Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.147
Filtrer
1.
J Am Chem Soc ; 146(37): 25490-25500, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39226482

RÉSUMÉ

The emergence of lysosome-targeting chimeras (LYTACs), which represents a promising strategy for membrane protein degradation based on lysosomal pathways, has attracted much attention in disease intervention and treatment. However, the expression level of commonly used lysosome-targeting receptors (LTRs) varies in different cell lines, thus limiting the broad applications of LYTACs. To overcome this difficulty, we herein report the development of integrin α3ß1 (ITGA3B1)-facilitated bispecific aptamer chimeras (ITGBACs) as a platform for the degradation of membrane proteins. ITGBACs consist of two aptamers, one targeting ITGA3B1 and another binding to the membrane-associated protein of interest (POI), effectively transporting the POI into lysosomes for degradation. Our findings demonstrate that ITGBACs effectively eliminate pathological membrane proteins, such as CD71 and PTK7, inducing significant cell-cycle arrest and apoptosis and markedly inhibiting tumor growth in tumor-bearing mice models. Therefore, this work provides a novel and versatile membrane protein degradation platform, offering a promising targeted therapy based on tumor-specific LTRs.


Sujet(s)
Aptamères nucléotidiques , Récepteurs à la transferrine , Humains , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/pharmacologie , Animaux , Souris , Récepteurs à la transferrine/métabolisme , Protéines membranaires/métabolisme , Protéolyse/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/composition chimique , Intégrine alpha3 bêta1/métabolisme , Lignée cellulaire tumorale , Antigènes CD/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Récepteurs à activité tyrosine kinase
2.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39273097

RÉSUMÉ

Iron is a vital element involved in a plethora of metabolic activities. Mammalian systemic iron homeostasis is mainly modulated by hepcidin, the synthesis of which is regulated by a number of proteins, including the hemochromatosis-associated proteins Hfe and Transferrin Receptor 2 (TfR2). Macrophages play versatile functions in iron homeostasis by storing iron derived from the catabolism of erythrocytes and supplying iron required for erythropoiesis. The absence of Hfe in macrophages causes a mild iron deficiency in aged mice and leads to an overproduction of the iron exporter Ferroportin 1 (Fpn1). Conversely, TfR2 gene silencing in macrophages does not influence systemic iron metabolism but decreases transcription of the macrophage Fpn1 in adult mice and modulates their immune response. This study investigated cellular and systemic iron metabolism in adult and aged male mice with macrophage-specific Hfe and TfR2 silencing (double knock-out, DKO). Serum iron parameters were significantly modified in aged animals, and significant differences were found in hepatic hepcidin transcription at both ages. Interestingly, splenic iron content was low in adult DKOs and splenic Fpn1 transcription was significantly increased in DKO animals at both ages, while the protein amount does not reflect the transcriptional trend. Additionally, DKO macrophages were isolated from mice bone marrow (BMDMs) and showed significant variations in the transcription of iron genes and protein amounts in targeted mice compared to controls. Specifically, Tranferrin Receptor 1 (TfR1) increased in DKO adult mice BMDMs, while the opposite is observed in the cells of aged DKO mice. Fpn1 transcript was significantly decreased in the BMDMs of adult DKO mice, while the protein was reduced at both ages. Lastly, a significant increase in Erythropoietin production was evidenced in aged DKO mice. Overall, our study reveals that Hfe and TfR2 in macrophages regulate hepatic Hepc production and affect iron homeostasis in the spleen and BMDMs, leading to an iron deficiency in aged animals that impairs their erythropoiesis.


Sujet(s)
Protéine de l'hémochromatose , Fer , Macrophages , Souris knockout , Récepteurs à la transferrine , Rate , Animaux , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Rate/métabolisme , Fer/métabolisme , Macrophages/métabolisme , Souris , Mâle , Protéine de l'hémochromatose/génétique , Protéine de l'hémochromatose/métabolisme , Moelle osseuse/métabolisme , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Hepcidines/métabolisme , Hepcidines/génétique , Souris de lignée C57BL , Homéostasie , Foie/métabolisme
3.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39273300

RÉSUMÉ

The hemojuvelin-hepcidin regulatory axis may play a key role in the iron metabolism both systemically and locally. There is a pressing need to evaluate this tightly regulated network of iron parameters and their potential impact on the development of ischemic stroke (IS). We aimed to assess iron metabolism biomarkers in patients after IS, evaluating changes over time and considering their clinical features. We studied 45 patients diagnosed with IS. We assessed major iron metabolism parameters, such as hepcidin, soluble hemojuvelin (sHJV), soluble transferrin receptor (sTfR), and ferritin, using immunoenzymathic methods at two time points: on admission and on the 7th day post IS. We found increased ferritin levels on the 7th day post IS compared to admission, and this was observed in the entire study group (p = 0.03) and in the subgroup treated with thrombolysis (p = 0.02). The hepcidin levels, on the other hand, showed a significant decrease on the 7th day, though this difference was only evident in the entire study group (p = 0.04). We also discovered significantly elevated sHJV levels in patients with PACI stroke compared to other stroke locations, both on admission and on the 7th day post IS (p < 0.05). Significantly higher sHJV levels were observed in patients treated with thrombolysis compared to those receiving conventional treatment, regardless of the time point (p < 0.0001 and p = 0.0002, respectively). Our study revealed changes in the iron metabolism parameters during stroke. The patients with anterior cerebral infarction and those treated with thrombolysis presented significantly elevated sHJV levels.


Sujet(s)
Marqueurs biologiques , Protéines liées au GPI , Protéine de l'hémochromatose , Hepcidines , Fer , Accident vasculaire cérébral ischémique , Récepteurs à la transferrine , Humains , Fer/métabolisme , Fer/sang , Mâle , Femelle , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/sang , Sujet âgé , Hepcidines/métabolisme , Hepcidines/sang , Protéine de l'hémochromatose/métabolisme , Protéine de l'hémochromatose/génétique , Études prospectives , Adulte d'âge moyen , Récepteurs à la transferrine/métabolisme , Protéines liées au GPI/métabolisme , Protéines liées au GPI/sang , Ferritines/sang , Ferritines/métabolisme , Sujet âgé de 80 ans ou plus
4.
Mol Med ; 30(1): 140, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251905

RÉSUMÉ

BACKGROUND: Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac4C) is mediated by the ac4C "writer", N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. METHODS: Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe2+, glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. RESULTS: The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac4C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. CONCLUSION: NAT10-mediated ac4C acetylation of TFRC aggravated SPI through promoting ferroptosis.


Sujet(s)
Ferroptose , Récepteurs à la transferrine , Sepsie , Sepsie/métabolisme , Sepsie/complications , Sepsie/étiologie , Acétylation , Animaux , Humains , Rats , Mâle , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Femelle , Lésion pulmonaire/métabolisme , Lésion pulmonaire/étiologie , Lésion pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Acetyltransferases/métabolisme , Acetyltransferases/génétique , Adulte d'âge moyen , Antigènes CD/métabolisme , Antigènes CD/génétique , Cytidine/analogues et dérivés , Cytidine/pharmacologie , Lignée cellulaire , /métabolisme , /étiologie , /anatomopathologie , Rat Sprague-Dawley , Survie cellulaire
5.
CNS Neurosci Ther ; 30(9): e70054, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39306799

RÉSUMÉ

OBJECTIVES: This study aims to elucidate the role of Fe2+ overload in kainic acid (KA)-induced excitotoxicity, investigate the involvement of ferritinophagy selective cargo receptor NCOA4 in the pathogenesis of excitotoxicity. METHODS: Western blotting was used to detect the expression of FTH1, NCOA4, Lamp2, TfR, FPN, and DMT1 after KA stereotaxic injection into the unilateral striatum of mice. Colocalization of Fe2+ with lysosomes in KA-treated primary cortical neurons was observed by using confocal microscopy. Desferrioxamine (DFO) was added to chelate free iron, a CCK8 kit was used to measure cell viability, and the Fe2+ levels were detected by FerroOrange. BODIPY C11 was used to determine intracellular lipid reactive oxygen species (ROS) levels, and the mRNA levels of PTGS2, a biomarker of ferroptosis, were measured by fluorescent quantitative PCR. 3-Methyladenine (3-MA) was employed to inhibit KA-induced activation of autophagy, and changes in ferritinophagy-related protein expression and the indicated biomarkers of ferroptosis were detected. Endogenous NCOA4 was knocked down by lentivirus transfection, and cell viability and intracellular Fe2+ levels were observed after KA treatment. RESULTS: Western blot results showed that the expression of NCOA4, DMT1, and Lamp2 was significantly upregulated, while FTH1 was downregulated, but there were no significant changes in TfR and FPN. The fluorescence results indicated that KA enhanced the colocalization of free Fe2+ with lysosomes in neurons. DFO intervention could effectively rescue cell damage, reduce intracellular lipid peroxidation, and decrease the increased transcript levels of PTGS2 caused by KA. Pretreatment with 3-MA effectively reversed KA-induced ferritinophagy and ferroptosis. Endogenous interference with NCOA4 significantly improved cell viability and reduced intracellular free Fe2+ levels in KA-treated cells. CONCLUSION: KA-induced excitotoxicity activates ferritinophagy, and targeting ferritinophagy effectively inhibits downstream ferroptosis. Interference with NCOA4 effectively attenuates KA-induced neuronal damage. This study provides a potential therapeutic target for excitotoxicity related disease conditions.


Sujet(s)
Ferritines , Ferroptose , Neurones , Coactivateurs de récepteurs nucléaires , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Souris , Ferritines/métabolisme , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Autophagie/effets des médicaments et des substances chimiques , Autophagie/physiologie , Souris de lignée C57BL , Mâle , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Espèces réactives de l'oxygène/métabolisme , Cellules cultivées , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Fer/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/métabolisme , Protéine de membrane-2 associée au lysosome/métabolisme , Protéine de membrane-2 associée au lysosome/génétique , Système y+ de transport d'acides aminés , Transporteurs de cations
6.
CNS Neurosci Ther ; 30(9): e70018, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39252474

RÉSUMÉ

OBJECTIVE: Nowadays, the prevalence of cognitive impairment in women has gradually increased, especially in postmenopausal women. There were few studies on the mechanistic effects of iron exposure on neurotoxicity in postmenopausal women. The aim of this study is to investigate the effect of iron accumulation on cognitive ability in ovariectomized mice and its possible mechanism and to provide a scientific basis for the prevention of cognitive dysfunction in postmenopausal women. METHODS: Female C57BL/6N ovariectomized model mice were induced with ferric citrate (FAC). The mice were randomly divided into 5 groups: control, sham, ovariectomized (Ovx), Ovx + 50 mg/kg FAC (Ovx + l), and Ovx + 100 mg/kg FAC (Ovx + h). The impact of motor and cognitive function was verified by a series of behavioral tests. The levels of serum iron parameters, malondialdehyde, and superoxide dismutase were measured. The ultrastructure of mice hippocampal microglia was imaged by transmission electron microscopy. The differential expression of hippocampal proteins was analyzed by Tandem Mass Tag labeling. RESULTS: Movement and cognitive function in Ovx + l/Ovx + h mice were significantly decreased compared to control and Sham mice. Then, iron exposure caused histopathological changes in the hippocampus of mice. In addition, proteomic analysis revealed that 29/27/41 proteins were differentially expressed in the hippocampus when compared by Ovx vs. Sham, Ovx + l vs. Ovx, as well as Ovx + h vs. Ovx + l groups, respectively. Moreover, transferrin receptor protein (TFR1) and divalent metal transporter 1 (DMT1) protein expression were significantly increased in the iron accumulation mice model with ovariectomy. CONCLUSION: Iron exposure could cause histopathological damage in the hippocampus of ovariectomised mice and, by altering hippocampal proteomics, particularly the expression of hippocampal iron metabolism-related proteins, could further influence cognitive impairment in ovariectomized mice.


Sujet(s)
Modèles animaux de maladie humaine , Composés du fer III , Hippocampe , Fer , Souris de lignée C57BL , Ovariectomie , Animaux , Femelle , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Souris , Fer/métabolisme , Composés du fer III/toxicité , Composés du fer III/pharmacologie , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Dysfonctionnement cognitif/métabolisme , Troubles de la cognition/anatomopathologie , Troubles de la cognition/induit chimiquement , Superoxide dismutase/métabolisme , Transporteurs de cations/métabolisme , Récepteurs à la transferrine/métabolisme
7.
Cell Death Dis ; 15(9): 655, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39242574

RÉSUMÉ

Studies have indicated that RAB17 expression levels are associated with tumor malignancy, and RAB17 is more highly expressed in endometrial cancer (EC) tissues than in peritumoral tissues. However, the roles and potential mechanisms of RAB17 in EC remain undefined. The present study confirmed that the expression of RAB17 facilitates EC progression by suppressing cellular ferroptosis-like alterations. Mechanistically, RAB17 attenuated ferroptosis in EC cells by inhibiting transferrin receptor (TFRC) protein expression in a ubiquitin proteasome-dependent manner. Because EC is a blood-deprived tumor with a poor energy supply, the relationship between RAB17 and hypoglycemia was investigated. RAB17 expression was increased in EC cells incubated in low-glucose medium. Moreover, low-glucose medium limited EC cell ferroptosis and promoted EC progression through the RAB17-TFRC axis. The in vitro results were corroborated by in vivo studies and clinical data. Overall, the present study revealed that increased RAB17 promotes the survival of EC cells during glucose deprivation by inhibiting the onset of TFRC-dependent ferroptosis.


Sujet(s)
Évolution de la maladie , Tumeurs de l'endomètre , Ferroptose , Récepteurs à la transferrine , Protéines G rab , Animaux , Femelle , Humains , Souris , Antigènes CD , Lignée cellulaire tumorale , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/génétique , Ferroptose/génétique , Glucose/métabolisme , Souris nude , Protéines G rab/métabolisme , Protéines G rab/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique
8.
Sci Rep ; 14(1): 21164, 2024 09 10.
Article de Anglais | MEDLINE | ID: mdl-39256468

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, presenting a persisting global health burden. Neutrophils have a double-edged role in tumor progression exhibiting both pro-tumor and anti-tumor functions. CD71, also known as transferrin receptor 1, performs a critical role in cellular iron uptake and is highly expressed on proliferating cells, and especially on activated immune cells. CD71 is known to be elevated in various types of solid cancers and is associated with poor prognosis, however, the expression of CD71 on neutrophils in PDAC and its potential clinical impact is still unknown. Therefore, we analyzed CD71 on circulating neutrophils in PDAC and clinical control patients and found a significant increased expression in PDAC patients. High expression of CD71 on neutrophils in PDAC patients was associated with reduced outcome compared to low expression. CD71 on neutrophils correlated positively with the levels of proinflammatory cytokines IL-6, IFN-γ, and growth factor ligands CD40-L, and BAFF in plasma of PDAC patients. Finally, we have demonstrated that high expression of CD71 on neutrophils was also associated with an increased expression of CD39 and CD25 on circulating T-cells. Based on our findings, we hypothesize that CD71 on neutrophils is associated with tumor progression in PDAC. Further studies are required to investigate the distinct functionality of CD71 expressing neutrophils and their potential clinical application.


Sujet(s)
Antigènes CD , Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , Granulocytes neutrophiles , Tumeurs du pancréas , Récepteurs à la transferrine , Humains , Granulocytes neutrophiles/métabolisme , Récepteurs à la transferrine/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/sang , Mâle , Antigènes CD/métabolisme , Femelle , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/sang , Pronostic , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/sang , Adulte d'âge moyen , Sujet âgé , Apyrase/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Métastase tumorale , Cytokines/métabolisme , Cytokines/sang
9.
Food Chem Toxicol ; 192: 114926, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39147356

RÉSUMÉ

Valproic acid (VPA), a common antiepileptic drug, can cause liver steatosis after long-term therapy. However, an impact of ferroptosis on VPA-induced liver steatosis has not been investigated. In the study, treatment with VPA promoted ferroptosis in the livers of mice by elevating ferrous iron (Fe2+) levels derived from the increased absorption by transferrin receptor 1 (TFR1) and the decreased storage by ferritin (FTH1 and FTL), disrupting the redox balance via reduced levels of solute carrier family 7 member 11 (SLC7A11), glutathione (GSH), and glutathione peroxidase 4 (GPX4), and augmenting acyl-CoA synthetase long-chain family member 4 (ACSL4) -mediated lipid peroxide generation, accompanied by enhanced liver steatosis. All the changes were significantly reversed by co-treatment with an iron-chelating agent, deferoxamine mesylate (DFO) and a ferroptosis inhibitor, ferrostatin-1 (Fer-1). Similarly, the increases in Fe2+, TFR1, and ACSL4 levels, as well as the decreases in GSH, GPX4, and ferroportin (FPN) levels, were detected in VPA-treated HepG2 cells. These changes were also attenuated after co-treatment with Fer-1. It demonstrates that ferroptosis promotes VPA-induced liver steatosis through iron overload, inhibition of the GSH-GPX4 axis, and upregulation of ACSL4. It offers a potential therapy targeting ferroptosis for patients with liver steatosis following VPA treatment.


Sujet(s)
Coenzyme A ligases , Stéatose hépatique , Ferroptose , Phospholipid hydroperoxide glutathione peroxidase , Récepteurs à la transferrine , Acide valproïque , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Acide valproïque/toxicité , Souris , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Humains , Stéatose hépatique/induit chimiquement , Stéatose hépatique/métabolisme , Mâle , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Fer/métabolisme , Souris de lignée C57BL , Ferritines/métabolisme , Glutathion/métabolisme , Cellules HepG2 , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme
10.
Int Immunopharmacol ; 140: 112787, 2024 Oct 25.
Article de Anglais | MEDLINE | ID: mdl-39088914

RÉSUMÉ

Myasthenia gravis (MG) is a multifaceted autoimmune disorder affecting the postsynaptic neuromuscular junction. In this study, we examined CD4+ and CD8+ T lymphocyte levels and ratios within peripheral blood mononuclear cells (PBMCs) in MG patients. Additionally, we assessed lymphocytes for the expression of CD71, which functions as a transferrin receptor mediating the uptake of iron into the cells. Building on recent discussions regarding CD20 depletion treatments in MG, we also scrutinized lymphocytes for CD20 expression. Comparative analyses were conducted among healthy controls, newly diagnosed MG patients, those undergoing pyridostigmine treatment alone, and MG patients receiving combination therapies. In the patients, the ratio of CD3+CD4+ T lymphocytes to CD3+ T lymphocytes was found to be decreased compared to the healthy controls, while the ratio of CD3+CD8+ cells to CD3+CD4+ cells increased. An increase in the percentage of CD71-expressing lymphocytes was observed in MG patients compared to the healthy control group, while CD20+ lymphocytes exhibited no statistical changes. Moreover, heightened serum lipid peroxidation levels were found in MG patients. These results suggest a possible relationship between iron metabolism, levels of CD71-expressing cells, and lipid peroxidation in MG. Conversely, pyridostigmine treatment reduced the levels of CD71-expressing cells and lipid peroxidation, suggesting potential immunomodulatory and antioxidant impacts of pyridostigmine in MG, either directly or indirectly.


Sujet(s)
Antigènes CD , Peroxydation lipidique , Myasthénie , Bromure de pyridostigmine , Récepteurs à la transferrine , Humains , Myasthénie/sang , Myasthénie/immunologie , Myasthénie/traitement médicamenteux , Mâle , Femelle , Antigènes CD/métabolisme , Adulte d'âge moyen , Adulte , Récepteurs à la transferrine/métabolisme , Bromure de pyridostigmine/usage thérapeutique , Études de cohortes , Sujet âgé , Lymphocytes T CD8+/immunologie , Lymphocytes T CD4+/immunologie , Fer/métabolisme
11.
Int J Pharm ; 663: 124570, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39134291

RÉSUMÉ

A multi-component paclitaxel (PTX) -loaded ß-elemene nanoemulsion by transferrin modification (Tf-PE-MEs) was developed to enhance non-small-cell lung cancer (NSCLC) treatment. After transferrin modification, the particle size of Tf-PE-MEs was (14.87 ± 1.84) nm, and the zeta potential was (-10.19 ± 0.870) mV, respectively. In vitro experiments showed that Tf-PE-MEs induced massive apoptosis in A549 cells, indicating that it had significant cytotoxicity to A549 cells. Through transferrin modification, Tf-PE-MEs accumulated at the tumor site efficiently with overexpressed transferrin receptor (TfR) on the surface of A549 cells. This will allow increasing PTX and ß-elemene concentration in the target cells, enhancing the therapeutic effect. Compared to PTX alone, Tf-PE-MEs displayed good anti-tumor efficacy and diminished systemic toxicity in vivo studies. With favourable therapeutic potential, this study provides a new strategy for the combined anticancer treatment of non-small cell lung cancer.


Sujet(s)
Antinéoplasiques d'origine végétale , Apoptose , Carcinome pulmonaire non à petites cellules , Émulsions , Tumeurs du poumon , Nanoparticules , Paclitaxel , Sesquiterpènes , Transferrine , Paclitaxel/administration et posologie , Paclitaxel/pharmacologie , Paclitaxel/composition chimique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Humains , Transferrine/composition chimique , Transferrine/administration et posologie , Tumeurs du poumon/traitement médicamenteux , Animaux , Cellules A549 , Sesquiterpènes/pharmacologie , Sesquiterpènes/composition chimique , Sesquiterpènes/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Antinéoplasiques d'origine végétale/administration et posologie , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/composition chimique , Souris de lignée BALB C , Souris nude , Récepteurs à la transferrine/métabolisme , Taille de particule , Souris , Lignée cellulaire tumorale , Mâle , Libération de médicament , Survie cellulaire/effets des médicaments et des substances chimiques
12.
Eur J Pharm Biopharm ; 203: 114468, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39173936

RÉSUMÉ

Lipid nanoparticles (LNPs) are promising delivery systems with the ability to deliver small interfering RNA (siRNA) and messenger RNA (mRNA) in diseased tissues and intracellular sites of action. However, delivery to non-hepatic tissues via systemic administration remains challenging. Antibody modification of LNPs is a hopeful approach for improving their selectivity to target tissues. The conventional method of antibody modification via thiol-maleimide linkage is concerned with reduced recognition efficiency of the disease-related target molecules owing to variations in antibody orientation on the surface of the LNPs. In this study, we developed a novel adapter lipopeptide for antibody modification of LNPs via the Fc-region. Here, we selected RI7-217, an anti-transferrin receptor antibody, as the ligand. Through optimization of spacer peptides, we found a FcBP-EKGG-lipid exhibits high water-dispersibility for post-insertion method to LNPs. We prepared RI7-217-modified LNPs by modifying LNPs with FcBP-EKGG-lipids and mixing the antibodies. We found that the luciferase protein expression of RI7-217-modified LNPs was significantly enhanced in an antibody-specific manner against transferrin receptor-expressing U-87 MG cells. This information would be valuable in the development of antibody-modified LNPs for cell-selective targeting.


Sujet(s)
Lipides , Nanoparticules , ARN messager , Récepteurs à la transferrine , Récepteurs à la transferrine/immunologie , Récepteurs à la transferrine/métabolisme , Nanoparticules/composition chimique , Lipides/composition chimique , Humains , Petit ARN interférent/administration et posologie , Fragments Fc des immunoglobulines , Liposomes
13.
Sci Transl Med ; 16(760): eadi2245, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39141703

RÉSUMÉ

Antisense oligonucleotides (ASOs) are promising therapeutics for treating various neurological disorders. However, ASOs are unable to readily cross the mammalian blood-brain barrier (BBB) and therefore need to be delivered intrathecally to the central nervous system (CNS). Here, we engineered a human transferrin receptor 1 (TfR1) binding molecule, the oligonucleotide transport vehicle (OTV), to transport a tool ASO across the BBB in human TfR knockin (TfRmu/hu KI) mice and nonhuman primates. Intravenous injection and systemic delivery of OTV to TfRmu/hu KI mice resulted in sustained knockdown of the ASO target RNA, Malat1, across multiple mouse CNS regions and cell types, including endothelial cells, neurons, astrocytes, microglia, and oligodendrocytes. In addition, systemic delivery of OTV enabled Malat1 RNA knockdown in mouse quadriceps and cardiac muscles, which are difficult to target with oligonucleotides alone. Systemically delivered OTV enabled a more uniform ASO biodistribution profile in the CNS of TfRmu/hu KI mice and greater knockdown of Malat1 RNA compared with a bivalent, high-affinity TfR antibody. In cynomolgus macaques, an OTV directed against MALAT1 displayed robust ASO delivery to the primate CNS and enabled more uniform biodistribution and RNA target knockdown compared with intrathecal dosing of the same unconjugated ASO. Our data support systemically delivered OTV as a potential platform for delivering therapeutic ASOs across the BBB.


Sujet(s)
Barrière hémato-encéphalique , Oligonucléotides antisens , ARN long non codant , Récepteurs à la transferrine , Animaux , Humains , Souris , Transport biologique , Barrière hémato-encéphalique/métabolisme , Techniques de knock-down de gènes , Macaca fascicularis , Oligonucléotides antisens/pharmacocinétique , Oligonucléotides antisens/administration et posologie , Récepteurs à la transferrine/métabolisme , ARN long non codant/métabolisme , ARN long non codant/génétique , Distribution tissulaire
14.
Eur J Pharm Biopharm ; 203: 114473, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39186959

RÉSUMÉ

Hydroxychloroquine sulfate (HCQ) is currently being repurposed for cancer treatment. The antitumor mechanism of HCQ is inhibition of cellular autophagy, but its therapeutic potential is severely limited by poor solubility, lack of tumor targeting and lower cellular uptake. Therefore, utilization of human H-chain apoferritin (HFn) composed only of heavy subunits is an attractive approach for tumor targeting drug delivery. This study focused on pH-triggered encapsulation of HCQ within the inner cavity of HFn to form HFn@HCQ nanoparticles for tumor-targeted drug delivery. Characterization using a range of techniques has been used to confirm the successful establishment of HFn@HCQ. HFn@HCQ exhibited pH-responsive release behavior, with almost no drug release at pH 7.4, but 80% release at pH 5.0. Owing to its intrinsic binding to transferrin receptor 1 (TfR1), HFn@HCQ was significantly internalized through TfR1-mediated endocytosis, with a 4.4-fold difference of internalization amount across cell lines. Additionally, HFn@HCQ enhanced the antitumor effect against four different cancer cell lines when compared against HCQ alone, especially in TfR1 high-expressing cells, where the inhibitory effect was 3-fold higher than free HCQ. The autophagy inhibition of HFn@HCQ has been demonstrated, which is a major pathway to induce cancer cell death. According to current findings, HFn based drug delivery is a promising strategy to target and kill TfR1 overexpressing tumor cells.


Sujet(s)
Antinéoplasiques , Apoferritines , Autophagie , Libération de médicament , Repositionnement des médicaments , Hydroxychloroquine , Nanoparticules , Humains , Hydroxychloroquine/pharmacologie , Hydroxychloroquine/composition chimique , Hydroxychloroquine/administration et posologie , Autophagie/effets des médicaments et des substances chimiques , Repositionnement des médicaments/méthodes , Apoferritines/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Nanoparticules/composition chimique , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments/méthodes , Concentration en ions d'hydrogène , Récepteurs à la transferrine/métabolisme , Tumeurs/traitement médicamenteux , Vecteurs de médicaments/composition chimique , Endocytose/effets des médicaments et des substances chimiques
15.
PeerJ ; 12: e17842, 2024.
Article de Anglais | MEDLINE | ID: mdl-39131609

RÉSUMÉ

Background: Ferroptosis is a non-apoptotic iron-dependent form of cell death implicated in various cancer pathologies. However, its precise role in tumor growth and progression of cervical cancer (CC) remains unclear. Transferrin receptor protein 1 (TFRC), a key molecule associated with ferroptosis, has been identified as influencing a broad range of pathological processes in different cancers. However, the prognostic significance of TFRC in CC remains unclear. The present study utilized bioinformatics to explore the significance of the ferroptosis-related gene TFRC in the progression and prognosis of CC. Methods: We obtained RNA sequencing data and corresponding clinical information on patients with CC from The Cancer Genome Atlas (TCGA), Genotype Tissue Expression (GTEx) and Gene Expression Omnibus (GEO) databases. Using least absolute shrinkage and selection operator (LASSO) Cox regression, we then generated a multigene signature of five ferroptosis-related genes (FRGs) for the prognostic prediction of CC. We investigated the relationship between TFRC gene expression and immune cell infiltration by employing single-sample GSEA (ssGSEA) analysis. The potential functional role of the TFRC gene was evaluated through gene set enrichment analysis (GSEA). Immunohistochemistry and qPCR was employed to assess TFRC mRNA and protein expression in 33 cases of cervical cancer. Furthermore, the relationship between TFRC mRNA expression and overall survival (OS) was investigated in patients. Results: CC samples had significantly higher TFRC gene expression levels than normal tissue samples. Higher TFRC gene expression levels were strongly associated with higher cancer T stages and OS events. The findings of multivariate analyses illustrated that the OS in CC patients with high TFRC expression is shorter than in patients with low TFRC expression. Significant increases were observed in the levels of TFRC mRNA and protein expression in patients diagnosed with CC. Conclusion: Increased TFRC expression in CC was associated with disease progression, an unfavorable prognosis, and dysregulated immune cell infiltration. In addition, it highlights ferroptosis as a promising therapeutic target for CC.


Sujet(s)
Ferroptose , Récepteurs à la transferrine , Microenvironnement tumoral , Tumeurs du col de l'utérus , Humains , Femelle , Ferroptose/génétique , Récepteurs à la transferrine/génétique , Récepteurs à la transferrine/métabolisme , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/mortalité , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Pronostic , Régulation de l'expression des gènes tumoraux , Antigènes CD/génétique , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme
16.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125916

RÉSUMÉ

Understanding the role of iron in ethanol-derived hepatic stress could help elucidate the efficacy of dietary or clinical interventions designed to minimize liver damage from chronic alcohol consumption. We hypothesized that normal levels of iron are involved in ethanol-derived liver damage and reduced dietary iron intake would lower the damage caused by ethanol. We used a pair-fed mouse model utilizing basal Lieber-DeCarli liquid diets for 22 weeks to test this hypothesis. In our mouse model, chronic ethanol exposure led to mild hepatic stress possibly characteristic of early-stage alcoholic liver disease, seen as increases in liver-to-body weight ratios. Dietary iron restriction caused a slight decrease in non-heme iron and ferritin (FeRL) expression while it increased transferrin receptor 1 (TfR1) expression without changing ferroportin 1 (FPN1) expression. It also elevated protein lysine acetylation to a more significant level than in ethanol-fed mice under normal dietary iron conditions. Interestingly, iron restriction led to an additional reduction in nicotinamide adenine dinucleotide (NAD+) and NADH levels. Consistent with this observation, the major mitochondrial NAD+-dependent deacetylase, NAD-dependent deacetylase sirtuin-3 (SIRT3), expression was significantly reduced causing increased protein lysine acetylation in ethanol-fed mice at normal and low-iron conditions. In addition, the detection of superoxide dismutase 1 and 2 levels (SOD1 and SOD2) and oxidative phosphorylation (OXPHOS) complex activities allowed us to evaluate the changes in antioxidant and energy metabolism regulated by ethanol consumption at normal and low-iron conditions. We observed that the ethanol-fed mice had mild liver damage associated with reduced energy and antioxidant metabolism. On the other hand, iron restriction may exacerbate certain activities of ethanol further, such as increased protein lysine acetylation and reduced antioxidant metabolism. This metabolic change may prove a barrier to the effectiveness of dietary reduction of iron intake as a preventative measure in chronic alcohol consumption.


Sujet(s)
Antioxydants , Métabolisme énergétique , Éthanol , Animaux , Souris , Acétylation/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Antioxydants/métabolisme , Mâle , Fer/métabolisme , Superoxide dismutase-1/métabolisme , Superoxide dismutase-1/génétique , Superoxide dismutase/métabolisme , Lysine/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Récepteurs à la transferrine/métabolisme , Sirtuine-3/métabolisme , Sirtuine-3/génétique , NAD/métabolisme , Ferritines/métabolisme , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/anatomopathologie , Maladies alcooliques du foie/étiologie
17.
Biophys Chem ; 314: 107308, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39208499

RÉSUMÉ

The human transmembrane protein Transferrin Receptor-1 is regarded as a promising target for the systemic delivery of therapeutic agents, particularly of nucleic acid therapeutics, such as short double stranded RNAs. This ubiquitous receptor is involved in cellular iron uptake, keeping intracellular homeostasis. It is overexpressed in multiple cancer cell types and is internalized via clathrin-mediated endocytosis. In previous studies, a human transferrin receptor-1 RNA aptamer, identified as TR14 ST1-3, was shown to be capable of effectively internalizing into cells in culture and to deliver small, double stranded RNAs in vitro and in vivo, via systemic administration. To understand, at the molecular level, the aptamer binding to the receptor and the impact of conjugation with the therapeutic RNA, a multi-level in silico protocol was employed, including protein-aptamer docking, molecular dynamics simulations and free energy calculations. The competition for the binding pocket, between the aptamer and the natural ligand human Transferrin, was also evaluated. The results show that the aptamer binds to the same region as Transferrin, with residues from the helical domain showing a critical role. Moreover, the conjugation to the therapeutic RNA, was shown not to affect aptamer binding. Overall, this study provides an atomic-level understanding of aptamer association to human Transferrin Receptor-1 and of its conjugation with a short model-therapeutic RNA, providing also important clues for futures studies aiming to deliver other oligonucleotide-based therapeutics via Transferrin Receptor.


Sujet(s)
Aptamères nucléotidiques , Récepteurs à la transferrine , Humains , Antigènes CD , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/métabolisme , Sites de fixation , Simulation numérique , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Liaison aux protéines , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/composition chimique , ARN/métabolisme , ARN/composition chimique , Thermodynamique , Transferrine/composition chimique , Transferrine/métabolisme
18.
Int J Biol Macromol ; 278(Pt 2): 134777, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39153669

RÉSUMÉ

Glioblastoma (GBM) represents a formidable challenge in oncology, characterized by aggressive proliferation and poor prognosis. Iron metabolism plays a critical player in GBM progression, with dysregulated iron uptake and utilization contributing to tumor growth and therapeutic resistance. Iron's pivotal role in DNA synthesis, oxidative stress, and angiogenesis underscores its significance in GBM pathogenesis. Elevated expression of iron transporters, such as transferrin receptor 1 (TfR1), highlights the tumor's reliance on iron for survival. Innovative treatment strategies targeting iron dysregulation hold promise for overcoming therapeutic challenges in GBM management. Approaches such as iron chelation therapies, induction of ferroptosis to nanoparticle-based drug delivery systems exploit iron-dependent vulnerabilities, offering avenues for enhance treatment efficacy and improve patient outcomes. As research advances, understanding the complexities of iron-mediated carcinogenesis provides a foundation for developing precision medicine approaches tailored to combat GBM effectively. This review explores the intricate relationship between iron metabolism and GBM, elucidating its multifaceted implications and therapeutic opportunities. By consolidating the latest insights into iron metabolism in GBM, this review underscores its potential as a therapeutic target for improving patient care in combination with the standard of care approach.


Sujet(s)
Ferroptose , Glioblastome , Fer , Récepteurs à la transferrine , Humains , Récepteurs à la transferrine/métabolisme , Fer/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Animaux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Antigènes CD/métabolisme , Antigènes CD/génétique , Agents chélateurs du fer/usage thérapeutique , Agents chélateurs du fer/pharmacologie
19.
Medicine (Baltimore) ; 103(32): e39256, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121332

RÉSUMÉ

Iron metabolism plays an important role in insulin resistance, and the triglyceride-glucose (TyG) index has been proposed in recent years as a more accessible and cost-effective marker for insulin resistance. This study aims to evaluate the association between iron metabolism markers, including ferritin (FER), transferrin (TRF), and transferrin receptor (TFR), and the TyG index. A total of 6524 Chinese individuals aged between 18 and 75 years were included in this study. Multivariable linear models were used to investigate the association between FER, TRF, and TFR levels, and the TyG index. Further subgroup analyses stratified by age and sex were also performed. There was a positive association between FER and TRF levels and the TyG index in all 3 multivariable linear regression models, regardless of stratification by sex and age. Additionally, TFR was positively associated with the TyG index among females and those aged ≥45 years, but not among males and those aged <45 years. Our findings reveal a positive association between FER and TRF levels and the TyG index in a Chinese population, while the association between TFR levels and the TyG index showed different patterns depending on age and gender.


Sujet(s)
Marqueurs biologiques , Glycémie , Ferritines , Fer , Enquêtes nutritionnelles , Récepteurs à la transferrine , Transferrine , Triglycéride , Humains , Mâle , Femelle , Adulte d'âge moyen , Adulte , Chine , Études transversales , Triglycéride/sang , Récepteurs à la transferrine/sang , Ferritines/sang , Sujet âgé , Marqueurs biologiques/sang , Transferrine/analyse , Transferrine/métabolisme , Fer/sang , Fer/métabolisme , Adolescent , Glycémie/analyse , Glycémie/métabolisme , Jeune adulte , Insulinorésistance
20.
Commun Biol ; 7(1): 1011, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154074

RÉSUMÉ

The acquisition of ectopic fibroblast growth factor receptor 1 (FGFR1) expression is well documented in prostate cancer (PCa) progression, notably in conferring tumor growth advantage and facilitating metastasis. However, how FGFR1 contributes to PCa progression is not fully revealed. Here we report that ectopic FGFR1 in PCa cells promotes transferrin receptor 1 (TFR1) expression and expands the labile iron pool (LIP), and vice versa. We further demonstrate that FGFR1 stabilizes iron regulatory proteins 2 (IRP2) and therefore, upregulates TFR1 via promoting IRP2 binding to the IRE of TFR1. Deletion of FGFR1 in DU145 cells decreases the LIP, which potentiates the anticancer efficacy of iron chelator. Intriguingly, forced expression of IRP2 in FGFR1 depleted cells reinstates TFR1 expression and LIP, subsequently restoring the tumorigenicity of the cells. Together, our results here unravel a new mechanism by which FGFR1 drives PCa progression and suggest a potential novel target for PCa therapy.


Sujet(s)
Homéostasie , Protéine-2 de régulation du fer , Fer , Tumeurs de la prostate , Récepteur FGFR1 , Humains , Récepteur FGFR1/métabolisme , Récepteur FGFR1/génétique , Mâle , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Fer/métabolisme , Protéine-2 de régulation du fer/métabolisme , Protéine-2 de régulation du fer/génétique , Lignée cellulaire tumorale , Animaux , Protéolyse , Souris , Régulation de l'expression des gènes tumoraux , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Antigènes CD
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE