Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 251: 114527, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36628874

RESUMEN

The aims of this study were to evaluated the effect and underlying mechanism of Gandankang (GDK) aqueous extract in alleviating the acute liver injury induced by carbon tetrachloride (CCl4) in vivo and in vitro. Mice were divided into 5 groups (n = 8) for acute (Groups: control, 0.3 % CCl4, BD (Bifendate), 1.17, 2.34 and 4.68 mg/kg GDK) liver injury study. 10 µL/g CCl4 with corn oil were injected interperitoneally (i.p) expect the control group. HepG2 cells were used in vitro study. The results showed GDK can effectively inhibit liver damage and restore the structure and function of the liver. In mechanism, GDK inhibited CCl4-induced liver fibrosis and blocked the NF-κB pathway to effectively inhibit the hepatic inflammatory response; and inhibited CCl4-induced oxidative stress by upregulating the Keap1/Nrf2 pathway-related proteins and promoting the synthesis of several antioxidants. Additionally, it inhibited ferroptosis in the liver by regulating the expression of ACSl4 and GPX4. GDK reduced lipid peroxide generation in vitro by downregulating the production of reactive oxygen species and Fe2+ aggregation, thereby inhibiting ferroptosis and alleviating CCl4-induced hepatocyte injury. In conclusion, we describe the potential complex mechanism underlying the effect of GDK against acute liver injury.


Asunto(s)
Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas , Ratones , Animales , Tetracloruro de Carbono/toxicidad , Tetracloruro de Carbono/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Hígado , Antioxidantes/metabolismo , Estrés Oxidativo , Transducción de Señal , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo
2.
Mol Biol Rep ; 49(5): 3955-3964, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35235160

RESUMEN

PURPOSE: Cellular responses following cerebral ischemia/reperfusion injury are critical to recovery and survival after ischemic stroke. Understanding of these cellular responses can help the design of therapies to protect brain tissue and promote recovery after stroke. One of these cellular responses may be mediated by the AKT (protein kinase B) signal transduction pathway. This study was aimed to investigate the cerebral ischemia-induced alterations of AKT signaling and the upstream molecular pathways. METHODS: We modeled cerebral ischemia by middle cerebral artery occlusion in 2-3-month-old male C57BL/6J mice and then analyze the brain samples by using quantitative Western blots and phosphorylation/activation-dependent kinase antibodies. Cerebral ischemia was confirmed by staining of brain slices with 1% 2,3,5-triphenyltetrazolium chloride (TTC) and Nissl, as well as neurological assessments of the mice 24 h after ischemia-reperfusion surgery. RESULTS: We found marked downregulation of AKT within 12 h of cerebral ischemia/reperfusion, which leads to overactivation of glycogen synthase kinase-3ß (GSK-3ß). Furthermore, we found that the downregulation of AKT was mediated by downregulation of mTORC2 (the complex 2 of the mechanistic target of rapamycin) instead of its common upstream kinases, phosphatidylinositol 3-kinase and phosphoinositide-dependent kinase-1. CONCLUSION: Our findings provide new insight into the cellular responses to ischemia/reperfusion brain injury and will help develop new treatments targeting the AKT signaling pathway for the treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica , Glucógeno Sintasa Quinasa 3 beta , Accidente Cerebrovascular Isquémico , Proteínas Proto-Oncogénicas c-akt , Daño por Reperfusión , Serina-Treonina Quinasas TOR , Animales , Isquemia Encefálica/metabolismo , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
3.
Neurocrit Care ; 37(1): 91-101, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35137354

RESUMEN

BACKGROUND: Inhaled argon (iAr) has shown promising therapeutic efficacy for acute ischemic stroke and has exhibited impressive advantages over other inert gases as a neuroprotective agent. However, the optimal dose, duration, and time point of iAr for acute ischemic stroke are unknown. Here, we explored variable iAr schedules and evaluated the neuroprotective effects of acute iAr administration on lesion volume, brain edema, and neurological function in a mouse model of cerebral ischemic/reperfusion injury. METHODS: Adult ICR (Institute of Cancer Research) mice were randomly subjected to sham, moderate (1.5 h), or severe (3 h) transient middle cerebral artery occlusion (tMCAO). One hour after tMCAO, the mice were randomized to variable iAr protocols or air. General and focal deficit scores were assessed during double-blind treatment. Infarct volume, overall recovery, and brain edema were analyzed 24 h after cerebral ischemic/reperfusion injury. RESULTS: Compared with those in the tMCAO-only group, lesion volume (p < 0.0001) and neurologic outcome (general, p < 0.0001; focal, p < 0.0001) were significantly improved in the group administered iAr 1 h after stroke onset (during ischemia). Short-term argon treatment (1 or 3 h) significantly improved the infarct volume (1 vs. 24 h, p < 0.0001; 3 vs. 24 h, p < 0.0001) compared with argon inhalation for 24 h. The concentration of iAr was confirmed to be a key factor in improving focal neurological outcomes relative to that in the tMCAO group, with higher concentrations of iAr showing better effects. Additionally, even though ischemia research has shown an increase in cerebral damage proportional to the ischemia time, argon administration showed significant neuroprotective effects on infarct volume (p < 0.0001), neurological deficits (general, p < 0.0001; focal, p < 0.0001), weight recovery (p < 0.0001), and edema (p < 0.0001) in general, particularly in moderate stroke. CONCLUSIONS: Timely iAr administration during ischemia showed optimal neurological outcomes and minimal infarct volumes. Moreover, an appropriate duration of argon administration was important for better neuroprotective efficacy. These findings may provide vital guidance for using argon as a neuroprotective agent and moving to clinical trials in acute ischemic stroke.


Asunto(s)
Edema Encefálico , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Daño por Reperfusión , Accidente Cerebrovascular , Animales , Ratones , Argón/farmacología , Argón/uso terapéutico , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Ratones Endogámicos ICR , Fármacos Neuroprotectores/farmacología , Distribución Aleatoria , Daño por Reperfusión/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico
4.
Ecotoxicol Environ Saf ; 245: 114118, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36174321

RESUMEN

Mori fructus aqueous extracts (MFAEs) have been used as a traditional Chinese medicine for thousands of years with the function of strengthening the liver and tonifying the kidney. However, its inner mechanism to alleviative renal injury is unclear. To investigate the attenuation of MFAEs on nephrotoxicity and uncover its potential molecular mechanism, we established a nephrotoxicity model induced by carbon tetrachloride (CCl4). The mice were randomly divided into control group, CCl4 model group (10% CCl4), CCl4 + low and high MFAEs groups (10% CCl4 + 100 mg/kg and 200 mg/kg MFAEs). We found that MFAEs decreased the kidney index of mice, restored the pathological changes of renal structure induced by CCl4, reduced cystatin C, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule 1 (Kim-1) blood urea nitrogen and creatinine contents in serum, promoted the nuclear transportation of Nrf2 (nuclear factor erythroid derived 2 like 2), elevated the expression of HO-1 (heme oxygenase 1), GPX4 (glutathione peroxidase 4), SLC7A11 (solute carrier family 7 member 11), ZO-1 (zonula occludens-1) and Occludin, suppressed the expression of Keap1 (kelch-like ECH-associated protein 1), HMGB1 (High Mobility Group Protein 1), ACSL4 (acyl-CoA synthetase long chain family member 4) and TXNIP (thioredoxin interacting protein), upregulated the flora of Akkermansia, Anaerotruncus, Clostridium_sensu_stricto, Ihubacter, Alcaligenes, Dysosmobacter, and downregulated the flora of Clostridium_XlVa, Helicobacter, Paramuribaculum. Overlapped with Disbiome database, Clostridium_XlVa, Akkermansia and Anaerotruncus may be the potential genera treated with renal injury. It indicated that MFAEs could ameliorate kidney injury caused by CCl4 via Nrf2 signaling.


Asunto(s)
Microbioma Gastrointestinal , Proteína HMGB1 , Animales , Tetracloruro de Carbono/metabolismo , Tetracloruro de Carbono/toxicidad , Coenzima A/metabolismo , Creatinina , Cistatina C/metabolismo , Proteína HMGB1/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Riñón/metabolismo , Ligasas/metabolismo , Lipocalina 2/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Ocludina/metabolismo , Estrés Oxidativo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Tiorredoxinas/metabolismo
5.
J Nanobiotechnology ; 18(1): 141, 2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33008409

RESUMEN

As a main element in the hard metal industry, cobalt is one of the major components of human metal implants. Cobalt-containing implants, especially joint prostheses used for artificial joint replacement, can be corroded due to the complex physiological environment in vivo, producing a large number of nanoscale cobalt particles (Cobalt Nanoparticles, CoNPs). These CoNPs can be first accumulated around the implant to cause adverse local reactions and then enter into the blood vessels followed by reaching the liver, heart, brain, kidney, and other organs through systematic circulation, which leads to multi-system toxicity symptoms. To ensure the long-term existence of cobalt-containing implants in the body, it is urgently required to find out a safe and effective detoxification drug. Herein, we have demonstrated that CoNPs could induce the ferroptosis-like cell death through the enhancement of intracellular reactive oxygen species (ROS) level, cytoplasmic Fe2+ level, lipid peroxidation, and consumption of reduced glutathione (GSH) as well as inhibition of glutathione peroxidase 4 (GPX4) activity. Importantly, α-lipoic acid (ALA), a natural antioxidant with the capability to scavenge free radicals and chelate toxic metals, was found to efficiently alleviate the adverse effects of CoNPs. The present study illustrates a new mechanism of CoNPs mediated by ferroptosis-like cytotoxicity and discloses an effective method for the detoxification of CoNPs by employing the natural antioxidant of ALA, providing a basis for further in vivo detoxification study.


Asunto(s)
Muerte Celular/efectos de los fármacos , Cobalto/toxicidad , Ferroptosis/efectos de los fármacos , Nanopartículas del Metal/uso terapéutico , Ácido Tióctico/farmacología , Células 3T3 , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Cobalto/química , Humanos , Inactivación Metabólica , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/metabolismo , Ácido Tióctico/química
6.
J Appl Clin Med Phys ; 20(1): 293-307, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30508275

RESUMEN

OBJECTIVE: Multislice computed tomography (MSCT) has been used for diagnosis of small intestinal diseases. However, the radiation dose is a big problem. This study was to investigate whether CARE Dose 4D combined with sinogram-affirmed iterative reconstruction (SAFIRE) can provide better image quality at a lower dose for imaging small intestinal diseases compared to MSCT. METHODS: The noise reduction ability of SAFIRE was assessed by scanning the plain water mold using SOMATOM Definition Flash double-source spiral CT. CT images at each stage of radiography for 239 patients were obtained. The patients were divided into groups A and B were based on different tube voltage and current or the image recombination methods. The images were restructured using with filtered back projection (FBP) and SAFIRE (S1-S5). The contrast noise ratio (CNR), CT Dose index (CTDI), subjective scoring, and objective scoring were compared to obtain the best image and reformation parameters at different stages of CT. RESULTS: Twenty-six restructuring patterns of tube voltage and current were obtained by FBP and SAFIRE. The average radiation dose using CARE Dose 4D combined with SAFIRE (S4-S5) reduced approximately 74.85% compared to conditions where the tube voltage of 100 kV and tube current of 131 mAs for patients with MSCT small intestinal CT enterography at plain CT scan, arterial stage, small intestine, and portal venous phase. The objective and subjective scoring were all significantly different among groups A and B at each stage. CONCLUSIONS: Combination of CARE Dose 4D and SAFIRE is shown to decrease the radiation dose while maintaining image quality.


Asunto(s)
Algoritmos , Tomografía Computarizada Cuatridimensional/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Enfermedades Intestinales/diagnóstico por imagen , Intestino Delgado/diagnóstico por imagen , Tomografía Computarizada Multidetector/métodos , Interpretación de Imagen Radiográfica Asistida por Computador/métodos , Femenino , Humanos , Enfermedades Intestinales/patología , Enfermedades Intestinales/radioterapia , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Masculino , Persona de Mediana Edad , Dosificación Radioterapéutica
7.
Rev Esp Enferm Dig ; 111(12): 941-945, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31718212

RESUMEN

BACKGROUND: interleukin-37 (IL-37) is as a natural suppressor of the innate inflammatory and immune responses. It has also been reported to be involved in carcinogenesis and metastasis. The present case-control study was designed to investigate the role of serum levels of IL-37 in patients with gastric cancer. METHODS: serum IL-37 levels were determined using the enzyme-linked immunosorbent assay in 180 patients diagnosed with gastric cancer and 100 healthy controls. The association between IL-37 levels and clinical factors was assessed. Univariate and multivariate analyses were performed to investigate the prognostic significance of these parameters in gastric cancer. RESULTS: serum IL-37 levels in gastric cancer patients (5.606 ± 0.837 pg/ml) were significantly higher than those in healthy controls (2.364 ± 0.210 pg/ml, p < 0.001). High serum IL-37 levels were related to a poorly differentiated histologic type (p = 0.046) and advanced T stage (p = 0.003). The Kaplan-Meier survival analysis indicated that the high-IL-37 group had a poorer overall survival and progression-free survival (overall survival [OS]: 39.0 months vs 13.0 months, p < 0.001, progression-free survival [PFS]: 25.0 months vs 10.0 months, p < 0.001). Multivariate analyses showed serum IL-37 to be an independent prognostic factor for gastric cancer patients (OS: hazard ratios [HR] = 1.842, 95% CI: 1.190-2.854, p = 0.006; PFS: HR = 1.547, 95% CI: 1.014-2.359, p = 0.043). CONCLUSIONS: in conclusion, serum IL-37 levels were associated with poor overall survival and progression-free survival in gastric cancer patients. IL-37 may be a potential predictor of prognosis in gastric cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Interleucina-1/sangre , Neoplasias Gástricas/sangre , Anciano , Análisis de Varianza , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Supervivencia sin Progresión , Curva ROC , Estudios Retrospectivos , Sensibilidad y Especificidad , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología
8.
Stroke ; 47(1): 187-95, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26564104

RESUMEN

BACKGROUND AND PURPOSE: Our previous study has defined a role of TP53-induced glycolysis and apoptosis regulator in neuroprotection against ischemic injury through increasing the flow of pentose phosphate pathway. We hypothesized that the pentose phosphate pathway product nicotinamide adenine dinucleotide phosphate (NADPH) could be a novel drug for treatment of ischemic stroke. METHODS: The NADPH was given before, at the onset, or after stroke onset with single or repeated intravenous (mice and rats) or intraperitoneal injections (monkey). The short- and long-term therapeutic effects of NADPH were evaluated in male adult ICR mice (total=614) with transient middle cerebral artery occlusion, in male adult Sprague-Dawley rats (total=114) with permanent middle cerebral artery occlusion, and in male adult rhesus monkey (total=12) with thrombotic middle cerebral artery occlusion. RESULTS: Administration of NADPH led to a dramatic increase in the levels of ATP and reduced form of glutathione, whereas it decreased the levels of reactive oxygen species. NADPH significantly reduced infarct volume, improved poststroke survival, and recovery of neurological functions in mouse and rat models of stroke. Robust neuroprotection of a single dose of NADPH was seen when it was administered within 5 hours after reperfusion; however, repeat administration of NADPH twice a day for 7 days starting 24 hours after the onset of stroke also offered therapeutic effects. Pretreatment with NADPH also significantly improved the outcome of stroke insult. CONCLUSIONS: Administration of exogenous NADPH significantly protected neurons against ischemia/reperfusion-induced injury in 2 rodent stroke models. Thus, NADPH might be a promising drug candidate for treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , NADP/administración & dosificación , Vía de Pentosa Fosfato/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Administración Intravenosa , Animales , Isquemia Encefálica/patología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología
9.
J Neurosci ; 34(22): 7458-71, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24872551

RESUMEN

TP53-induced glycolysis and apoptosis regulator (TIGAR) inhibits glycolysis and increases the flow of pentose phosphate pathway (PPP), which generates NADPH and pentose. We hypothesized that TIGAR plays a neuroprotective role in brain ischemia as neurons do not rely on glycolysis but are vulnerable to oxidative stress. We found that TIGAR was highly expressed in brain neurons and was rapidly upregulated in response to ischemia/reperfusion insult in a TP53-independent manner. Overexpression of TIGAR in normal mice with lentivirus reduced ischemic neuronal injury, whereas lentivirus-mediated TIGAR knockdown aggravated it. In cultured primary neurons, increasing TIGAR expression reduced oxygen and glucose deprivation (OGD)/reoxygenation-induced injury, whereas decreasing its expression worsened the injury. The glucose 6-phosphate dehydrogenase was upregulated in mouse and cellular models of stroke, and its upregulation was further enhanced by overexpression of TIGAR. Supplementation of NADPH also reduced ischemia/reperfusion brain injury and alleviated TIGAR knockdown-induced aggravation of ischemic injury. In animal and cellular stroke models, ischemia/reperfusion increased mitochondrial localization of TIGAR. OGD/reoxygenation-induced elevation of ROS, reduction of GSH, dysfunction of mitochondria, and activation of caspase-3 were rescued by overexpression of TIGAR or supplementation of NADPH, while knockdown of TIGAR aggravated these changes. Together, our results show that TIGAR protects ischemic brain injury via enhancing PPP flux and preserving mitochondria function, and thus may be a valuable therapeutic target for ischemic brain injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Redes y Vías Metabólicas/fisiología , Proteínas/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Isquemia Encefálica/prevención & control , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/metabolismo , Mitocondrias/patología , Mitocondrias/fisiología , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/fisiología , Fármacos Neuroprotectores/metabolismo , Monoéster Fosfórico Hidrolasas
10.
Dig Dis Sci ; 60(7): 1967-76, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25652145

RESUMEN

AIM: This study was to investigate the effects and mechanisms of miR-362-3p on regulation of gastric cancer (GC) cell metastasis potential. METHODS: We detected miR-362-3p level in GC and adjacent normal tissues and investigated the relationship with clinicopathological factors. Next, we analyzed the level of miR-362-3p expression and CD82 in different differentiated GC cells compared with a normal gastric mucosa cell by RT-PCR and Western blot. Dual-luciferase reporter assay and Western blot confirmed a direct interaction between miR-362-3p and CD82 3'UTR. After miR-362-3p and CD82 were silenced in GC cells, we compared the transfected GC cells migration and invasion capacity by transwell assay. In addition, we detected the effects on cells angiogenesis by tube formation assay. Western blot was used to detect the impact of CD82 and miR-362-3p on epithelial-to-mesenchymal transition markers in treated GC cells. RESULTS: Level of miR-362-3p expression was much higher in GC cells than in normal gastric mucosa cell, and miR-362-3p expression negatively correlated with CD82 mRNA expression in these cell lines. Furthermore, miR-362-3p expression induced [corrected] GC cell metastasis capacity by suppression of CD82 expression. Level of miR-362-3p may mediate E-cadherin, N-cadherin, and vimentin expression in GC cells. CONCLUSION: This study illuminated that downregulation of miR-362-3p along with the upregulation of CD82 in GC cells resulted in the inhibition of GC migration and invasion. Thus, our results suggested that miR-362-3p or CD82 can be exploited as a new potential target for control of GC in the future.


Asunto(s)
Proteína Kangai-1/metabolismo , MicroARNs/metabolismo , Interferencia de ARN , Neoplasias Gástricas/metabolismo , Anticuerpos , Movimiento Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Proteína Kangai-1/genética , Masculino , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Estómago/citología
11.
BMC Complement Altern Med ; 14: 461, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25465226

RESUMEN

BACKGROUND: Naja naja atra venom (NNAV) displays diverse pharmacological actions including analgesia, anti-inflammation and immune regulation.In this study, we investigated the effects of NNAV on pulmonary fibrosis and its mechanisms of action. METHODS: To determine if Naja naja atra venom (NNAV) can produce beneficial effects on pulmonary fibrosis, two marine models of pulmonary fibrosis were produced with bleomycin (BLM) and lipopolysaccharide (LPS). NNAV (30, 90, 270 µg/kg) was orally administered once a day started five days before BLM and LPS until to the end of experiment. The effects of NNAV treatment on pulmonary injury were evaluated with arterial blood gas analysis, hydroxyproline (HYP) content assessment and HE/Masson staining. The effects of NNAV treatment on inflammatory related cytokines, fibrosis related TGF-ß/Smad signaling pathway and oxidative stress were examined. RESULTS: The results showed that NNAV improved the lung gas-exchange function and attenuated the fibrotic lesions in lung. NNAV decreased IL-1ß and TNF-α levels in serum in both pulmonary fibrosis models. NNAV inhibited the activation of NF-κB in LPS-induced and TGF-ß/Smad pathway in BLM-induced pulmonary fibrosis. Additionally, NNAV also increased the levels of SOD and GSH and reduced the levels of MDA in BLM-induced pulmonary fibrosis model. CONCLUSIONS: The present study indicates that NNAV attenuates LPS- and BLM-induced lung fibrosis. Its mechanisms of action are associated with inhibiting inflammatory response and oxidative stress. The study suggests that NNAV might be a potential therapeutic drug for treatment of pulmonary fibrosis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Venenos Elapídicos/uso terapéutico , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Bleomicina , Venenos Elapídicos/farmacología , Elapidae , Femenino , Fibrosis , Hidroxiprolina/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , FN-kappa B/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
12.
Front Pharmacol ; 15: 1377235, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38783961

RESUMEN

Protein glycosylation is an extensively studied field, with the most studied forms being oxygen or nitrogen-linked N-acetylglucosamine (O-GlcNAc or N-GlcNAc) glycosylation. Particular residues on proteins are targeted by O-GlcNAcylation, which is among the most intricate post-translational modifications. Significantly contributing to an organism's proteome, it influences numerous factors affecting protein stability, function, and subcellular localization. It also modifies the cellular function of target proteins that have crucial responsibilities in controlling pathways related to the central nervous system, cardiovascular homeostasis, and other organ functions. Under conditions of acute stress, changes in the levels of O-GlcNAcylation of these proteins may have a defensive function. Nevertheless, deviant O-GlcNAcylation nullifies this safeguard and stimulates the advancement of several ailments, the prognosis of which relies on the cellular milieu. Hence, this review provides a concise overview of the function and comprehension of O-GlcNAcylation in ischemia diseases, aiming to facilitate the discovery of new therapeutic targets for efficient treatment, particularly in patients with diabetes.

13.
J Alzheimers Dis ; 94(2): 651-668, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334605

RESUMEN

BACKGROUND: At least one-third of Alzheimer's disease (AD) patients have cerebrovascular abnormalities, micro- and macro-infarctions, and ischemic white matter alterations. Stroke prognosis impacts AD development due to vascular disease. Hyperglycemia can readily produce vascular lesions and atherosclerosis, increasing the risk of cerebral ischemia. Our previous research has demonstrated that protein O-GlcNAcylation, a dynamic and reversible post-translational modification, provides protection against ischemic stroke. However, the role of O-GlcNAcylation in the exacerbation of cerebral ischemia injury due to hyperglycemia remains to be elucidated. OBJECTIVE: In this study, we explored the role and underlying mechanism of protein O-GlcNAcylation in the exacerbation of cerebral ischemia injury caused by hyperglycemia. METHODS: High glucose-cultured brain microvascular endothelial (bEnd3) cells were injured by oxygen-glucose deprivation. Cell viability was used as the assay result. Stroke outcomes and hemorrhagic transformation incidence were assessed in mice after middle cerebral artery occlusion under high glucose and streptozotocin-induced hyperglycemic conditions. Western blot estimated that O-GlcNAcylation influenced apoptosis levels in vitro and in vivo. RESULTS: In in vitro analyses showed that Thiamet-G induces upregulation of protein O-GlcNAcylation, which attenuates oxygen-glucose deprivation/R-induce injury in bEnd3 cells cultured under normal glucose conditions, while aggravated it under high glucose conditions. In in vivo analyses, Thiamet-G exacerbated cerebral ischemic injury and induced hemorrhagic transformation, accompanied by increased apoptosis. While blocking protein O-GlcNAcylation with 6-diazo-5-oxo-L-norleucine alleviated cerebral injury of ischemic stroke in different hyperglycemic mice. CONCLUSION: Overall, our study highlights the crucial role of O-GlcNAcylation in exacerbating cerebral ischemia injury under conditions of hyperglycemia. O-GlcNAcylation could potentially serve as a therapeutic target for ischemic stroke associated with AD.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Hiperglucemia , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratones , Animales , Isquemia Encefálica/metabolismo , Accidente Cerebrovascular/complicaciones , Hiperglucemia/complicaciones , Infarto de la Arteria Cerebral Media/complicaciones , Glucosa/metabolismo , Oxígeno/metabolismo , Lesiones Encefálicas/complicaciones , Accidente Cerebrovascular Isquémico/complicaciones
14.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 24(10): 600-3, 2012 Oct.
Artículo en Zh | MEDLINE | ID: mdl-23040776

RESUMEN

OBJECTIVE: To analyze the value of automated acid-base mapping on diagnose and treatment of patients with community acquired pneumonia (CAP) in emergency department. METHODS: According to medical history, pulmonary function test, diagnosing guideline of chronic obstructive pulmonary disease (COPD), 111 patients with CAP were divided into two groups: single CAP group (n=56) and COPD complicated with CAP group [acute exacerbation of chronic obstructive pulmonary disease (AECOPD) group, n=55]. After enquiring medical history, arterial blood samples were drawn for blood gas analysis and automated acid-base mapping was analyzed. RESULTS: Arterial blood gas analysis showed arterial carbon dioxide partial pressure (PaCO(2)), HCO(3)(-), base excess of AECOPD group were obviously higher than those in CAP group (PaCO(2): 7.714±2.414 kPa vs. 5.896±1.308 kPa, HCO(3)(-): 30.767±7.185 mmol/L vs. 25.014±3.043 mmol/L, BE: 4.345±5.371 mmol/L vs. -0.354±3.180 mmol/L, all P<0.01). Automated acid-base mapping showed acid-base disturbance of AECOPD group was 89.1% and CAP group was 66.1%. Chi-square analysis were done for patients of normal (10.9%, 33.9%), acute respiratory acidosis (12.7%, 14.3%), chronic respiratory acidosis (49.1%, 10.7%), respiratory alkalosis (7.3%, 14.3%), metabolic acidosis (12.7%, 17.9%), metabolic alkalosis (12.7%, 8.9%) between AECOPD group and CAP group, and statistical significance was found between AECOPD group and single CAP group (χ (2)=24.421, P=0.001). Advanced Chi-square analysis for patients of normal, acute respiratory acidosis, respiratory alkalosis, metabolic acidosis, metabolic alkalosis were done and showed no statistical difference (χ (2)=5.280, P=0.260). It is indicated chronic respiratory acidosis occurrences rate in AECOPD patients was higher than single CAP patients. CONCLUSIONS: Our study demonstrated that automated acid-base mapping may be helpful for emergency physician to rapidly recognize multi-acid-base disturbance in patients with CAP, and to promptly identify acute or chronic phase of respiratory disease.


Asunto(s)
Análisis de los Gases de la Sangre/métodos , Infecciones Comunitarias Adquiridas/sangre , Neumonía/sangre , Enfermedad Pulmonar Obstructiva Crónica/sangre , Equilibrio Ácido-Base , Anciano , Anciano de 80 o más Años , Infecciones Comunitarias Adquiridas/diagnóstico , Infecciones Comunitarias Adquiridas/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neumonía/diagnóstico , Neumonía/terapia , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/terapia
15.
Brain Res Bull ; 178: 133-143, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808323

RESUMEN

Folic acid (FA) supplementation in early pregnancy is recommended to protect against birth defects. But excess FA has exhibited neurodevelopmental toxicity. We previously reported that the mice treated with 2.5-fold the dietary requirement of FA one week before mating and throughout pregnancy and lactation displayed abnormal behaviors in the offspring. Here we found the levels of non-phosphorylated ß-catenin (active) were increased in the brains of weaning and adult FA-exposed offspring. Meanwhile, demethylation of protein phosphatase 2 A catalytic subunit (PP2Ac), which suppresses its enzyme activity in regulatory subunit dependent manner, was significantly inhibited. Among the upstream regulators of ß-catenin, PI3K/Akt/GSK-3ß but not Wnt signaling was stimulated in FA-exposed brains only at weaning. In mouse neuroblastoma N2a cells, knockdown of PP2Ac or leucine carboxyl methyltransferase-1 (LCMT-1), or overexpression of PP2Ac methylation-deficient mutant decreased ß-catenin dephosphorylation. These results suggest that excess FA may activate ß-catenin via suppressing PP2Ac demethylation, providing a novel mechanism for the influence of FA on neurodevelopment.


Asunto(s)
Encéfalo/efectos de los fármacos , Suplementos Dietéticos , Ácido Fólico/farmacología , Complejo Vitamínico B/farmacología , beta Catenina/efectos de los fármacos , Factores de Edad , Animales , Femenino , Ácido Fólico/administración & dosificación , Masculino , Ratones , Embarazo , Factores Sexuales , Complejo Vitamínico B/administración & dosificación , Destete
16.
J Cell Physiol ; 226(7): 1915-25, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21506122

RESUMEN

The molecular basis for induction of apoptosis in melanoma cells by vincristine remains unknown. Here we tested the potential involvement of AMP-activated protein kinase (AMPK) in this process. We found for the first time that vincristine induces AMPK activation (AMPKα, Thr 172) and Acetyl-CoA carboxylase (ACC, Ser 79) (a downstream molecular target of AMPK) phosphorylation in cultured melanoma cells in vitro. Reactive oxygen species (ROS) dependent LKB1 activation serves as the upstream signal for AMPK activation. AMPK inhibitor (compound C) or AMPKα siRNA knockdown inhibits vincristine induced B16 melanoma cell apoptosis, while AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-riboside (AICAR) enhances it. AMPK activation is involved in vincristine induced p53 phosphorylation and stabilization, the latter is known to mediate melanoma cell apoptosis. Further, activation of AMPK by vincristine inhibits mTOR Complex 1 (mTORC1) in B16 melanoma cells, which serves as another important mechanism to induce melanoma cell apoptosis. Our study provides new insights into understanding the cellular and molecular mechanisms of vincristine induced cancer cell death/apoptosis. We suggest that combining AMPK activator AICAR with vincristine may have potential to be used as a new therapeutic intervention against melanoma.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Melanoma Experimental/enzimología , Vincristina/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Acetil-CoA Carboxilasa/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática , Activadores de Enzimas/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Complejos Multiproteicos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Ribonucleótidos/farmacología , Serina-Treonina Quinasas TOR , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo
17.
Cancer Invest ; 29(10): 645-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22085268

RESUMEN

The aim of our meta-analysis was to assess the association between UGT1A7 polymorphisms and cancer risk. Case?control studies containing available polymorphic alleles (UGT1A7*1,*2,*3, and*4) and genotypes categorized according to enzymatic activity (High, Intermediate, and Low) were chosen to assess this association. Twenty-one case?control studies were identified. Meta-analysis indicated that UGT1A7 had a significant effect on cancer risk. In subgroup analysis, a significantly increased risk was associated with East Asians, hepatocellular cancer, and colorectal cancer. This meta-analysis suggested that there is a cancer risk associated with UGT1A7*3, Intermediate, and Low activity UGT1A7 genotypes, which is most evident in Asian individuals.


Asunto(s)
Predisposición Genética a la Enfermedad , Glucuronosiltransferasa/genética , Neoplasias/etiología , Polimorfismo Genético , Estudios de Casos y Controles , Genotipo , Humanos , Neoplasias/genética , Sesgo de Publicación , Riesgo
18.
Exp Neurol ; 343: 113785, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34153323

RESUMEN

BACKGROUND/AIM: Stroke is among the most common causes of disability and death in highly developed countries and China. We sought to study the role of oleanolic acid in cerebral ischemia-reperfusion injury. METHODS: Middle cerebral artery occlusion (MCAO) was performed to induce cerebral ischemia-reperfusion injury in mice. For the short-term effects of oleanolic acid (OA) against MCAO, mice administrated with OA (6 mg/kg /d) for 3 days before the injury were evaluated the infarct volume, neurological scores, blood brain barrier permeability and oxidative stress level, while for the long-term effects, MCAO mice were injected daily with OA for 6 weeks, followed by assessments of motor function, behavior and cerebral infarction area. RESULTS: Pretreatment of oleanolic acid alleviated MCAO-induced ischemia-reperfusion injury as indicated by the significant decreases in cerebral infarction area and neurological symptom score at 24 h post injury, Evans blue leakage, expression of matrix metalloproteinase 9 (MMP9) and occludin, dihydroethidium fluorescence, and block malonaldehyde generation. In the long run, OA significantly reduced brain loss, enhanced the motor function, promoted the recovery of nerve function, and improved the learning and memory ability 9 weeks after the ischemia-reperfusion injury. OA also inhibited astrocytes proliferation and microglia activation, promoted the expression of synapse-related proteins, and increased the number of DCX+ cells in the hippocampus. CONCLUSIONS: OA exhibits both short-term and long-term protective effects against the cerebral ischemia-reperfusion injury in mice. The short-term protective mechanism is related to the anti-oxidation of blood-brain barrier, while the long-term protective effect lies in neuroglia modulation, promotion of synaptic connection and neuroregeneration.


Asunto(s)
Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Ácido Oleanólico/administración & dosificación , Daño por Reperfusión/prevención & control , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteína Doblecortina , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos ICR , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Resultado del Tratamiento
19.
Biosci Rep ; 41(1)2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33345265

RESUMEN

Cobalt nanoparticles (CoNPs) released from hip joint implants are known to have a toxic effect on several organs probably through increasing reactive oxygen species (ROS). Ferrous ion (Fe2+) is well-known to enhance oxidative stress by catalysing the production of ROS. However, in our pilot study, we found that Fe2+ conversely inhibited the ROS production induced by CoNPs. To elucidate the underlying mechanism, the present study treated vascular endothelial HUVEC and HMEC-1 cells with CoNPs alone or in combination with ferrous lactate [Fe(CH3CHOHCOO)2], ferrous succinate [Fe(CH2COO)2], and ferrous chloride (FeCl2). CoNP toxicity was evaluated by measuring cell viability, rate of apoptosis and lactose dehydrogenase (LDH) release, and intracellular ROS levels. Treatment with CoNPs decreased cell viability, LDH release, and ROS production and increased apoptosis. CoNPs increased hypoxia-inducible factor-1α (HIF-1α) protein level and mRNA levels of vascular endothelial growth factor (VEGF) and glucose transporter 1 (GLUT1) downstream of HIF-1α signalling. Silencing HIF-1α attenuated CoNP toxicity, as seen by recovery of cell viability, LDH release, and ROS levels and reduced apoptosis. CoNPs caused a pronounced reduction of Fe2+ in cells, but supplementation with Fe(CH3CHOHCOO)2, Fe(CH2COO)2, and FeCl2 restored Fe2+ levels and inhibited HIF-1α activation. Moreover, all three Fe2+-containing agents conferred protection from CoNPs; Fe(CH3CHOHCOO)2 and Fe(CH2COO)2 more effectively than FeCl2. In summary, the present study revealed that CoNPs exert their toxicity on human vascular endothelial cells by depleting intracellular Fe2+ level, which causes activation of HIF-1α signalling. Supplements of Fe2+, especially in the form of Fe(CH3CHOHCOO)2 and Fe(CH2COO)2, mitigated CoNP toxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Cobalto/química , Endotelio Vascular/efectos de los fármacos , Compuestos Ferrosos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Nanopartículas del Metal/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Compuestos Ferrosos/química , Compuestos Ferrosos/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Nanopartículas del Metal/química , Transducción de Señal/efectos de los fármacos
20.
Biomed Pharmacother ; 138: 111477, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33765582

RESUMEN

Protein O-GlcNAcylation is a dynamic post-translational protein modification that regulates fundamental cellular functions in both normal physiology and diseases. The levels of protein O-GlcNAcylation are determined by flux of the hexosamine biosynthetic pathway (HBP), which is a branch of glycolysis, and are directly controlled by a pair of enzymes: O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). An increase in protein O-GlcNAcylation has been shown to have protective effects on ischemia-related insults in the heart and brain. To determine whether O-GlcNAcylation plays a beneficial role in ischemia-reperfusion (IR)-induced intestinal injury, we used pharmacological manipulation of O-GlcNAc to induce loss- and gain-of-function conditions and evaluated the viability and apoptosis of intestinal epithelioid cells in an in vitro oxygen-glucose deprivation (OGD) model and tissue injury grade in a small intestinal ischemia-reperfusion (SIIR) mouse model. We found that 1) Upregulation of O-GlcNAcylation induced by glucosamine (GlcN, increase in HBP flux) or thiamet G (an OGA inhibitor) enhanced intestinal cell survival in the OGD model. In contrast, downregulation of O-GlcNAcylation induced by DON (due to a reduction in HBP flux) or OMSI-1 (an OGT inhibitor) made the cells more susceptible to hypoxia injury. 2) Reducing the increase in O-GlcNAcylation levels with a combination of either GlcN with DON or thiamet G with OMSI-1 partly canceled its protective effect on OGD-induced cell injury. 3) In the in vivo SIIR mouse model, GlcN augmented intestinal protein O-GlcNAcylation and significantly alleviated intestinal injury by inhibiting cell apoptosis. These results indicate that acute increases in protein O-GlcNAcylation confer protection against intestinal ischemia insults, suggesting that O-GlcNAcylation, as an endogenous stress sensor, could be a universal protective mechanism and could be a potential therapeutic target for intestinal ischemic disease.


Asunto(s)
Glucosa/deficiencia , Hipoxia/metabolismo , Intestino Delgado/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Animales , Línea Celular , Hipoxia/patología , Hipoxia/prevención & control , Intestino Delgado/patología , Masculino , Ratones , Ratones Endogámicos ICR , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA