Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Anticancer Res ; 41(12): 5997-6001, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34848453

RESUMEN

BACKGROUND/AIM: Rhenium(I)-diselenoether (Re-diSe) is a drug under development for the treatment of metastatic cancers, with selective inhibitory effects on MDA-MB231 cancer cells compared to normal HEK-293 cells, and with greater effects than its diselenide (di-Se) ligand. Rhenium (Re) compounds inhibit cathepsins, which are important proteolytic enzymes in cancer. This study investigated the effects of Re-diSe and di-Se on the production of cathepsins B and S in MDA-MB231 malignant and HEK-293 normal cells and their inhibitory effects following treatment with different doses for 72 h. MATERIALS AND METHODS: Elisa tests were used to assay the amount of cathepsins B and S in the medium of cultures. RESULTS: Re-diSe, but not diSe affected the viability of malignant cells and the expression of cathepsins B and S. CONCLUSION: To the best of our knowledge, this is the first demonstration that Re-diSe may decrease the production of cathepsins B and S in cancer cells at doses as low as 10 µM.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Catepsina B/biosíntesis , Catepsinas/biosíntesis , Renio , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Estructura Molecular , Renio/química
2.
Inflammation ; 44(5): 2006-2017, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34037897

RESUMEN

Psoriasis is a common inflammatory dermatology disease. Strongly expressed serum amyloid A (SAA) promotes psoriasis exacerbation through inducing IL-17 secretion. What's more, SAA can stimulate the release of cathepsin B. The current work was performed to demonstrate the specific effects of cathepsin B silencing on inflammatory response, proliferation, and differentiation of IL-17A and SAA-induced keratinocytes and to report the precise role of cathepsin B in psoriasis-like lesion. HaCaT keratinocytes received treatment with IL-17A (0, 10, 50, 100 ng/ml) or SAA (0, 1, 5, 10, 20 µg/ml) for 24 h to establish psoriasis-like keratinocytes model. HaCaT keratinocytes were transfected with small interfering RNA (siRNA)-cathepsin B for the functional experiments. Cathepsin B mRNA and protein levels were separately assessed by performing RT-qPCR and Western blot analysis. Then, CCK-8 for detection of cell proliferative capacity and Western blot assay for detection of Ki67 and PCNA expression were adopted to evaluate the influence of silenced cathepsin B on proliferation of IL-17A/SAA-induced HaCaT keratinocytes. Furthermore, IL-6, IL-1ß, TNF-α, and p-NF-κB p65 were detected to assess the effects of cathepsin B knockdown on inflammatory response in IL-17A/SAA-induced HaCaT keratinocytes. In addition, assessment of KRT10, FLG, and LOR levels were applied to analyze the function of cathepsin B silencing on differentiation of IL-17A/SAA-induced HaCaT keratinocytes. Cathepsin B expression is distinctly elevated in IL-17A/SAA-induced HaCaT keratinocytes. IL-17A or SAA treatment enhanced proliferation, promoted the release of inflammatory factors, and arrested differentiation in HaCaT keratinocytes. Furthermore, downregulation of cathepsin B reduced proliferation, suppressed inflammatory response, and boosted differentiation in IL-17A/SAA-induced HaCaT keratinocytes. To sum up, cathepsin B silencing rescued excessive proliferation and inflammatory response and scarce differentiation in HaCaT keratinocytes induced by IL-17A and SAA. These findings prompted that cathepsin B might be a promising therapeutic target for psoriasis-like lesion, which helps to develop an anti-psoriatic agent.


Asunto(s)
Catepsina B/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Interleucina-17/toxicidad , Queratinocitos/efectos de los fármacos , Psoriasis/prevención & control , Proteína Amiloide A Sérica/toxicidad , Catepsina B/biosíntesis , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Queratinocitos/metabolismo , Psoriasis/inducido químicamente , Psoriasis/metabolismo
3.
J Clin Pathol ; 74(2): 84-90, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32467319

RESUMEN

AIM: Cathepsins are proteases that regulate a wide range of physiological processes, including protein turnover, cell signalling and antigen presentation. Recent studies have shown that cathepsins are highly upregulated in many types of tumours. Of the 15 cathepsins in humans, cathepsins V and S are abundantly expressed in the thymus, and we previously showed that the immunostaining of these cathepsins could serve as diagnostic markers for thymic epithelial tumours. However, little is known about the expression of other cathepsins in thymic epithelial tumours. To determine the diagnostic implications of cathepsins, we performed immunohistochemical analysis of cathepsin B (CTB), cathepsin D (CTD) and cathepsin K (CTK), all of which have been reported to correlate with the progression of squamous cell carcinoma. METHODS: The association between cathepsin expression and clinicopathological features was evaluated in 122 cases of thymoma and thymic carcinoma. RESULTS: CTB and CTD were frequently expressed in type A and type AB thymomas. In contrast, CTB and CTD were significantly less common in type B thymomas than in type A or AB thymomas. In type AB thymomas, the expression of CTB correlated with histological features, and was found predominantly in the type A component. Notably, CTK was expressed most commonly in thymic carcinomas, and patients who died of the disease showed increased expression of CTK. CONCLUSIONS: The expression of CTB and CTD correlated with the histological subtype of thymoma. In addition, the expression of CTK appears to be useful for the diagnosis of thymic carcinomas and as a prognostic marker.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Catepsina B/biosíntesis , Catepsina D/biosíntesis , Catepsina K/biosíntesis , Neoplasias Glandulares y Epiteliales/patología , Neoplasias del Timo/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias del Timo/metabolismo , Adulto Joven
4.
Sci Rep ; 9(1): 491, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679571

RESUMEN

Pulmonary fibrosis is a progressive disease characterized by a widespread accumulation of myofibroblasts and extracellular matrix components. Growing evidences support that cysteine cathepsins, embracing cathepsin B (CatB) that affects TGF-ß1-driven Smad pathway, along with their extracellular inhibitor cystatin C, participate in myofibrogenesis. Here we established that curcumin, a potent antifibrotic drug used in traditional Asian medicine, impaired the expression of both α-smooth muscle actin and mature TGF-ß1 and inhibited the differentiation of human lung fibroblasts (CCD-19Lu cells). Curcumin induced a compelling upregulation of CatB and CatL. Conversely cystatin C was downregulated, which allowed the recovery of the peptidase activity of secreted cathepsins and the restoration of the proteolytic balance. Consistently, the amount of both insoluble and soluble type I collagen decreased, reaching levels similar to those observed for undifferentiated fibroblasts. The signaling pathways activated by curcumin were further examined. Curcumin triggered the expression of nuclear peroxisome proliferator-activated receptor γ (PPARγ). Contrariwise PPARγ inhibition, either by an antagonist (2-chloro-5-nitro-N-4-pyridinyl-benzamide) or by RNA silencing, restored TGF-ß1-driven differentiation of curcumin-treated CCD-19Lu cells. PPARγ response element (PPRE)-like sequences were identified in the promoter regions of both CatB and CatL. Finally, we established that the transcriptional induction of CatB and CatL depends on the binding of PPARγ to PPRE sequences as a PPARγ/Retinoid X Receptor-α heterodimer.


Asunto(s)
Catepsina B/biosíntesis , Catepsina L/biosíntesis , Diferenciación Celular/efectos de los fármacos , Curcumina/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Pulmón/metabolismo , PPAR gamma/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Línea Celular , Humanos
5.
ACS Appl Mater Interfaces ; 10(48): 41056-41069, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30387987

RESUMEN

Intracellular activation of nanomaterials within cancer cells presents a powerful means to enhance anticancer specificity and efficacy. In light of upregulated lysosomal protease cathepsin-B (CathB) in many types of invasive cancer cells, herein, we exploit CathB-catalyzed biodegradation of acetylated rapeseed protein isolate (ARPI) to design polymer-drug nanocomplexes that can produce proapoptotic peptides in situ and synergize chemotherapy. ARPI forms nanocomplexes with chitosan (CS) and anticancer drug doxorubicin (DOX) [DOX-ARPI/CS nanoparticles (NPs)] by ionic self-assembly. The dual acidic pH- and CathB-responsive properties of the nanocomplexes and CathB-catalyzed biodegradation of ARPI enable efficient lysosomal escape and nuclei trafficking of released DOX, resulting in elevated cytotoxicity in CathB-overexpressing breast cancer cells. The ARPI-derived bioactive peptides exhibit synergistic anticancer effect with DOX by regulating pro- and antiapoptotic-relevant proteins ( p53, Bax, Bcl-2, pro-caspase-3) at mitochondria. In an orthotopic breast tumor model of CathB-overexpressing breast cancer, DOX-ARPI/CS NPs remarkably inhibit tumor growth, enhance tumor cell apoptosis and prolong host survival without eliciting any systemic toxicity. These results suggest that exploitation of multifunctional biomaterials to specifically produce anticancer agents inside cancer cells and trigger drug release to the subcellular target sites is a promising strategy for designing effective synergistic nanomedicines with minimal off-target toxicity.


Asunto(s)
Brassica rapa/química , Neoplasias de la Mama , Catepsina B/biosíntesis , Doxorrubicina , Portadores de Fármacos , Nanoestructuras , Proteínas de Neoplasias/metabolismo , Proteínas de Almacenamiento de Semillas , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Femenino , Humanos , Células MCF-7 , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Proteínas de Almacenamiento de Semillas/química , Proteínas de Almacenamiento de Semillas/farmacocinética , Proteínas de Almacenamiento de Semillas/farmacología
6.
Comp Biochem Physiol B Biochem Mol Biol ; 221-222: 18-28, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29649577

RESUMEN

Cathepsin B is a lysosomal proteolytic enzyme that has been suggested to play a role in pathological processes of immune system. In this study, the full-length cDNA sequence of cathepsin B transcript in the giant river prawn Macrobrachium rosenbergii (MrCTSB) was obtained from 454 pyrosequencing of cDNAs from hepatopancreas and muscle. It was 1158 bp in length, containing an open reading frame (ORF) of 987 bp corresponding to 328 amino acids. The predicted molecular mass and pI of MrCTSB protein was 36.04 kDa and 4.73. The major characteristics of MrCTSB protein consisted of a propeptide of C1 peptidase family at the N-terminus and a cysteine protease (Pept_C1) domain at the C-terminus. The 3-dimentional structure of MrCTSB was constructed by computer-assisted homology modeling. The folding of MrCTSB was highly conserved to human CTSB structure and the modeled MrCTSB displayed characteristics of cysteine proteinases superfamily. The docking study was performed to investigate binding interactions between known inhibitors against MrCTSB. Known inhibitors were oriented in the groove of catalytic site cleft. They bound to subsites from S2, S1, S1', and S2', respectively, with key residues in each subsite. Challenge of juvenile prawns with Aeromonas hydrophila revealed that the MrCTSB transcript in hepatopancreas significantly increased at 60-96 h post injection (hpi). This suggested that MrCTSB may play roles in innate immunity of M. rosenbergii. Our results provide useful information for a more comprehensive study in immune-related functions of MrCTSB.


Asunto(s)
Aeromonas hydrophila , Proteínas de Artrópodos , Catepsina B , Regulación Enzimológica de la Expresión Génica , Palaemonidae , Animales , Proteínas de Artrópodos/biosíntesis , Proteínas de Artrópodos/genética , Catepsina B/biosíntesis , Catepsina B/genética , Biología Computacional , Palaemonidae/enzimología , Palaemonidae/genética , Palaemonidae/microbiología
7.
Cancer Res ; 78(10): 2524-2535, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29510992

RESUMEN

CHIP/STUB1 ubiquitin ligase is a negative co-chaperone for HSP90/HSC70, and its expression is reduced or lost in several cancers, including breast cancer. Using an extensive and well-annotated breast cancer tissue collection, we identified the loss of nuclear but not cytoplasmic CHIP to predict more aggressive tumorigenesis and shorter patient survival, with loss of CHIP in two thirds of ErbB2+ and triple-negative breast cancers (TNBC) and in one third of ER+ breast cancers. Reduced CHIP expression was seen in breast cancer patient-derived xenograft tumors and in ErbB2+ and TNBC cell lines. Ectopic CHIP expression in ErbB2+ lines suppressed in vitro oncogenic traits and in vivo xenograft tumor growth. An unbiased screen for CHIP-regulated nuclear transcription factors identified many candidates whose DNA-binding activity was up- or downregulated by CHIP. We characterized myeloid zinc finger 1 (MZF1) as a CHIP target, given its recently identified role as a positive regulator of cathepsin B/L (CTSB/L)-mediated tumor cell invasion downstream of ErbB2. We show that CHIP negatively regulates CTSB/L expression in ErbB2+ and other breast cancer cell lines. CTSB inhibition abrogates invasion and matrix degradation in vitro and halts ErbB2+ breast cancer cell line xenograft growth. We conclude that loss of CHIP remodels the cellular transcriptome to unleash critical pro-oncogenic pathways, such as the matrix-degrading enzymes of the cathepsin family, whose components can provide new therapeutic opportunities in breast and other cancers with loss of CHIP expression.Significance: These findings reveal a novel targetable pathway of breast oncogenesis unleashed by the loss of tumor suppressor ubiquitin ligase CHIP/STUB1. Cancer Res; 78(10); 2524-35. ©2018 AACR.


Asunto(s)
Catepsina B/metabolismo , Catepsina L/metabolismo , Transformación Celular Neoplásica/genética , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Catepsina B/biosíntesis , Catepsina L/biosíntesis , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Células MCF-7 , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/mortalidad , Ubiquitina-Proteína Ligasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Reprod Dev ; 84(1): 67-75, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27862569

RESUMEN

Growth factors synthesized by ovarian somatic cells affect cumulus cell expansion and oocyte maturation in vitro. Fibroblast growth factor 10 (FGF10), for example, is a known regulator of mammalian cumulus-oocyte complex maturation. In this study, we investigated the effects of 0, 5, 10, 50, and 100 ng/mL FGF10 (5F, 10F, 50F, and 100F, respectively) on in vitro cumulus cell expansion, oocyte maturation, and embryo development. The percentage of fully expanded cumulus cells at the oocyte's metaphase-II (MII) stage was significantly higher in the 10F-treated group than in the control. Transcript abundance of the cumulus cell expansion-related gene encoding hyaluronian synthase 2 (HAS2) in cumulus cells at oocyte germinal vesicle breakdown (GVBD) was significantly higher in the 10F- and 50F-treated groups compared to untreated controls, whereas the mRNA abundance of the protease cathepsin B (CTSB) at the oocyte MII stage was remarkably decreased in the 10F-treated group. The percentage of oocytes with normal spindles was greater in the 10F- and 50F-treated group at GVBD than in the other groups; the 5F-, 10F-, and 100F-treated groups were higher than the control; and the 50F-treated group was highest at MII. The abundance of GDF9 and BMP15 transcript at GVBD and BMP15 and CCNB1 transcripts at MII increased in the 10F-treated group. Cleavage rate, blastocyst formation rate, and total cell number were significantly higher in the 5F- to 50F-treated groups. These results demonstrate that FGF10 markedly improves cumulus cell expansion, oocyte maturation, and subsequent embryo development. Mol. Reprod. Dev. 84: 67-75, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Células del Cúmulo/metabolismo , Factor 10 de Crecimiento de Fibroblastos/farmacología , Oocitos/metabolismo , Animales , Proteína Morfogenética Ósea 15/biosíntesis , Catepsina B/biosíntesis , Células Cultivadas , Células del Cúmulo/citología , Femenino , Factor 9 de Diferenciación de Crecimiento/biosíntesis , Hialuronano Sintasas/biosíntesis , Oocitos/citología , Porcinos
9.
Biochim Biophys Acta ; 1851(10): 1304-1316, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26143381

RESUMEN

During autophagy, autophagosomes fuse with lysosomes to degrade damaged organelles and misfolded proteins. Breakdown products are released into the cytosol and contribute to energy and metabolic building block supply, especially during starvation. Lipophagy has been defined as the autophagy-mediated degradation of lipid droplets (LDs) by lysosomal acid lipase. Adipose triglyceride lipase (ATGL) is the major enzyme catalyzing the initial step of lipolysis by hydrolyzing triglycerides (TGs) in cytosolic LDs. Consequently, most organs and cells, including macrophages, lacking ATGL accumulate TGs, resulting in reduced intracellular free fatty acid concentrations. Macrophages deficient in hormone-sensitive lipase (H0) lack TG accumulation albeit reduced in vitro TG hydrolase activity. We hypothesized that autophagy is activated in lipase-deficient macrophages to counteract their energy deficit. We therefore generated mice lacking both ATGL and HSL (A0H0). Macrophages from A0H0 mice showed 73% reduced neutral TG hydrolase activity, resulting in TG-rich LD accumulation. Increased expression of cathepsin B, accumulation of LC3-II, reduced expression of p62 and increased DQ-BSA dequenching suggest intact autophagy and functional lysosomes in A0H0 macrophages. Markedly decreased acid TG hydrolase activity and lipid flux independent of bafilomycin A1 treatment, however, argue against effective lysosomal degradation of LDs in A0H0 macrophages. We conclude that autophagy of proteins and cell organelles but not of LDs is active as a compensatory mechanism to circumvent and balance the reduced availability of energy substrates in A0H0 macrophages.


Asunto(s)
Autofagia/fisiología , Lipólisis/fisiología , Macrófagos Peritoneales/metabolismo , Triglicéridos/metabolismo , Animales , Autofagia/efectos de los fármacos , Catepsina B/biosíntesis , Catepsina B/genética , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Lipasa/genética , Lipasa/metabolismo , Lipólisis/efectos de los fármacos , Lisosomas/enzimología , Lisosomas/genética , Macrólidos/farmacología , Macrófagos Peritoneales/citología , Ratones , Ratones Mutantes , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Triglicéridos/genética
10.
J Mol Cell Cardiol ; 86: 32-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26163874

RESUMEN

AIMS: Macrophage inflammation response is important in the pathogenesis of atherosclerosis. We investigated the role and mechanism of cellular repressor of E1A-stimulated genes (CREG) in regulating TNF-α induced inflammation response in macrophages and explore whether CREG might be a therapeutic target for atherosclerosis. METHOD AND RESULTS: Immunostaining and western blotting showed that expression of CREG was reduced in human atherosclerotic coronary artery. In vivo experiments demonstrated that supplementation of recombinant CREG protein to ApoE(-/-) mice fed with high fat diet alleviated aortic atherosclerosis development and inflammation. In vitro, macrophage from ApoE(-/-) mice fed with high fat diet had lower level of CREG compared to control mice fed with normal diet. Immunohistochemical staining and western blotting further confirmed that CREG inhibited inflammatory response of macrophages induced by TNF-α. Supplementation of exogenous recombinant CREG protein or CREG gene silencing showed that CREG promoted autophagy in TNF-α treated macrophages. The use of autophagy inhibitors, 3-methyladenine and bafilomycin A, identified that CREG attenuated TNF-α induced inflammation by activate autophagy. In addition, supplementation of exogenous CREG protein stimulated expression and maturity of cathepsin B and cathepsin L and induced lysosome formation, whereas CREG deficiency reduced lysosomal formation. CONCLUSION: CREG inhibits inflammation and promotes autophagy mediated by lysosome formation; it might be a potential therapeutic target in atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Inflamación/genética , Proteínas Represoras/biosíntesis , Factor de Necrosis Tumoral alfa/administración & dosificación , Animales , Apolipoproteínas E/genética , Aterosclerosis/patología , Catepsina B/biosíntesis , Catepsina L/biosíntesis , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Lisosomas/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Proteínas Represoras/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
Hum Mol Genet ; 24(15): 4198-211, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25926625

RESUMEN

Saposin (Sap) C deficiency is a rare variant form of Gaucher disease caused by impaired Sap C expression or accelerated degradation, and associated with accumulation of glucosylceramide and other lipids in the endo/lysosomal compartment. No effective therapies are currently available for the treatment of Sap C deficiency. We previously reported that a reduced amount and enzymatic activity of cathepsin (Cath) B and Cath D, and defective autophagy occur in Sap C-deficient fibroblasts. Here, we explored the use of two compounds, BCM-95, a curcumin derivative, and (2-hydroxypropyl)-ß-cyclodextrin (HP-ß-CD), to improve lysosomal function of Sap C-deficient fibroblasts. Immunofluorescence and biochemical studies documented that each compound promotes an increase of the expression levels and activities of Cath B and Cath D, and efficient clearance of cholesterol (Chol) and ceramide (Cer) in lysosomes. We provide evidence that BCM-95 and HP-ß-CD enhance lysosomal function promoting autophagic clearance capacity and lysosome reformation. Our findings suggest a novel pharmacological approach to Sap C deficiency directed to treat major secondary pathological aspects in this disorder.


Asunto(s)
Curcumina/efectos adversos , Enfermedad de Gaucher/tratamiento farmacológico , Saposinas/genética , beta-Ciclodextrinas/administración & dosificación , Autofagia/efectos de los fármacos , Catepsina B/biosíntesis , Catepsina B/genética , Catepsina D/biosíntesis , Catepsina D/genética , Curcumina/análogos & derivados , Fibroblastos/metabolismo , Fibroblastos/patología , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/patología , Glucosilceramidas/metabolismo , Humanos , Lisosomas/genética , Lisosomas/patología , Saposinas/deficiencia
12.
Braz. dent. j ; 25(6): 502-507, Nov-Dec/2014. tab
Artículo en Inglés | LILACS | ID: lil-732258

RESUMEN

This aim of this study was to assess the ability of manual or rotary instrumentation associated with photodynamic therapy (PDT) to reduce Enterococcus faecalis using three combinations of light/photosensitizers: toluidine blue O/laser, fuchsin/halogen light and fuchsin/LED. Twenty deciduous molars were selected and contaminated with Enterococcus faecalis (McFarland 0.5 scale). Working length determination was performed by visual method. The teeth were randomly divided into two groups: G1 (n=10): manual instrumentation (Kerr-type files) and G2 (n=10): rotary instrumentation (ProTaper system). The bacteria were collected three times using sterile paper cones compatible with the anatomic diameter of the root canal for 30 s before and after instrumentation and after PDT. The samples were diluted in peptone water, seeded on blood agar plates and incubated in an oven at 37 °C for colony-forming units counting. The decrease of E. faecalis counts after instrumentation and after PDT was compared using the Wilcoxon test, t-test and Kruskal Wallis test. A significant reduction of E. faecalis occurred after manual and rotary instrumentation and after PDT using the three combinations of light/photosensitizer (p<0.05). It may be concluded that both rotary and manual instrumentation reduced E. faecalis. Fuchsin with halogen light or LED irradiation and toluidine blue O with laser irradiation can be used to reduce E. faecalis in root canals of primary molars. PDT can be used as an adjuvant to conventional endodontic treatment.


O objetivo do presente estudo foi avaliar a redução de Enterococcus faecalis após instrumentação manual ou rotatória associada à terapia fotodinâmica (PDT) utilizando 3 combinações luz/fotossensibilizante: azul de toluidina O/laser, fucsina/luz halógena e fucsina/LED. Foram selecionados 20 molares decíduos que foram contaminados com Enterococcus faecalis (escala 0,5 de McFarland). A odontometria foi feita através do método visual. Os dentes foram divididos aleatoriamente em dois grupos: G1 (n=10): instrumentação manual (limas tipo Kerr) e G2 (n=10): instrumentação rotatória (sistema ProTaper). Foram realizadas coletas com cone de papel estéril compatível com o diâmetro anatômico do canal durante 30 s antes e após a instrumentação e a PDT. As amostras foram diluídas em água peptonada, semeadas em placas de agar-sangue e incubadas em estufa a 37 °C para contagem das unidades formadoras de colônias. As comparações antes da redução de E. faecalis após a instrumentação e após a realização da PDT foram realizadas pelo teste de Wilcoxon, teste t e Kruskal Wallis. Houve redução significante de E. faecalis após a instrumentação manual ou rotatória e após realização da PDT com as três combinações de luz/fotossensibilizante (p<0,05). Pode-se concluir que a instrumentação rotatória e manual acarretou a redução de E. faecalis. A fucsina irradiada com luz halógena ou led e o azul de toluidina irradiado com laser podem ser utilizados para redução de E. faecalis do sistema de canais radiculares de molares decíduos. A terapia fotodinâmica pode ser utilizada como coadjuvante ao tratamento endodôntico convencional.


Asunto(s)
Animales , Ratones , Fosfatasa Ácida/biosíntesis , Catepsina B/biosíntesis , Leucina/análogos & derivados , Leupeptinas/farmacología , Melanoma Experimental/enzimología , Oligopéptidos/farmacología , Pepstatinas/farmacología , Péptido Hidrolasas/biosíntesis , Inhibidores de Proteasas/farmacología , Inducción Enzimática , Leucina/farmacología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Células Tumorales Cultivadas
13.
J Biol Chem ; 289(31): 21716-26, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939850

RESUMEN

The induction of inflammatory cytokines such as IL-1ß is associated with the progression of human immunodeficiency virus, type 1 (HIV-1) disease or AIDS. Unlike most inflammatory cytokines that are regulated by NF-κB at the transcriptional level, production of mature IL-1ß also depends on inflammasome activation. The mechanism by which HIV-1 induces pro-IL-1ß expression and activates inflammasomes to cleave pro-IL-1ß into its bioactive form is not clearly defined. We report here that HIV-1 infection in human monocytes efficiently induced IL-1ß expression and inflammasome activation. Toll-like receptor 8 (TLR8) was required for inducing pro-IL-1ß expression, whereas the NLRP3 inflammasome was required for IL-1ß maturation and release. Furthermore, the lysosomal protease cathepsin B and HIV-1 induced production of reactive oxygen species were critical for HIV-induced inflammasome activation and IL-1ß production. HIV-1 entry, reverse transcription, and integration were all required for both pro-IL-1ß expression and inflammasome activation. Finally, we show that HIV-1-derived RNA was sufficient to induce both pro-IL-1ß expression and inflammasome activation. We conclude that HIV-1 infection induced the expression of pro-IL-1ß via TLR8-mediated mechanisms and activated caspase-1 through the NLRP3 inflammasome to cleave pro-IL-1ß into bioactive IL-1ß. These findings help to elucidate mechanisms of HIV-1 disease progression and identify novel targets for treating HIV-1 induced inflammation and immune activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por VIH/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/biosíntesis , Monocitos/metabolismo , Receptor Toll-Like 8/fisiología , Catepsina B/biosíntesis , Técnicas de Silenciamiento del Gen , VIH-1/genética , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR , ARN Viral/genética , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 8/genética
14.
Int J Mol Sci ; 15(4): 5807-20, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24714089

RESUMEN

Cathepsin B is one of the major lysosomal cysteine proteases involved in neuronal protein catabolism. This cathepsin is released after traumatic injury and increases neuronal death; however, release of cystatin C, a cathepsin inhibitor, appears to be a self-protective brain response. Here we describe the effect of cystatin C intracerebroventricular administration in rats prior to inducing a traumatic brain injury. We observed that cystatin C injection caused a dual response in post-traumatic brain injury recovery: higher doses (350 fmoles) increased bleeding and mortality, whereas lower doses (3.5 to 35 fmoles) decreased bleeding, neuronal damage and mortality. We also analyzed the expression of cathepsin B and cystatin C in the brains of control rats and of rats after a traumatic brain injury. Cathepsin B was detected in the brain stem, cerebellum, hippocampus and cerebral cortex of control rats. Cystatin C was localized to the choroid plexus, brain stem and cerebellum of control rats. Twenty-four hours after traumatic brain injury, we observed changes in both the expression and localization of both proteins in the cerebral cortex, hippocampus and brain stem. An early increase and intralysosomal expression of cystatin C after brain injury was associated with reduced neuronal damage.


Asunto(s)
Lesiones Encefálicas/mortalidad , Lesiones Encefálicas/patología , Catepsina B/biosíntesis , Cistatina C/farmacología , Animales , Apoptosis , Tronco Encefálico/metabolismo , Catepsina B/metabolismo , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Plexo Coroideo/metabolismo , Cistatina C/biosíntesis , Hemorragia/inducido químicamente , Hipocampo/metabolismo , Masculino , Neuronas/patología , Ratas , Ratas Wistar
15.
Oncol Rep ; 31(2): 940-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24337203

RESUMEN

Degradation of the extracellular matrix (ECM) is a critical step of tumor cell invasion and requires protease-dependent proteolysis focalized at the invadopodia where the proteolysis of the ECM occurs. Most of the extracellular proteases belong to serine- or metallo-proteases and the invadopodia is where protease activity is regulated. While recent data looking at global protease activity in the growth medium reported that their activity and role in invasion is dependent on Na+/H+ exchanger 1 (NHE1)-driven extracellular acidification, there is no data on this aspect at the invadopodia, and an open question remains whether this acid extracellular pH (pHe) activation of proteases in tumor cells occurs preferentially at invadopodia. We previously reported that the NHE1 is expressed in breast cancer invadopodia and that the NHE1­dependent acidification of the peri-invadopodial space is critical for ECM proteolysis. In the present study, using, for the first time, in situ zymography analysis, we demonstrated a concordance between NHE1 activity, extracellular acidification and protease activity at invadopodia to finely regulate ECM digestion. We demonstrated that: (i) ECM proteolysis taking place at invadopodia is driven by acidification of the peri-invadopodia microenvironment; (ii) that the proteases have a functional pHe optimum that is acidic; (iii) more than one protease is functioning to digest the ECM at these invadopodial sites of ECM proteolysis; and (iv) lowering pHe or inhibiting the NHE1 increases protease secretion while blocking protease activity changes NHE1 expression at the invadopodia.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Transporte de Catión/metabolismo , Extensiones de la Superficie Celular/metabolismo , Matriz Extracelular/metabolismo , Péptido Hidrolasas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Antiarrítmicos/farmacología , Catepsina B/antagonistas & inhibidores , Catepsina B/biosíntesis , Catepsina B/metabolismo , Proteínas de Transporte de Catión/antagonistas & inhibidores , Línea Celular Tumoral , Matriz Extracelular/patología , Femenino , Guanidinas/farmacología , Humanos , Concentración de Iones de Hidrógeno , Metaloproteinasa 14 de la Matriz/biosíntesis , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Invasividad Neoplásica/patología , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Sulfonas/farmacología , Tiofenos/farmacología
16.
Free Radic Biol Med ; 65: 1398-1407, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24103565

RESUMEN

Protein disulfide isomerase (PDI) and its homologs are oxidoreductases facilitating protein folding in the ER. Endo-PDI (also termed ERp46) is highly expressed in endothelial cells. It belongs to the PDI family but its physiological function is largely unknown. We studied the role of Endo-PDI in endothelial angiogenic responses. Stimulation of human umbilical vein endothelial cells (with TNFα (10ng/ml) increased ERK1/2 phosphorylation. This effect was largely attenuated by Endo-PDI siRNA, whereas JNK and p38 MAP kinase phosphorylation was Endo-PDI independent. Similarly, TNFα-stimulated NF-κB signaling determined by IκBα degradation as well as TNFα-induced ICAM expression was unaffected by Endo-PDI siRNA. The action of Endo-PDI was not mediated by extracellular thiol exchange or cell surface PDI as demonstrated by nonpermeative inhibitors and PDI-neutralizing antibody. Moreover, exogenously added PDI failed to restore ERK1/2 activation after Endo-PDI knockdown. This suggests that Endo-PDI acts intracellularly potentially by maintaining the Ras/Raf/MEK/ERK pathway. Indeed, knockdown of Endo-PDI attenuated Ras activation measured by G-LISA and Raf phosphorylation. ERK activation influences gene expression by the transcriptional factor AP-1, which controls MMP-9 and cathepsin B, two proteases required for angiogenesis. TNFα-stimulated MMP-9 and cathepsin B induction was reduced by silencing of Endo-PDI. Accordingly, inhibition of cathepsin B or Endo-PDI siRNA blocked the TNFα-stimulated angiogenic response in the spheroid outgrowth assays. Moreover ex vivo tube formation and in vivo Matrigel angiogenesis in response to TNFα were attenuated by Endo-PDI siRNA. In conclusion, our study establishes Endo-PDI as a novel, important mediator of AP-1-driven gene expression and endothelial angiogenic function.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Neovascularización Fisiológica/fisiología , Proteína Disulfuro Isomerasas/metabolismo , Factor de Transcripción AP-1/genética , Factor de Necrosis Tumoral alfa/farmacología , Inductores de la Angiogénesis/antagonistas & inhibidores , Inductores de la Angiogénesis/farmacología , Catepsina B/antagonistas & inhibidores , Catepsina B/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Células Cultivadas , Retículo Endoplásmico , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas I-kappa B , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 9 de la Matriz/biosíntesis , NADPH Oxidasas , Inhibidor NF-kappaB alfa , Fosforilación , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Pliegue de Proteína , Interferencia de ARN , ARN Interferente Pequeño , Esferoides Celulares , Reductasa de Tiorredoxina-Disulfuro , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas ras/genética
17.
Free Radic Biol Med ; 65: 1155-1163, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23999505

RESUMEN

Advanced glycation end product (AGE)-modified proteins are formed by the nonenzymatic glycation of free amino groups of proteins and, along with lipofuscin (a highly oxidized aggregate of covalently cross-linked proteins, sugars, and lipids), have been found to accumulate during aging and in several age-related diseases. As the in vivo effects of diet-derived AGEs or lipofuscin remain elusive, we sought to study the impact of oral administration of glucose-, fructose-, or ribose-modified albumin or of artificial lipofuscin in a genetically tractable model organism. We report herein that continuous feeding of young Drosophila flies with culture medium enriched in AGEs or in lipofuscin resulted in reduced locomotor performance and in accelerated rates of AGE-modified proteins and carbonylated proteins accumulation in the somatic tissues and hemolymph of flies, as well as in a significant reduction of flies health span and life span. These phenotypic effects were accompanied by reduced proteasome peptidase activities in both the hemolymph and the somatic tissues of flies and higher levels of oxidative stress; furthermore, oral administration of AGEs or lipofuscin in flies triggered an upregulation of the lysosomal cathepsin B, L activities. Finally, RNAi-mediated cathepsin D knockdown reduced flies longevity and significantly augmented the deleterious effects of AGEs and lipofuscin, indicating that lysosomal cathepsins reduce the toxicity of diet-derived AGEs or lipofuscin. Our in vivo studies demonstrate that chronic ingestion of AGEs or lipofuscin disrupts proteostasis and accelerates the functional decline that occurs with normal aging.


Asunto(s)
Envejecimiento/efectos de los fármacos , Drosophila melanogaster/metabolismo , Productos Finales de Glicación Avanzada/farmacología , Lipofuscina/farmacología , Pliegue de Proteína/efectos de los fármacos , Albúminas/química , Animales , Animales Modificados Genéticamente , Catepsina B/biosíntesis , Catepsina B/metabolismo , Catepsina D/genética , Catepsina L/biosíntesis , Catepsina L/metabolismo , Dieta , Fructosa/química , Glucosa/química , Productos Finales de Glicación Avanzada/administración & dosificación , Productos Finales de Glicación Avanzada/química , Glicosilación , Lipofuscina/administración & dosificación , Lipofuscina/química , Longevidad/efectos de los fármacos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Interferencia de ARN , ARN Interferente Pequeño , Ribosa/química , Regulación hacia Arriba
18.
Mol Cell Biol ; 33(21): 4308-20, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24001769

RESUMEN

Sorting-related receptor with A-type repeats (SORLA) is a sorting receptor for the amyloid precursor protein (APP) that prevents breakdown of APP into Aß peptides, a hallmark of Alzheimer's disease (AD). Several cytosolic adaptors have been shown to interact with the cytoplasmic domain of SORLA, thereby controlling intracellular routing of SORLA/APP complexes in cell lines. However, the relevance of adaptor-mediated sorting of SORLA for amyloidogenic processes in vivo remained unexplored. We focused on the interaction of SORLA with phosphofurin acidic cluster sorting protein 1 (PACS1), an adaptor that shuttles proteins between the trans-Golgi network (TGN) and endosomes. By studying PACS1 knockdown in neuronal cell lines and investigating transgenic mice expressing a PACS1-binding-defective mutant form of SORLA, we found that disruption of SORLA and PACS1 interaction results in the inability of SORLA/APP complexes to sort to the TGN in neurons and in increased APP processing in the brain. Loss of PACS1 also impairs the proper expression of the cation-independent mannose 6-phosphate receptor and its target cathepsin B, a protease that breaks down Aß. Thus, our data identified the importance of PACS1-dependent protein sorting for amyloidogenic-burden control via both SORLA-dependent and SORLA-independent mechanisms.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/enzimología , Catepsina B/biosíntesis , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Relacionadas con Receptor de LDL/química , Proteínas de Transporte de Membrana/química , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Neuronas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Receptor IGF Tipo 2/metabolismo , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
19.
Oncol Rep ; 30(4): 1681-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23877402

RESUMEN

Ac­Phe­Lys­PABC­DOX (PDOX) is a smart doxorubicin (DOX) prodrug designed to decrease toxicities while maintaining the potent anticancer effects of DOX. The present study aimed to elucidate the molecular mechanisms of action of PDOX using MGC­803 gastric cancer cells as a model. The cells were treated with both PDOX and DOX, and cytotoxicities, cell cycle analysis, reactive oxygen species (ROS) generation, mitochondrial damage and ERK1/2 signaling pathway alterations were studied. Abundant cathepsin B expression was observed in the MGC­803 cells, and treatment with PDOX and DOX triggered dose­dependent cytotoxicity and resulted in a significant reduction in cell viability. IC50 of PDOX and DOX was 14.9 and 4.9 µM, respectively. Both PDOX and DOX significantly decreased p­ERK1/2, increased ROS generation, reduced mitochondrial membrane potential, caused mitochondrial swelling and arrested the cell cycle at the G2/S phase, and these effects were more pronounced for PDOX than for DOX. PDOX and DOX have different mechanisms of action, particularly the mitochondria­centered intrinsic apoptosis involving reactive oxidative stress and the ERK1/2 signaling pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/análogos & derivados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oligopéptidos/farmacología , Antibióticos Antineoplásicos/farmacología , Catepsina B/biosíntesis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Dilatación Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
20.
Oncol Rep ; 30(2): 723-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23708264

RESUMEN

The molecular mechanism involved in the metastasis of endometrial cancer (EC) remains unclear. The lysosomal cysteine protease Cathepsin B has been implicated in the progression of various human tumors. In the present study, we assessed the expression of Cathepsin B and its functions in EC. Immunohistochemistry was used to examine Cathepsin B expression in 76 paraffin-embedded endometrial tumor tissues. Lentiviral packing short hairpin RNA (shRNA) was transfected into HEC-1A cells to build a stable Cathepsin B knockdown cell line. The cellular levels of Cathepsin B mRNA and protein were detected by real-time PCR and western immunoblotting. The functions of Cathepsin B in EC cells were measured by MTT, migration and invasion assays. In additon, tumorigenicity assays were established in nude mice to study tumor growth in vivo. The results of our study showed that Cathepsin B was overexpressed in EC tissues compared with normal endometrium and endometrial atypical hyperplasia. Depletion of Cathepsin B in vitro inhibited cell proliferation, migration and invasion. Tumor formation assays confirmed that suppression of Cathepsin B inhibited the proliferation potential of HEC-1A cells in vivo, demonstrated by lower proliferation rates. These results suggest that Cathepsin B may act as an oncogene in EC, with the potential to provide a new therapeutic target for treating endometrial malignancy.


Asunto(s)
Catepsina B/deficiencia , Catepsina B/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Animales , Pruebas de Carcinogenicidad/métodos , Catepsina B/biosíntesis , Catepsina B/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/metabolismo , Hiperplasia Endometrial/patología , Neoplasias Endometriales/metabolismo , Endometrio/metabolismo , Endometrio/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...