes bruns/métabolisme , Adipocytes bruns/effets des médicaments et des substances chimiques , Thiamine/pharmacologie , Thiamine/métabolisme , Choline/métabolisme , Choline/pharmacologie , Protéine-1 de découplage/métabolisme , Protéine-1 de découplage/génétique , Lipolyse/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Thermogenèse/effets des médicaments et des substances chimiques , Adipogenèse/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Cellules cultivées , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
6.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125904

RÉSUMÉ

α-glucosidase, a pharmacological target for type 2 diabetes mellitus (T2DM), is present in the intestinal brush border membrane and catalyzes the hydrolysis of sugar linkages during carbohydrate digestion. Since α-glucosidase inhibitors (AGIs) modulate intestinal metabolism, they may influence oxidative stress and glycolysis inhibition, potentially addressing intestinal dysfunction associated with T2DM. Herein, we report on a study of an ortho-carbonyl substituted hydroquinone series, whose members differ only in the number and position of methyl groups on a common scaffold, on radical-scavenging activities (ORAC assay) and correlate them with some parameters obtained by density functional theory (DFT) analysis. These compounds' effect on enzymatic activity, their molecular modeling on α-glucosidase, and their impact on the mitochondrial respiration and glycolysis of the intestinal Caco-2 cell line were evaluated. Three groups of compounds, according their effects on the Caco-2 cells metabolism, were characterized: group A (compounds 2, 3, 5, 8, 9, and 10) reduces the glycolysis, group B (compounds 1 and 6) reduces the basal mitochondrial oxygen consumption rate (OCR) and increases the extracellular acidification rate (ECAR), suggesting that it induces a metabolic remodeling toward glycolysis, and group C (compounds 4 and 7) increases the glycolysis lacking effect on OCR. Compounds 5 and 10 were more potent as α-glucosidase inhibitors (AGIs) than acarbose, a well-known AGI with clinical use. Moreover, compound 5 was an OCR/ECAR inhibitor, and compound 10 was a dual agent, increasing the proton leak-driven OCR and inhibiting the maximal electron transport flux. Additionally, menadione-induced ROS production was prevented by compound 5 in Caco-2 cells. These results reveal that slight structural variations in a hydroquinone scaffold led to diverse antioxidant capability, α-glucosidase inhibition, and the regulation of mitochondrial bioenergetics in Caco-2 cells, which may be useful in the design of new drugs for T2DM and metabolic syndrome.


Sujet(s)
Antioxydants , Métabolisme énergétique , Inhibiteurs des glycoside hydrolases , Hydroquinones , alpha-Glucosidase , Humains , Cellules Caco-2 , alpha-Glucosidase/métabolisme , Inhibiteurs des glycoside hydrolases/pharmacologie , Inhibiteurs des glycoside hydrolases/composition chimique , Antioxydants/pharmacologie , Antioxydants/composition chimique , Antioxydants/métabolisme , Hydroquinones/pharmacologie , Hydroquinones/composition chimique , Métabolisme énergétique/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
7.
Signal Transduct Target Ther ; 9(1): 216, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39143065

RÉSUMÉ

Third-generation EGFR tyrosine kinase inhibitors (TKIs), exemplified by osimertinib, have demonstrated promising clinical efficacy in the treatment of non-small cell lung cancer (NSCLC). Our previous work has identified ASK120067 as a novel third-generation EGFR TKI with remarkable antitumor effects that has undergone New Drug Application (NDA) submission in China. Despite substantial progress, acquired resistance to EGFR-TKIs remains a significant challenge, impeding the long-term effectiveness of therapeutic approaches. In this study, we conducted a comprehensive investigation utilizing high-throughput proteomics analysis on established TKI-resistant tumor models, and found a notable upregulation of branched-chain amino acid transaminase 1 (BCAT1) expression in both osimertinib- and ASK120067-resistant tumors compared with the parental TKI-sensitive NSCLC tumors. Genetic depletion or pharmacological inhibition of BCAT1 impaired the growth of resistant cells and partially re-sensitized tumor cells to EGFR TKIs. Mechanistically, upregulated BCAT1 in resistant cells reprogrammed branched-chain amino acid (BCAA) metabolism and promoted alpha ketoglutarate (α-KG)-dependent demethylation of lysine 27 on histone H3 (H3K27) and subsequent transcriptional derepression of glycolysis-related genes, thereby enhancing glycolysis and promoting tumor progression. Moreover, we identified WQQ-345 as a novel BCAT1 inhibitor exhibiting antitumor activity both in vitro and in vivo against TKI-resistant lung cancer with high BCAT1 expression. In summary, our study highlighted the crucial role of BCAT1 in mediating resistance to third-generation EGFR-TKIs through epigenetic activation of glycolysis in NSCLC, thereby supporting BCAT1 as a promising therapeutic target for the treatment of TKI-resistant NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Récepteurs ErbB , Glycolyse , Tumeurs du poumon , Inhibiteurs de protéines kinases , Transaminases , Humains , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transaminases/génétique , Transaminases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Souris , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Acrylamides/pharmacologie , Animaux , Dérivés de l'aniline/pharmacologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Indoles , Pyrimidines
8.
Int J Nanomedicine ; 19: 7831-7850, 2024.
Article de Anglais | MEDLINE | ID: mdl-39105099

RÉSUMÉ

Purpose: Compared with traditional photothermal therapy (PTT, >50°C), mild PTT (≤45°C) is a promising strategy for tumor therapy with fewer adverse effects. Unfortunately, its anti-tumor efficacy is hampered by thermoresistance induced by overexpression of heat shock proteins (HSPs). In our previous study, we found bufalin (BU) is a glycolysis inhibitor that depletes HSPs, which is expected to overcome thermotolerance of tumor cells. In this study, BU-loaded multifunctional nanoparticles (NPs) were developed for enhancing the mild PTT of colorectal cancer (CRC). Methods: Fe3O4 NPs coated with the polydopamine (PDA) shell modified with polyethylene glycol (PEG) and cyclic arginine-glycyl-aspartic peptide (cRGD) for loading BU (Fe3O4@PDA-PEG-cRGD/BU NPs) were developed. The thermal variations in Fe3O4@PDA-PEG-cRGD/BU NPs solution under different conditions were measured. Glycolysis inhibition was evaluated by measuring the glucose uptake, extracellular lactate, and intracellular adenosine triphosphate (ATP) levels. The cellular cytotoxicity of Fe3O4@PDA-PEG-cRGD/BU NPs was analyzed using a cell counting kit-8 assay, Calcein-AM/PI double staining, and flow cytometry in HCT116 cells. The magnetic resonance imaging (MRI) performance and anti-tumor therapeutic efficacy of Fe3O4@PDA-PEG-cRGD/BU NPs were evaluated in HCT116-tumor bearing mice. Results: Fe3O4@PDA-PEG-cRGD/BU NPs had an average diameter of 260.4±3.5 nm, the zeta potential of -23.8±1.6 mV, the drug loading rate of 1.1%, which had good thermal stability, photothermal conversion efficiencies and MRI performance. In addition, the released BU not only killed tumor cells but also interfered with glycolysis by targeting the steroid receptor coactivator 3 (SRC-3)/HIF-1α pathway, preventing intracellular ATP synthesis, and combating HSP-dependent tumor thermoresistance, ultimately strengthening the thermal sensitivity toward mild PTT both in vitro and in vivo. Conclusion: This study provides a highly effective strategy for enhancing the therapeutic effects of mild PTT toward tumors.


Sujet(s)
Bufanolide , Tumeurs colorectales , Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Thérapie photothermique , Animaux , Bufanolide/pharmacologie , Bufanolide/composition chimique , Bufanolide/pharmacocinétique , Humains , Glycolyse/effets des médicaments et des substances chimiques , Tumeurs colorectales/thérapie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/traitement médicamenteux , Thérapie photothermique/méthodes , Souris , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Indoles/composition chimique , Indoles/pharmacologie , Polyéthylène glycols/composition chimique , Polymères/composition chimique , Souris de lignée BALB C , Lignée cellulaire tumorale , Souris nude , Cellules HCT116 , Nanoparticules de magnétite/composition chimique , Nanoparticules/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Arch Microbiol ; 206(9): 370, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115561

RÉSUMÉ

Staphylococcus xylosus has emerged as a bovine mastitis pathogen with increasing drug resistance, resulting in substantial economic impacts. This study utilized iTRAQ analysis to investigate the mechanisms driving resistance evolution in S. xylosus under ceftiofur sodium stress. Findings revealed notable variations in the expression of 143 proteins, particularly glycolysis-related proteins (TpiA, Eno, GlpD, Ldh) and peptidoglycan (PG) hydrolase Atl. Following the induction of ceftiofur sodium resistance in S. xylosus, the emergence of resistant strains displaying characteristics of small colony variants (SCVs) was observed. The transcript levels of TpiA, Eno, GlpD and Ldh were up-regulated, TCA cycle proteins (ICDH, MDH) and Atl were down-regulated, lactate content was increased, and NADH concentration was decreased in SCV compared to the wild strain. That indicates a potential role of carbon metabolism, specifically PG hydrolysis, glycolysis, and the TCA cycle, in the development of resistance to ceftiofur sodium in S. xylosus.


Sujet(s)
Antibactériens , Carbone , Céphalosporines , Résistance bactérienne aux médicaments , Staphylococcus , Céphalosporines/pharmacologie , Céphalosporines/métabolisme , Antibactériens/pharmacologie , Staphylococcus/effets des médicaments et des substances chimiques , Staphylococcus/génétique , Staphylococcus/métabolisme , Carbone/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Animaux , Bovins , Glycolyse/effets des médicaments et des substances chimiques , Cycle citrique , Mammite bovine/microbiologie , Infections à staphylocoques/microbiologie , Tests de sensibilité microbienne , Femelle
10.
Cells ; 13(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39120291

RÉSUMÉ

A substantial challenge in human brain aging is to find a suitable model to mimic neuronal aging in vitro as accurately as possible. Using directly converted neurons (iNs) from human fibroblasts is considered a promising tool in human aging since it retains the aging-associated mitochondrial donor signature. Still, using iNs from aged donors can pose certain restrictions due to their lower reprogramming and conversion efficacy than those from younger individuals. To overcome these limitations, our study aimed to establish an in vitro neuronal aging model mirroring features of in vivo aging by acute exposure on young iNs to either human stress hormone cortisol or the mitochondrial stressor rotenone, considering stress as a trigger of in vivo aging. The impact of rotenone was evident in mitochondrial bioenergetic properties by showing aging-associated deficits in mitochondrial respiration, cellular ATP, and MMP and a rise in glycolysis, mitochondrial superoxide, and mitochondrial ROS; meanwhile, cortisol only partially induced an aging-associated mitochondrial dysfunction. To replicate the in vivo aging-associated mitochondrial dysfunctions, using rotenone, a mitochondrial complex I inhibitor, proved to be superior to the cortisol model. This work is the first to use stress on young iNs to recreate aging-related mitochondrial impairments.


Sujet(s)
Mitochondries , Neurones , Roténone , Humains , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Roténone/pharmacologie , Vieillissement , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Hydrocortisone/métabolisme , Espèces réactives de l'oxygène/métabolisme , Donneurs de tissus , Glycolyse/effets des médicaments et des substances chimiques , Adénosine triphosphate/métabolisme
11.
BMC Cardiovasc Disord ; 24(1): 381, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044140

RÉSUMÉ

BACKGROUND: Metabolic abnormalities and immune inflammation are deeply involved in pulmonary vascular remodelling and the development of pulmonary hypertension (PH). However, the regulatory mechanisms of glycolysis in macrophages are still elusive. Cumulative evidence indicates that ß-catenin plays a crucial role in metabolic reprogramming. This study aimed to investigate the effect of ß-catenin on macrophage glycolysis in PH. METHODS: LPS-induced BMDMs were generated via in vitro experiments. A monocrotaline (MCT)-induced PH rat model was established, and the ß-catenin inhibitor XAV939 was administered in vivo. The role of ß-catenin in glycolysis was analysed. The degree of pulmonary vascular remodelling was measured. RESULTS: ß-catenin was significantly increased in both in vitro and in vivo models. In LPS-induced BMDMs, ß-catenin increased the levels of hexokinase 2 (HK2), phosphofructokinase (PFK), M2-pyruvate kinase (PKM2), lactate dehydrogenase (LDH), and lactate (LA) and the expression of inflammatory cytokines and promoted PASMC proliferation and migration in vitro. XAV939 decreased the level of glycolysis and downregulated the expression of inflammatory cytokines in vivo. MCT promoted pulmonary arterial structural remodelling and right ventricular hypertrophy, and XAV939 alleviated these changes. CONCLUSIONS: Our findings suggest that ß-catenin is involved in the development of PH by promoting glycolysis and the inflammatory response in macrophages. Inhibition of ß-catenin could improve the progression of PH.


Sujet(s)
Modèles animaux de maladie humaine , Glycolyse , Hypertension pulmonaire , Macrophages , Monocrotaline , Artère pulmonaire , Rat Sprague-Dawley , Remodelage vasculaire , bêta-Caténine , Animaux , Glycolyse/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme , Hypertension pulmonaire/induit chimiquement , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/physiopathologie , Mâle , Remodelage vasculaire/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Artère pulmonaire/métabolisme , Artère pulmonaire/effets des médicaments et des substances chimiques , Artère pulmonaire/physiopathologie , Artère pulmonaire/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Transduction du signal , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/physiopathologie , Hypertrophie ventriculaire droite/induit chimiquement , Médiateurs de l'inflammation/métabolisme , Rats , Mouvement cellulaire/effets des médicaments et des substances chimiques
12.
Theranostics ; 14(9): 3509-3525, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948065

RÉSUMÉ

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


Sujet(s)
Néovascularisation choroïdienne , Cellules endothéliales , Glycolyse , Animaux , Glycolyse/effets des médicaments et des substances chimiques , Souris , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Néovascularisation choroïdienne/traitement médicamenteux , Néovascularisation choroïdienne/métabolisme , Humains , Protéine-1 de liaison à la boîte Y/métabolisme , Protéine-1 de liaison à la boîte Y/génétique , Inhibiteurs de l'angiogenèse/pharmacologie , Hexokinase/métabolisme , Hexokinase/génétique , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Rétinopathie diabétique/traitement médicamenteux , Rétinopathie diabétique/métabolisme , Rétinopathie diabétique/génétique , Cellules endothéliales de la veine ombilicale humaine , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme
13.
Cell Rep ; 43(7): 114488, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39002124

RÉSUMÉ

Neuroinflammation is a prominent feature of Alzheimer's disease (AD). Activated microglia undergo a reprogramming of cellular metabolism necessary to power their cellular activities during disease. Thus, selective targeting of microglial immunometabolism might be of therapeutic benefit for treating AD. In the AD brain, the levels of microglial hexokinase 2 (HK2), an enzyme that supports inflammatory responses by promoting glycolysis, are significantly increased. In addition, HK2 displays non-metabolic activities that extend its inflammatory role beyond glycolysis. The antagonism of HK2 affects microglial phenotypes and disease progression in a gene-dose-dependent manner. HK2 complete loss fails to improve pathology by exacerbating inflammation, while its haploinsufficiency reduces pathology in 5xFAD mice. We propose that the partial antagonism of HK2 is effective in slowing disease progression by modulating NF-κB signaling through its cytosolic target, IKBα. The complete loss of HK2 affects additional inflammatory mechanisms related to mitochondrial dysfunction.


Sujet(s)
Maladie d'Alzheimer , Évolution de la maladie , Hexokinase , Microglie , Hexokinase/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Animaux , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/anatomopathologie , Souris , Humains , Facteur de transcription NF-kappa B/métabolisme , Souris transgéniques , Transduction du signal , Inhibiteur alpha de NF-KappaB/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Inflammation/anatomopathologie , Inflammation/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Dosage génique
14.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38985581

RÉSUMÉ

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Sujet(s)
Cardiotoxicité , Doxorubicine , Métabolisme énergétique , Acides gras , Glucose , Oxydoréduction , Animaux , Doxorubicine/toxicité , Glucose/métabolisme , Acides gras/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Cycle citrique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cardiopathies/induit chimiquement , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/prévention et contrôle , Cardiopathies/physiopathologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/anatomopathologie , Mitochondries du myocarde/enzymologie , Souris knockout , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Transporteur de glucose de type 4/métabolisme , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables
15.
Sci Rep ; 14(1): 16561, 2024 07 17.
Article de Anglais | MEDLINE | ID: mdl-39020066

RÉSUMÉ

Characteristic volatile organic compounds (VOCs) are anticipated to be used for the identification of lung cancer cells. However, to date, consistent biomarkers of VOCs in lung cancer cells have not been obtained through direct comparison between cancer and healthy groups. In this study, we regulated the glycolysis, a common metabolic process in cancer cells, and employed solid phase microextraction gas chromatography mass spectrometry (SPME-GC-MS) combined with untargeted analysis to identify the characteristic VOCs shared by cancer cells. The VOCs released by three types of lung cancer cells (A549, PC-9, NCI-H460) and one normal lung epithelial cell (BEAS-2B) were detected using SPME-GC-MS, both in their resting state and after treatment with glycolysis inhibitors (2-Deoxy-D-glucose, 2-DG/3-Bromopyruvic acid, 3-BrPA). Untargeted analysis methods were employed to compare the VOC profiles between each type of cancer cell and normal cells before and after glycolysis regulation. Our findings revealed that compared to normal cells, the three types of lung cancer cells exhibited three common differential VOCs in their resting state: ethyl propionate, acetoin, and 3-decen-5-one. Furthermore, under glycolysis control, a single common differential VOC-acetoin was identified. Notably, acetoin levels increased by 2.60-3.29-fold in all three lung cancer cell lines upon the application of glycolysis inhibitors while remaining relatively stable in normal cells. To further elucidate the formation mechanism of acetoin, we investigated its production by blocking glutaminolysis. This interdisciplinary approach combining metabolic biochemistry with MS analysis through interventional synthetic VOCs holds great potential for revolutionizing the identification of lung cancer cells and paving the way for novel cytological examination techniques.


Sujet(s)
Chromatographie gazeuse-spectrométrie de masse , Glycolyse , Tumeurs du poumon , Composés organiques volatils , Humains , Composés organiques volatils/métabolisme , Composés organiques volatils/analyse , Glycolyse/effets des médicaments et des substances chimiques , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Lignée cellulaire tumorale , Cellules A549 , Microextraction en phase solide
16.
J Proteome Res ; 23(8): 3682-3695, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39037832

RÉSUMÉ

Dental caries is a chronic oral infectious disease, and Streptococcus mutans (S. mutans) plays an important role in the formation of dental caries. Trans-cinnamaldehyde (CA) exhibits broad-spectrum antibacterial activity; however, its target and mechanism of action of CA on S. mutans needs to be further explored. In this study, it was verified that CA could inhibit the growth and biofilm formation of S. mutans. Further proteomic analysis identified 33, 55, and 78 differentially expressed proteins (DEPs) in S. mutans treated with CA for 1, 2, and 4 h, respectively. Bioinformatics analysis showed that CA interfered with carbohydrate metabolism, glycolysis, pyruvate metabolism, and the TCA cycle, as well as amino acid metabolism of S. mutans. Protein interactions suggested that pyruvate dehydrogenase (PDH) plays an important role in the antibacterial effect of CA. Moreover, the upstream and downstream pathways related to PDH were verified by various assays, and the results proved that CA not only suppressed the glucose and sucrose consumption and inhibited glucosyltransferase (GTF) and lactate dehydrogenase (LDH) activities but also decreased the ATP production. Interestingly, the protein interaction, qRT-PCR, and molecular docking analysis showed that PDH might be the target of CA to fight S. mutans. In summary, the study shows that CA interferes with the carbohydrate metabolism of bacteria by inhibiting glycolysis and the tricarboxylic acid (TCA) cycle via binding to PDH, which verifies that PDH is a potential target for the development of new drugs against S. mutans.


Sujet(s)
Acroléine , Métabolisme glucidique , Simulation de docking moléculaire , Complexe du pyruvate déshydrogénase , Streptococcus mutans , Streptococcus mutans/effets des médicaments et des substances chimiques , Streptococcus mutans/génétique , Streptococcus mutans/enzymologie , Acroléine/pharmacologie , Acroléine/analogues et dérivés , Acroléine/métabolisme , Métabolisme glucidique/effets des médicaments et des substances chimiques , Complexe du pyruvate déshydrogénase/métabolisme , Complexe du pyruvate déshydrogénase/antagonistes et inhibiteurs , Antibactériens/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Biofilms/effets des médicaments et des substances chimiques , Biofilms/croissance et développement , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/antagonistes et inhibiteurs , Protéomique/méthodes , Caries dentaires/microbiologie , Cycle citrique/effets des médicaments et des substances chimiques , Adénosine triphosphate/métabolisme
17.
Respir Res ; 25(1): 291, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080660

RÉSUMÉ

Acute lung injury (ALI) is characterized by an unregulated inflammatory reaction, often leading to severe morbidity and ultimately death. Excessive inflammation caused by M1 macrophage polarization and pyroptosis has been revealed to have a critical role in ALI. Recent study suggests that glycolytic reprogramming is important in the regulation of macrophage polarization and pyroptosis. However, the particular processes underlying ALI have yet to be identified. In this study, we established a Lipopolysaccharide(LPS)-induced ALI model and demonstrated that blocking glycolysis by using 2-Deoxy-D-glucose(2-DG) significantly downregulated the expression of M1 macrophage markers and pyroptosis-related genes, which was consistent with the in vitro results. Furthermore, our research has revealed that Phosphoglycerate Kinase 1(PGK1), an essential enzyme in the glycolysis pathway, interacts with NOD-, LRR- and pyrin domain-containing protein 3(NLRP3). We discovered that LPS stimulation improves the combination of PGK1 and NLRP3 both in vivo and in vitro. Interestingly, the absence of PGK1 reduces the phosphorylation level of NLRP3. Based on in vitro studies with mice bone marrow-derived macrophages (BMDMs), we further confirmed that siPGK1 plays a protective role by inhibiting macrophage pyroptosis and M1 macrophage polarization. The PGK1 inhibitor NG52 suppresses the occurrence of excessive inflammation in ALI. In general, it is plausible to consider a therapeutic strategy that focuses on modulating the relationship between PGK1 and NLRP3 as a means to mitigate the activation of inflammatory macrophages in ALI.


Sujet(s)
Lésion pulmonaire aigüe , Macrophages , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Phosphoglycerate kinase , Pyroptose , Pyroptose/physiologie , Pyroptose/effets des médicaments et des substances chimiques , Animaux , Phosphoglycerate kinase/métabolisme , Phosphoglycerate kinase/génétique , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/enzymologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Souris , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/enzymologie , Glycolyse/physiologie , Glycolyse/effets des médicaments et des substances chimiques , Mâle , Lipopolysaccharides/toxicité , Souris knockout , Cellules cultivées
18.
Chem Biol Interact ; 399: 111141, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-38992767

RÉSUMÉ

Mitochondrial Pyruvate Carrier 1 (MPC1) is localized on mitochondrial outer membrane to mediate the transport of pyruvate from cytosol to mitochondria. It is also well known to act as a tumor suppressor. Hexavalent chromium (Cr (VI)) contamination poses a global challenge due to its high toxicity and carcinogenesis. This research was intended to probe the potential mechanism of MPC1 in the effect of Cr (VI)-induced carcinogenesis. First, Cr (VI)-treatments decreased the expression of MPC1 in vitro and in vivo. Overexpression of MPC1 inhibited Cr (VI)-induced glycolysis and migration in A549 cells. Then, high mobility group A2 (HMGA2) protein strongly suppressed the transcription of MPC1 by binding to its promoter, and HMGA2/MPC1 axis played an important role in oxidative phosphorylation (OXPHOS), glycolysis and cell migration. Furthermore, endoplasmic reticulum (ER) stress made a great effect on the interaction between HMGA2 and MPC1. Finally, the mammalian target of the rapamycin (mTOR) was determined to mediate MPC1-regulated OXPHOS, aerobic glycolysis and cell migration. Collectively, our data revealed a novel HMGA2/MPC-1/mTOR signaling pathway to promote cell growth via facilitating the metabolism reprogramming from OXPHOS to aerobic glycolysis, which might be a potential therapy for cancers.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Chrome , Glycolyse , Protéine HMGA2 , Transporteurs d'acides monocarboxyliques , Transduction du signal , Sérine-thréonine kinases TOR , Humains , Sérine-thréonine kinases TOR/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Protéine HMGA2/métabolisme , Protéine HMGA2/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Chrome/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Cellules A549 , Souris , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Souris nude , Protéines de transport membranaire/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Lignée cellulaire tumorale , Protéines de transport de la membrane mitochondriale
19.
Brain Res Bull ; 215: 111032, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39029715

RÉSUMÉ

BACKGROUND: Buyang Huanwu Decoction (BHD) is widely used in Chinese clinical practice for the treatment and prevention of ischemic cerebral vascular diseases. This study was designed to investigate the effects of BHD on ischemic stroke (IS) and its underlying mechanism. METHODS: The middle cerebral artery occlusion (MCAO) rat model and oxygen-glucose deprivation and reoxygenation (OGD/R) rat brain microvascular endothelial cell (RBMVEC) models were established. Brain infarction size and neurological score were calculated following MCAO surgery. Evans blue was used to measure blood brain barrier (BBB) permeability. Cell counting kit-8 (CCK-8) and TUNEL assays were performed to evaluate the cell viability and apoptosis of RBMVECs. Dual-luciferase reporter assay was used to analyze the transcriptional activities of apoptosis-related genes. RESULTS: Results showed that higher infarction volume, neurological scores, and BBB permeability in the MCAO group rats were reduced after BHD treatment. Drug serum (DS) treatment had no impact on the normal RBMVECs' cell viability and cell apoptosis. Besides, DS treatment decreased the lactate production, glucose uptake, and extracellular acidification rate in normal and OGD/R-induced RBMVECs. DS treatment downregulated the protein levels of pan-lysine lactylation (kla), histone H3 lysine 18 lactylation (H3K18la), and the transcriptional of apoptotic protease activating factor-1 (Apaf-1) in OGD/R-treated RBMVECs. In addition, Apaf-1 overexpression decreased cell viability and increased apoptosis and glycolysis activity of OGD/R-treated RBMVECs. CONCLUSION: In summary, BHD inhibited glycolysis and apoptosis via suppressing the pan-kla and H3K18la protein levels and the Apaf-1 transcriptional activity, thus restraining the progression of IS.


Sujet(s)
Apoptose , Médicaments issus de plantes chinoises , Cellules endothéliales , Glycolyse , Accident vasculaire cérébral ischémique , Rat Sprague-Dawley , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Rats , Mâle , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Glucose/métabolisme , Microvaisseaux/effets des médicaments et des substances chimiques , Microvaisseaux/métabolisme , Modèles animaux de maladie humaine
20.
J Transl Med ; 22(1): 672, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033271

RÉSUMÉ

BACKGROUND: T cells play a pivotal role in chemotherapy-triggered anti-tumor effects. Emerging evidence underscores the link between impaired anti-tumor immune responses and resistance to paclitaxel therapy in triple-negative breast cancer (TNBC). Tumor-related endothelial cells (ECs) have potential immunoregulatory activity. However, how ECs regulate T cell activity during TNBC chemotherapy remains poorly understood. METHODS: Single-cell analysis of ECs in patients with TNBC receiving paclitaxel therapy was performed using an accessible single-cell RNA sequencing (scRNA-seq) dataset to identify key EC subtypes and their immune characteristics. An integrated analysis of a tumor-bearing mouse model, immunofluorescence, and a spatial transcriptome dataset revealed the spatial relationship between ECs, especially Tumor necrosis factor receptor (TNFR) 2+ ECs, and CD8+ T cells. RNA sequencing, CD8+ T cell proliferation assays, flow cytometry, and bioinformatic analyses were performed to explore the immunosuppressive function of TNFR2 in ECs. The downstream metabolic mechanism of TNFR2 was further investigated using RNA sequencing, cellular glycolysis assays, and western blotting. RESULTS: In this study, we identified an immunoregulatory EC subtype, characterized by enhanced TNFR2 expression in non-responders. By a mouse model of TNBC, we revealed a dynamic reduction in the proportion of the CD8+ T cell-contacting tumor vessels that could co-localize spatially with CD8+ T cells during chemotherapy and an increased expression of TNFR2 by ECs. TNFR2 suppresses glycolytic activity in ECs by activating NF-κB signaling in vitro. Tuning endothelial glycolysis enhances programmed death-ligand (PD-L) 1-dependent inhibitory capacity, thereby inducing CD8+ T cell suppression. In addition, TNFR2+ ECs showed a greater spatial affinity for exhausted CD8+ T cells than for non-exhausted CD8+ T cells. TNFR2 blockade restores impaired anti-tumor immunity in vivo, leading to the loss of PD-L1 expression by ECs and enhancement of CD8+ T cell infiltration into the tumors. CONCLUSIONS: These findings reveal the suppression of CD8+ T cells by ECs in chemoresistance and indicate the critical role of TNFR2 in driving the immunosuppressive capacity of ECs via tuning glycolysis. Targeting endothelial TNFR2 may serve as a potent strategy for treating TNBC with paclitaxel.


Sujet(s)
Lymphocytes T CD8+ , Résistance aux médicaments antinéoplasiques , Cellules endothéliales , Glycolyse , Récepteur au facteur de nécrose tumorale de type II , Tumeurs du sein triple-négatives , Récepteur au facteur de nécrose tumorale de type II/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Animaux , Humains , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Lignée cellulaire tumorale , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Souris , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE