Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.461
Filtrar
1.
Biomaterials ; 312: 122760, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39163825

RESUMEN

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


Asunto(s)
Artritis Reumatoide , Desoxiglucosa , Dexametasona , Glucosa , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Animales , Glucosa/metabolismo , Dexametasona/farmacología , Dexametasona/uso terapéutico , Ratones , Desoxiglucosa/farmacología , Inflamación/tratamiento farmacológico , Glucólisis/efectos de los fármacos , Polímeros/química , Ácido Hialurónico/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Masculino , Humanos , Proliferación Celular/efectos de los fármacos
2.
Chem Biol Drug Des ; 104(3): e14624, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39317696

RESUMEN

In the pentose phosphate pathway, dehydroepiandrosterone (DHEA) uncompetitively inhibits glucose-6-phosphate dehydrogenase (G6PD), reducing NADPH production and increasing oxidative stress, which can influence the onset and/or progression of several diseases, including cancer. 2-Deoxy-D-glucose (2-DG), a glucose mimetic, competes with glucose for cellular uptake, inhibiting glycolysis and competing with glucose-6-phosphate (G-6-P) for G6PD activity. In this study, we report that DHEA-α-2-DG (5), an α-covalent conjugate of DHEA and 2-DG, exhibits better anticancer activity than DHEA, 2-DG, DHEA +2-DG, and polydatin in MCF-7 cells, and reduces NADPH/NADP+ ratio in cellular assays. In vitro enzyme kinetics and molecular docking studies showed that 5 uncompetitively inhibits human G6PD activity and binds to the structural NADP+ site but not to the catalytic NADP+ site. Further combining 5 with the FDA-approved drug tamoxifen enhanced its cytotoxicity against MCF-7 cells, suggesting that it could serve as a candidate for combination of drug strategies.


Asunto(s)
Antineoplásicos , Deshidroepiandrosterona , Desoxiglucosa , Glucosafosfato Deshidrogenasa , Simulación del Acoplamiento Molecular , Humanos , Glucosafosfato Deshidrogenasa/metabolismo , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Deshidroepiandrosterona/farmacología , Deshidroepiandrosterona/química , Células MCF-7 , Desoxiglucosa/farmacología , Desoxiglucosa/química , Desoxiglucosa/análogos & derivados , Desoxiglucosa/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , NADP/metabolismo , Tamoxifeno/farmacología , Tamoxifeno/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Cinética
3.
Sci Rep ; 14(1): 20368, 2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223206

RESUMEN

Local inflammatory microenvironment in the early stage of myocardial infarction (MI) severely impaired cardiac recovery post-MI. Macrophages play a pivotal role in this process. A classical glycolytic inhibitor, 2-Deoxy-Glucose (2-DG), has been found to regulate the excessive pro-inflammatory macrophage polarization in the infarcted myocardium. This study investigated the effect of 2-DG-loaded chitosan/gelatin composite patch on the infarct microenvironment post-MI and its impact on cardiac repair. The results showed that the 2-DG patch significantly inhibited the expression of inflammatory cytokines, alleviated reactive oxygen species (ROS) accumulation, repressed the proinflammatory polarization of macrophages, attenuated local inflammatory microenvironment in the ischemic hearts, as well as improved cardiac function, reduced scar size, and promoted angiogenesis post-MI. In terms of mechanism, 2-DG exerts anti-inflammatory effects through inhibiting the NF-κB signaling pathway and reducing the assembly and activation of the NLRP3 inflammasome. These findings suggest that 2-DG composite patch may represent a promising therapeutic strategy for cardiac repair after MI.


Asunto(s)
Desoxiglucosa , Infarto del Miocardio , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Desoxiglucosa/farmacología , Desoxiglucosa/administración & dosificación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratones , Masculino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal/efectos de los fármacos , Quitosano/farmacología , Quitosano/química , Gelatina/química , Citocinas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Ratones Endogámicos C57BL
4.
Biol Direct ; 19(1): 73, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182128

RESUMEN

Hematopoietic stem cells (HSCs) exhibit significant functional and metabolic alterations within the lung cancer microenvironment, contributing to tumor progression and immune evasion by increasing differentiation into myeloid-derived suppressor cells (MDSCs). Our aim is to analyze the metabolic transition of HSCs from glycolysis to oxidative phosphorylation (OXPHOS) in lung cancer and determine its effects on HSC functionality. Using a murine Lewis Lung Carcinoma lung cancer model, we conducted metabolic profiling of long-term and short-term HSCs, as well as multipotent progenitors, comparing their metabolic states in normal and cancer conditions. We measured glucose uptake using 2-[N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)Amino]-2-Deoxyglucose (2-NBDG) and assessed levels of lactate, acetyl-coenzyme A, and ATP. Mitochondrial functionality was evaluated through flow cytometry, alongside the impact of the glucose metabolism inhibitor 2-DG on HSC differentiation and mitochondrial activity. HSCs under lung cancer conditions showed increased glucose uptake and lactate production, with an associated rise in OXPHOS activity, marking a metabolic shift. Treatment with 2-DG led to decreased T-HSCs and MDSCs and an increased red blood cell count, highlighting its potential to influence metabolic and differentiation pathways in HSCs. This study provides novel insights into the metabolic reprogramming of HSCs in lung cancer, emphasizing the critical shift from glycolysis to OXPHOS and its implications for the therapeutic targeting of cancer-related metabolic pathways.


Asunto(s)
Desoxiglucosa , Glucólisis , Células Madre Hematopoyéticas , Neoplasias Pulmonares , Fosforilación Oxidativa , Animales , Desoxiglucosa/metabolismo , Desoxiglucosa/análogos & derivados , Ratones , Neoplasias Pulmonares/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Reprogramación Celular , Diferenciación Celular
5.
Cell Signal ; 123: 111351, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39159908

RESUMEN

In autosomal dominant polycystic kidney disease (ADPKD) there is cyst growth in the kidneys that leads to chronic kidney disease often requiring dialysis or kidney transplantation. There is enhanced aerobic glycolysis (Warburg effect) in the cyst lining epithelial cells that contributes to cyst growth. The glucose mimetic, 2-Deoxy-d-glucose (2-DG) inhibits glycolysis. The effect of early and late administration of 2-DG on cyst growth and kidney function was determined in Pkd1RC/RC mice, a hypomorphic PKD model orthologous to human disease. Early administration of 2-DG resulted in decreased kidney weight, cyst index, cyst number and cyst size, but no change in kidney function. 2-DG decreased proliferation. a major mediator of cyst growth, of cells lining the cyst. Late administration of 2-DG did not have an effect on cyst growth or kidney function. To determine mechanisms of decreased proliferation, an array of mTOR and autophagy proteins was measured in the kidney. 2-DG suppressed autophagic flux in Pkd1RC/RC kidneys and decreased autophagy proteins, ATG3, ATG5 and ATG12-5. 2-DG had no effect on p-mTOR or p-S6 (mTORC1) and decreased p-AMPK. 2-DG decreased p-4E-BP1, p-c-Myc and p-ERK that are known to promote proliferation and cyst growth in PKD. 2-DG decreased p-AKTS473, a marker of mTORC2. So the role of mTORC2 in cyst growth was determined. Knockout of Rictor (mTORC2) in Pkd1 knockout mice did not change the PKD phenotype. In summary, 2-DG decreases proliferation in cells lining the cyst and decreases cyst growth by decreasing proteins that are known to promote proliferation. In conclusion, the present study reinforces the therapeutic potential of 2-DG for use in patients with ADPKD.


Asunto(s)
Proliferación Celular , Desoxiglucosa , Modelos Animales de Enfermedad , Riñón , Riñón Poliquístico Autosómico Dominante , Animales , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Desoxiglucosa/farmacología , Desoxiglucosa/uso terapéutico , Ratones , Proliferación Celular/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Riñón/efectos de los fármacos , Canales Catiónicos TRPP/metabolismo , Autofagia/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Biomacromolecules ; 25(9): 6164-6180, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39164913

RESUMEN

Prostate cancer (PC) is the fifth leading cause of cancer-related deaths among men worldwide. Prostate-specific membrane antigen (PSMA), a molecular target of PC, is clinically used for the treatment and diagnosis of PC using radioligand approaches. However, no PSMA-based chemotherapies have yet been approved by the FDA. Here, we present a novel therapeutic approach using PSMA-targeted 2-deoxyglucose-dendrimer (PSMA-2DG-D) for targeted delivery of a potent tyrosine kinase inhibitor, cabozantinib (Cabo), selectively to PC cells. PSMA-2DG-D demonstrates intracellular localization in PSMA (+) PC cells through PSMA-mediated internalization. This PSMA-specific targeting translates to enhanced efficacy of Cabo compared to the free drug when conjugated to PSMA-2DG-D. Furthermore, systemically administered fluorescently labeled PSMA-2DG-D-Cy5 specifically targets PSMA (+) tumors with minimal off-target accumulation in the PC3-PIP tumor xenograft mouse model. This demonstrates that the PSMA-2DG-D platform is a promising new delivery system for potent chemotherapeutics, where systemic side effects are a significant concern.


Asunto(s)
Antígenos de Superficie , Dendrímeros , Desoxiglucosa , Glutamato Carboxipeptidasa II , Neoplasias de la Próstata , Piridinas , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Animales , Ratones , Desoxiglucosa/farmacología , Desoxiglucosa/química , Piridinas/química , Piridinas/administración & dosificación , Piridinas/farmacología , Glutamato Carboxipeptidasa II/metabolismo , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Dendrímeros/química , Antígenos de Superficie/metabolismo , Anilidas/farmacología , Anilidas/administración & dosificación , Anilidas/farmacocinética , Anilidas/química , Nanomedicina/métodos , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos , Sistemas de Liberación de Medicamentos/métodos
7.
Sheng Li Xue Bao ; 76(4): 517-525, 2024 Aug 25.
Artículo en Chino | MEDLINE | ID: mdl-39192785

RESUMEN

The aim of this study was to investigate whether the protective effect of 2-deoxyglucose (2-DG) on lung ischemia/reperfusion (I/R) injury is mediated by inhibiting nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-mediated pyroptosis in rats. Male Sprague-Dawley rats were randomly divided into control group, 2-DG group, lung I/R injury group (I/R group) and 2-DG+I/R group. 2-DG (0.7 g/kg) was intraperitoneally injected 1 h prior to lung ischemia. The tissue structure was measured under light microscope. Lung injury parameters were detected. The contents of malondialdehyde (MDA), myeloperoxidase (MPO) and lactate were determined by commercially available kits. ELISA was used to detect the levels of IL-1ß and IL-18. Western blot, qRT-PCR and immunofluorescence staining were used to measure the expression changes of glycolysis and pyroptosis related indicators. The results showed that there was no significant difference in the parameters between the control group and the 2-DG group. However, the lung injury parameters, oxidative stress response, lactic acid content, IL-1ß, and IL-18 levels were significantly increased in the I/R group. The protein expression levels of glycolysis and pyroptosis related indicators including hexokinase 2 (HK2), pyruvate kinase 2 (PKM2), NLRP3, Gasdermin superfamily member GSDMD-N, cleaved-Caspase1, cleaved-IL-1ß and cleaved-IL-18, and the gene expression levels of HK2, PKM2 and NLRP3 were markedly up-regulated in the I/R group compared with those in the control group. The expression of HK2 and NLRP3 was also increased detected by immunofluorescence staining. Compared with the I/R group, the 2-DG+I/R group exhibited significantly improved alveolar structure and inflammatory infiltration, reduced lung injury parameters, and decreased expression of glycolysis and pyroptosis related indicators. These results suggest that 2-DG protects against lung I/R injury possibly by inhibiting NLRP3-mediated pyroptosis in rats.


Asunto(s)
Desoxiglucosa , Pulmón , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Ratas Sprague-Dawley , Daño por Reperfusión , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Masculino , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Ratas , Pulmón/metabolismo , Pulmón/patología , Desoxiglucosa/farmacología , Interleucina-1beta/metabolismo , Interleucina-18/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/prevención & control , Lesión Pulmonar/etiología , Estrés Oxidativo
8.
J Agric Food Chem ; 72(34): 19131-19142, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39145730

RESUMEN

Fructose occurs in foods and as a metabolite in vivo. It can be degraded, leading to the formation of reactive carbonyl compounds, which may influence food properties and have an impact on health. The present study performed an in-depth qualitative and quantitative profiling of fructose degradation products. Thus, the α-dicarbonyl compounds 3-deoxyglucosone, glucosone, methylglyoxal, glyoxal, hydroxypyruvaldehyde, threosone, 3-deoxythreosone, and 1-desoxypentosone and the monocarbonyl compounds formaldehyde, acetaldehyde, glycolaldehyde, glyceraldehyde, and dihydroxyacetone were detected in fructose solutions incubated at 37 °C. Quantitative profiling after 7 days revealed 4.6-271.6-fold higher yields of all degradation products from fructose compared to glucose. Except for 3-deoxyglucosone, the product formation appeared to be metal dependent, indicating oxidative pathways. CaCl2 and MgCl2 partially reduced fructose degradation. Due to its high reactivity compared to glucose, particularly toward metal-catalyzed pathways, fructose may be a strong contributor to sugar degradation and Maillard reaction in foods and in vivo.


Asunto(s)
Fructosa , Glucosa , Fructosa/química , Fructosa/metabolismo , Glucosa/metabolismo , Glucosa/química , Reacción de Maillard , Oxidación-Reducción , Glioxal/química , Glioxal/metabolismo , Desoxiglucosa/análogos & derivados
9.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(6): 875-882, 2024 Jun 06.
Artículo en Chino | MEDLINE | ID: mdl-38955736

RESUMEN

Objective: To explore the relationship between serum 1, 5-dehydratoglucitol (1, 5-AG) level and insulin resistance, microvascular complications in patients with type 2 diabetes mellitus (T2DM). Methods: The clinical data of 836 patients with T2DM admitted to the Changsha Central Hospital Affiliated to University of South China from May to December 2023 were retrospectively and cross-sectionally analyzed. Serum 1, 5-AG levels were detected by pyranose oxidase method. According to the microvascular complications (diabetic peripheral neuropathy, diabetic nephropathy, diabetic retinopathy), the patients were divided into simple group (no microvascular complications, n=490), complication group 1 (1 microvascular complications, n=217), and complication group 2 (2 or more microvascular complications, n=129). The relationship between serum 1, 5-AG level and the related indicators of insulin resistance in T2DM patients were explored by Spearman correlation analysis, and the influencing factors of microvascular complications in T2DM patients were explored by multiple ordered logistic regression analysis. Results: The levels of FBG(fasting blood glucose) [(7.37±0.56) mmol/L], FINS(fasting insulin) [(11.34±1.86) mU/L] and HOMA-IR(homeostatic model assessment of insulin resistance) (0.96±0.31) in simple group were lower than those in complication group 1 [(8.37±1.02) mmol/L, (16.26±2.32) mU/L, (1.32±0.41)], complication group 2 [(10.25±2.13) mmol/L, (18.53±2.67) mU/L, (1.54±0.44)], and FBG, FINS and HOMA-IR in complication group 1 were lower than those in complication group 2, and the differences were statistically significant (F=537.470, 791.690, 136.340, P<0.001). Serum 1, 5-AG level in simple group [77.16 (16.30, 128.07) µg/ml] was higher than that in complication group 1 [51.05 (14.67, 63.18) µg/ml] and complication group 2 [30.42 (12.53, 47.26) µg/ml], and the serum level of 1, 5-AG in complication group 1 was higher than that in complication group 2, and the difference was statistically significant (H=210.020, P<0.001). The results of Spearman correlation analysis showed that serum 1, 5-AG level was negatively correlated with FBG, FINS and HOMA-IR in T2DM patients (r=-0.431, -0.372, -0.546, P<0.001). The results of multiple ordered logistic regression analysis showed that Longer duration of diabetes (OR=2.261, 95%CI: 1.564-3.269), increased HbA1c (OR=2.040, 95%CI: 1.456-2.858), and increased HOMA-IR (OR=2.158, 95%CI: 1.484-3.137) and decreased 1, 5-AG (OR=2.512, 95%CI: 1.691-3.732) were independent risk factors for microvascular complications in T2DM patients (P<0.05). The results of ROC curve analysis showed that the area under the curve of serum 1, 5-AG in the identification of one microvascular complication was 0.763 (95%CI: 0.731-0.795), and the area under the curve of serum 1, 5-AG in the identification of two or more microvascular complications was 0.730 (95%CI: 0.692-0.767). Conclusion: Serum 1, 5-AG level is negatively correlated with insulin resistance in T2DM patients. Low serum 1, 5-AG level may be an independent risk factor for microvascular complications in T2DM patients.


Asunto(s)
Desoxiglucosa , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Estudios Transversales , Estudios Retrospectivos , Desoxiglucosa/sangre , Desoxiglucosa/análogos & derivados , Glucemia , Masculino , Femenino , Insulina/sangre , Persona de Mediana Edad , Angiopatías Diabéticas/sangre
10.
J Control Release ; 372: 715-727, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38955253

RESUMEN

Chemo-immunotherapy holds the advantage of specific antitumor effects by activating cytotoxic lymphocyte cells (CTLs) immune response. However, multiple barriers have limited the outcomes partly due to tumor-cell-mediated exhaustion of CTLs in the immunosuppressive tumor microenvironment (iTME). Here, we rationally designed a simple-yet-versatile Ca2+ nanogenerator to modulate iTME for enhancing 2-deoxyglucose (2-DG) mediated chemo-immunotherapy. Briefly, after 2-DG chemotherapy, CaO2 nanoparticles coated with EL4 cell membrane (denoted as CaNP@ECM) could preferentially accumulate in tumor tissue via adhesion between LFA-1 on EL4 cell membrane and ICAM-1 on inflamed endothelial cell in tumor tissues and display a series of benefits for CTLs: i) Increasing glucose availability of CTLs while reducing lactic acid secretion through Ca2+ overloading mediated inhibition of tumor cell glycolysis, as well as relieving hypoxia; ii) Reversing CTLs exhaustion via TGF-ß1 scavenging and PD-L1 blockade through PD-1 and TGF-ß1R on EL4 cell membrane; iii) Boosting tumor immunotherapy via immunologic death (ICD) of tumor cells induced by Ca2+ overloading. We demonstrate that the multi-modal Ca2+ nanogenerator rescues T cells from exhaustion and inhibits tumor growth both in vitro and in vivo. More importantly, the study also facilitate the development of glucose metabolism inhibition-based tumor immunotherapy via Ca2+ overloading.


Asunto(s)
Calcio , Desoxiglucosa , Inmunoterapia , Ratones Endogámicos C57BL , Nanopartículas , Microambiente Tumoral , Animales , Inmunoterapia/métodos , Desoxiglucosa/farmacología , Desoxiglucosa/administración & dosificación , Nanopartículas/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Calcio/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Ratones , Femenino , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Óxidos , Humanos , Ratones Endogámicos BALB C , Agotamiento de Células T , Compuestos de Calcio
11.
Cell Biochem Biophys ; 82(3): 2285-2296, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38824236

RESUMEN

Fatty acid synthase (FASN) catalyzes the rate-limiting step of cellular lipogenesis. FASN expression is upregulated in various types of cancer cells, implying that FASN is a potential target for cancer therapy. 2-Deoxy-D-glucose (2-DG) specifically targets cancer cells by inhibiting glycolysis and glucose metabolism, resulting in multiple anticancer effects. However, whether the effects of 2-DG involve lipogenic metabolism remains to be elucidated. We investigated the effect of 2-DG administration on FASN expression in HeLa human cervical cancer cells. 2-DG treatment for 24 h decreased FASN mRNA and protein levels and suppressed the activity of an exogenous rat Fasn promoter. The use of a chemical activator or inhibitors or of a mammalian expression plasmid showed that neither AMPK nor the Sp1 transcription factor is responsible for the inhibitory effect of 2-DG on FASN expression. Administration of thapsigargin, an endoplasmic reticulum (ER) stress inducer, or 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF), a site 1 protease inhibitor, mimicked the inhibitory effect of 2-DG on FASN expression. 2-DG did not further decrease FASN expression in the presence of thapsigargin or AEBSF. Site 1 protease mediates activation of ATF6, an ER stress mediator, as well as sterol regulatory element-binding protein 1 (SREBP1), a robust transcription factor for FASN. Administration of 2-DG or thapsigargin for 24 h suppressed activation of ATF6 and SREBP1, as did AEBSF. We speculated that these effects of 2-DG or thapsigargin are due to feedback inhibition via increased GRP78 expression following ER stress. Supporting this, exogenous overexpression of GRP78 in HeLa cells suppressed SREBP1 activation and Fasn promoter activity. These results suggest that 2-DG suppresses FASN expression via an ER stress-dependent pathway, providing new insight into the molecular basis of FASN regulation in cancer.


Asunto(s)
Desoxiglucosa , Regulación hacia Abajo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Animales , Humanos , Ratas , Factor de Transcripción Activador 6/metabolismo , Factor de Transcripción Activador 6/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Desoxiglucosa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Acido Graso Sintasa Tipo I/metabolismo , Acido Graso Sintasa Tipo I/genética , Ácido Graso Sintasas/metabolismo , Ácido Graso Sintasas/genética , Células HeLa , Regiones Promotoras Genéticas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Tapsigargina/farmacología
12.
Free Radic Biol Med ; 222: 317-330, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38944213

RESUMEN

Mitochondrial transporters facilitate the translocation of metabolites between the cytoplasm and mitochondria and are critical for mitochondrial functional integrity. Although many mitochondrial transporters are associated with metabolic diseases, how they regulate mitochondrial function and their metabolic contributions at the cellular level are largely unknown. Here, we show that mitochondrial thiamine pyrophosphate (TPP) transporter SLC25A19 is required for mitochondrial respiration. SLC25A19 deficiency leads to reduced cell viability, increased integrated stress response (ISR), enhanced glycolysis and elevated cell sensitivity to 2-deoxyglucose (2-DG) treatment. Through a series of biochemical assays, we found that the depletion of mitochondrial NADH is the primary cause of the impaired mitochondrial respiration in SLC25A19 deficient cells. We also showed involvement of SLC25A19 in regulating the enzymatic activities of complexes I and III, the tricarboxylic acid (TCA) cycle, malate-aspartate shuttle and amino acid metabolism. Consistently, addition of idebenone, an analog of coenzyme Q10, restores mitochondrial respiration and cell viability in SLC25A19 deficient cells. Together, our findings provide new insight into the functions of SLC25A19 in mitochondrial and cellular physiology, and suggest that restoring mitochondrial respiration could be a novel strategy for treating SLC25A19-associated disorders.


Asunto(s)
Homeostasis , Mitocondrias , NAD , Humanos , Células A549 , Respiración de la Célula , Supervivencia Celular , Ciclo del Ácido Cítrico , Desoxiglucosa/farmacología , Desoxiglucosa/metabolismo , Glucólisis , Células K562 , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , NAD/metabolismo , Ubiquinona/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacología
13.
Acta Biomater ; 184: 368-382, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908417

RESUMEN

Starvation therapy aims to "starve" tumor cells by cutting off their nutritional supply. However, due to the complex and varied energy metabolism of tumors, targeting a single nutrient supply often fails to yield significant therapeutic benefits. This study proposes a tumor energy cocktail therapy that combines metformin, an oxidative phosphorylation inhibitor, with 2-deoxy-d-glucose (2-DG), a glycolysis inhibitor, to target tumor cells. To minimize the dosage of both drugs, we have developed a drug delivery strategy that prepared metformin as a nanoderivative, denoted as MA-dots. These MA-dots not only preserve the antitumor properties of metformin but also serve as a targeted delivery platform for 2-DG, ensuring its direct reach to the tumor site. Upon reaching the acidic tumor environment, the composite disintegrates, releasing 2-DG to inhibit glycolysis by targeting hexokinase 2 (HK2), the key enzyme in glycolysis, while MA-dots inhibit mitochondrial OXPHOS. This dual action significantly reduces ATP production in tumor cells, leading to apoptosis. In human lung tumor cells, the half-maximal inhibitory concentration (IC50) of 2-DG@MA-dots was significantly lower than that of either metformin or 2-DG alone, showing a nearly 100-fold and 30-fold reduction in IC50 values to 11.78 µg mL-1, from 1159 µg mL-1 and 351.20 µg mL-1, respectively. In studies with A549 tumor-bearing mice, the combination of low-dose 2-DG and metformin did not impede tumor growth, whereas 2-DG@MA-dots markedly decreased tumor volume, with the mean final tumor volume in the combination treatment group being approximately 89 times greater than that in the 2-DG@MA-dot group. STATEMENT OF SIGNIFICANCE: Metformin is a promising antitumor agent capable of modulating mitochondrial oxidative phosphorylation to inhibit cancer growth. However, its antitumor efficacy is limited when used alone due to compensatory energy mechanisms. Hence, we introduced glycolysis inhibitor 2-deoxy-d-glucose (2-DG) to inhibit an alternative tumor energy pathway. In our study, we developed a drug delivery strategy using metformin-derived nanomedicine (MA-dots) to load 2-DG. This approach enables the co-delivery of both drugs and their synergistic effect at the tumor site, disrupting both energy pathways and introducing an innovative "energy cocktail therapy".


Asunto(s)
Desoxiglucosa , Metformina , Humanos , Desoxiglucosa/farmacología , Metformina/farmacología , Metformina/uso terapéutico , Animales , Ratones , Ratones Desnudos , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Hexoquinasa/metabolismo , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Células A549 , Antineoplásicos/farmacología , Antineoplásicos/química
14.
Food Chem ; 456: 139965, 2024 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-38852460

RESUMEN

Pasteurisation and spray drying are critical steps to ensure the safety and shelf-life of formulae, but these treatments also induce formation of some potentially harmful Maillard reaction products. In this study, the occurrence of potentially harmful Maillard reaction products and proximate compositions in different commercial formulae were analysed. Our results showed that infant formulae had significantly higher concentrations of furosine, Nε-(carboxymethyl)lysine (CML) and Nε-(carboxyethyl)lysine (CEL) than follow-on/toddler formula. Specialty formulae had higher concentrations of glyoxal and CML than other types of formulae. Correlation analysis indicated that concentrations of 5-hydroxymethylfurfural, 3-deoxyglucosone, CML and CEL were closely related to fat contents. These results provided insight into concentrations of potentially harmful Maillard reaction products in different types of formulae and provide a theoretical basis for further optimisation of processing.


Asunto(s)
Fórmulas Infantiles , Lisina , Reacción de Maillard , Fórmulas Infantiles/química , Fórmulas Infantiles/análisis , Lisina/química , Lisina/análogos & derivados , Lisina/análisis , Humanos , Furaldehído/análogos & derivados , Furaldehído/análisis , Furaldehído/química , Glioxal/química , Glioxal/análisis , Lactante , Desoxiglucosa/análogos & derivados , Desoxiglucosa/química , Desoxiglucosa/análisis
15.
Biomed Pharmacother ; 176: 116841, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38834004

RESUMEN

Metastasis is the leading cause of cancer-related deaths, making the development of novel, more effective therapies imperative to alleviate patient suffering. Metabolic switching is a hallmark of cancer cells that facilitates metastasis. Cancer cells obtain most of their energy and intermediate metabolites, which are required to proliferate and metastasize, through aerobic glycolysis. Previous work from our laboratory has shown that Caveolin-1 (CAV1) expression in cancer cells promotes glycolysis and metastasis. Here, we sought to determine if limiting glycolysis reduced CAV1-enhanced metastasis and to identify the mechanism(s) involved. We evaluated the effects of the glycolysis inhibitor 2-deoxy-D-glucose (2-DG) in metastatic melanoma and breast cancer cell lines expressing or not CAV1. Non-cytotoxic concentrations of 2-DG (1 mM) inhibited the migration of B16-F10 melanoma and MDA-MB-231 breast cancer cells. CAV1-mediated activation of Src/Akt signaling was required for CAV1-enhanced migration and was blocked in the presence of 2-DG. Moreover, inhibition of Akt reduced CAV1-enhanced lung metastasis of B16-F10 cells. Collectively, these findings highlight the importance of CAV1-induced metabolic reprogramming for metastasis and point towards possible therapeutic approaches to prevent metastatic disease by inhibiting glycolysis and Src/Akt signaling.


Asunto(s)
Caveolina 1 , Movimiento Celular , Desoxiglucosa , Glucólisis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Familia-src Quinasas , Caveolina 1/metabolismo , Glucólisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Humanos , Línea Celular Tumoral , Ratones , Movimiento Celular/efectos de los fármacos , Desoxiglucosa/farmacología , Femenino , Metástasis de la Neoplasia , Melanoma Experimental/patología , Melanoma Experimental/metabolismo , Melanoma Experimental/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Ratones Endogámicos C57BL
16.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891879

RESUMEN

One aspect of ovarian tumorigenesis which is still poorly understood is the tumor-stroma interaction, which plays a major role in chemoresistance and tumor progression. Cancer-associated fibroblasts (CAFs), the most abundant stromal cell type in the tumor microenvironment, influence tumor growth, metabolism, metastasis, and response to therapy, making them attractive targets for anti-cancer treatment. Unraveling the mechanisms involved in CAFs activation and maintenance is therefore crucial for the improvement of therapy efficacy. Here, we report that CAFs phenoconversion relies on the glucose-dependent inhibition of autophagy. We show that ovarian cancer cell-conditioning medium induces a metabolic reprogramming towards the CAF-phenotype that requires the autophagy-dependent glycolytic shift. In fact, 2-deoxy-D-glucose (2DG) strongly hampers such phenoconversion and, most importantly, induces the phenoreversion of CAFs into quiescent fibroblasts. Moreover, pharmacological inhibition (by proline) or autophagy gene knockdown (by siBECN1 or siATG7) promotes, while autophagy induction (by either 2DG or rapamycin) counteracts, the metabolic rewiring induced by the ovarian cancer cell secretome. Notably, the nutraceutical resveratrol (RV), known to inhibit glucose metabolism and to induce autophagy, promotes the phenoreversion of CAFs into normal fibroblasts even in the presence of ovarian cancer cell-conditioning medium. Overall, our data support the view of testing autophagy inducers for targeting the tumor-promoting stroma as an adjuvant strategy to improve therapy success rates, especially for tumors with a highly desmoplastic stroma, like ovarian cancer.


Asunto(s)
Autofagia , Fibroblastos Asociados al Cáncer , Glucosa , Neoplasias Ováricas , Humanos , Femenino , Autofagia/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/tratamiento farmacológico , Glucosa/metabolismo , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Resveratrol/farmacología , Medios de Cultivo Condicionados/farmacología , Desoxiglucosa/farmacología , Glucólisis/efectos de los fármacos
17.
Theranostics ; 14(8): 3221-3245, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38855177

RESUMEN

The availability of non-invasive drug delivery systems capable of efficiently transporting bioactive molecules across the blood-brain barrier to specific cells at the injury site in the brain is currently limited. Delivering drugs to neurons presents an even more formidable challenge due to their lower numbers and less phagocytic nature compared to other brain cells. Additionally, the diverse types of neurons, each performing specific functions, necessitate precise targeting of those implicated in the disease. Moreover, the complex synthetic design of drug delivery systems often hinders their clinical translation. The production of nanomaterials at an industrial scale with high reproducibility and purity is particularly challenging. However, overcoming this challenge is possible by designing nanomaterials through a straightforward, facile, and easily reproducible synthetic process. Methods: In this study, we have developed a third-generation 2-deoxy-glucose functionalized mixed layer dendrimer (2DG-D) utilizing biocompatible and cost-effective materials via a highly facile convergent approach, employing copper-catalyzed click chemistry. We further evaluated the systemic neuronal targeting and biodistribution of 2DG-D, and brain delivery of a neuroprotective agent pioglitazone (Pio) in a pediatric traumatic brain injury (TBI) model. Results: The 2DG-D exhibits favorable characteristics including high water solubility, biocompatibility, biological stability, nanoscale size, and a substantial number of end groups suitable for drug conjugation. Upon systemic administration in a pediatric mouse model of traumatic brain injury (TBI), the 2DG-D localizes in neurons at the injured brain site, clears rapidly from off-target locations, effectively delivers Pio, ameliorates neuroinflammation, and improves behavioral outcomes. Conclusions: The promising in vivo results coupled with a convenient synthetic approach for the construction of 2DG-D makes it a potential nanoplatform for addressing brain diseases.


Asunto(s)
Dendrímeros , Desoxiglucosa , Sistemas de Liberación de Medicamentos , Neuronas , Animales , Dendrímeros/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Desoxiglucosa/farmacología , Desoxiglucosa/farmacocinética , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ratones , Pioglitazona/farmacología , Pioglitazona/administración & dosificación , Pioglitazona/farmacocinética , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encefalopatías/tratamiento farmacológico , Humanos , Modelos Animales de Enfermedad , Distribución Tisular , Masculino
18.
Nat Commun ; 15(1): 4665, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821965

RESUMEN

Minimally invasive thermal therapy is a successful alternative treatment to surgery in solid tumors with high complete ablation rates, however, tumor recurrence remains a concern. Central memory CD8+ T cells (TCM) play important roles in protection from chronic infection and cancer. Here we find, by single-cell RNA analysis of human breast cancer samples, that although the memory phenotype of peripheral CD8+ T cells increases slightly after microwave ablation (MWA), the metabolism of peripheral CD8+ T cells remains unfavorable for memory phenotype. In mouse models, glycolysis inhibition by 2-deoxy-D-glucose (2DG) in combination with MWA results in long-term anti-tumor effect via enhancing differentiation of tumor-specific CD44hiCD62L+CD8+ TCM cells. Enhancement of CD8+ TCM cell differentiation determined by Stat-1, is dependent on the tumor-draining lymph nodes (TDLN) but takes place in peripheral blood, with metabolic remodeling of CD8+ T cells lasting the entire course of the the combination therapy. Importantly, in-vitro glycolysis inhibition in peripheral CD8+ T cells of patients with breast or liver tumors having been treated with MWA thrice leads to their differentiation into CD8+ TCM cells. Our work thus offers a potential strategy to avoid tumor recurrence following MWA therapy and lays down the proof-of-principle for future clinical trials.


Asunto(s)
Neoplasias de la Mama , Linfocitos T CD8-positivos , Diferenciación Celular , Glucólisis , Memoria Inmunológica , Microondas , Glucólisis/efectos de los fármacos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Diferenciación Celular/efectos de los fármacos , Ratones , Femenino , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Microondas/uso terapéutico , Desoxiglucosa/farmacología , Desoxiglucosa/uso terapéutico , Línea Celular Tumoral , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Células T de Memoria/inmunología , Células T de Memoria/metabolismo
19.
J Food Sci ; 89(6): 3455-3468, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38700315

RESUMEN

Excessive accumulation of advanced glycation end products (AGEs) in the body is associated with diabetes and its complications. In this study, we aimed to explore the potential and mechanism of coffee leaf extract (CLE) in inhibiting the generation of AGEs and their precursors in an in vitro glycation model using bovine serum albumin and glucose (BSA-Glu) for the first time. High-performance liquid chromatography analysis revealed that CLE prepared with ultrasound pretreatment (CLE-U) contained higher levels of trigonelline, mangiferin, 3,5-dicaffeoylquinic acid, and γ-aminobutyric acid than CLE without ultrasound pretreatment (CLE-NU). The concentrations of these components, along with caffeine and rutin, were dramatically decreased when CLE-U or CLE-NU was incubated with BSA-Glu reaction mixture. Both CLE-U and CLE-NU exhibited a dose-dependent inhibition of fluorescent AGEs, carboxymethyllysine, fructosamine, 5-hydroxymethylfurfural, 3-deoxyglucosone, glyoxal, as well as protein oxidation products. Notably, CLE-U exhibited a higher inhibitory capacity compared to CLE-NU. CLE-U effectively quenched fluorescence intensity and increased the α-helix structure of the BSA-Glu complex. Molecular docking results suggested that the key bioactive compounds present in CLE-U interacted with the arginine residues of BSA, thereby preventing its glycation. Overall, this research sheds light on the possible application of CLE as a functional ingredient in combating diabetes by inhibiting the generation of AGEs.


Asunto(s)
Productos Finales de Glicación Avanzada , Extractos Vegetales , Hojas de la Planta , Albúmina Sérica Bovina , Extractos Vegetales/farmacología , Extractos Vegetales/química , Hojas de la Planta/química , Albúmina Sérica Bovina/química , Coffea/química , Alcaloides/farmacología , Furaldehído/análogos & derivados , Furaldehído/farmacología , Fructosamina , Cromatografía Líquida de Alta Presión , Glioxal , Glucosa/metabolismo , Simulación del Acoplamiento Molecular , Glicosilación/efectos de los fármacos , Ácido Quínico/análogos & derivados , Ácido Quínico/farmacología , Rutina/farmacología , Lisina/análogos & derivados , Cafeína/farmacología , Desoxiglucosa/análogos & derivados , Desoxiglucosa/farmacología , Xantonas
20.
Biomed Pharmacother ; 175: 116776, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38788546

RESUMEN

Choroidal neovascularization (CNV), characterized as a prominent feature of wet age-related macular degeneration (AMD), is a primary contributor to visual impairment and severe vision loss globally, while the prevailing treatments are often unsatisfactory. The development of conventional treatment strategies has largely been based on the understanding that the angiogenic switch of endothelial cells is dictated by angiogenic growth factors alone. Even though treatments targeting vascular endothelial growth factor (VEGF), like Ranibizumab, are widely administered, more than half of the patients still exhibit inadequate or null responses, emphasizing the imperative need for solutions to this problem. Here, aiming to explore therapeutic strategies from a novel perspective of endothelial cell metabolism, a biocompatible nanomedicine delivery system is constructed by loading RGD peptide-modified liposomes with 2-deoxy-D-glucose (RGD@LP-2-DG). RGD@LP-2-DG displayed good targeting performance towards endothelial cells and excellent in vitro and in vivo inhibitory effects on neovascularization were demonstrated. Moreover, our mechanistic studies revealed that 2-DG interfered with N-glycosylation, leading to the inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) and its downstream signaling. Notably, the remarkable inhibitory effect on neovascularization and biocompatibility of RGD@LP-2-DG render it a highly promising and clinically translatable therapeutic candidate for the treatment of wet AMD and other angiogenic diseases, particularly in patients who are unresponsive to currently available treatments.


Asunto(s)
Neovascularización Coroidal , Desoxiglucosa , Liposomas , Nanomedicina , Oligopéptidos , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Degeneración Macular Húmeda , Oligopéptidos/química , Animales , Humanos , Nanomedicina/métodos , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/patología , Neovascularización Coroidal/metabolismo , Degeneración Macular Húmeda/tratamiento farmacológico , Degeneración Macular Húmeda/metabolismo , Desoxiglucosa/farmacología , Desoxiglucosa/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA