Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Toxicol Pathol ; 37(1): 39-43, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38283374

RESUMEN

Ameloblastoma is a locally aggressive tumor derived from the odontogenic epithelium of the developing tooth germ. It is rarely reported in experimental Sprague-Dawley (SD) rats. In this 90-day percutaneous repeated-dose toxicity study, mandibular nodules were observed from day 56 to 90. Upon necropsy, a well-demarcated nodule, approximately 1.2×1.0×1.0 cm, was found attached to the mandibular bone, alongside the unerupted left incisor. Histopathologically, the epithelial cells formed islands, nests, or anastomosing strands. The epithelial islands were surrounded by a peripheral layer of tall columnar cells with antibasilar nuclei arranged in a palisading pattern. Several mitotic cells were observed. Some epithelial islands displayed cystic degenerative changes and squamous metaplasia. Necrotic tissue with inflammatory cell infiltration was observed at the tumor margins. Immunohistochemically, the neoplastic epithelial islands and mesenchymal components exhibited positivity for pan-cytokeratin and vimentin, respectively. Based on these features, the case was diagnosed as follicular ameloblastoma in an SD rat.

2.
J Immunol ; 203(9): 2532-2544, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31548328

RESUMEN

Dual oxidase 2 (DUOX2) generates H2O2 that plays a critical role in both host defense and chronic inflammation. Previously, we demonstrated that the proinflammatory mediators IFN-γ and LPS enhance expression of DUOX2 and its maturation factor DUOXA2 through STAT1- and NF-κB‒mediated signaling in human pancreatic cancer cells. Using a panel of colon and pancreatic cancer cell lines, we now report the induction of DUOX2/DUOXA2 mRNA and protein expression by the TH2 cytokine IL-4. IL-4 activated STAT6 signaling that, when silenced, significantly decreased induction of DUOX2. Furthermore, the TH17 cytokine IL-17A combined synergistically with IL-4 to increase DUOX2 expression in both colon and pancreatic cancer cells mediated, at least in part, by signaling through NF-κB. The upregulation of DUOX2 was associated with a significant increase in the production of extracellular H2O2 and DNA damage-as indicated by the accumulation of 8-oxo-dG and γH2AX-which was suppressed by the NADPH oxidase inhibitor diphenylene iodonium and a DUOX2-specific small interfering RNA. The clinical relevance of these experiments is suggested by immunohistochemical, microarray, and quantitative RT-PCR studies of human colon and pancreatic tumors demonstrating significantly higher DUOX2, IL-4R, and IL-17RA expression in tumors than in adjacent normal tissues; in pancreatic adenocarcinoma, increased DUOX2 expression is adversely associated with overall patient survival. These data suggest a functional association between DUOX2-mediated H2O2 production and induced DNA damage in gastrointestinal malignancies.


Asunto(s)
Neoplasias del Colon/metabolismo , Daño del ADN , Oxidasas Duales/genética , Peróxido de Hidrógeno/metabolismo , Interleucina-17/farmacología , Interleucina-4/farmacología , Neoplasias Pancreáticas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/patología , Humanos , FN-kappa B/fisiología , Oxidación-Reducción , Neoplasias Pancreáticas/patología , Receptores de Interleucina-4/fisiología , Factor de Transcripción STAT6/fisiología , Transducción de Señal , Regulación hacia Arriba
3.
J Biol Chem ; 292(19): 7866-7887, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28330872

RESUMEN

Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O2̇̄, is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decrease NOX1 expression stably in HT-29 human colon cancer cells. The 80-90% decrease in NOX1 expression achieved by RNAi produced a significant decline in ROS production and a G1/S block that translated into a 2-3-fold increase in tumor cell doubling time without increased apoptosis. The block at the G1/S checkpoint was associated with a significant decrease in cyclin D1 expression and profound inhibition of mitogen-activated protein kinase (MAPK) signaling. Decreased steady-state MAPK phosphorylation occurred concomitant with a significant increase in protein phosphatase activity for two colon cancer cell lines in which NOX1 expression was knocked down by RNAi. Diminished NOX1 expression also contributed to decreased growth, blood vessel density, and VEGF and hypoxia-inducible factor 1α (HIF-1α) expression in HT-29 xenografts initiated from NOX1 knockdown cells. Microarray analysis, supplemented by real-time PCR and Western blotting, revealed that the expression of critical regulators of cell proliferation and angiogenesis, including c-MYC, c-MYB, and VEGF, were down-regulated in association with a decline in hypoxic HIF-1α protein expression downstream of silenced NOX1 in both colon cancer cell lines and xenografts. These studies suggest a role for NOX1 in maintaining the proliferative phenotype of some colon cancers and the potential of NOX1 as a therapeutic target in this disease.


Asunto(s)
Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Colon/metabolismo , Ciclina D1/metabolismo , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , NADPH Oxidasa 1 , Trasplante de Neoplasias , Fenotipo , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Mol Carcinog ; 56(12): 2643-2662, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28762556

RESUMEN

NADPH oxidase 5 (NOX5) generated reactive oxygen species (ROS) have been implicated in signaling cascades that regulate cancer cell proliferation. To evaluate and validate NOX5 expression in human tumors, we screened a broad range of tissue microarrays (TMAs), and report substantial overexpression of NOX5 in malignant melanoma and cancers of the prostate, breast, and ovary. In human UACC-257 melanoma cells that possesses high levels of functional endogenous NOX5, overexpression of NOX5 resulted in enhanced cell growth, increased numbers of BrdU positive cells, and increased γ-H2AX levels. Additionally, NOX5-overexpressing (stable and inducible) UACC-257 cells demonstrated increased normoxic HIF-1α expression and decreased p27Kip1 expression. Similarly, increased normoxic HIF-1α expression and decreased p27Kip1 expression were observed in stable NOX5-overexpressing clones of KARPAS 299 human lymphoma cells and in the human prostate cancer cell line, PC-3. Conversely, knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased cell growth, decreased HIF-1α expression, and increased p27Kip1 expression. Likewise, in an additional human melanoma cell line, WM852, and in PC-3 cells, transient knockdown of endogenous NOX5 resulted in increased p27Kip1 and decreased HIF-1α expression. Knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased Akt and GSK3ß phosphorylation, signaling pathways known to modulate p27Kip1 levels. In summary, our findings suggest that NOX5 expression in human UACC-257 melanoma cells could contribute to cell proliferation due, in part, to the generation of high local concentrations of extracellular ROS that modulate multiple pathways that regulate HIF-1α and networks that signal through Akt/GSK3ß/p27Kip1 .


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , NADPH Oxidasa 5/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , NADPH Oxidasa 5/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN
5.
Clin Sci (Lond) ; 128(12): 863-75, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25818486

RESUMEN

The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1-5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.


Asunto(s)
NADPH Oxidasas/metabolismo , Neoplasias/enzimología , Neoplasias Hematológicas/enzimología , Humanos , NADPH Oxidasas/genética , NADPH Oxidasas/fisiología , Lesiones Precancerosas/enzimología , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
6.
J Immunol ; 190(4): 1859-72, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23296709

RESUMEN

Pancreatitis is associated with release of proinflammatory cytokines and reactive oxygen species and plays an important role in the development of pancreatic cancer. We recently demonstrated that dual oxidase (Duox)2, an NADPH oxidase essential for reactive oxygen species-related, gastrointestinal host defense, is regulated by IFN-γ-mediated Stat1 binding to the Duox2 promoter in pancreatic tumor lines. Because LPS enhances the development and invasiveness of pancreatic cancer in vivo following TLR4-related activation of NF-κB, we examined whether LPS, alone or combined with IFN-γ, regulated Duox2. We found that upregulation of TLR4 by IFN-γ in BxPC-3 and CFPAC-1 pancreatic cancer cells was augmented by LPS, resulting in activation of NF-κB, accumulation of NF-κB (p65) in the nucleus, and increased binding of p65 to the Duox2 promoter. TLR4 silencing with small interfering RNAs, as well as two independent NF-κB inhibitors, attenuated LPS- and IFN-γ-mediated Duox2 upregulation in BxPC-3 cells. Induction of Duox2 expression by IFN-γ and LPS may result from IFN-γ-related activation of Stat1 acting in concert with NF-κB-related upregulation of Duox2. Sustained extracellular accumulation of H(2)O(2) generated by exposure to both LPS and IFN-γ was responsible for an ∼50% decrease in BxPC-3 cell proliferation associated with a G(1) cell cycle block, apoptosis, and DNA damage. We also demonstrated upregulation of Duox expression in vivo in pancreatic cancer xenografts and in patients with chronic pancreatitis. These results suggest that inflammatory cytokines can interact to produce a Duox-dependent pro-oxidant milieu that could increase the pathologic potential of pancreatic inflammation and pancreatic cancer cells.


Asunto(s)
Interferón gamma/fisiología , Lipopolisacáridos/fisiología , Proteínas de la Membrana/biosíntesis , NADPH Oxidasas/biosíntesis , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular Tumoral , Enfermedad Crónica , Oxidasas Duales , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/enzimología , Pancreatitis/enzimología , Pancreatitis/inmunología , Pancreatitis/metabolismo , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 4/fisiología , Células Tumorales Cultivadas
7.
Chem Biol Interact ; 380: 110511, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37120125

RESUMEN

Carteolol is a commonly-used topical medication for primary open-angle glaucoma. However, long-term and frequent ocular application of carteolol entails its residuals at low concentration in the aqueous humor for a long duration and may exert latent toxicity in the human corneal endothelial cells (HCEnCs). Here, we treated the HCEnCs in vitro with 0.0117% carteolol for 10 days. Thereafter, we removed the cartelolol and normally cultured the cells for 25 days to investigate the chronical toxicity of carteolol and the underlying mechanism. The results exhibited that 0.0117% carteolol induces senescent features in the HCEnCs, such as increased senescence-associated ß-galactosidase positive rates, enlarged relative cell area and upregulated p16INK4A and senescence-associated secretory phenotypes, including IL-1α, TGF-ß1, IL-10, TNF-α, CCL-27, IL-6 and IL-8, as well as decreased Lamin B1 expression and cell viability and proliferation. Thereby, further exploration demonstrated that the carteolol activates ß-arrestin-ERK-NOX4 pathway to increase reactive oxygen species (ROS) production that imposes oxidative stress on energetic metabolism causing a vicious cycle between declining ATP and increasing ROS production and downregulation of NAD+ resulting in metabolic disturbance-mediated senescence of the HCEnCs. The excess ROS also impair DNA to activate the DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with diminished poly(ADP-Ribose) polymerase (PARP) 1, a NAD+-dependent enzyme for DNA damage repair, resulting in cell cycle arrest and subsequent DDR-mediated senescence. Taken together, carteolol induces excess ROS to trigger HCEnC senescence via metabolic disturbance and DDR pathway.


Asunto(s)
Carteolol , Glaucoma de Ángulo Abierto , Humanos , Especies Reactivas de Oxígeno/metabolismo , Senescencia Celular , Transducción de Señal/fisiología , Células Endoteliales/metabolismo , beta-Arrestinas/metabolismo , NAD/metabolismo , NADPH Oxidasa 4/metabolismo
8.
Free Radic Biol Med ; 205: 262-274, 2023 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-37330147

RESUMEN

Pro-inflammatory cytokines upregulate the expression of the H2O2-producing NADPH oxidase dual oxidase 2 (DUOX2)2 which, when elevated, adversely affects survival from pancreatic ductal adenocarcinoma (PDAC). Because the cGAS-STING pathway is known to initiate pro-inflammatory cytokine expression following uptake of exogenous DNA, we examined whether activation of cGAS-STING could play a role in the generation of reactive oxygen species by PDAC cells. Here, we found that a variety of exogenous DNA species markedly increased the production of cGAMP, the phosphorylation of TBK1 and IRF3, and the translocation of phosphorylated IRF3 into the nucleus, leading to a significant, IRF3-dependent enhancement of DUOX2 expression, and a significant flux of H2O2 in PDAC cells. However, unlike the canonical cGAS-STING pathway, DNA-related DUOX2 upregulation was not mediated by NF-κB. Although exogenous IFN-ß significantly increased Stat1/2-associated DUOX2 expression, intracellular IFN-ß signaling that followed cGAMP or DNA exposure did not itself increase DUOX2 levels. Finally, DUOX2 upregulation subsequent to cGAS-STING activation was accompanied by the enhanced, normoxic expression of HIF-1α and VEGF-A as well as DNA double strand cleavage, suggesting that cGAS-STING signaling may support the development of an oxidative, pro-angiogenic microenvironment that could contribute to the inflammation-related genetic instability of pancreatic cancer.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias Pancreáticas , Humanos , Oxidasas Duales/genética , Oxidasas Duales/metabolismo , Peróxido de Hidrógeno/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Transducción de Señal , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , ADN/metabolismo , Citocinas , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral
9.
J Biol Chem ; 286(14): 12245-56, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21321110

RESUMEN

Dual oxidase 2 is a member of the NADPH oxidase (Nox) gene family that plays a critical role in the biosynthesis of thyroid hormone as well as in the inflammatory response of the upper airway mucosa and in wound healing, presumably through its ability to generate reactive oxygen species, including H2O2. The recently discovered overexpression of Duox2 in gastrointestinal malignancies, as well as our limited understanding of the regulation of Duox2 expression, led us to examine the effect of cytokines and growth factors on Duox2 in human tumor cells. We found that exposure of human pancreatic cancer cells to IFN-γ (but not other agents) produced a profound up-regulation of the expression of Duox2, and its cognate maturation factor DuoxA2, but not other members of the Nox family. Furthermore, increased Duox2/DuoxA2 expression was closely associated with a significant increase in the production of both intracellular reactive oxygen species and extracellular H2O2. Examination of IFN-γ-mediated signaling events demonstrated that in addition to the canonical Jak-Stat1 pathway, IFN-γ activated the p38-MAPK pathway in pancreatic cancer cells, and both played an important role in the induction of Duox2 by IFN-γ. Duox2 up-regulation following IFN-γ exposure is also directly associated with the binding of Stat1 to elements of the Duox2 promoter. Our findings suggest that the pro-inflammatory cytokine IFN-γ initiates a Duox2-mediated reactive oxygen cascade in human pancreatic cancer cells; reactive oxygen species production in this setting could contribute to the pathophysiologic characteristics of these tumors.


Asunto(s)
Interferón gamma/farmacología , Proteínas de la Membrana/metabolismo , NADPH Oxidasas/metabolismo , Factor de Transcripción STAT1/metabolismo , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Oxidasas Duales , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/metabolismo , Proteínas de la Membrana/genética , Microscopía Confocal , NADPH Oxidasas/genética , Compuestos Onio/farmacología , Reacción en Cadena de la Polimerasa , Unión Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/genética
10.
J Photochem Photobiol B ; 235: 112568, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36137302

RESUMEN

The human corneal endothelial cells (HCEnCs) play a vital role in the maintenance of corneal transparency and visual acuity. In our daily life, HCEnCs are inevitably exposed to ultraviolet B (UVB) radiation leading to decreases of visual acuity and corneal transparency resulting in visual loss eventually. Therefore, understanding the UVB-induced cytotoxicity in HCEnCs is of importance for making efficient strategies to protect our vision from UVB-damage. However, in-depth knowledge about UVB-induced cytotoxicity in HCEnCs is missing. Herein, we pulse-irradiated the HCEnCs in vitro with 150 mJ/cm2 UVB (the environmental dose) at each subculture for 4 passages to explore the insights into UVB-induced phototoxicity. The results showed that the UVB-treated HCEnCs exhibit typical senescent characteristics, including significantly enlarged relative cell area, increased senescence-associated ß-galactosidase positive staining, and upregulated p16INK4A and senescence associated secretory phenotypes (SASPs) such as CCL-27, IL-1α/6/8/10, TGF-ß1 and TNF-α, as well as decreased cell proliferation and Lamin B1 expression, and translocation of Lamin B1. Furthermore, we explored the causative mechanisms of senescence and found that 150 mJ/cm2 UVB pulse-irradiation impairs DNA to activate DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with downregulated DNA repair enzyme PARP1, leading to cell cycle arrest resulting in DDR-mediated senescence. Meanwhile, UVB pulse-irradiation also elicits a consistent increase of ROS production to aggravate DNA damage and impose oxidative stress on energy metabolism leading to metabolic disturbance resulting in metabolic disturbance-mediated senescence. Altogether, the repeated pulse-irradiation of 150 mJ/cm2 UVB induces HCEnC senescence via both DDR pathway and energy metabolism disturbance.


Asunto(s)
Senescencia Celular , Daño del ADN , Células Endoteliales , Estrés Oxidativo , Rayos Ultravioleta , Células Cultivadas , Senescencia Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de la radiación , Humanos , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta/efectos adversos , beta-Galactosidasa/metabolismo
11.
Nanomaterials (Basel) ; 12(24)2022 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-36558272

RESUMEN

Ultrathin porous Co3O4 nanosheets are synthesized successfully, the thickness of which is about three unit-cell dimensions. The enhanced oxygen evolution reaction (OER) performance and electronic interaction between Co3O4 and Au is firstly reported in Co3O4 ultrathin porous nanosheets by physically mixing with Au nanoparticles. With the loading of the Au nanoparticles, the current density of ultrathin porous Co3O4 nanosheets is enhanced from 9.97 to 14.76 mA cm-2 at an overpotential of 0.5 V, and the overpotential required for 10 mA cm-2 decreases from 0.51 to 0.46 V, smaller than that of commercial IrO2 (0.54 V). Furthermore, a smaller Tafel slope and excellent durability are also obtained. Raman spectra, XPS measurement, and X-ray absorption near edge structure spectra (XANES) show that the enhanced OER ascribed to a higher Co2+/Co3+ ratio and quicker charge transfer due to the electronic interaction between Au and ultrathin Co3O4 nanosheets with low-coordinated surface, and Co2+ ions are beneficial for the formation of CoOOH active sites.

12.
Fish Shellfish Immunol ; 31(6): 1059-64, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21963856

RESUMEN

To understand the defense mechanisms of Crustacean animals, brine shrimp Artemia sinica prophenoloxidase (AsproPO) cDNA was cloned and its expression at early developmental stages was examined by reverse-transcription PCR (RT-PCR) and semi-quantitative RT-PCR, and activity of phenoloxidase (PO) at different developmental stages was further detected by using l-3,4-dihydroxyphenylalanine (l-DOPA) as a specific substrate in this study. It was found that the full-length of AsproPO cDNA is 2125 bp and it contains an open reading frame of 2100 bp encoding a protein of 699 amino acids. The deduced amino acid sequence of AsproPO has two putative copper binding sites highly conserved in Arthropods. Semi-quantitative RT-PCR analyses showed that the gene of AsproPO expressed at Emergence, Instar I and Instar II stages but did not at 0 h and 6 h stages. Activity measurement showed that PO activity could only be detected at Instar II stage but the other measured stages. All these implied that Artemia proPO immune system was complexly modulated during early development.


Asunto(s)
Artemia/enzimología , Artemia/inmunología , Catecol Oxidasa/genética , Catecol Oxidasa/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Regulación del Desarrollo de la Expresión Génica/inmunología , Estadios del Ciclo de Vida/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión/genética , Cobre/metabolismo , ADN Complementario/genética , Levodopa/metabolismo , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
13.
Acta Biochim Biophys Sin (Shanghai) ; 43(9): 722-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21757454

RESUMEN

Phenoloxidase from Artemia sinica (AsPO) was purified by Superdex 200 gel-filtration and Q Sepharose fast flow ion-exchange chromatography, and its properties were characterized biochemically and enzymatically by using L-dihydroxyphenylalanine (L-DOPA) as the specific substrate. Results showed that AsPO was isolated as a monomeric protein of 125.5 kDa in molecular mass. The optimal pH value and temperature are 7.0 and 50°C, respectively, for its PO activity. The AsPO had an apparent K(m) value of 4.2 mM on L-DOPA, and 10.9 mM on catechol, respectively. Oxidase inhibitor on PO activity showed that the AsPO was extremely sensitive to ascorbic acid, sodium sulfite, and citric acid; and was very sensitive to cysteine, benzoic acid, and 1-phenyl-2-thiourea. Combined with its specific enzyme activity on L-DOPA and catechol, it can be concluded that AsPO is most probably a typical catechol-type O-diphenoloxidase. Its PO activity was also sensitive to metal ions and chelators, and 20 mM DETC-inhibited PO activity was obviously recovered by 15 mM Cu(2+), indicating that AsPO is most probably a copper-containing metalloenzyme. All these data about specific substrate, sensitivity to oxidase inhibitor metal ions and chelators indicate that the AsPO has the properties of a catechol-type copper-containing O-diphenoloxidase that functions as a vital humoral factor in host defense via melaninization as in other Crustaceans.


Asunto(s)
Artemia/enzimología , Monofenol Monooxigenasa/aislamiento & purificación , Monofenol Monooxigenasa/metabolismo , Animales , Ácido Ascórbico/farmacología , Ácido Benzoico/farmacología , Biocatálisis/efectos de los fármacos , Catecol Oxidasa/metabolismo , Catecoles/metabolismo , Ácido Cítrico/farmacología , Cobre/química , Cisteína/farmacología , Electroforesis en Gel de Poliacrilamida , Pruebas de Enzimas , Inhibidores Enzimáticos/farmacología , Concentración de Iones de Hidrógeno , Cinética , Levodopa/metabolismo , Peso Molecular , Monofenol Monooxigenasa/química , Feniltiourea/farmacología , Especificidad por Sustrato , Sulfitos/farmacología , Temperatura
14.
Life Sci ; 277: 119453, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33798551

RESUMEN

Limbal stem cells (LSCs) are crucial for corneal transparency and vision. Any damages to LSCs might lead to limbal stem cell deficiency resulting in corneal opacification and even blindness. Here, we investigated the cytotoxicity of timolol and its underlying mechanisms in rabbit LSCs (rLSCs) in vitro. High concentrations of 0.5% and 0.25% timolol induced necroptosis in rLSCs to upregulate receptor interacting protein kinase (RIPK)1, RIPK3, mixed lineage kinase domain-like (MLKL) and phosphorylated MLKL along with downregulation of caspase-8 and caspase-2 within 4 h. While, median concentrations of 0.125% to 0.0625% timolol induced apoptosis in the rLSCs within 28 h. The apoptotic mechanism in the median-concentration timolol-treated rLSCs is probably via extrinsic apoptosis pathway by activating caspase-2, caspase-8 and caspase-3 and intrinsic apoptosis pathway triggered by excessive generation of ROS and subsequent DNA damage to upregulate Bax and Bad, downregulate Bcl-2 and Bcl-xL, subsequently disrupt mitochondrial membrane potential, cytosolically translocate cytochrome c and apoptosis-inducing factor, and activate caspase-9. In addition, low concentration of 0.03125% timolol induced senescence in the rLSCs by elevating ROS level and increasing number of senescence associated ß-galactosidase positive cells at 28 h. Our findings reveal that timolol induces necroptosis, apoptosis and senescence concentration-dependently in rLSCs in vitro.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Apoptosis , Senescencia Celular , Limbo de la Córnea/patología , Necroptosis , Células Madre/patología , Timolol/farmacología , Animales , Técnicas In Vitro , Limbo de la Córnea/efectos de los fármacos , Limbo de la Córnea/metabolismo , Masculino , Fosforilación , Conejos , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/metabolismo
15.
Naunyn Schmiedebergs Arch Pharmacol ; 393(1): 77-88, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31420720

RESUMEN

Norfloxacin, a frequently used ocular antibiotic, might have cytotoxic effect on human corneal endothelial cells (HCECs), subsequently damage the cornea and finally impair human vision. However, the possible mechanisms of cytotoxicity of norfloxacin to HCEC line are unclear. Herein, we investigated the cytotoxicity of norfloxacin and its underlying cellular and molecular mechanisms using in vitro cultured non-transfected HCECs and verified the cytotoxicity with cat corneal endothelium in vivo. In the present study, the cytotoxicity of norfloxacin in the in vitro cultured HCECs was recognized by causing abnormal morphology such as cell shrinkage and detachment from plate bottom, and decline of viability of in vitro cultured HCECs. Then, its cytotoxicity was verified by inducing reduction of cell density and morphological abnormality of in vivo cat corneal endothelial cells. Furthermore, the cytotoxicity of norfloxacin in HCECs was corroborated as apoptosis by elevation of plasma membrane permeability, S phase arrest, phosphatidylserine externalization, DNA fragmentation, and apoptotic body formation in in vitro cultured HCECs and apoptosis-like swollen cells in the in vivo model. Moreover, norfloxacin induced extrinsic death receptor-mediated apoptosis pathway by activating caspase-2/-8/-3 and intrinsic mitochondrion-dependent apoptosis pathway by downregulating anti-apoptotic Bcl-2 and upregulating of pro-apoptotic Bad, which disrupted mitochondrial transmembrane potential, subsequently upregulated cytoplasmic cytochrome c and apoptosis-inducing factor and finally activated caspase-9/-3. Generally, norfloxacin induces HCE cell apoptosis via a death receptor-mediated and mitochondrion-dependent signaling pathway.


Asunto(s)
Antibacterianos/farmacología , Córnea/citología , Células Endoteliales/efectos de los fármacos , Norfloxacino/farmacología , Animales , Apoptosis/efectos de los fármacos , Gatos , Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN , Células Endoteliales/patología , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Electrónica de Transmisión
16.
Curr Eye Res ; 45(9): 1065-1073, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32090638

RESUMEN

PURPOSE: To provide scientific data for clinical practice in making strategies for accelerating corneal endothelial wound healing, we investigated the impact of UVA on the corneal endothelial wound healing process and the underlying mechanism using an in vitro cell model. MATERIALS AND METHODS: An in vitro cell model for corneal endothelial wound healing was established by scratching the in vitro cultured human corneal endothelial cell (HCEnC) confluent layer. Then, we investigated the impacts of UVA irradiation and Ascorbic acid-2-phosphate (Asc-2p) on the wound healing process of the in vitro HCEnC model by examining wound-healing index, F-actin+ rate, Ki-67+ rate, and ROS production. RESULTS: After scratching, the Ki-67+ and F-actin+ HCEnCs occupied the scratching gap. Furthermore, the F-actin+ rates were significantly higher than Ki-67+ rates in the wound closure area. After irradiated with UVA, the wound-healing indexes, Ki-67+ rates and F-actin+ rates of the wound-healing model significantly reduced, whereas the ROS production significantly increased in a dose-dependent manner. Pretreatment with Asc-2p significantly reduced the ROS production as well as increased the wound-healing indexes, Ki-67+rates and F-actin+ rates of the UVA irradiated wound-healing model. CONCLUSION: The migration of HCEnC plays a major role in the wound healing process of the established cell model, which is like the wound healing process in vivo. UVA decreases the wound closure of the in vitro HCEnC model dose-dependently, while antioxidant Asc-2p can attenuate the damage to UVA to HCEnCs probably via reducing ROS to improve their migration.


Asunto(s)
Endotelio Corneal/efectos de la radiación , Rayos Ultravioleta , Cicatrización de Heridas/efectos de la radiación , Actinas/metabolismo , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/farmacología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Corneal/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo
17.
PLoS One ; 15(5): e0233208, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32428030

RESUMEN

To facilitate functional investigation of the role of NADPH oxidase 1 (NOX1) and associated reactive oxygen species in cancer cell signaling, we report herein the development and characterization of a novel mouse monoclonal antibody that specifically recognizes the C-terminal region of the NOX1 protein. The antibody was validated in stable NOX1 overexpression and knockout systems, and demonstrates wide applicability for Western blot analysis, confocal microscopy, flow cytometry, and immunohistochemistry. We employed our NOX1 antibody to characterize NOX1 expression in a panel of 30 human colorectal cancer cell lines, and correlated protein expression with NOX1 mRNA expression and superoxide production in a subset of these cells. Although a significant correlation between oncogenic RAS status and NOX1 mRNA levels could not be demonstrated in colon cancer cell lines, RAS mutational status did correlate with NOX1 expression in human colon cancer surgical specimens. Immunohistochemical analysis of a comprehensive set of tissue microarrays comprising over 1,200 formalin-fixed, paraffin-embedded tissue cores from human epithelial tumors and inflammatory disease confirmed that NOX1 is overexpressed in human colon and small intestinal adenocarcinomas, as well as adenomatous polyps, compared to adjacent, uninvolved intestinal mucosae. In contradistinction to prior studies, we did not find evidence of NOX1 overexpression at the protein level in tumors versus histologically normal tissues in prostate, lung, ovarian, or breast carcinomas. This study constitutes the most comprehensive histopathological characterization of NOX1 to date in cellular models of colon cancer and in normal and malignant human tissues using a thoroughly evaluated monoclonal antibody. It also further establishes NOX1 as a clinically relevant therapeutic target in colorectal and small intestinal cancer.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias del Colon/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Intestino Delgado/enzimología , NADPH Oxidasa 1/biosíntesis , Proteínas de Neoplasias/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Células CACO-2 , Neoplasias del Colon/genética , Células HT29 , Humanos , Intestino Delgado/patología , Modelos Biológicos , NADPH Oxidasa 1/genética , Proteínas de Neoplasias/genética
18.
Toxicology ; 428: 152305, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31605733

RESUMEN

In the present study, the toxicity of phenylephrine, a selective α1-adrenergic receptor agonist, in corneal epithelial cells and its underlying mechanisms were investigated using an in vitro model of human corneal epithelial cells (HCEPCs) and an in vivo model of New Zealand white rabbit corneas. The HCEPCs treated with phenylephrine at concentrations from 10% to 0.078125% displayed abnormal morphology, decline of cell viability and elevation of plasma membrane permeability time- and dose-dependently. Moreover, 10%-1.25% phenylephrine induce necrosis characteristics of marginalization and uneven distribution of chromatin through up-regulation of RIPK1, RIPK3 and MLKL along with inactivation of caspase-8 and caspase-2, whereas 0.625% phenylephrine induced condensed chromatin, S phase arrest, phosphatidylserine externalization, DNA fragmentation and apoptotic body formation in the HCECs through activation of caspase-2, -8, -9 and -3 as well as down-regulation of Bcl-2, up-regulation of Bad, ΔΨm disruption and release of cytochrome c and AIF into cytosol. At last, 10% phenylephrine induced destruction of the corneal epithelia and apoptosis of corneal epithelial cells in rabbit corneas. In conclusion, 10% to 1.25% phenylephrine cause necroptosis via RIPK1-RIPK3-MLKL axis and 0.625% phenylephrine induce apoptosis via a mitochondrion-dependent and death receptor-mediated signal pathway in HCEPCs.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/toxicidad , Apoptosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Necroptosis/efectos de los fármacos , Fenilefrina/toxicidad , Animales , Ciclo Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Córnea/citología , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Humanos , Masculino , Conejos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factores de Tiempo
19.
Oxid Med Cell Longev ; 2018: 4967318, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30116483

RESUMEN

The introduction of intracameral anaesthesia by injection of lidocaine has become popular in cataract surgery for its inherent potency, rapid onset, tissue penetration, and efficiency. However, intracameral lidocaine causes corneal thickening, opacification, and corneal endothelial cell loss. Herein, we investigated the effects of lidocaine combined with sodium ferulate, an antioxidant with antiapoptotic and anti-inflammatory properties, on lidocaine-induced damage of corneal endothelia with in vitro experiment of morphological changes and cell viability of cultured human corneal endothelial cells and in vivo investigation of corneal endothelial cell density and central corneal thickness of cat eyes. Our finding indicates that sodium ferulate from 25 to 200 mg/L significantly reduced 2 g/L lidocaine-induced toxicity to human corneal endothelial cells, and 50 mg/L sodium ferulate recovered the damaged human corneal endothelial cells to normal growth status. Furthermore, 100 mg/L sodium ferulate significantly inhibited lidocaine-induced corneal endothelial cell loss and corneal thickening in cat eyes. In conclusion, sodium ferulate protects human corneal endothelial cells from lidocaine-induced cytotoxicity and attenuates corneal endothelial cell loss and central corneal thickening of cat eyes after intracameral injection with lidocaine. It is likely that the antioxidant effect of sodium ferulate reduces the cytotoxic and inflammatory corneal reaction during intracameral anaesthesia.


Asunto(s)
Ácidos Cumáricos/uso terapéutico , Endotelio Corneal/efectos de los fármacos , Lidocaína/efectos adversos , Animales , Gatos , Ácidos Cumáricos/farmacología , Humanos , Masculino
20.
Oncotarget ; 8(24): 38113-38135, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28498822

RESUMEN

Human colon cancers express higher levels of NADPH oxidase 1 [NOX1] than adjacent normal epithelium. It has been suggested that reactive oxygen species [ROS] derived from NOX1 contribute to DNA damage and neoplastic transformation in the colon, particularly during chronic inflammatory stress. However, the mechanism(s) underlying increased NOX1 expression in malignant tumors or chronic inflammatory states involving the intestine are poorly characterized. We examined the effects of two pro-inflammatory cytokines, IL-4 and IL-13, on the regulation of NOX1. NOX1 expression was increased 4- to 5-fold in a time- and concentration-dependent manner by both cytokines in human colon cancer cell lines when a functional Type II IL-4 receptor was present. Increased NOX1 transcription following IL-4/IL-13 exposure was mediated by JAK1/STAT6 signaling, was associated with a ROS-related inhibition of protein tyrosine phosphatase activity, and was dependent upon activation and specific binding of GATA3 to the NOX1 promoter. NOX1-mediated ROS production increased cell cycle progression through S-phase leading to a significant increase in cellular proliferation. Evaluation of twenty pairs of surgically-resected colon cancers and their associated uninvolved adjacent colonic epithelium demonstrated a significant increase in the active form of NOX1, NOX1-L, in tumors compared to normal tissues, and a significant correlation between the expression levels of NOX1 and the Type II IL-4 receptor in tumor and the uninvolved colon. These studies imply that NOX1 expression, mediated by IL-4/IL-13, could contribute to an oxidant milieu capable of supporting the initiation or progression of colonic cancer, suggesting a role for NOX1 as a therapeutic target.


Asunto(s)
Neoplasias del Colon/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , NADPH Oxidasa 1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/patología , Humanos , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA