Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 875
Filtrar
1.
Neurosurg Rev ; 47(1): 679, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39317802

RESUMEN

Wu et al. (2021) investigated the neuroprotective effects of hypoxia preconditioning (HPC) in a rat model of traumatic brain injury (TBI). The study demonstrated that HPC enhances brain resilience to TBI by upregulating glucose transporters GLUT1 and GLUT3 through the HIF-1α signaling pathway. Comprehensive molecular and histological analyses confirmed increased expression of these transporters, correlating with reduced neuronal apoptosis and cerebral edema. The robust methodology, including rigorous statistical validation and time-course assessments, underscores HPC's potential therapeutic role in mitigating neuronal loss and improving glucose transport post-injury. However, the study could be strengthened by incorporating additional preconditioning controls, comparative analyses with other neuroprotective strategies, and exploring downstream metabolic effects in greater detail. Furthermore, expanding the research to include diverse animal models and examining sex-dependent responses would enhance the generalizability and translational relevance of the findings. Future studies should also integrate metabolic flux analysis and advanced imaging techniques to further elucidate HPC's mechanisms of action.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Glucosa , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neuronas , Transducción de Señal , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/terapia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratas , Glucosa/metabolismo , Transducción de Señal/fisiología , Neuronas/metabolismo , Precondicionamiento Isquémico/métodos , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo
2.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 1997-2009, 2024 Jul 25.
Artículo en Chino | MEDLINE | ID: mdl-39044571

RESUMEN

Glucose uptake by lymphocytes is dependent on the facilitative glucose transporters (GLUT1, GLUT3, GLUT4, and GLUT6) of the GLUT family and the Na+-coupled glucose transporter SGLT1. GLUTs and SGLTs are widely expressed in mammals, and their expression and functions may affect cell development, homeostasis, activation, and differentiation. This article details the important functions of several GLUTs and SGLTs in lymphocytes and points out that glucose transporters play a key role in supplying energy for lymphocytes, maintaining intracellular glucose homeostasis, and improving the efficiency of immune responses, which reflect their key roles in signal transduction. Probing into the effects of glucose transporters on lymphocyte functions will help to decipher the functioning mechanisms of lymphocytes in diseases. Furthermore, this paper prospects the application values of glucose transporters in lymphocytes from molecular biology, aiming to provide better strategies for the clinical treatment of lymphocyte-related diseases and promote the research and development of targeted therapeutic drugs.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa , Linfocitos , Linfocitos/inmunología , Linfocitos/metabolismo , Humanos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Glucosa/metabolismo , Animales , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética
3.
FASEB J ; 38(13): e23800, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38979931

RESUMEN

Insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM), has emerged as a pathological feature in Alzheimer's disease (AD). Given the shared role of insulin resistance in T2DM and AD, repurposing peripheral insulin sensitizers is a promising strategy to preserve neuronal insulin sensitivity and prevent AD. 1-Deoxynojirimycin (DNJ), a bioactive iminosugar, exhibited insulin-sensitizing effects in metabolic tissues and was detected in brain tissue post-oral intake. However, its impact on brain and neuronal insulin signaling has not been described. Here, we investigated the effect of DNJ treatment on insulin signaling and AD markers in insulin-resistant human SK-N-SH neuroblastoma, a cellular model of neuronal insulin resistance. Our findings show that DNJ increased the expression of insulin signaling genes and the phosphorylation status of key molecules implicated in insulin resistance (Y1146-pIRß, S473-pAKT, S9-GSK3B) while also elevating the expression of glucose transporters Glut3 and Glut4, resulting in higher glucose uptake upon insulin stimuli. DNJ appeared to mitigate the insulin resistance-driven increase in phosphorylated tau and Aß1-42 levels by promoting insulin-induced phosphorylation of GSK3B (a major tau kinase) and enhancing mRNA expression of the insulin-degrading enzyme (IDE) pivotal for insulin and Aß clearance. Overall, our study unveils probable mechanisms underlying the potential benefits of DNJ for AD, wherein DNJ attenuates tau and amyloid pathologies by reversing neuronal insulin resistance. This provides a scientific basis for expanding the use of DNJ-containing products for neuroprotective purposes and prompts further research into compounds with similar mechanisms of action.


Asunto(s)
1-Desoxinojirimicina , Enfermedad de Alzheimer , Resistencia a la Insulina , Neuronas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Humanos , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/análogos & derivados , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Línea Celular Tumoral , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Transportador de Glucosa de Tipo 4/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Fosforilación/efectos de los fármacos , Biomarcadores/metabolismo
4.
Nutrients ; 16(14)2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39064806

RESUMEN

Glucose is the primary energy source for most mammalian cells and its transport is affected by a family of facilitative glucose transporters (GLUTs) encoded by the SLC2 gene. GLUT1 and GLUT3, highly expressed isoforms in the blood-brain barrier and neuronal membranes, respectively, are associated with multiple neurodevelopmental disorders including epilepsy, dyslexia, ADHD, and autism spectrum disorder (ASD). Dietary therapies, such as the ketogenic diet, are widely accepted treatments for patients with the GLUT1 deficiency syndrome, while ameliorating certain symptoms associated with GLUT3 deficiency in animal models. A ketogenic diet, high-fat diet, and calorie/energy restriction during prenatal and postnatal stages can also alter the placental and brain GLUTs expression with long-term consequences on neurobehavior. This review focuses primarily on the role of diet/energy perturbations upon GLUT isoform-mediated emergence of neurodevelopmental and neurodegenerative disorders.


Asunto(s)
Encéfalo , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 3 , Trastornos del Neurodesarrollo , Placenta , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Humanos , Embarazo , Encéfalo/metabolismo , Placenta/metabolismo , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Trastornos del Neurodesarrollo/etiología , Animales , Fenómenos Fisiologicos Nutricionales Maternos , Dieta Cetogénica , Dieta Alta en Grasa/efectos adversos , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Glucosa/metabolismo
5.
Cells ; 13(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38994998

RESUMEN

Iron is often accumulated in the liver during pathological conditions such as cirrhosis and cancer. Elevated expression of glucose transporters GLUT1 and GLUT3 is associated with reduced overall survival in patients with hepatocellular carcinoma. However, it is not known whether iron can regulate glucose transporters and contribute to tumor proliferation. In the present study, we found that treatment of human liver cell line HepG2 with ferric ammonium citrate (FAC) resulted in a significant upregulation of GLUT3 mRNA and protein in a dose-dependent manner. Similarly, iron accumulation in mice fed with high dietary iron as well as in mice injected intraperitoneally with iron dextran enhanced the GLUT3 expression drastically in the liver. We demonstrated that iron-induced hepatic GLUT3 upregulation is mediated by the LKB1/AMPK/CREB1 pathway, and this activation was reversed when treated with iron chelator deferiprone. In addition, inhibition of GLUT3 using siRNA prevented iron-mediated increase in the expression of cell cycle markers and cellular hyperproliferation. Furthermore, exogenous sodium beta-hydroxybutyrate treatment prevented iron-mediated hepatic GLUT3 activation both in vitro and in vivo. Together, these results underscore the importance of iron, AMPK, CREB1 and GLUT3 pathways in cell proliferation and highlight the therapeutic potential of sodium beta-hydroxybutyrate in hepatocellular carcinoma with high GLUT3 expression.


Asunto(s)
Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Transportador de Glucosa de Tipo 3 , Hierro , Hígado , Proliferación Celular/efectos de los fármacos , Animales , Humanos , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Células Hep G2 , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hierro/metabolismo , Masculino , Proteínas Quinasas Activadas por AMP/metabolismo , Compuestos de Amonio Cuaternario/farmacología , Compuestos Férricos/farmacología , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética
6.
Cancer Lett ; 598: 217109, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39002692

RESUMEN

Gemcitabine serves as a first-line chemotherapeutic treatment for pancreatic cancer (PC), but it is prone to rapid drug resistance. Increasing the sensitivity of PC to gemcitabine has long been a focus of research. Fasting interventions may augment the effects of chemotherapy and present new options. SIRT7 is known to link metabolism with various cellular processes through post-translational modifications. We found upregulation of SIRT7 in PC cells is associated with poor prognosis and gemcitabine resistance. Cross-analysis of RNA-seq and ATAC-seq data suggested that GLUT3 might be a downstream target gene of SIRT7. Subsequent investigations demonstrated that SIRT7 directly interacts with the enhancer region of GLUT3 to desuccinylate H3K122. Our group's another study revealed that GLUT3 can transport gemcitabine in breast cancer cells. Here, we found GLUT3 KD reduces the sensitivity of PC cells to gemcitabine, and SIRT7 KD-associated gemcitabine-sensitizing could be reversed by GLUT3 KD. While fasting mimicking induced upregulation of SIRT7 expression in PC cells, knocking down SIRT7 enhanced sensitivity to gemcitabine through upregulating GLUT3 expression. We further confirmed the effect of SIRT7 deficiency on the sensitivity of gemcitabine under fasting conditions using a mouse xenograft model. In summary, our study demonstrates that SIRT7 can regulate GLUT3 expression by binding to its enhancer and altering H3K122 succinylation levels, thus affecting gemcitabine sensitivity in PC cells. Additionally, combining SIRT7 knockdown with fasting may improve the efficacy of gemcitabine. This unveils a novel mechanism by which SIRT7 influences gemcitabine sensitivity in PC and offer innovative strategies for clinical combination therapy with gemcitabine.


Asunto(s)
Desoxicitidina , Resistencia a Antineoplásicos , Gemcitabina , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 3 , Neoplasias Pancreáticas , Sirtuinas , Regulación hacia Arriba , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Humanos , Sirtuinas/genética , Sirtuinas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Animales , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Ratones , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Antimetabolitos Antineoplásicos/farmacología , Técnicas de Silenciamiento del Gen , Ratones Desnudos , Femenino
7.
Mol Biol Rep ; 51(1): 727, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861014

RESUMEN

BACKGROUND: The presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and its receptor in various testicular cells and spermatozoa suggests a potential role in enhancing spermatogonial and postmeiotic cell development. Moreover, GM-CSF activates the pivotal pathways implicated in sperm motility regulation and glucose metabolism. However, the impact of GM-CSF on testicular biopsies from patients with obstructive azoospermia (OA) remains unexplored. Therefore, this study aimed to investigate the in vitro effects of GM-CSF on the expression of genes related to glucose transporters and signaling pathways, sperm motility, and viability in testicular biopsies. METHODS AND RESULTS: Following testicular sperm extraction from 20 patients diagnosed with OA, each sample was divided into two parts: the experimental samples were incubated with medium containing 2 ng/ml GM-CSF at 37 °C for 60 min, and the control samples were incubated with medium without GM-CSF. Subsequently, the oocytes retrieved from the partner were injected with sperm from the treatment and control groups. The sperm parameters (motility and viability), the expression levels of sperm motility-related genes (PIK3R1, PIK3CA, and AKT1), and the expression levels of sperm energy metabolism-related genes (GLUT1, GLUT3, and GLUT14) were assessed. Furthermore, the fertilization and day 3 embryo development rate and embryo quality were evaluated. Compared with those in the nontreated group, the motility parameters and the mRNA expression levels of PIK3R1, AKT1, and GLUT3 in testicular sperm supplemented with GM-CSF were significantly greater (p < 0.05). However, no significant differences in the mRNA expression of PIK3CA, GLUT1, or GLUT14 were detected. According to the ICSI results, compared with the control group, the GM-CSF treatment group exhibited significantly greater fertilization rates (p = 0.027), Day 3 embryo development rate (p = 0.001), and proportions of good-quality embryos (p = 0.002). CONCLUSIONS: GM-CSF increased the expression of genes related to motility and the energy metabolism pathway and effectively promoted the motility of testis-extracted spermatozoa, consequently yielding positive clinical outcomes.


Asunto(s)
Azoospermia , Metabolismo Energético , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Inyecciones de Esperma Intracitoplasmáticas , Motilidad Espermática , Adulto , Femenino , Humanos , Masculino , Azoospermia/genética , Azoospermia/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Transducción de Señal/efectos de los fármacos , Motilidad Espermática/efectos de los fármacos , Motilidad Espermática/genética , Espermatozoides/efectos de los fármacos , Testículo/metabolismo , Testículo/efectos de los fármacos
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(5): 894-903, 2024 May 20.
Artículo en Chino | MEDLINE | ID: mdl-38862447

RESUMEN

OBJECTIVE: To investigate the effect of Jiaotaiwan on brain insulin-PI3K/AKT pathway in a mouse model of Alzheimer's disease (AD). METHODS: Fifty 3-month-old male APP/PS1 double transgenic mice were randomized into AD model group, low-, medium- and high-dose Jiaotaiwan treatment groups, and donepezil treatment group. Cognitive functions of the mice were assessed using water maze and open field tests, and neuronal pathologies were observed with HE staining and Nissl staining; immunohistochemistry was used to detect amyloid Aß deposition in the brain. Fasting serum insulin levels of the mice were measured, and the expressions of Aß42, insulin-PI3K/AKT pathway components and downstream glucose transporters in the brain tissue were detected with RT-qPCR and Western blotting. RESULTS: The AD mouse models exhibited obvious impairment of learning and memory abilities, significantly reduced hippocampal neurons, and obvious Aß amyloid plaques in the brain tissue with increased Aß42 protein expression (P < 0.05) and insulin resistance index, decreased hippocampal PI3K expressions, lowered expressions of AKT and InR, reduced expressions of GLUT1, GLUT3, and GLUT4, and increased expression of GSK3ß in both the hippocampus and cortex. Treatment with Jiaotaiwan and donepezil both effectively improved memory ability of the mouse models, increased the number of hippocampal neurons, reduced Aß amyloid plaques and increased the expressions of PI3K, AKT, InR, GLUT1, GLUT3 and GLUT4 in the hippocampus and cortex. CONCLUSION: Jiaotaiwan improves learning and memory abilities of APP/PS1 double transgenic mice and delay the development of AD by activating the PI3K/AKT pathway and regulating the expression levels of its downstream GLUTs in the brain.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos , Glucosa , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Masculino , Medicamentos Herbarios Chinos/farmacología , Glucosa/metabolismo , Hipocampo/metabolismo , Péptidos beta-Amiloides/metabolismo , Insulina/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Memoria/efectos de los fármacos , Transportador de Glucosa de Tipo 3/metabolismo , Neuronas/metabolismo
9.
CNS Neurosci Ther ; 30(6): e14806, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38887182

RESUMEN

AIM: Glucose-dependent insulinotropic polypeptide (GIP) is a ligand of glucose-dependent insulinotropic polypeptide receptor (GIPR) that plays an important role in the digestive system. In recent years, GIP has been regarded as a hormone-like peptide to regulate the local metabolic environment. In this study, we investigated the antioxidant role of GIP on the neuron and explored the possible mechanism. METHODS: Cell counting Kit-8 (CCK-8) was used to measure cell survival. TdT-mediated dUTP Nick-End Labeling (TUNEL) was used to detect apoptosis in vitro and in vivo. Reactive oxygen species (ROS) levels were probed with 2', 7'-Dichloro dihydrofluorescein diacetate (DCFH-DA), and glucose intake was detected with 2-NBDG. Immunofluorescence staining and western blot were used to evaluate the protein level in cells and tissues. Hematoxylin-eosin (HE) staining, immunofluorescence staining and tract-tracing were used to observe the morphology of the injured spinal cord. Basso-Beattie-Bresnahan (BBB) assay was used to evaluate functional recovery after spinal cord injury. RESULTS: GIP reduced the ROS level and protected cells from apoptosis in cultured neurons and injured spinal cord. GIP facilitated wound healing and functional recovery of the injured spinal cord. GIP significantly improved the glucose uptake of cultured neurons. Meanwhile, inhibition of glucose uptake significantly attenuated the antioxidant effect of GIP. GIP increased glucose transporter 3 (GLUT3) expression via up-regulating the level of hypoxia-inducible factor 1α (HIF-1α) in an Akt-dependent manner. CONCLUSION: GIP increases GLUT3 expression and promotes glucose intake in neurons, which exerts an antioxidant effect and protects neuronal cells from oxidative stress both in vitro and in vivo.


Asunto(s)
Polipéptido Inhibidor Gástrico , Transportador de Glucosa de Tipo 3 , Glucosa , Neuronas , Estrés Oxidativo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Traumatismos de la Médula Espinal , Animales , Polipéptido Inhibidor Gástrico/farmacología , Polipéptido Inhibidor Gástrico/metabolismo , Glucosa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Masculino , Células Cultivadas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
10.
J Nutr Biochem ; 132: 109675, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38945454

RESUMEN

The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.


Asunto(s)
Encéfalo , Dieta Alta en Grasa , Trastornos de la Memoria , Efectos Tardíos de la Exposición Prenatal , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Embarazo , Encéfalo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Fenómenos Fisiologicos Nutricionales Maternos , Resistencia a la Insulina , Ratas Wistar , Glucosa/metabolismo , Factores Sexuales , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética
11.
Int. j. morphol ; 42(3): 805-813, jun. 2024. ilus, tab
Artículo en Inglés | LILACS | ID: biblio-1564593

RESUMEN

SUMMARY: Glucose has an essential role in the proliferation and survival of testicular tissue. Glucose transporters (GLUTs) are responsible for glucose uptake across cell membranes. In the present work, two main isoforms GLUT1 and GLUT3 were investigated in the testes of Laboratory mouse (BALB/c), Lesser Egyptian jerboa (Jaculus jaculus), Golden hamster (Mesocricetus auratus), and Desert Hedgehog (Paraechinus aethiopicus). Immunofluorescent localization of GLUT1 and GLUT3 showed considerable species differences. The lowest expression of GLUT1 and GLUT3 was localized in the testis of Laboratory mouse (BALB/c), the highest GLUT1 localization was detected in the testis of Lesser Egyptian jerboa (Jaculus jaculus), and the highest GLUT3 immunofluorescent localization was observed in the testis of Hedgehog (Paraechinus aethiopicus). The results imply that GLUT3 is the principal glucose transporter in the studied testes, which is related to species differences. The different immunolocalization of GLUT in examined testes suggests using various transport systems for energy gain in different species.


La glucosa tiene un papel esencial en la proliferación y supervivencia del tejido testicular. Los transportadores de glucosa (GLUT) son responsables de la absorción de glucosa a través de las membranas celulares. En el presente trabajo, se investigaron dos isoformas principales GLUT1 y GLUT3 en los testículos de un ratón de laboratorio (BALB/c), un jerbo egipcio menor (Jaculus jaculus), un hámster dorado (Mesocricetus auratus) y un erizo del desierto (Paraechinus aethiopicus). La localización inmunofluorescente de GLUT1 y GLUT3 mostró diferencias considerables entre especies. La expresión más baja de GLUT1 y GLUT3 se localizó en el testículo del ratón de laboratorio (BALB/c), la localización más alta de GLUT1 se detectó en el testículo del jerbo egipcio menor (Jaculus jaculus) y la localización inmunofluorescente de GLUT3 más alta se observó en el testículo de Erizo (Paraechinus aethiopicus). Los resultados implican que GLUT3 es el principal transportador de glucosa en los testículos estudiados, lo que está relacionado con diferencias entre especies. La diferente inmunolocalización de GLUT en los testículos examinados sugiere el uso de varios sistemas de transporte para ganar energía en diferentes especies.


Asunto(s)
Animales , Testículo/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Mamíferos , Ratones Endogámicos BALB C
12.
Cell Death Dis ; 15(5): 376, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811531

RESUMEN

The tumor margin as the invasive front has been proven to be closely related to the progression and metastasis of oral squamous cell carcinoma (OSCC). However, how tumor cells in the marginal region obtain the extra energy needed for tumor progression is still unknown. Here, we used spatial metabolomics and the spatial transcriptome to identify enhanced energy metabolism in the tumor margin of OSCC and identified that the downregulation of Ras-related glycolysis inhibitor and calcium channel regulator (RRAD) in tumor cells mediated this process. The absence of RRAD enhanced the ingestion of glucose and malignant behaviors of tumor cells both in vivo and in vitro. Mechanically, the downregulation of RRAD promoted the internal flow of Ca2+ and elevated its concentration in the nucleus, which resulted in the activation of the CAMKIV-CREB1 axis to induce the transcription of the glucose transporter GLUT3. GLUT inhibitor-1, as an inhibitor of GLUT3, could suppress this vigorous energy metabolism and malignant behaviors caused by the downregulation of RRAD. Taken together, our study revealed that enhanced energy metabolism in the tumor margin mediated by RRAD promotes the progression of OSCC and proved that GLUT3 is a potential target for future treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Progresión de la Enfermedad , Metabolismo Energético , Neoplasias de la Boca , Humanos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Animales , Línea Celular Tumoral , Ratones , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Ratones Endogámicos BALB C , Glucosa/metabolismo , Calcio/metabolismo , Glucólisis
13.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38731997

RESUMEN

Glucose, the primary energy substrate for fetal oxidative processes and growth, is transferred from maternal to fetal circulation down a concentration gradient by placental facilitative glucose transporters. In sheep, SLC2A1 and SLC2A3 are the primary transporters available in the placental epithelium, with SLC2A3 located on the maternal-facing apical trophoblast membrane and SLC2A1 located on the fetal-facing basolateral trophoblast membrane. We have previously reported that impaired placental SLC2A3 glucose transport resulted in smaller, hypoglycemic fetuses with reduced umbilical artery insulin and glucagon concentrations, in addition to diminished pancreas weights. These findings led us to subject RNA derived from SLC2A3-RNAi (RNA interference) and NTS-RNAi (non-targeting sequence) fetal pancreases to qPCR followed by transcriptomic analysis. We identified a total of 771 differentially expressed genes (DEGs). Upregulated pathways were associated with fat digestion and absorption, particularly fatty acid transport, lipid metabolism, and cholesterol biosynthesis, suggesting a potential switch in energetic substrates due to hypoglycemia. Pathways related to molecular transport and cell signaling in addition to pathways influencing growth and metabolism of the developing pancreas were also impacted. A few genes directly related to gluconeogenesis were also differentially expressed. Our results suggest that fetal hypoglycemia during the first half of gestation impacts fetal pancreas development and function that is not limited to ß cell activity.


Asunto(s)
Hipoglucemia , Páncreas , Placenta , Interferencia de ARN , Transcriptoma , Embarazo , Animales , Femenino , Placenta/metabolismo , Ovinos , Páncreas/metabolismo , Páncreas/embriología , Hipoglucemia/genética , Hipoglucemia/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Feto/metabolismo , Desarrollo Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Glucosa/metabolismo , Perfilación de la Expresión Génica
14.
Sci Rep ; 14(1): 12051, 2024 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802412

RESUMEN

GDM, as a metabolic disease during pregnancy, regulates GLUT3 translocation by AMPK, thereby affecting glucose uptake in trophoblasts. It provides a new research idea and therapeutic target for alleviating intrauterine hyperglycemia in GDM. STZ was used to construct GDM mice, inject AICAR into pregnant mice, and observe fetal and placental weight; flow cytometry was employed for the detection of glucose uptake by primary trophoblast cells; immunofluorescence was applied to detect the localization of GLUT3 and AMPK in placental tissue; Cocofal microscope was used to detect the localization of GLUT3 in trophoblast cells;qRT-PCR and Western blot experiments were carried out to detect the expression levels of GLUT3 and AMPK in placental tissue; CO-IP was utilized to detect the interaction of GLUT3 and AMPK. Compared with the normal pregnancy group, the weight of the fetus and placenta of GDM mice increased (P < 0.001), and the ability of trophoblasts to take up glucose decreased (P < 0.001). In addition, AMPK activity in trophoblasts and membrane localization of GLUT3 in GDM mice were down-regulated compared with normal pregnant mice (P < 0.05). There is an interaction between GLUT3 and AMPK. Activating AMPK in trophoblasts can up-regulate the expression of GLUT3 membrane protein in trophoblasts of mice (P < 0.05) and increase the glucose uptake of trophoblasts (P < 0.05). We speculate that inhibition of AMPK activity in GDM mice results in aberrant localization of GLUT3, which in turn attenuates glucose uptake by placental trophoblast cells. AICAR activates AMPK to increase the membrane localization of GLUT3 and improve the glucose uptake capacity of trophoblasts.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Diabetes Gestacional , Transportador de Glucosa de Tipo 3 , Glucosa , Transducción de Señal , Trofoblastos , Animales , Trofoblastos/metabolismo , Femenino , Embarazo , Glucosa/metabolismo , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 3/genética , Diabetes Gestacional/metabolismo , Placenta/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Ribonucleótidos/farmacología
15.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119715, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583782

RESUMEN

Ovarian cancer (OvCa) is characterized by early metastasis and high mortality rates, underscoring the need for deeper understanding of these aspects. This study explores the role of glucose transporter 3 (GLUT3) driven by zinc finger E-box-binding homeobox 1 (ZEB1) in OvCa progression and metastasis. Specifically, this study explored whether ZEB1 promotes glycolysis and assessed the potential involvement of GLUT3 in this process in OvCa cells. Our findings revealed that ZEB1 and GLUT3 were excessively expressed and closely correlated in OvCa. Mechanistically, ZEB1 activates the transcription of GLUT3 by binding to its promoter region. Increased expression of GLUT3 driven by ZEB1 dramatically enhances glycolysis, and thus fuels Warburg Effect to promote OvCa progression and metastasis. Consistently, elevated ZEB1 and GLUT3 expression in clinical OvCa is correlated with poor prognosis, reinforcing the profound contribution of ZEB1-GLUT3 axis to OvCa. These results suggest that activation of GLUT3 expression by ZEB1 is crucial for the proliferation and metastasis of OvCa via fueling glycolysis, shedding new light on OvCa treatment.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 3 , Neoplasias Ováricas , Activación Transcripcional , Efecto Warburg en Oncología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Humanos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Línea Celular Tumoral , Glucólisis/genética , Animales , Proliferación Celular/genética , Ratones , Regiones Promotoras Genéticas , Ratones Desnudos
16.
PLoS One ; 19(4): e0301724, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38625978

RESUMEN

BACKGROUNDS: Oral squamous cell carcinoma is a malignant tumor of the head and neck, and its molecular mechanism remains to be explored. METHODS: By analyzing the OSCC data from the TCGA database, we found that SLC2A3 is highly expressed in OSCC patients. The expression level of SLC2A3 was verified by RT-PCR and western blotting in OSCC cell lines. The function of SLC2A3 in OSCC cell lines and Lactic acid in SLC2A3-knockdown OSCC cells were detected by colony formation, CCK8, transwell, and wound healing assays. The effect of SLC2A3 on tumor growth and metastasis was tested in vivo. GSEA and Western blot were used to analyze and validate tumor phenotypes and signaling pathway molecules. RESULTS: We analyzed OSCC datasets in the TCGA database and found that SLC2A3 had abnormally high expression and was associated with poor prognosis. We also found that oral squamous cell carcinoma cells had increased proliferation, migration, invasion, EMT phenotype, and glycolysis due to SLC2A3 overexpression. Conversely, SLC2A3 knockdown had the opposite effect. Our in vivo experiments confirmed that SLC2A3 overexpression promoted tumor growth and metastasis while knockdown inhibited it. We also observed that high SLC2A3 expression led to EMT and the activation of the TGF-ß signaling pathway, while knockdown inhibited it. Interestingly, exogenous lactic acid restored the EMT, proliferation, migration, and invasion abilities of oral cancer cells inhibited by knocking down SLC2A3. CONCLUSIONS: Our study reveals that SLC2A3 expression was up-regulated in OSCC. SLC2A3 activates the TGF-ß signaling pathway through lactic acid generated from glycolysis, thus regulating the biological behavior of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/patología , Proliferación Celular , Transducción de Señal , Neoplasias de Cabeza y Cuello/genética , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 3/genética
17.
Respir Res ; 25(1): 158, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38594707

RESUMEN

BACKGROUND: Airway remodelling plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Epithelial-mesenchymal transition (EMT) is a significant process during the occurrence of airway remodelling. Increasing evidence suggests that glucose transporter 3 (GLUT3) is involved in the epithelial mesenchymal transition (EMT) process of various diseases. However, the role of GLUT3 in EMT in the airway epithelial cells of COPD patients remains unclear. METHODS: We detected the levels of GLUT3 in the peripheral lung tissue of COPD patients and cigarette smoke (CS)-exposed mice. Two Gene Expression Omnibus GEO datasets were utilised to analyse GLUT3 gene expression profiles in COPD. Western blot and immunofluorescence were used to detect GLUT3 expression. In addition, we used the AAV9-GLUT3 inhibitor to reduce GLUT3 expression in the mice model. Masson's staining and lung function measurement were used detect the collagen deposition and penh in the mice. A cell study was performed to confirm the regulatory effect of GLUT3. Inhibition of GLUT3 expression with siRNA, Western blot, and immunofluorescence were used to detect the expression of E-cadherin, N-cadherin, vimentin, p65, and ZEB1. RESULTS: Based on the GEO data set analysis, GLUT3 expression in COPD patients was higher than in non-smokers. Moreover, GLUT3 was highly expressed in COPD patients, CS exposed mice, and BEAS-2B cells treated with CS extract (CSE). Further research revealed that down-regulation of GLUT3 significantly alleviated airway remodelling in vivo and in vitro. Lung function measurement showed that GLUT3 reduction reduced airway resistance in experimental COPD mice. Mechanistically, our study showed that reduction of GLUT3 inhibited CSE-induced EMT by down-regulating the NF-κB/ZEB1 pathway. CONCLUSION: We demonstrate that CS enhances the expression of GLUT3 in COPD and further confirm that GLUT3 may regulate airway remodelling in COPD through the NF-κB/ZEB1 pathway; these findings have potential value in the diagnosis and treatment of COPD. The down-regulation of GLUT3 significantly alleviated airway remodelling and reduced airway resistance in vivo. Our observations uncover a key role of GLUT3 in modulating airway remodelling and shed light on the development of GLUT3-targeted therapeutics for COPD.


Asunto(s)
Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Ratones , Animales , FN-kappa B/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Fumar Cigarrillos/efectos adversos , Transportador de Glucosa de Tipo 3/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transición Epitelial-Mesenquimal , Células Epiteliales/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
18.
Cell Biochem Funct ; 42(3): e4001, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38571370

RESUMEN

Carbonic anhydrase 8 (CA8) is a member of the α-carbonic anhydrase family but does not catalyze the reversible hydration of carbon dioxide. In the present study, we examined the effects of CA8 on two human colon cancer cell lines, SW480 and SW620, by suppressing CA8 expression through shRNA knockdown. Our results showed that knockdown of CA8 decreased cell growth and cell mobility in SW620 cells, but not in SW480 cells. In addition, downregulated CA8 resulted in a significant decrease of glucose uptake in both SW480 and SW620 cells. Interestingly, stable downregulation of CA8 decreased phosphofructokinase-1 expression but increased glucose transporter 3 (GLUT3) levels in SW620 cells. However, transient downregulation of CA8 fails to up-regulate GLUT3 expression, indicating that the increased GLUT3 observed in SW620-shCA8 cells is a compensatory effect. In addition, the interaction between CA8 and GLUT3 was evidenced by pull-down and IP assays. On the other hand, we showed that metformin, a first-line drug for type II diabetes patients, significantly inhibited cell migration of SW620 cells, depending on the expressions of CA8 and focal adhesion kinase. Taken together, our data demonstrate that when compared to primary colon cancer SW480 cells, metastatic colon cancer SW620 cells respond differently to downregulated CA8, indicating that CA8 in more aggressive cancer cells may play a more important role in controlling cell survival and metformin response. CA8 may affect glucose metabolism- and cell invasion-related molecules in colon cancer, suggesting that CA8 may be a potential target in future cancer therapy.


Asunto(s)
Anhidrasas Carbónicas , Neoplasias del Colon , Neoplasias Colorrectales , Diabetes Mellitus Tipo 2 , Metformina , Humanos , Transportador de Glucosa de Tipo 3/genética , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/metabolismo , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Glucosa , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo
19.
Cancer Lett ; 588: 216784, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38458594

RESUMEN

Glycolytic metabolism is a hallmark of pancreatic ductal adenocarcinoma (PDAC), and tumor-associated stromal cells play important roles in tumor metabolism. We previously reported that tumor-associated macrophages (TAMs) facilitate PDAC progression. However, little is known about whether TAMs are involved in regulating glycolysis in PDAC. Here, we found a positive correlation between CD68+ TAM infiltration and FDG maximal standardized uptake (FDG SUVmax) on PET-CT images of PDAC. We discovered that the glycolytic gene set was prominently enriched in the high TAM infiltration group through Gene Set Enrichment Analysis using The Cancer Genome Atlas database. Mechanistically, TAMs secreted IL-8 to promote GLUT3 expression in PDAC cells, enhancing tumor glycolysis both in vitro and in vivo, whereas this effect could be blocked by the IL-8 receptor inhibitor reparixin. Furthermore, IL-8 promoted the translocation of phosphorylated STAT3 into the nucleus to activate the GLUT3 promoter. Overall, we demonstrated that TAMs boosted PDAC cell glycolysis through the IL-8/STAT3/GLUT3 signaling pathway. Our cumulative findings suggest that the abrogation of TAM-induced tumor glycolysis by reparixin might exhibit an antitumor impact and offer a potential therapeutic target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Sulfonamidas , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Fluorodesoxiglucosa F18/uso terapéutico , Tomografía Computarizada por Tomografía de Emisión de Positrones , Macrófagos/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Transducción de Señal , Glucólisis , Línea Celular Tumoral , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
20.
Cannabis Cannabinoid Res ; 9(3): 766-780, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38364116

RESUMEN

Introduction: Cannabis use is increasing among pregnant people, and cannabidiol (CBD), a constituent of cannabis, is often perceived as "natural" and "safe" as it is non-intoxicating. In utero, cannabis exposure is associated with negative health outcomes, including fetal growth restriction (FGR). The placenta supplies oxygen and nutrients to the fetus, and alterations in placental development can lead to FGR. While there has been some investigation into the effects of Δ9-THC, there has been limited investigation into the impacts of in utero gestational CBD exposure on the placenta. Methods: This study used histological and transcriptomic analysis of embryonic day (E)19.5 rat placentas from vehicle and CBD (3 mg/kg intraperitoneal injection) exposed pregnancies (E6.5-18.5). Results: The study revealed that pups from CBD-exposed pregnancies were 10% smaller, with the placentae displaying a decreased fetal blood space perimeter-to-area ratio. The transcriptomic analysis supported compromised angiogenesis and blood vessel formation with downregulated biological processes, including tube morphogenesis, angiogenesis, blood vessel morphogenesis, blood vessel development and vasculature development. Further, the CBD-exposed placentas displayed changed expression of glucose transporters (decreased GLUT1 and GR expression and increased GLUT3 expression). Transcriptomic analysis further revealed upregulated biological processes associated with metabolism. Finally, histological and transcriptomic analysis revealed altered cell populations within the placenta, specifically to syncytiotrophoblast layer II and endothelial cells. Conclusion: Together these results suggest that the structural changes in CDB-exposed placentae, including the altered expression of nutrient transporters and the changes to the placental fetal vasculature, may underlie the reduced fetal growth.


Asunto(s)
Cannabidiol , Retardo del Crecimiento Fetal , Placenta , Embarazo , Animales , Femenino , Placenta/efectos de los fármacos , Placenta/metabolismo , Cannabidiol/farmacología , Cannabidiol/toxicidad , Ratas , Retardo del Crecimiento Fetal/inducido químicamente , Desarrollo Fetal/efectos de los fármacos , Ratas Sprague-Dawley , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...