Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(9)2022 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35563057

RESUMEN

Ketamine-associated cystitis is characterized by suburothelial inflammation and urothelial cell death. Norketamine (NK), the main metabolite of ketamine, is abundant in urine following ketamine exposure. NK has been speculated to exert toxic effects in urothelial cells, similarly to ketamine. However, the molecular mechanisms contributing to NK-induced urothelial cytotoxicity are almost unclear. Here, we aimed to investigate the toxic effects of NK and the potential mechanisms underlying NK-induced urothelial cell injury. In this study, NK exposure significantly reduced cell viability and induced apoptosis in human urinary bladder epithelial-derived RT4 cells that NK (0.01-0.5 mM) exhibited greater cytotoxicity than ketamine (0.1-3 mM). Signals of mitochondrial dysfunction, including mitochondrial membrane potential (MMP) loss and cytosolic cytochrome c release, were found to be involved in NK-induced cell apoptosis and death. NK exposure of cells also triggered the expression of endoplasmic reticulum (ER) stress-related proteins including GRP78, CHOP, XBP-1, ATF-4 and -6, caspase-12, PERK, eIF-2α, and IRE-1. Pretreatment with 4-phenylbutyric acid (an ER stress inhibitor) markedly prevented the expression of ER stress-related proteins and apoptotic events in NK-exposed cells. Additionally, NK exposure significantly activated JNK, ERK1/2, and p38 signaling and increased intracellular calcium concentrations ([Ca2+]i). Pretreatment of cells with both PD98059 (an ERK1/2 inhibitor) and BAPTA/AM (a cell-permeable Ca2+ chelator), but not SP600125 (a JNK inhibitor) and SB203580 (a p38 inhibitor), effectively suppressed NK-induced mitochondrial dysfunction, ER stress-related signals, and apoptotic events. The elevation of [Ca2+]i in NK-exposed cells could be obviously inhibited by BAPTA/AM, but not PD98059. Taken together, these findings suggest that NK exposure exerts urothelial cytotoxicity via a [Ca2+]i-regulated ERK1/2 activation, which is involved in downstream mediation of the mitochondria-dependent and ER stress-triggered apoptotic pathway, consequently resulting in urothelial cell death. Our findings suggest that regulating [Ca2+]i/ERK signaling pathways may be a promising strategy for treatment of NK-induced urothelial cystitis.


Asunto(s)
Cistitis , Ketamina , Apoptosis , Estrés del Retículo Endoplásmico , Femenino , Humanos , Ketamina/análogos & derivados , Ketamina/farmacología , Sistema de Señalización de MAP Quinasas , Masculino , Mitocondrias/metabolismo
2.
Int J Mol Sci ; 23(5)2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35270009

RESUMEN

Methylmercury (MeHg), a long-lasting organic pollutant, is known to induce cytotoxic effects in mammalian cells. Epidemiological studies have suggested that environmental exposure to MeHg is linked to the development of diabetes mellitus (DM). The exact molecular mechanism of MeHg-induced pancreatic ß-cell cytotoxicity is still unclear. Here, we found that MeHg (1-4 µM) significantly decreased insulin secretion and cell viability in pancreatic ß-cell-derived RIN-m5F cells. A concomitant elevation of mitochondrial-dependent apoptotic events was observed, including decreased mitochondrial membrane potential and increased proapoptotic (Bax, Bak, p53)/antiapoptotic (Bcl-2) mRNA ratio, cytochrome c release, annexin V-Cy3 binding, caspase-3 activity, and caspase-3/-7/-9 activation. Exposure of RIN-m5F cells to MeHg (2 µM) also induced protein expression of endoplasmic reticulum (ER) stress-related signaling molecules, including C/EBP homologous protein (CHOP), X-box binding protein (XBP-1), and caspase-12. Pretreatment with 4-phenylbutyric acid (4-PBA; an ER stress inhibitor) and specific siRNAs for CHOP and XBP-1 significantly inhibited their expression and caspase-3/-12 activation in MeHg-exposed RIN-mF cells. MeHg could also evoke c-Jun N-terminal kinase (JNK) activation and reactive oxygen species (ROS) generation. Antioxidant N-acetylcysteine (NAC; 1mM) or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox; 100 µM) markedly prevented MeH-induced ROS generation and decreased cell viability in RIN-m5F cells. Furthermore, pretreatment of cells with SP600125 (JNK inhibitor; 10 µM) or NAC (1 mM) or transfection with JNK-specific siRNA obviously attenuated the MeHg-induced JNK phosphorylation, CHOP and XBP-1 protein expression, apoptotic events, and insulin secretion dysfunction. NAC significantly inhibited MeHg-activated JNK signaling, but SP600125 could not effectively reduce MeHg-induced ROS generation. Collectively, these findings demonstrate that the induction of ROS-activated JNK signaling is a crucial mechanism underlying MeHg-induced mitochondria- and ER stress-dependent apoptosis, ultimately leading to ß-cell death.


Asunto(s)
Estrés del Retículo Endoplásmico , Compuestos de Metilmercurio , Animales , Apoptosis , Caspasa 3/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Mamíferos/metabolismo , Compuestos de Metilmercurio/farmacología , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
3.
Int J Mol Sci ; 22(9)2021 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922211

RESUMEN

4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), a major active metabolite of bisphenol A (BPA), is generated in the mammalian liver. Some studies have suggested that MBP exerts greater toxicity than BPA. However, the mechanism underlying MBP-induced pancreatic ß-cell cytotoxicity remains largely unclear. This study demonstrated the cytotoxicity of MBP in pancreatic ß-cells and elucidated the cellular mechanism involved in MBP-induced ß-cell death. Our results showed that MBP exposure significantly reduced cell viability, caused insulin secretion dysfunction, and induced apoptotic events including increased caspase-3 activity and the expression of active forms of caspase-3/-7/-9 and PARP protein. In addition, MBP triggered endoplasmic reticulum (ER) stress, as indicated by the upregulation of GRP 78, CHOP, and cleaved caspase-12 proteins. Pretreatment with 4-phenylbutyric acid (4-PBA; a pharmacological inhibitor of ER stress) markedly reversed MBP-induced ER stress and apoptosis-related signals. Furthermore, exposure to MBP significantly induced the protein phosphorylation of JNK and AMP-activated protein kinase (AMPK)α. Pretreatment of ß-cells with pharmacological inhibitors for JNK (SP600125) and AMPK (compound C), respectively, effectively abrogated the MBP-induced apoptosis-related signals. Both JNK and AMPK inhibitors also suppressed the MBP-induced activation of JNK and AMPKα and of each other. In conclusion, these findings suggest that MBP exposure exerts cytotoxicity on ß-cells via the interdependent activation of JNK and AMPKα, which regulates the downstream apoptotic signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fenoles/toxicidad , Animales , Supervivencia Celular , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratas , Transducción de Señal
4.
Am J Emerg Med ; 38(6): 1296.e1-1296.e3, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31956048

RESUMEN

A 64-year-old woman presented with coma, seizure, and lactic acidosis after ingesting 80 yam bean seeds. This rotenone-containing seeds cause cellular asphyxia via blockage of the mitochondrial electron transport. Subsequent oxidative stress results in the formation of lipid peroxidation (LPO). Rotenone analysis via liquid chromatography mass spectrometry revealed the following: 31,590 ng/mL in cooked yam bean seed and 100 ng/mL in the blood. We attempted to use N-acetylcysteine to alleviate oxidative stress and documented the continuous decline in the plasma concentration of LPO.


Asunto(s)
Pachyrhizus/efectos adversos , Rotenona/análisis , Acidosis Láctica/complicaciones , Acidosis Láctica/etiología , Coma/etiología , Femenino , Humanos , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Rotenona/efectos adversos , Rotenona/sangre , Convulsiones/etiología
5.
Mol Carcinog ; 57(11): 1599-1607, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30074275

RESUMEN

Triple negative breast cancer (TNBC) is aggressive with a worse prognosis. We have recently shown that bitter melon extract (BME) treatment was more effective in inhibition of TNBC tumor growth in mouse models as compared to ER positive breast tumor growth. Aberrant dysregulation of lipid metabolism is associated with breast cancer progression, however, anti-cancer mechanism of BME linking lipid metabolism in breast cancer growth remains unexplored. Here, we observed that accumulation of esterified cholesterol was reduced in BME treated TNBC cell lines as compared to control cells. We next evaluated expression levels of acyl-CoA: cholesterol acyltransferase 1 (ACAT-1) in TNBC cells treated with BME. Our results demonstrated that BME treatment inhibited ACAT-1 expression in TNBC cells. Subsequently, we found that sterol regulatory element-binding proteins-1 and -2, and FASN was significantly reduced in BME treated TNBC cell lines. Low-density lipoprotein receptor was also downregulated in BME treated TNBC cells as compared to control cells. We further demonstrated that BME feeding reduced tumor growth in TNBC mammospheres implanted into NSG mice, and inhibits ACAT-1 expression. To our knowledge, this is the first report demonstrating BME suppresses TNBC cell growth through ACAT-1 inhibition, and have potential for additional therapeutic regimen against human breast cancer.


Asunto(s)
Colesterol/metabolismo , Momordica charantia/química , Extractos Vegetales/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Esterificación/efectos de los fármacos , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Extractos Vegetales/química , Receptores de LDL/genética , Receptores de LDL/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Regul Toxicol Pharmacol ; 92: 67-74, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29155076

RESUMEN

Steady-fiber granule (SFG) is a functional food mixture that is composed of four major ingredients, resistant maltodextrin, white kidney bean (Phaseolus vulgaris) extract, mulberry leaf (Morus alba L.) extract, and niacin-bound chromium complex. This study focused on determining the safety of SFG. Genotoxicity and 28-day oral toxicity were evaluated. SFG did not induce mutagenicity in the bacterial reverse mutation assay using five Salmonella typhimurium strains (TA98, TA100, TA102, TA1535, and TA1537) in the presence or absence of metabolic activation (S9 system). SFG also did not induce clastogenic effects in Chinese hamster ovary cells with or without S9 treatment. Similarly, SFG did not induce genotoxicity in a micronucleus test conducted with mice. A dose-dependent 28-day oral toxicity assessment of SFG for rats revealed no significant effects on mortality, body weight, selected organ weights, and behavior. Evaluations of hematology, clinical biochemistry, and histopathology showed no adverse effects in rats treated with SFG. These results suggest that SFG has no significant mutagenic or toxic properties, and the no observed adverse effect level of SFG was defined as at least 5000 mg/kg/day orally for 28 days for male and female rats.


Asunto(s)
Alimentos Funcionales/efectos adversos , Morus/efectos adversos , Ácidos Nicotínicos/efectos adversos , Compuestos Organometálicos/efectos adversos , Phaseolus/efectos adversos , Extractos Vegetales/efectos adversos , Hojas de la Planta/efectos adversos , Polisacáridos/efectos adversos , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Células CHO , Cricetulus , Femenino , Masculino , Ratones , Ratones Endogámicos ICR , Pruebas de Mutagenicidad/métodos , Mutación/efectos de los fármacos , Niacina/efectos adversos , Ácidos Nicotínicos/administración & dosificación , Compuestos Organometálicos/administración & dosificación , Extractos Vegetales/administración & dosificación , Polisacáridos/administración & dosificación , Ratas , Ratas Wistar
7.
Toxicol Appl Pharmacol ; 294: 54-64, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26806093

RESUMEN

Molybdenum (Mo), a well-known toxic environmental and industrial pollutant, causes adverse health effects and diseases in humans and has received attention as a potential risk factor for DM. However, the roles of Mo in the mechanisms of the toxicological effects in pancreatic ß-cells are mostly unclear. In this study, the results revealed dysfunction of insulin secretion and apoptosis in the pancreatic ß-cell-derived RIN-m5F cells and the isolated mouse islets in response to Mo. These effects were accompanied by a mitochondria-dependent apoptotic signals including a decreased in the MMP, an increase in cytochrome c release, and the activation of caspase cascades and PARP. In addition, ER stress was triggered as indicated by several key molecules of the UPR. Furthermore, exposure to Mo induced the activation of ERK1/2, JNK, AMPKα, and GSK3-α/ß. Pretreatment with specific pharmacological inhibitors (in RIN-m5F cells and isolated mouse islets) of JNK (SP600125) and AMPK (Compound C) or transfection with si-RNAs (in RIN-m5F cells) specific to JNK and AMPKα effectively prevented the Mo-induced apoptosis and related signals, but inhibitors of ERK1/2 and GSK3-α/ß (PD98059 and LiCl, respectively) did not reverse the Mo-induced effects. Additionally, both the inhibitors and specific si-RNAs could suppress the Mo-induced phosphorylation of JNK and AMPKα each other. Taken together, these results suggest that Mo exerts its cytotoxicity on pancreatic ß-cells by inducing dysfunction and apoptosis via interdependent JNK and AMPK activation downstream-regulated mitochondrial-dependent and ER stress-triggered apoptosis pathways.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Quinasas Janus/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Molibdeno/farmacología , Animales , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos
8.
Int J Mol Sci ; 16(1): 924-49, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25561239

RESUMEN

Lipid metabolism is regulated by multiple signaling pathways, and generates a variety of bioactive lipid molecules. These bioactive lipid molecules known as signaling molecules, such as fatty acid, eicosanoids, diacylglycerol, phosphatidic acid, lysophophatidic acid, ceramide, sphingosine, sphingosine-1-phosphate, phosphatidylinositol-3 phosphate, and cholesterol, are involved in the activation or regulation of different signaling pathways. Lipid metabolism participates in the regulation of many cellular processes such as cell growth, proliferation, differentiation, survival, apoptosis, inflammation, motility, membrane homeostasis, chemotherapy response, and drug resistance. Bioactive lipid molecules promote apoptosis via the intrinsic pathway by modulating mitochondrial membrane permeability and activating different enzymes including caspases. In this review, we discuss recent data in the fields of lipid metabolism, lipid-mediated apoptosis, and cancer therapy. In conclusion, understanding the underlying molecular mechanism of lipid metabolism and the function of different lipid molecules could provide the basis for cancer cell death rationale, discover novel and potential targets, and develop new anticancer drugs for cancer therapy.


Asunto(s)
Apoptosis , Metabolismo de los Lípidos , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ácido Graso Sintasas/metabolismo , Glicerofosfolípidos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , MicroARNs/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingolípidos/metabolismo
9.
Curr Res Toxicol ; 6: 100155, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38379848

RESUMEN

Paraquat (PQ), a toxic and nonselective bipyridyl herbicide, is one of the most extensively used pesticides in agricultural countries. In addition to pneumotoxicity, the liver is an important target organ for PQ poisoning in humans. However, the mechanism of PQ in hepatotoxicity remains unclear. In this study, we found that exposure of rat hepatic H4IIE cells to PQ (0.1-2 mM) induced significant cytotoxicity and apoptosis, which was accompanied by mitochondria-dependent apoptotic signals, including loss of mitochondrial membrane potential (MMP), cytosolic cytochrome c release, and changes in the Bcl-2/Bax mRNA ratio. Moreover, PQ (0.5 mM) exposure markedly induced JNK and ERK1/2 activation, but not p38-MAPK. Blockade of JNK and ERK1/2 signaling by pretreatment with the specific pharmacological inhibitors SP600125 and PD98059, respectively, effectively prevented PQ-induced cytotoxicity, mitochondrial dysfunction, and apoptotic events. Additionally, PQ exposure stimulated significant oxidative stress-related signals, including reactive oxygen species (ROS) generation and intracellular glutathione (GSH) depletion, which could be reversed by the antioxidant N-Acetylcysteine (NAC). Buffering the oxidative stress response with NAC also effectively abrogated PQ-induced hepatotoxicity, MMP loss, apoptosis, and phosphorylation of JNK and ERK1/2 protein, however, the JNK or ERK inhibitors did not suppress ROS generation in PQ-treated cells. Collectively, these results demonstrate that PQ exposure induces hepatic cell toxicity and death via an oxidative stress-dependent JNK/ERK activation-mediated downstream mitochondria-regulated apoptotic pathway.

10.
Eur J Pharmacol ; 977: 176676, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38815787

RESUMEN

Wogonin, a vital bioactive compound extracted from the medicinal plant, Scutellaria baicalensis, has been wildly used for its potential in mitigating the progression of chronic diseases. Chronic kidney disease (CKD) represents a significant global health challenge due to its high prevalence, morbidity and mortality rates, and associated complications. This study aimed to assess the potential of wogonin in attenuating renal fibrosis and to elucidate the underlying molecular mechanisms using a unilateral ureteral obstruction (UUO) mouse model as a CKD mimic. Male mice, 8 weeks old, underwent orally administrated of either 50 mg/kg/day of wogonin or positive control of 5 mg/kg/day candesartan following UUO surgery. NRK52E cells were exposed to tumor growth factors-beta (TGF-ß) to evaluate the anti-fibrotic effects of wogonin. The results demonstrated that wogonin treatment effectively attenuated TGF-ß-induced fibrosis markers in NRK-52E cells. Additionally, administration of wogonin significantly improved histopathological alterations and downregulated the expression of pro-fibrotic factors (Fibronectin, α-smooth muscle actin, Collagen IV, E-cadherin, and TGF-ß), oxidative stress markers (Catalase, superoxide dismutase 2, NADPH oxidase 4, and thioredoxin reductase 1), inflammatory molecules (Cyclooxygenase-2 and TNF-α), and the infiltration of neutrophils and macrophages in UUO mice. Furthermore, wogonin treatment mitigated endoplasmic reticulum (ER) stress-associated molecular markers (GRP78, GRP94, ATF4, CHOP, and the caspase cascade) and suppressed apoptosis. The findings indicate that wogonin treatment ameliorates key fibrotic aspects of CKD by attenuating ER stress-related apoptosis, inflammation, and oxidative stress, suggesting its potential as a future therapeutic target.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Fibrosis , Flavanonas , Obstrucción Ureteral , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología , Obstrucción Ureteral/tratamiento farmacológico , Flavanonas/farmacología , Flavanonas/uso terapéutico , Apoptosis/efectos de los fármacos , Masculino , Ratones , Línea Celular , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Factor de Crecimiento Transformador beta/metabolismo , Ratas , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/patología , Ratones Endogámicos C57BL
11.
Exp Cell Res ; 318(10): 1075-85, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22472346

RESUMEN

The glomerular capillary wall, composed of endothelial cells, the glomerular basement membrane and the podocytes, is continually subjected to hemodynamic force arising from tractional stress due to blood pressure and shear stress due to blood flow. Exposure of glomeruli to abnormal hemodynamic force such as hyperfiltration is associated with glomerular injury and progressive renal disease, and the conversion of mechanical stimuli to chemical signals in the regulation of the process is poorly understood in podocytes. By examining DNA fragmentation, apoptotic nuclear changes and cytochrome c release, we found that shear stress induced cell apoptosis in cultured podocytes. Meanwhile, podocytes exposed to shear stress also stimulated c-Src phosphorylation, phospholipase D (PLD) activation and mammalian target of rapamycin (mTOR) signaling. Using the antibodies against c-Src, PLD(1), and PLD(2) to perform reciprocal co-immunoprecipitations and in vitro PLD activity assay, our data indicated that c-Src interacted with and activated PLD(1) but not PLD(2). The inhibition of shear stress-induced c-Src phosphorylation by PP(2) (a specific inhibitor of c-Src kinase) resulted in reduced PLD activity. Phosphatidic acid, produced by shear stress-induced PLD activation, stimulated mTOR signaling, and caused podocyte hypertrophy and apoptosis.


Asunto(s)
Apoptosis , Fosfolipasa D/metabolismo , Podocitos/fisiología , Proteínas Tirosina Quinasas/metabolismo , Estrés Fisiológico , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Células Cultivadas , Ratones , Fosfatidilcolinas/metabolismo , Fosforilación , Podocitos/enzimología , Podocitos/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Resistencia al Corte , Transducción de Señal , Familia-src Quinasas
12.
Toxicol In Vitro ; 86: 105483, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36252918

RESUMEN

Chlorpyrifos (CPF) is one of the most abundant and widely used organophosphate pesticides for agricultural, industrial, and household purposes in the world. Epidemiological studies have reported that CPF can induce neurotoxic impairments in mammalian, which is linked to an important risk factor for development of neurodegenerative diseases (NDs). However, limited information is available on CPF-induced neurotoxicity, with the underlying exact mechanism remains unclear. In this study, CPF exposure (10-400 µM) significantly reduced Neuro-2a cell viability and induced apoptotic events, including the increase in caspase-3 activity, apoptotic cell population, and cleavage of caspase-3/-7 and PARP. Exposure of Neuro-2a cells to CPF also triggered CHOP activation. Transfection with CHOP-specific siRNA markedly suppressed the expression of CHOP, and attenuated cytotoxicity and apoptotic events in CPF-exposed Neuro-2a cells. Furthermore, CPF exposure obviously evoked the phosphorylation of Akt as well as ROS generation in a time-dependent manner. Pretreatment with LY294002 (an Akt inhibitor) effectively attenuated the CPF-induced Akt phosphorylation, CHOP activation, and apoptotic events, but not that ROS production. Of note, buffering the ROS generation with antioxidant N-acetylcysteine effectively prevented the CPF-induced ROS generation, CHOP activation, and apoptotic events, but not that the Akt phosphorylation. Collectively, these findings indicate that CPF exposure exerts neuronal cytotoxicity via the independent pathways of ROS generation and Akt activation downstream-regulated CHOP-triggered apoptosis, ultimately leading to neuronal cell death.


Asunto(s)
Cloropirifos , Animales , Cloropirifos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Caspasa 3/metabolismo , Estrés Oxidativo , Muerte Celular , Apoptosis , Mamíferos/metabolismo
13.
J Biol Chem ; 286(3): 1828-35, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21084311

RESUMEN

The Ca(2+)-sensing receptor (CaR) regulates salt and water transport in the kidney as demonstrated by the association of gain of function CaR mutations with a Bartter syndrome-like, salt-wasting phenotype, but the precise mechanism for this effect is not fully established. We found previously that the CaR interacts with and inactivates an inwardly rectifying K(+) channel, Kir4.1, which is expressed in the distal nephron that contributes to the basolateral K(+) conductance, and in which loss of function mutations are associated with a complex phenotype that includes renal salt wasting. We now find that CaR inactivates Kir4.1 by reducing its cell surface expression. Mutant CaRs reduced Kir4.1 cell surface expression and current density in HEK-293 cells in proportion to their signaling activity. Mutant, activated Gα(q) reduced cell surface expression and current density of Kir4.1, and these effects were blocked by RGS4, a protein that blocks signaling via Gα(i) and Gα(q). Other α subunits had insignificant effects. Knockdown of caveolin-1 blocked the effect of Gα(q) on Kir4.1, whereas knockdown of the clathrin heavy chain had no effect. CaR had no comparable effect on the renal outer medullary K(+) channel, an apical membrane distal nephron K(+) channel that is internalized by clathrin-coated vesicles. Co-immunoprecipitation studies showed that the CaR and Kir4.1 physically associate with caveolin-1 in HEK cells and in kidney extracts. Thus, the CaR decreases cell surface expression of Kir4.1 channels via a mechanism that involves Gα(q) and caveolin. These results provide a novel molecular basis for the inhibition of renal NaCl transport by the CaR.


Asunto(s)
Regulación de la Expresión Génica , Nefronas/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal , Cloruro de Sodio/metabolismo , Animales , Transporte Biológico/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Cadenas Pesadas de Clatrina/genética , Cadenas Pesadas de Clatrina/metabolismo , Vesículas Cubiertas por Clatrina/genética , Vesículas Cubiertas por Clatrina/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Ratas , Receptores Sensibles al Calcio/genética , Desequilibrio Hidroelectrolítico/genética , Desequilibrio Hidroelectrolítico/metabolismo
14.
J Biomed Biotechnol ; 2012: 254582, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22888198

RESUMEN

Cinnabar, a naturally occurring mercuric sulfide (HgS), has long been used in Chinese mineral medicine for more than 2000 years. Although mercury is well-known for its toxicity, whether cinnabar induces neurotoxicity, especially in infants and children, is unknown. The purpose of this study was to explore the neurotoxic effects of low-dose of cinnabar (10 mg/kg/day) on developing mice. The results revealed neurobehavioral defects in F1-C-Cin group, which were associated with Hg accumulation, increased NO(x) levels in whole blood, and Na(+)/K(+)-ATPase activities in brain tissues. F1- and F2-Cin-V groups were found to increase brain Hg contents and prominent neurobehavioral defects compared with F1-C-V group, suggesting that the fetal brain was more susceptible to irreversible effects for cinnabar-induced damage. Moreover, F1- and F2-Cin-Cin groups had severely neurobehavioral dysfunctions, closely correlated with the further alteration of NO(x) levels and Na(+)/K(+)-ATPase activities than F1- and F2-C-Cin groups. Effects in F2-Cin-Cin group were more significant than those in F1-Cin-Cin group. In conclusion, this study demonstrates that exposure to low-dose of cinnabar during the perinatal and developmental stages results in irreversible and severe injuries of the neurotoxicity in offspring, and NO(x) and Na(+)/K(+)-ATPase activities may exist potential and useful biomarkers for neurotoxicity-induced by low-doses of mercuric compounds.


Asunto(s)
Compuestos de Mercurio/administración & dosificación , Compuestos de Mercurio/toxicidad , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/patología , Neurotoxinas/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Animales Recién Nacidos , Umbral Auditivo/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Femenino , Audición/efectos de los fármacos , Tamaño de la Camada , Locomoción/efectos de los fármacos , Masculino , Mercurio/sangre , Ratones , Ratones Endogámicos ICR , Óxido Nítrico/sangre , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Sueño/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Tiempo
15.
Arch Toxicol ; 86(6): 923-33, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22622864

RESUMEN

A pool of myoblasts available for myogenesis is important for skeletal muscle size. The decreased number of skeletal muscle fibers could be due to the decreased myoblast proliferation or cytotoxicity. Identification of toxicants that regulate myoblast apoptosis is important in skeletal muscle development or regeneration. Here, we investigate the cytotoxic effect and its possible mechanisms of arsenic trioxide (As(2)O(3)) on myoblasts. C2C12 myoblasts underwent apoptosis in response to As(2)O(3) (1-10 µM), accompanied by increased Bax/Bcl-2 ratio, decreased mitochondria membrane potential, increased cytochrome c release, increased caspase-3/-9 activity, and increased poly (ADP-ribose) polymerase (PARP) cleavage. Moreover, As(2)O(3) triggered the endoplasmic reticulum (ER) stress indentified through several key molecules of the unfolded protein response, including glucose-regulated protein (GRP)-78, GRP-94, PERK, eIF2α, ATF6, and caspase-12. Pretreatment with antioxidant N-acetylcysteine (NAC, 0.5 mM) dramatically suppressed the increases in reactive oxygen species (ROS), lipid peroxidation, ER stress, caspase cascade activity, and apoptosis in As(2)O(3) (10 µM)-treated myoblasts. Furthermore, As(2)O(3) (10 µM) effectively decreased the phosphorylation of Akt, which could be reversed by NAC. Over-expression of constitutive activation of Akt (c.a. Akt) also significantly attenuated As(2)O(3)-induced myoblast apoptosis. Taken together, these results suggest that As(2)O(3) may exert its cytotoxicity on myoblasts by inducing apoptosis through a ROS-induced mitochondrial dysfunction, ER stress, and Akt inactivation signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsénico/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mioblastos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Trióxido de Arsénico , Arsenicales , Línea Celular , Citotoxinas/toxicidad , Humanos , Mioblastos/metabolismo , Óxidos/toxicidad , Transducción de Señal/efectos de los fármacos
16.
Int J Mol Sci ; 13(10): 12349-66, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23202902

RESUMEN

Mercury is a toxic heavy metal that is an environmental and industrial pollutant throughout the world. Mercury exposure leads to many physiopathological injuries in mammals. However, the precise toxicological effects of mercury on pancreatic islets in vivo are still unclear. Here, we investigated whether mercuric compounds can induce dysfunction and damage in the pancreatic islets of mice, as well as the possible mechanisms involved in this process. Mice were treated with methyl mercuric chloride (MeHgCl, 2 mg/kg) and mercuric chloride (HgCl(2), 5 mg/kg) for more than 2 consecutive weeks. Our results showed that the blood glucose levels increased and plasma insulin secretions decreased in the mice as a consequence of their exposure. A significant number of TUNEL-positive cells were revealed in the islets of mice that were treated with mercury for 2 consecutive weeks, which was accompanied by changes in the expression of the mRNA of anti-apoptotic (Bcl-2, Mcl-1, and Mdm-2) and apoptotic (p53, caspase-3, and caspase-7) genes. Moreover, plasma malondialdehyde (MDA) levels increased significantly in the mice after treatment with mercuric compounds for 2 consecutive weeks, and the generation of reactive oxygen species (ROS) in the pancreatic islets also markedly increased. In addition, the mRNA expression of genes related to antioxidation, including Nrf2, GPx, and NQO1, were also significantly reduced in these islets. These results indicate that oxidative stress injuries that are induced by mercuric compounds can cause pancreatic islets dysfunction and apoptosis in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Cloruro de Mercurio/toxicidad , Compuestos de Metilmercurio/toxicidad , Animales , Glucemia/análisis , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 7/genética , Caspasa 7/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Insulina/sangre , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/sangre , Cloruro de Mercurio/química , Compuestos de Metilmercurio/química , Ratones , Ratones Endogámicos ICR , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Materials (Basel) ; 15(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35208076

RESUMEN

The influence of alloying element Mg on Na and Sr modifying Al-7Si hypoeutectic alloys was investigated. The residual content of Na and the morphology of modified eutectic silicon were characterized. It was found that the alloying element Mg had an enhanced effect on the uptake of sodium in the Al-7Si hypoeutectic alloy modified by the Na-contained modifier. Moreover, the morphology of eutectic silicon of the modified Al-7Si alloys was significantly different from that of Al-7Si-0.4Mg alloys in the present research. When the addition of the modifier is enough, both modifiers could entirely modify the eutectic silicon phase of Al-7Si alloys, while incompletely modified eutectic silicon was observed in both Na-modified and Sr-modified Al-7Si-0.4Mg alloy. It was observed that there was an adhering relationship between the partially modified eutectic silicon with Mg-rich phases. According to the results, it can be proposed that the addition of Mg will affect the solidification behavior of alloys, thereby, leading to the incomplete modification of eutectic silicon phases.

18.
Oncol Lett ; 23(3): 78, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35111247

RESUMEN

Tongue squamous cell carcinoma (SCC) is a most common type of oral cancer. Due to its highly invasive nature and poor survival rate, the development of effective pharmacological therapeutic agents is urgently required. Quercetin (3,3',4',5,7-pentahydroxyflavone) is a polyphenolic flavonoid found in plants and is an active component of Chinese herbal medicine. The present study investigated the pharmacological effects and possible mechanisms of quercetin on apoptosis of the tongue SCC-derived SAS cell line. Following treatment with quercetin, cell viability was assessed via the MTT assay. Apoptotic and necrotic cells, mitochondrial transmembrane potential and caspase-3/7 activity were analyzed via flow cytometric analyses. A caspase-3 activity assay kit was used to detect the expression of caspase-3 activity. Western blot analysis was performed to examine the expression levels of proteins associated with the MAPKs, AMPKα, GSK3-α/ß and caspase-related signaling pathways. The results revealed that quercetin induced morphological alterations and decreased the viability of SAS cells. Quercetin also increased apoptosis-related Annexin V-FITC fluorescence and caspase-3 activity, and induced mitochondria-dependent apoptotic signals, including a decrease in mitochondrial transmembrane potential and Bcl-2 protein expression, and an increase in cytosolic cytochrome c, Bax, Bak, cleaved caspase-3, cleaved caspase-7 and cleaved poly (ADP-ribose) polymerase protein expression. Furthermore, quercetin significantly increased the protein expression levels of phosphorylated (p)-ERK, p-JNK1/2 and p-GSK3-α/ß, but not p-p38 or p-AMPKα in SAS cells. Pretreatment with the pharmacological JNK inhibitor SP600125 effectively reduced the quercetin-induced apoptosis-related signals, as well as p-ERK1/2 and p-GSK3-α/ß protein expression. Both ERK1/2 and GSK3-α/ß inhibitors, PD98059 and LiCl, respectively, could significantly prevent the quercetin-induced phosphorylation of ERK1/2 and GSK3-α/ß, but not JNK activation. Taken together, these results suggested that quercetin may induce tongue SCC cell apoptosis via the JNK-activation-regulated ERK1/2 and GSK3-α/ß-mediated mitochondria-dependent apoptotic signaling pathway.

19.
Am J Physiol Cell Physiol ; 300(3): C397-405, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21123730

RESUMEN

The mechanical properties of tissues and cells including renal glomeruli are important determinants of their differentiated state, function, and responses to injury but are not well characterized or understood. Understanding glomerular mechanics is important for understanding renal diseases attributable to abnormal expression or assembly of structural proteins and abnormal hemodynamics. We use atomic force microscopy (AFM) and a new technique, capillary micromechanics, to measure the elastic properties of rat glomeruli. The Young's modulus of glomeruli was 2,500 Pa, and it was reduced to 1,100 Pa by cytochalasin and latunculin, and to 1,400 Pa by blebbistatin. Cytochalasin or latrunculin reduced the F/G actin ratios of glomeruli but did not disrupt their architecture. To assess glomerular biomechanics in disease, we measured the Young's moduli of glomeruli from two mouse models of primary glomerular disease, Col4a3(-/-) mice (Alport model) and Tg26(HIV/nl) mice (HIV-associated nephropathy model), at stages where glomerular injury was minimal by histopathology. Col4a3(-/-) mice express abnormal glomerular basement membrane proteins, and Tg26(HIV/nl) mouse podocytes have multiple abnormalities in morphology, adhesion, and cytoskeletal structure. In both models, the Young's modulus of the glomeruli was reduced by 30%. We find that glomeruli have specific and quantifiable biomechanical properties that are dependent on the state of the actin cytoskeleton and nonmuscle myosins. These properties may be altered early in disease and represent an important early component of disease. This increased deformability of glomeruli could directly contribute to disease by permitting increased distension with hemodynamic force or represent a mechanically inhospitable environment for glomerular cells.


Asunto(s)
Módulo de Elasticidad/fisiología , Elasticidad/fisiología , Enfermedades Renales/fisiopatología , Glomérulos Renales/fisiología , Glomérulos Renales/fisiopatología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Autoantígenos/genética , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/genética , Módulo de Elasticidad/efectos de los fármacos , Elasticidad/efectos de los fármacos , Enfermedades Renales/patología , Glomérulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley
20.
Int J Cancer ; 128(12): 2833-42, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20734387

RESUMEN

Ca(2+) is a ubiquitous cellular signal which plays a central role in the regulation of cell function. To understand aberrant signaling through the Ca(2+)-sensing receptor (CaR) in prostate cancer cells, we compared expression of CaR signaling components in human nonmalignant prostate epithelial cells and several prostate cancer cell lines, as well as normal human prostate and prostate tumor specimens. We found that levels of the CaR, Gα(12) and p115RhoGEF expression are significantly up-regulated in more tumorigenic prostate cancer cells and prostate tumor specimens. By silencing CaR, Gα(12), p115RhoGEF or choline kinase (ChoK) expression, analyzing the change in lipid profiles, blocking signaling pathways using chemical inhibitors, and co-immunoprecipitating the relevant signaling proteins, we demonstrate that p115RhoGEF, a regulator of G protein signaling (RGS) with GAP activity for Gα(12/13) and with guanine nucleotide exchange activity for the small G protein Rho, plays an important role in the regulation of Ca(o)(2+)-induced ChoK activation and cell proliferation in more tumorigenic prostate cancer cell lines. The results demonstrate an important role of p115RhoGEF in prostate tumorigenesis and provide a potential target of cancer therapeutics.


Asunto(s)
Calcio/metabolismo , Colina Quinasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Humanos , Inmunoprecipitación , Masculino , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA