Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
1.
ACS Appl Mater Interfaces ; 16(7): 8484-8498, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38334265

RESUMEN

Malignant tumors are still one of the most deadly diseases that threaten human life and health. However, developing new drugs is challenging due to lengthy trials, funding constraints, and regulatory approval procedures. Consequently, researchers have devoted themselves to transforming some clinically approved old drugs into antitumor drugs with certain active ingredients, which have become an attractive alternative. Disulfiram (DSF), an antialcohol medication, can rapidly metabolize in the physiological environment into diethyldithiocarbamate (DTC) which can readily react with Cu2+ ions in situ to form the highly toxic bis(N,N-diethyldithiocarbamate)-copper(II) (CuET) complex. In this study, DSF is loaded into mesoporous dopamine nanocarriers and surface-chelated with tannin and Cu2+ to construct M-MDTC nanoprodrugs under the camouflage of K7 tumor cell membranes. After intravenous injection, M-MDTC nanoprodrugs successfully reach the tumor sites with the help of mediated cell membranes. Under slightly acidic pH and photothermal stimulation conditions, DSF and Cu2+ are simultaneously released, forming a highly toxic CuET to kill tumor cells in situ. The generated CuET can also induce immunogenic cell death of tumor cells, increase the proportion of CD86+ CD80+ cells, and promote dendritic cell maturation. In vitro and in vivo studies of M-MDTC nanoprodrugs have shown excellent tumor-cell-killing ability and solid tumor suppression. This approach enables in situ amplification of chemotherapy in the tumor microenvironment, achieving an effective antitumor treatment.


Asunto(s)
Cadaverina/análogos & derivados , Cobre , Neoplasias , Humanos , Línea Celular Tumoral , Cobre/farmacología , Cobre/uso terapéutico , Microambiente Tumoral , Biomimética , Disulfiram/farmacología , Ditiocarba/farmacología , Ditiocarba/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología
2.
Biomolecules ; 9(12)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779105

RESUMEN

Polyamines are involved in the regulation of many cellular functions and are promising biomarkers of numerous physiological conditions. Since the concentrations of these compounds in biological fluids are low, sample extraction is one of the most critical steps of their analysis. Here, we developed a comprehensive, sensitive, robust, and high-throughput LC-MS/MS stable-isotope dilution method for the simultaneous determination of 19 metabolites related to polyamine metabolism, including polyamines, acetylated and diacetylated polyamines, precursors, and catabolites from liquid biopsies. The sample extraction was optimized to remove interfering compounds and to reduce matrix effects, thus being useful for large clinical studies. The method consists of two-step liquid-liquid extraction with a Folch extraction and ethyl acetate partitioning combined with dansyl chloride derivatization. The developed method was applied to a small gender-related trial concerning human serum and urine samples from 40 obese subjects. Sex differences were found for cadaverine, putrescine, 1,3-diaminopropane, γ-aminobutyric acid, N8-acetylspermidine, and N-acetylcadaverine in urine; N1-acetylspermine in serum; and spermine in both serum and urine. The results demonstrate that the developed method can be used to analyze biological samples for the study of polyamine metabolism and its association with human diseases.


Asunto(s)
Cromatografía Liquida/métodos , Extracción Líquido-Líquido/métodos , Metaboloma , Obesidad/metabolismo , Poliaminas/metabolismo , Espectrometría de Masas en Tándem/métodos , Acetilación , Cadaverina/análogos & derivados , Cadaverina/sangre , Compuestos de Dansilo/química , Diaminas/sangre , Femenino , Humanos , Concentración de Iones de Hidrógeno , Biopsia Líquida , Masculino , Obesidad/sangre , Obesidad/orina , Poliaminas/sangre , Poliaminas/química , Poliaminas/orina , Putrescina/sangre , Caracteres Sexuales , Espermidina/análogos & derivados , Espermidina/sangre , Espermina/sangre , Espermina/orina , Ácido gamma-Aminobutírico/sangre
3.
Bioorg Chem ; 92: 103194, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31493706

RESUMEN

Cathepsin B plays key roles in tumor progression with its overexpression being associated with invasive and metastatic phenotypes and is a primary target of protease activated antibody-directed prodrug therapy. It therefore represents a potential therapeutic and diagnostic target and effort has been made to develop fluorescent probes to report on Cathepsin B activity in cells and animal models of cancer. We have designed, synthesized, and thoroughly evaluated four novel "turn on" probes that employ a lysosomotropic dansylcadaverine dye to report on Cathepsin B activity. Enzyme activity assays using a recombinant human enzyme and cancer cell lysates coupled with confocal microscopy experiments demonstrated that one of the probes, derivatized with the self-immolative prodrug linker p-aminobenzyl alcohol, can selectively report on Cathepsin B in biological samples including live cells.


Asunto(s)
Cadaverina/análogos & derivados , Catepsina B/análisis , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/metabolismo , Neoplasias/diagnóstico por imagen , Compuestos de Aminobifenilo/química , Cadaverina/síntesis química , Cadaverina/metabolismo , Catepsina B/metabolismo , Catepsina L/análisis , Catepsina L/metabolismo , Línea Celular Tumoral , Humanos , Hidrólisis , Cinética , Microscopía Confocal , Estructura Molecular , Imagen Óptica , Proteínas Recombinantes/análisis , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
4.
PLoS Negl Trop Dis ; 13(8): e0007610, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31449523

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging arbovirus known to cause chronic myalgia and arthralgia with high morbidity. CHIKV is now considered endemic in many countries across Asia and Africa. In this study, the susceptibility of various human, mammalian and mosquito cell lines to CHIKV infection was evaluated. CHIKV infection was found to be cell-type dependent and virus strain-specific. Furthermore, SJCRH30 (human rhabdomyosarcoma cell line) was showed to be highly permissive to CHIKV infection, with maximum production of infectious virions observed at 12 h.p.i. Pre-infection treatment of SJCRH30 with various inhibitors of endocytosis, including monodansylcadaverine (receptor-mediated endocytic inhibitor), dynasore (clathrin-mediated endocytic inhibitor), as well as filipin (caveolin-mediated endocytosis inhibitor), resulted in minimal inhibition of CHIKV infection. In contrast, dose-dependent inhibition of CHIKV infection was observed with the treatment of macropinocytosis inhibitor, 5-(N-ethyl-N-isopropyl)amiloride (EIPA). Furthermore, siRNA-mediated knockdown of sortin nexin 9 (SNX9) a protein involved in macropinosome formation, also resulted in a significant dose-dependent reduction in viral titre. By performing a virus entry assay, CHIKV particles were also observed to colocalize with FITC-dextran, a macropinosome marker. This study shows for the first time, that the infectious entry of CHIKV into human muscle cells is mediated by macropinocytosis. Together, the data from this study may pave the way for the development of specific inhibitors that target the entry process of CHIKV into cells.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Músculos/virología , Pinocitosis/fisiología , Internalización del Virus , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Cadaverina/análogos & derivados , Cadaverina/farmacología , Línea Celular , Supervivencia Celular , Virus Chikungunya/crecimiento & desarrollo , Clatrina/antagonistas & inhibidores , Endocitosis/efectos de los fármacos , Filipina/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Hidrazonas/farmacología , Cinética , Pinocitosis/efectos de los fármacos , Pinocitosis/genética , ARN Interferente Pequeño , Rabdomiosarcoma , Nexinas de Clasificación/genética , Carga Viral , Ensayo de Placa Viral
5.
Plant Physiol ; 179(2): 460-476, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30510038

RESUMEN

Inorganic phosphate (Pi) is often a limiting plant nutrient. In members of the Brassicaceae family, such as Arabidopsis (Arabidopsis thaliana), Pi deprivation reshapes root system architecture to favor topsoil foraging. It does so by inhibiting primary root extension and stimulating lateral root formation. Root growth inhibition from phosphate (Pi) deficiency is triggered by iron-stimulated, apoplastic reactive oxygen species generation and cell wall modifications, which impair cell-to-cell communication and meristem maintenance. These processes require LOW PHOSPHATE RESPONSE1 (LPR1), a cell wall-targeted ferroxidase, and PHOSPHATE DEFICIENCY RESPONSE2 (PDR2), the single endoplasmic reticulum (ER)-resident P5-type ATPase (AtP5A), which is thought to control LPR1 secretion or activity. Autophagy is a conserved process involving the vacuolar degradation of cellular components. While the function of autophagy is well established under nutrient starvation (C, N, or S), it remains to be explored under Pi deprivation. Because AtP5A/PDR2 likely functions in the ER stress response, we analyzed the effect of Pi limitation on autophagy. Our comparative study of mutants defective in the local Pi deficiency response, ER stress response, and autophagy demonstrated that ER stress-dependent autophagy is rapidly activated as part of the developmental root response to Pi limitation and requires the genetic PDR2-LPR1 module. We conclude that Pi-dependent activation of autophagy in the root apex is a consequence of local Pi sensing and the associated ER stress response, rather than a means for systemic recycling of the macronutrient.


Asunto(s)
Arabidopsis/fisiología , Autofagia/fisiología , Estrés del Retículo Endoplásmico/fisiología , Fosfatos/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Arabidopsis/citología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Estrés del Retículo Endoplásmico/genética , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Meristema/genética , Meristema/metabolismo , Mutación , Fosfitos/metabolismo , Células Vegetales , Raíces de Plantas/genética , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/metabolismo , Plantas Modificadas Genéticamente
6.
Amino Acids ; 50(7): 923-932, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29627904

RESUMEN

Microbial transglutaminase (TGase) has been successfully used to produce site-specific protein conjugates derivatized at the level of glutamine (Gln) or lysine (Lys) residues with diverse applications. Here, we study the drug human interferon ß-1a (IFN) as a substrate of TGase. The derivatization reaction was performed using carbobenzoxy-L-glutaminyl-glycine to modify Lys residues and dansylcadaverine for Gln residues. The 166 amino acids polypeptide chain of IFN ß-1a contains 11 Lys and 11 Gln residues potential sites of TGase derivatization. By means of mass spectrometry analyses, we demonstrate the highly selective derivatization of this protein by TGase at the level of Lys115 and as secondary site at the level of Lys33, while no reactive Gln residue was detected. Limited proteolysis experiments were performed on IFN to determine flexible regions of the protein under physiological conditions. Interestingly, primary and secondary sites of limited proteolysis and of TGase derivatization occur at the same regions of the polypeptide chain, indicating that the extraordinary selectivity of the TGase-mediated reaction is dictated by the conformational features of the protein substrate. We envisage that the TGase-mediated derivatization of IFN can be used to produce interesting derivatives of this important therapeutic protein.


Asunto(s)
Proteínas Bacterianas/química , Interferón beta-1a/química , Lisina/química , Procesamiento Proteico-Postraduccional , Streptomyces/enzimología , Transglutaminasas/química , Cadaverina/análogos & derivados , Cadaverina/química , Humanos
7.
Sci Rep ; 8(1): 2822, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434216

RESUMEN

Dysregulated autophagy is central to the pathogenesis and therapeutic development of cancer. However, how autophagy is regulated in cancer is not well understood and genes that modulate cancer autophagy are not fully defined. To gain more insights into autophagy regulation in cancer, we performed a large-scale RNA interference screen in K562 human chronic myeloid leukemia cells using monodansylcadaverine staining, an autophagy-detecting approach equivalent to immunoblotting of the autophagy marker LC3B or fluorescence microscopy of GFP-LC3B. By coupling monodansylcadaverine staining with fluorescence-activated cell sorting, we successfully isolated autophagic K562 cells where we identified 336 short hairpin RNAs. After candidate validation using Cyto-ID fluorescence spectrophotometry, LC3B immunoblotting, and quantitative RT-PCR, 82 genes were identified as autophagy-regulating genes. 20 genes have been reported previously and the remaining 62 candidates are novel autophagy mediators. Bioinformatic analyses revealed that most candidate genes were involved in molecular pathways regulating autophagy, rather than directly participating in the autophagy process. Further autophagy flux assays revealed that 57 autophagy-regulating genes suppressed autophagy initiation, whereas 21 candidates promoted autophagy maturation. Our RNA interference screen identifies identified genes that regulate autophagy at different stages, which helps decode autophagy regulation in cancer and offers novel avenues to develop autophagy-related therapies for cancer.


Asunto(s)
Autofagia/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Citometría de Flujo , Colorantes Fluorescentes , Ensayos Analíticos de Alto Rendimiento , Humanos , Células K562 , Microscopía Fluorescente , Interferencia de ARN , ARN Interferente Pequeño , Espectrometría de Fluorescencia
8.
Eur J Pharm Sci ; 111: 376-382, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29030178

RESUMEN

In this study, we present a targeted drug delivery system to improve intravesical therapy of bladder diseases. The drug delivery system consists of wheat germ agglutinin (WGA) to facilitate specific interaction with the surface of bladder cells and α-poly-(L)-glutamic acid (PGA) as polymeric backbone to increase the number of drug molecules per targeting moiety. Additionally, fluorescein cadaverine was coupled to PGA to visualise and track the delivery system. Using 5637 single cells and cell monolayers, the optimised F-PGA-WGA delivery system, with an approximate molecular weight of 670kDa, could convince with its promising cytoadhesive as well as cytoinvasive potential. Using the competitive inhibitor N, N', N″-triacetylchitotriose a specificity of the carbohydrate-mediated interaction between the cell and the delivery system of up to 98% was determined. F-PGA alone did not show any interaction with the cells. Moreover, a high drug loading of 77 molecules of the model drug Dansylcadaverine per backbone was achieved. Microscopic analysis further confirmed binding and uptake of the cytoadhesive polymer even after additional loading with the model drug. Combining the auspicious targeting properties of WGA with the high drug loading possibilities of the backbone might finally lead to an enhanced efficacy when used for intravesical therapy.


Asunto(s)
Cadaverina/análogos & derivados , Ácido Poliglutámico/química , Enfermedades de la Vejiga Urinaria/tratamiento farmacológico , Aglutininas del Germen de Trigo/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Materiales Biocompatibles , Transporte Biológico , Cadaverina/administración & dosificación , Cadaverina/química , Cadaverina/farmacocinética , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Urotelio/citología
9.
J Occup Health ; 60(2): 148-155, 2018 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-29187674

RESUMEN

OBJECTIVES: Indium compounds are used in manufacturing displays of mobile phones and televisions. However, these materials cause interstitial pneumonia in exposed workers. Animal experiments demonstrated that indium compounds caused lung cancer. Chronic inflammation is considered to play a role in lung carcinogenesis and fibrosis induced by particulate matters. 8-Nitroguanine (8-nitroG) is a mutagenic DNA lesion formed during inflammation and may participate in carcinogenesis. To clarify the mechanism of carcinogenesis, we examined 8-nitroG formation in indium-exposed cultured cells. METHODS: We treated RAW 264.7 mouse macrophages with indium oxide (In2O3) nanoparticles (primary diameter: 30-50 nm), and performed fluorescent immunocytochemistry to detect 8-nitroG. The extent of 8-nitroG formation was evaluated by quantitative image analysis. We measured the amount of nitric oxide (NO) in the culture supernatant of In2O3-treated cells by the Griess method. We also examined the effects of inhibitors of inducible NO synthase (iNOS) and endocytosis on In2O3-induced 8-nitroG formation. RESULTS: In2O3 significantly increased the intensity of 8-nitroG formation in RAW 264.7 cells in a dose-dependent manner. In2O3-induced 8-nitroG formation was observed at 2 h and further increased at 4 h, and the amount of NO released from In2O3-exposed cells was significantly increased at 2-4 h compared with the control. 8-NitroG formation was suppressed by 1400W (an iNOS inhibitor), methyl-ß-cyclodextrin and monodansylcadaverine (inhibitors of caveolae- and clathrin-mediated endocytosis, respectively). CONCLUSIONS: These results suggest that endocytosis and NO generation participate in indium-induced 8-nitroG formation. NO released from indium-exposed inflammatory cells may induce DNA damage in adjacent lung epithelial cells and contribute to carcinogenesis.


Asunto(s)
Daño del ADN/efectos de los fármacos , Guanina/análogos & derivados , Indio/farmacología , Macrófagos/efectos de los fármacos , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Cadaverina/análogos & derivados , Cadaverina/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Guanina/biosíntesis , Inmunohistoquímica , Ratones , Nanopartículas , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Tamaño de la Partícula , beta-Ciclodextrinas/farmacología
10.
PLoS One ; 12(9): e0185164, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28945762

RESUMEN

The insulin-like growth factor-1 receptor (IGF1R) mediates the biological actions of IGF1 and IGF2. The IGF1R is involved in both physiological and pathological activities and is usually overexpressed in most types of cancer. In addition to its classical mechanism of action, recent evidence has shown a nuclear presence of IGF1R, associated with novel genomic/transcriptional types of activities. The present study was aimed at evaluating the hypothesis that nuclear IGF1R localization is not restricted to cancer cells and might constitute a novel physiologically relevant regulatory mechanism. Our data shows that nuclear translocation takes place in a wide array of cells, including normal diploid fibroblasts. In addition, we provide evidence for a synergistic effect of a nuclear translocation blocker along with selective IGF1R inhibitors in terms of decreasing cell proliferation. Given the important role of the IGF1R in mitogenesis, the present results may be of translational relevance in cancer research. In conclusion, results are consistent with the concept that nuclear IGF1R fulfills important physiological and pathological roles.


Asunto(s)
Proliferación Celular/fisiología , Receptores de Somatomedina/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Cadaverina/análogos & derivados , Cadaverina/farmacología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Núcleo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Células Cultivadas , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Microscopía Confocal , Receptor IGF Tipo 1 , Receptores de Somatomedina/antagonistas & inhibidores , Receptores de Somatomedina/genética , Transducción de Señal
11.
PLoS One ; 12(9): e0184324, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28877265

RESUMEN

Glioblastoma is the most common and lethal malignant primary brain tumor for which the development of efficacious chemotherapeutic agents remains an urgent need. The anti-helminthic drug niclosamide, which has long been in use to treat tapeworm infections, has recently attracted renewed interest due to its apparent anticancer effects in a variety of in vitro and in vivo cancer models. However, the mechanism(s) of action remains to be elucidated. In the present study, we found that niclosamide induced cell toxicity in human glioblastoma cells corresponding with increased protein ubiquitination, ER stress and autophagy. In addition, niclosamide treatment led to down-regulation of Wnt/ß-catenin, PI3K/AKT, MAPK/ERK, and STAT3 pro-survival signal transduction pathways to further reduce U-87 MG cell viability. Taken together, these results provide new insights into the glioblastoma suppressive capabilities of niclosamide, showing that niclosamide can target multiple major cell signaling pathways simultaneously to effectively promote cell death in U-87 MG cells. Niclosamide constitutes a new prospect for a therapeutic treatment against human glioblastoma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Niclosamida/farmacología , Ubiquitinación , Antihelmínticos/química , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Cadaverina/análogos & derivados , Cadaverina/química , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/tratamiento farmacológico , Humanos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
12.
J Biol Chem ; 292(29): 12041-12053, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28546427

RESUMEN

Ubiquitous polyamine spermidine is not required for normal planktonic growth of Bacillus subtilis but is essential for robust biofilm formation. However, the structural features of spermidine required for B. subtilis biofilm formation are unknown and so are the molecular mechanisms of spermidine-stimulated biofilm development. We report here that in a spermidine-deficient B. subtilis mutant, the structural analogue norspermidine, but not homospermidine, restored biofilm formation. Intracellular biosynthesis of another spermidine analogue, aminopropylcadaverine, from exogenously supplied homoagmatine also restored biofilm formation. The differential ability of C-methylated spermidine analogues to functionally replace spermidine in biofilm formation indicated that the aminopropyl moiety of spermidine is more sensitive to C-methylation, which it is essential for biofilm formation, but that the length and symmetry of the molecule is not critical. Transcriptomic analysis of a spermidine-depleted B. subtilis speD mutant uncovered a nitrogen-, methionine-, and S-adenosylmethionine-sufficiency response, resulting in repression of gene expression related to purine catabolism, methionine and S-adenosylmethionine biosynthesis and methionine salvage, and signs of altered membrane status. Consistent with the spermidine requirement in biofilm formation, single-cell analysis of this mutant indicated reduced expression of the operons for production of the exopolysaccharide and TasA protein biofilm matrix components and SinR antagonist slrR Deletion of sinR or ectopic expression of slrR in the spermidine-deficient ΔspeD background restored biofilm formation, indicating that spermidine is required for expression of the biofilm regulator slrR Our results indicate that spermidine functions in biofilm development by activating transcription of the biofilm matrix exopolysaccharide and TasA operons through the regulator slrR.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/agonistas , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica , Polisacáridos Bacterianos/biosíntesis , Espermidina/metabolismo , Factores de Transcripción/agonistas , Adenosilmetionina Descarboxilasa/genética , Adenosilmetionina Descarboxilasa/metabolismo , Bacillus subtilis/citología , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Metionina/metabolismo , Metilación , Ciclo del Nitrógeno , Operón , Purinas/metabolismo , S-Adenosilmetionina/metabolismo , Análisis de la Célula Individual , Espermidina/análogos & derivados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
J Biol Chem ; 292(15): 6369-6380, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28258224

RESUMEN

In Drosophila, the final immune deficiency (IMD) pathway-dependent signal is transmitted through proteolytic conversion of the nuclear factor-κB (NF-κB)-like transcription factor Relish to the active N-terminal fragment Relish-N. Relish-N is then translocated from the cytosol into the nucleus for the expression of IMD-controlled genes. We previously demonstrated that transglutaminase (TG) suppresses the IMD pathway by polymerizing Relish-N to inhibit its nuclear translocation. Conversely, we also demonstrated that orally ingested synthetic amines, such as monodansylcadaverine (DCA) and biotin-labeled pentylamine, are TG-dependently incorporated into Relish-N, causing the nuclear translocation of modified Relish-N in gut epithelial cells. It remains unclear, however, whether polyamine-containing Relish-N retains transcriptional activity. Here, we used mass spectrometry analysis of a recombinant Relish-N modified with DCA by TG activity after proteolytic digestion and show that the DCA-modified Gln residues are located in the DNA-binding region of Relish-N. TG-catalyzed DCA incorporation inhibited binding of Relish-N to the Rel-responsive element in the NF-κB-binding DNA sequence. Subcellular fractionation of TG-expressing Drosophila S2 cells indicated that TG was localized in both the cytosol and nucleus. Of note, natural polyamines, including spermidine and spermine, competitively inhibited TG-dependent DCA incorporation into Relish-N. Moreover, in vivo experiments demonstrated that Relish-N was modified by spermine and that this modification reduced transcription of IMD pathway-controlled cecropin A1 and diptericin genes. These findings suggest that intracellular TG regulates Relish-N-mediated transcriptional activity by incorporating polyamines into Relish-N and via protein-protein cross-linking.


Asunto(s)
Cadaverina/análogos & derivados , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Factores de Transcripción/metabolismo , Transglutaminasas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Cadaverina/metabolismo , Línea Celular , Núcleo Celular/genética , Citosol/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster , Células Epiteliales/citología , Intestinos/citología , Dominios Proteicos , Factores de Transcripción/genética , Transglutaminasas/genética
14.
Artículo en Inglés | MEDLINE | ID: mdl-28224118

RESUMEN

Airway epithelial cell is often the initial site of attack by pathogens, and cell death is commonly caused by internalization of Mycobacterium tuberculosis (Mtb). However, the mechanism of interaction between epithelial cells and Mtb is not well understood. In this study, we investigated the role of the heparin-binding hemagglutinin (HBHA) protein of Mtb in the function of epithelial cells. In particular, the autophagy of A549 cells was determined based on microtubule-associated protein 1 light chain 3 alpha (LC3) activity. Autophagosome formation was detected by Monodansylcadaverine (MDC) staining and immune fluorescence staining of LC3. Autophagy could be significantly suppressed by HBHA protein. In addition, the LDH assay results showed that HBHA treatment could induce death on A549 cells. To explore the form of cell death, we detected the activity of caspase-3 and LDH release of A549 cells in the presence or absence of caspase inhibitor Z-VAD-FMK. Results demonstrated that HBHA treatment could induce apoptosis of A549 cells. To further confirm these results, we constructed the recombinant Mycobacterium smegmatis (MS) expressing HBHA (rMS-HBHA) and explored the influence of rMS-HBHA on the function of A549 cells. rMS-HBHA infection significantly inhibited LC3 expression and the maturation of autophagosomes in A549 cells. Subsequently, we infected A549 cells with MS and detected the viability of intracellular MS by CFU counts. rMS-HBHA showed higher survival and replication capacity in A549 cells than those of the wild-type MS. Finally, infection of A549 cells with rMS-HBHA caused further apoptosis. These findings suggested that rMS-HBHA could inhibit autophagy, promote its survival and replication within A549 cells, and subsequently induce apoptosis on infected cells to facilitate infection.


Asunto(s)
Autofagia/efectos de los fármacos , Lectinas/antagonistas & inhibidores , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Células A549 , Clorometilcetonas de Aminoácidos/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Cadaverina/análogos & derivados , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Recuento de Colonia Microbiana , Células Epiteliales/microbiología , Escherichia coli/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas de la Membrana/farmacología , Viabilidad Microbiana , Proteínas Asociadas a Microtúbulos/metabolismo , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/inmunología , Mycobacterium tuberculosis/genética , Proteínas Recombinantes , Factores de Virulencia
15.
Cell Death Differ ; 24(4): 660-671, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28186502

RESUMEN

Tumor necrosis factor-alpha (TNF-α) exists in two forms: secretory TNF-α (sTNF-α) and transmembrane TNF-α (tmTNF-α). Although both forms of TNF-α induce tumor cell apoptosis, tmTNF-α is able to kill tumor cells that are resistant to sTNF-α-mediated cytotoxicity, indicating their differences in signal transduction. Here, we demonstrate that internalization of TNFR1 is crucial for sTNF-α- but not for tmTNF-α-induced apoptosis. sTNF-α induces binding of tumor necrosis factor receptor type 1-associated death domain protein (TRADD) to the death domain (DD) of TNFR1 and subsequent activation of nuclear factor kappa B (NF-κB), and the formation of death-inducing signaling complexes (DISCs) in the cytoplasm after internalization. In contrast, tmTNF-α induces DISC formation on the membrane in a DD-independent manner. It leads to the binding of signal transducer and activator of transcription 1 (STAT1) to a region spanning amino acids 319-337 of TNFR1 and induces phosphorylation of serine at 727 of STAT1. The phosphorylation of STAT1 promotes its binding to TRADD, and thus recruits Fas-associated protein with DD (FADD) and caspase 8 to form DISC complexes. This STAT1-dependent signaling results in apoptosis but not NF-κB activation. STAT1-deficiency in U3A cells counteracts tmTNF-α-induced DISC formation and apoptosis. Conversely, reconstitution of STAT1 expression restores tmTNF-α-induced apoptotic signaling in the cell line. Consistently, tmTNF-α suppresses the growth of STAT1-containing HT1080 tumors, but not of STAT1-deficient U3A tumors in vivo. Our data reveal an unappreciated molecular mechanism of tmTNF-α-induced apoptosis and may provide a new clue for cancer therapy.


Asunto(s)
Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Cadaverina/análogos & derivados , Cadaverina/farmacología , Caspasa 8/metabolismo , Línea Celular , Proteína de Dominio de Muerte Asociada a Fas/antagonistas & inhibidores , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Células HEK293 , Humanos , Ratones , FN-kappa B/metabolismo , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Unión Proteica , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Factor de Transcripción STAT1/genética , Transducción de Señal/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/toxicidad
16.
Amino Acids ; 49(3): 567-583, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-26886924

RESUMEN

Tissue transglutaminase (TGase 2) is the most abundantly expressed enzyme of the transglutaminase family and involved in a large variety of pathological processes, such as neurodegenerative diseases, disorders related to autoimmunity and inflammation as well as tumor growth, progression and metastasis. As a result, TGase 2 represents an attractive target for drug discovery and development, which requires assays that allow for the characterization of modulating agents and are appropriate for high-throughput screening. Herein, we report a fluorescence anisotropy-based approach for the determination of TGase 2's transamidase activity, following the time-dependent increase in fluorescence anisotropy due to the enzyme-catalyzed incorporation of fluorescein- and rhodamine B-conjugated cadaverines 1-3 (acyl acceptor substrates) into N,N-dimethylated casein (acyl donor substrate). These cadaverine derivatives 1-3 were obtained by solid-phase synthesis. To allow efficient conjugation of the rhodamine B moiety, different linkers providing secondary amine functions, such as sarcosyl and isonipecotyl, were introduced between the cadaverine and xanthenyl entities in compounds 2 and 3, respectively, with acyl acceptor 3 showing the most optimal substrate properties of the compounds investigated. The assay was validated for the search of both irreversible and reversible TGase 2 inhibitors using the inactivators iodoacetamide and a recently published L-lysine-derived acrylamide and the allosteric binder GTP, respectively. In addition, the fluorescence anisotropy-based method was proven to be suitable for high-throughput screening (Z' factor of 0.86) and represents a non-radioactive and highly sensitive assay for determining the active TGase 2 concentration.


Asunto(s)
Cadaverina/análogos & derivados , Inhibidores Enzimáticos/química , Colorantes Fluorescentes/química , Proteínas de Unión al GTP/química , Ensayos Analíticos de Alto Rendimiento , Proteínas Recombinantes/química , Transglutaminasas/química , Animales , Cadaverina/síntesis química , Caseínas/química , Dominio Catalítico , Fluoresceína/síntesis química , Polarización de Fluorescencia/métodos , Colorantes Fluorescentes/síntesis química , Proteínas de Unión al GTP/antagonistas & inhibidores , Guanosina Trifosfato/química , Cobayas , Humanos , Yodoacetamida/química , Cinética , Hígado/química , Hígado/enzimología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Rodaminas/química , Técnicas de Síntesis en Fase Sólida , Transglutaminasas/antagonistas & inhibidores
17.
J Eukaryot Microbiol ; 64(3): 308-321, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27603757

RESUMEN

Autophagy is a degradative process by which eukaryotic cells digest their own components to provide aminoacids that may function as energy source under nutritional stress conditions. There is experimental evidence for autophagy in parasitic protists belonging to the family Trypanosomatidae. However, few proteins implicated in this process have been characterized so far in these parasites. Moreover, it has been shown that autophagy is involved in Trypanosoma cruzi differentiation and thus might have a role in pathogenicity. Here, we report the cloning and biochemical characterization of TcVps15. In addition, we demonstrate that TcVps15 interact with the PI3K TcVps34 and that both proteins associate with cellular membranes. Under nutritional stress conditions, TcVps15 and TcVps34 modify their subcellular distribution showing a partial co-localization in autophagosomes with TcAtg8.1 and using an active site TcVps15-mutated version (TcVps15-K219D-HA) we demonstrated that this relocalization depends on the TcVps15 catalytic activity. Overexpression of TcVps15-HA and TcVps15-K219D-HA also leads to increased accumulation of monodansylcadaverine (MDC) in autophagic vacuoles under nutritional stress conditions compared to wild-type cells. In addition, the MDC-specific activity shows to be significantly higher in TcVps15-HA overexpressing cells when compared with TcVps15-K219D-HA. Our results reveal for the first time a role of TcVps15 as a key regulator of TcVps34 enzymatic activity and implicate the TcVps15-Vps34 complex in autophagy in T. cruzi, exposing a new key pathway to explore novel chemotherapeutic targets.


Asunto(s)
Autofagia , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/metabolismo , Proteína de Clasificación Vacuolar VPS15/metabolismo , Animales , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Técnicas de Cultivo de Célula , Membrana Celular/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/genética , Fosfatidilinositol 3-Quinasas Clase III/fisiología , Clonación Molecular , ADN Protozoario , Pruebas de Enzimas , Regulación Enzimológica de la Expresión Génica , Estadios del Ciclo de Vida , Mutagénesis Sitio-Dirigida , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Análisis de Secuencia , Transfección , Trypanosoma cruzi/citología , Trypanosoma cruzi/genética , Técnicas del Sistema de Dos Híbridos , Proteína de Clasificación Vacuolar VPS15/genética , Proteína de Clasificación Vacuolar VPS15/fisiología , Vacuolas/metabolismo
18.
Exp Cell Res ; 348(2): 165-176, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27658567

RESUMEN

Clearance of the apoptotic cells by phagocytes plays pivotal roles in maintenance of tissue homeostasis, promotion of immunological tolerance and anti-inflammatory response. Recent studies show that autophagy is involved in phagocytosis of the apoptotic cells. However, contribution of autophagy to phagocytosis of the apoptotic cells by macrophages is not clearly defined. Here, we assessed cytoprotective effect of autophagy on clearance of the apoptotic cells. Apoptosis of murine splenic lymphocytes and human T-cell leukemia cells was induced with cyclophosphamide. After engulfment of the apoptotic cells, expression of Belin-1 and LC3 in macrophages was upregulated, the number of MDC-positive vesicles, LC3-positive autophagosomes and autophagic ultrastructures increased significantly. Autophagosome was fused with phagosome containing fragments of the nuclei or other debris of the apoptotic cells to form amphisome. Some cells in macrophages phagocytosing the apoptotic cells became apoptotic. After autophagy of macrophages was inhibited with 3-MA, viability and survival of macrophages reduced, phagocytosis of the apoptotic cells by macrophages deceased significantly. These results demonstrate that autophagy plays an important role in promoting clearance of the apoptotic cells by protecting macrophages from apoptosis during phagocytosis as well as degrading the contents of phagosomes via amphisome formation.


Asunto(s)
Apoptosis , Autofagia , Citoprotección , Macrófagos/citología , Fagocitosis , Animales , Beclina-1/genética , Beclina-1/metabolismo , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Supervivencia Celular , Vesículas Citoplasmáticas/metabolismo , Humanos , Células Jurkat , Linfocitos/citología , Macrófagos/ultraestructura , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley
19.
Nutr Metab Cardiovasc Dis ; 26(10): 922-30, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27378395

RESUMEN

BACKGROUND AND AIMS: Transglutaminase 2 (TGM2), a member of the transglutaminase family of enzymes, is a multifunctional protein involved in numerous events spanning from cell differentiation, to signal transduction, apoptosis, and wound healing. It is expressed in a variety of cells, macrophages included. Macrophage TGM2 promotes the clearance of apoptotic cells (efferocytosis) and emerging evidence suggests that defective efferocytosis contributes to the consequences of inflammation-associated diseases, including atherosclerotic lesion progression and its sequelae. Of interest, active TGM2 identified in human atherosclerotic lesions plays critical roles in plaque stability through effects on matrix cross-linking and TGFß activity. This study explores the mechanisms by which TGM2 controls efferocytosis in human macrophages. METHODS AND RESULTS: Herein we show that TGM2 increases progressively during monocyte differentiation towards macrophages and controls their efferocytic potential as well as morphology and viability. Two experimental approaches that took advantage of the inhibition of TGM2 activity and protein silencing give proof that TGM2 reduction significantly impairs macrophage efferocytosis. Among the mechanisms involved we highlighted a role of the receptors CD14 and SR-AI whose levels were markedly reduced by TGM2 inhibition. Conversely, CD36 receptor and αvß3 integrin levels were not influenced. Of note, lipid accumulation and IL-10 secretion were reduced in macrophages displaying defective efferocytosis. CONCLUSION: Overall, our data define a crucial role of TGM2 activity during macrophage differentiation via mechanisms involving CD14 and SR-AI receptors and show that TGM2 inhibition triggers a pro-inflammatory phenotype.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP/antagonistas & inhibidores , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Factores de Empalme Serina-Arginina/metabolismo , Transglutaminasas/antagonistas & inhibidores , Apoptosis , Cadaverina/análogos & derivados , Cadaverina/farmacología , Diferenciación Celular , Forma de la Célula , Supervivencia Celular , Técnicas de Cocultivo , Cistamina/farmacología , Relación Dosis-Respuesta a Droga , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Interleucina-10/metabolismo , Células Jurkat , Macrófagos/enzimología , Macrófagos/metabolismo , Fenotipo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Linfocitos T/patología , Factores de Tiempo , Transfección , Transglutaminasas/genética , Transglutaminasas/metabolismo
20.
Phytomedicine ; 23(9): 939-47, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27387402

RESUMEN

BACKGROUND: Psoralidin (PSO), a natural phenolic coumarin, was reported to have anti-cancer activities. PSO induced reactive oxygen species (ROS) generation in cancer cells. The role of ROS in its anti-cancer effect remains unclear. PURPOSE: This study was designed to investigate the potential roles of ROS in PSO-induced anti-cancer effect in MCF-7 breast cancer cells. METHODS: Effect of PSO on cancer cell proliferation was determined by MTT assay. Comet assay was used to determine DNA damage. Protein expression was detected by Western blotting. Autophagic vacuoles were detected by monodansylcadaverine (MDC) staining. ROS generation was measured by fluorescent probe. NOX4 localization was determined by immunofluorescence staining. RESULTS: PSO treatment caused proliferation inhibition in time- and dose- dependent manners, which was partially reversed by N-acetyl cysteine (NAC) and diphenyleneiodonium (DPI). PSO induced DNA damage and increased protein expression of γ-H2AX, phosphorylation of ATM, ATR, Chk1, and Chk2. PSO induced autophagy as evidenced by the accumulation of autophagic vacuoles and alterations of autophagic protein expression. PSO-induced cell death was enhanced by autophagy inhibitor chloroquine (CQ). Furthermore, PSO treatment induced ROS formation, which was reversed by NAC or DPI pretreatment. The expression of NOX4 was significantly enhanced by PSO. Both NAC and DPI could reverse PSO-induced DNA damage and autophagic responses. In addition, silencing NOX4 by siRNA inhibited PSO-induced ROS generation, DNA damage, and autophagy. CONCLUSIONS: Taken together, these results showed that PSO induced DNA damage and protective autophagy mediated by ROS generation in a NOX4-dependent manner in MCF-7 cells.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Autofagia/efectos de los fármacos , Benzofuranos/toxicidad , Neoplasias de la Mama/genética , Cumarinas/toxicidad , Daño del ADN , NADPH Oxidasas/metabolismo , Especies de Nitrógeno Reactivo , Apoptosis/efectos de los fármacos , Cadaverina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Colorantes Fluorescentes , Silenciador del Gen , Humanos , Células MCF-7 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , ARN Interferente Pequeño/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA