Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Chem Biol ; 17(12): 1305-1313, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34725510

RESUMEN

Triacsins are an intriguing class of specialized metabolites possessing a conserved N-hydroxytriazene moiety not found in any other known natural products. Triacsins are notable as potent acyl-CoA synthetase inhibitors in lipid metabolism, yet their biosynthesis has remained elusive. Through extensive mutagenesis and biochemical studies, we here report all enzymes required to construct and install the N-hydroxytriazene pharmacophore of triacsins. Two distinct ATP-dependent enzymes were revealed to catalyze the two consecutive N-N bond formation reactions, including a glycine-utilizing, hydrazine-forming enzyme (Tri28) and a nitrite-utilizing, N-nitrosating enzyme (Tri17). This study paves the way for future mechanistic interrogation and biocatalytic application of enzymes for N-N bond formation.


Asunto(s)
Coenzima A Ligasas/metabolismo , Streptomyces aureofaciens/enzimología , Streptomyces aureofaciens/genética , Triazenos/metabolismo , Biocatálisis , Escherichia coli/genética , Glicina/química , Hidrazinas/química , Metabolismo de los Lípidos , Lípidos/química , Nitritos/química , Triazenos/química
2.
Chem Biol Drug Des ; 97(2): 237-252, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32772433

RESUMEN

The efficient synthesis of molecular hybrids including a DNA-intercalating 9-anilinoacridine (9-AnA) core and a methyl triazene DNA-methylating moiety is described. Nucleophilic aromatic substitution (SN Ar) and electrophilic aromatic substitution (EAS) reactions using readily accessible starting materials provide a quick entry to novel bifunctional anticancer molecules. The chimeras were evaluated for their anticancer activity. Chimera 7b presented the highest antitumor activity at low micromolar IC50 values in antiproliferative assays performed with various cancer cell lines. In comparison, compound 7b outperformed DNA-intercalating drugs like amsacrine and AHMA. Mechanistic studies of chimera 7b suggest a dual mechanism of action: methylation of the DNA-repairing protein MGMT associated with the triazene structural portion and Topo II inhibition by intercalation of the acridine core.


Asunto(s)
Amsacrina/análogos & derivados , Antineoplásicos/síntesis química , Triazenos/química , Amsacrina/química , Amsacrina/metabolismo , Amsacrina/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/química , ADN/metabolismo , ADN-Topoisomerasas de Tipo II/química , ADN-Topoisomerasas de Tipo II/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Sustancias Intercalantes/química , Sustancias Intercalantes/metabolismo , Sustancias Intercalantes/farmacología , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/metabolismo , Triazenos/metabolismo , Triazenos/farmacología
3.
Cell Physiol Biochem ; 52(3): 397-407, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30845379

RESUMEN

BACKGROUND/AIMS: TNF-α-mediated pro-inflammatory phenotypic change in monocytes is known to be implicated in the pathogenesis of metabolic inflammation and insulin resistance. However, the mechanism by which TNF-α-induces inflammatory phenotypic shift in monocytes is poorly understood. Since long-chain acyl-CoA synthetase 1 (ACSL1) is associated with inflammatory monocytes/macrophages, we investigated the role of ACSL1 in the TNF-α-driven inflammatory phenotypic shift in the monocytes. METHODS: Monocytes (Human monocytic THP-1 cells) were stimulated with TNF-α. Inflammatory phenotypic markers (CD16, CD11b, CD11c and HLA-DR) expression was determined with real time RTPCR and flow cytometry. IL-1ß and MCP-1 were determined by ELISA. Signaling pathways were identified by using ACSL1 inhibitor, ACSL1 siRNA and NF-κB reporter monocytic cells. Phosphorylation of NF-κB was analyzed by western blotting and flow cytometry. RESULTS: Our data show that TNF-α induced significant increase in the expression of CD16, CD11b, CD11c and HLA-DR. Inhibition of ACSL1 activity in the cells with triacsin C significantly suppressed the expression of these inflammatory markers. Using ACSL-1 siRNA, we further demonstrate that TNF-α-induced inflammatory markers expression in monocytic cells requires ACSL1. In addition, IL-1b and MCP-1 production by TNF-α activated monocytic cells was significantly blocked by the inhibition of ACSL-1 activity. Interestingly, elevated NF-κB activity resulting from TNF-α stimulation was attenuated in ACSL1 deficient cells. CONCLUSION: Our findings provide an evidence that TNF-α-associated inflammatory polarization in monocytes is an ACSL1 dependent process, which indicates its central role in TNF-α-driven metabolic inflammation.


Asunto(s)
Coenzima A Ligasas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Quimiocina CCL2/análisis , Coenzima A Ligasas/antagonistas & inhibidores , Coenzima A Ligasas/genética , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Inflamación/metabolismo , Interleucina-1beta/análisis , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Triazenos/química , Triazenos/metabolismo
4.
Chembiochem ; 20(9): 1145-1149, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30589194

RESUMEN

Triacsins are a family of natural products having in common an N-hydroxytriazene moiety not found in any other known secondary metabolites. Though many studies have examined the biological activity of triacsins in lipid metabolism, their biosynthesis has remained unknown. Here we report the identification of the triacsin biosynthetic gene cluster in Streptomyces aureofaciens ATCC 31442. Bioinformatic analysis of the gene cluster led to the discovery of the tacrolimus producer Streptomyces tsukubaensis NRRL 18488 as a new triacsin producer. In addition to targeted gene disruption to identify necessary genes for triacsin production, stable isotope feeding was performed in vivo to advance the understanding of N-hydroxytriazene biosynthesis.


Asunto(s)
Familia de Multigenes , Triazenos/metabolismo , Biología Computacional , Inhibidores Enzimáticos/metabolismo , Enzimas/genética , Genes Bacterianos , Mutación , Streptomyces/genética , Streptomyces aureofaciens/genética
5.
J Pharm Sci ; 105(2): 729-733, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26458075

RESUMEN

Nitric oxide (NO), an endogenous gas that plays a versatile role in the physiological system, has the ability to increase the intestinal absorption of water-soluble compounds through the paracellular route. However, it remains unclear whether NO can enhance the absorption of hydrophobic drugs through the transcellular route. In this study, we examined the absorption-enhancing effect of NO on intestinal permeability of hydrophobic drugs in rat intestine. The pretreatment of rat gastrointestinal sacs with NOC7, a NO-releasing reagent, significantly increased the permeation of griseofulvin from mucosa to serosa in the sacs prepared from the duodenum, but not in those prepared from the other regions such as jejunum, ileum, and colon. The absorption-enhancing effect of NOC7 on the duodenal permeation varied depending on the hydrophobicity of the drugs used. Furthermore, NOC7 treatment was found to be apparently ineffective on the griseofulvin permeation in the duodenum pretreated with dithiothreitol (DTT) that was used as a mucus remover, even though the permeation was increased by pretreatment with DTT alone. These results suggest that NO increases the absorption of hydrophobic drugs through the transcellular route in the duodenum by modulating the mucus layer function.


Asunto(s)
Duodeno/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Absorción Intestinal/efectos de los fármacos , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Animales , Sinergismo Farmacológico , Duodeno/metabolismo , Absorción Intestinal/fisiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratas , Ratas Wistar , Triazenos/administración & dosificación , Triazenos/metabolismo
6.
Gut ; 65(9): 1416-26, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-25986942

RESUMEN

OBJECTIVE: Barrett's metaplasia might develop if GORD causes oesophageal squamous cells to convert into columnar cells. Acid and bile exposures upregulate columnar differentiation genes like CDX2 in oesophageal squamous cells, but it is not known if such exposures downregulate squamous differentiation genes like SOX2. In addition to acid and bile, patients with GORD also have high oesophageal concentrations of nitric oxide (NO). This study aims to determine how acid, bile salts and NO affect genes that influence oesophageal cell phenotype. DESIGN: Oesophageal squamous cells from patients with Barrett's oesophagus were exposed to acidic bile salts or NOC-9 (an NO donor). SOX2, p63 (squamous transcription factor) and CDX2 mRNAs were measured by quantitative RT-PCR. SOX2 and its regulatory Akt pathway proteins were evaluated by western blotting. S-nitrosylation by NO was blocked by dithiothreitol. Immunohistochemistry for SOX2 was performed on the oesophagus of rats with surgically induced GORD which were fed diets with and without nitrite supplementation. RESULTS: In oesophageal squamous cells, NO profoundly decreased SOX2 protein and caused a significantly greater decrease in SOX2 mRNA than did acidic bile salts. NO also decreased p63 and increased CDX2 expression. NO caused S-nitrosylation of Akt, blocking its phosphorylation. Akt pathway inhibition by LY294002 or Akt siRNA reduced SOX2 mRNA. Rats fed with nitrite-supplemented diets exhibited weaker SOX2 oesophageal staining than rats fed with normal diets. CONCLUSIONS: In oesophageal squamous cells, NO blocks SOX2 expression through Akt S-nitrosylation. NO also increases CDX2 and decreases p63 expression. By triggering molecular events preventing squamous differentiation while promoting intestinal differentiation, NO might contribute to Barrett's pathogenesis.


Asunto(s)
Esófago de Barrett , Factor de Transcripción CDX2/metabolismo , Células Epiteliales , Reflujo Gastroesofágico , Óxido Nítrico/metabolismo , Factores de Transcripción SOXB1/metabolismo , Triazenos/metabolismo , Animales , Esófago de Barrett/etiología , Esófago de Barrett/metabolismo , Esófago de Barrett/patología , Ácidos y Sales Biliares/metabolismo , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Esófago/patología , Reflujo Gastroesofágico/complicaciones , Reflujo Gastroesofágico/metabolismo , Reflujo Gastroesofágico/patología , Humanos , Masculino , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/fisiología
7.
Eur J Med Chem ; 70: 1-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24125877

RESUMEN

In this research work we report the synthesis of a new series of triazene prodrugs designed for Melanocyte-Directed Enzyme Prodrug Therapy (MDEPT). These compounds are derived from the N-acyltyrosine amino acid - a good enzyme substrate for the tyrosinase enzyme, which is significantly overexpressed in melanoma cells. We analysed their chemical stability and plasma enzymatic hydrolysis, and we also evaluated the release of the antitumoral drug in the presence of the tyrosinase. Subsequently, we performed the evaluation of the prodrug cytotoxicity in melanoma cell lines with different levels of tyrosinase activity. Prodrug 5c showed the highest cytotoxicity against melanoma cell lines, and this effect correlated well with the tyrosinase activity suggesting that prodrug cytotoxicity is tyrosinase-dependent.


Asunto(s)
Aminoácidos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Monofenol Monooxigenasa/metabolismo , Profármacos/metabolismo , Triazenos/química , Triazenos/síntesis química , Triazenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Melanoma/enzimología , Estructura Molecular , Profármacos/síntesis química , Profármacos/química , Profármacos/farmacología , Relación Estructura-Actividad , Triazenos/metabolismo
8.
Anticancer Drugs ; 23(5): 483-93, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22421369

RESUMEN

ZRBA1 is a quinazoline-based molecule termed 'combi-molecule' designed to block the epidermal growth factor receptor (EGFR) and further degrade to FD105, an EGFR inhibitor plus a DNA-alkylating agent. To augment the potency of ZRBA1, we designed JDE52, a bistriazene that, following degradation, was 'programmed' to yield higher concentrations of the free inhibitor FD105 and a more cytotoxic bifunctional DNA-damaging species. The results indicated that JDE52 was capable of inducing significant blockade of EGFR phosphorylation, DNA strand breaks and interstrand cross-links in human cells. The fluorescent property of FD105, the secondary inhibitor that both JDE52 and ZRBA1 are capable of releasing, has permitted the analysis of its levels in tumour cells by ultraviolet flow cytometry. It was found that JDE52 was indeed capable of significantly releasing higher levels of fluorescence (P<0.05) in human tumour cells when compared with ZRBA1. Apoptosis was triggered by JDE52 at a faster rate than ZRBA1 and led to higher levels of cell killing. The results in toto suggest that the superior potency of JDE52, when compared with ZRBA1, may be imputed to mechanisms associated with the generation of higher intracellular concentrations of FD105 and to the induction of DNA cross-links. These combined mechanisms (blockade of EGFR-tyrosine kinase and induction of cross-links) contributed to an accelerated rate of apoptosis by JDE52. This study conclusively demonstrated that designing molecules as prodrugs of high levels of quinazoline inhibitors of EGFR and bifunctional DNA cross-linking species is a valid strategy to enhance the potency of mixed EGFR-DNA-targeting combi-molecules.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Roturas del ADN/efectos de los fármacos , Diseño de Fármacos , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Triazenos/farmacología , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Microscopía Fluorescente , Estructura Molecular , Quinazolinas/química , Quinazolinas/metabolismo , Relación Estructura-Actividad , Triazenos/química , Triazenos/metabolismo
9.
Cell Biochem Biophys ; 60(1-2): 89-97, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21448667

RESUMEN

Nitric oxide (NO) limits formation of neointimal hyperplasia in animal models of arterial injury in large part by inhibiting vascular smooth muscle cell (VSMC) proliferation through cell cycle arrest. The ubiquitin-conjugating enzyme UbcH10 is responsible for ubiquitinating cell cycle proteins for proper exit from mitosis. We hypothesize that NO prevents VSMC proliferation, and hence neointimal hyperplasia, by decreasing levels of UbcH10. Western blotting and immunofluorescent staining showed that NO reduced UbcH10 levels in a concentration-dependent manner in VSMC harvested from the abdominal aortas of Sprague-Dawley rats. Treatment with NO or siRNA to UbcH10 decreased both UbcH10 levels and VSMC proliferation (P<0.001), while increasing UbcH10 levels by plasmid transfection or angiotensin II stimulation increased VSMC proliferation to 150% (P=0.008) and 212% (P=0.002) of control, respectively. Immunofluorescent staining of balloon-injured rat carotid arteries showed a ~4-fold increase in UbcH10 levels, which was profoundly decreased following treatment with NO. Western blotting of carotid artery lysates showed no UbcH10 in uninjured vessels, a substantial increase in the injury alone group, and a significant decrease in the injury+NO group (~3-fold reduction versus injury alone). Importantly, in vitro and in vivo, a marked increase in polyubiquitinated UbcH10 was observed in the NO-treated VSMC and carotid arteries, respectively, indicating that NO may be decreasing unmodified UbcH10 levels by increasing its ubiquitination. Central to our hypothesis, we report that NO decreases UbcH10 levels in VSMC in vitro and following arterial injury in vivo in association with increasing polyubiquitinated-UbcH10 levels. These changes in UbcH10 levels correlate with VSMC proliferation and neointimal hyperplasia, making UbcH10 a promising therapeutic target for inhibiting this proliferative disease.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Túnica Íntima/efectos de los fármacos , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Western Blotting , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/prevención & control , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hiperplasia , Masculino , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Neointima/metabolismo , Neointima/prevención & control , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Triazenos/metabolismo , Triazenos/farmacología , Túnica Íntima/metabolismo , Túnica Íntima/patología , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinación/efectos de los fármacos
10.
Mol Cancer Ther ; 9(4): 869-82, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20354119

RESUMEN

To monitor the subcellular distribution of mixed epidermal growth factor (EGF) receptor (EGFR)-DNA targeting drugs termed combi-molecules, we designed AL237, a fluorescent prototype, to degrade into a green fluorescent DNA damaging species and FD105, a blue fluorescent EGFR inhibitor. Here we showed that AL237 damaged DNA in the 12.5 to 50 mumol/L range. Despite its size, it blocked EGFR phosphorylation in an enzyme assay (IC(50) = 0.27 mumol/L) and in MDA-MB468 breast cancer cells in the same concentration range as for DNA damage. This translated into inhibition of extracellular signal-regulated kinase 1/2 or BAD phosphorylation and downregulation of DNA repair proteins (XRCC1, ERCC1). Having shown that AL237 was a balanced EGFR-DNA targeting molecule, it was used as an imaging probe to show that (a) green and blue colors were primarily colocalized in the perinuclear and partially in the nucleus in EGFR- or ErbB2-expressing cells, (b) the blue fluorescence associated with FD105, but not the green, was colocalized with anti-EGFR red-labeled antibody, (c) the green fluorescence of nuclei was significantly more intense in NIH 3T3 cells expressing EGFR or ErbB2 than in their wild-type counterparts (P < 0.05). Similarly, the growth inhibitory potency of AL237 was selectively stronger in the transfectants. In summary, the results suggest that AL237 diffuses into the cells and localizes abundantly in the perinuclear region and partially in the nucleus where it degrades into EGFR and DNA targeting species. This bystander-like effect translates into high levels of DNA damage in the nucleus. Sufficient quinazoline levels are released in the cells to block EGF-induced activation of downstream signaling. Mol Cancer Ther; 9(4); 869-82. (c)2010 AACR.


Asunto(s)
Daño del ADN , Compuestos de Dansilo/metabolismo , Compuestos de Dansilo/farmacología , Receptores ErbB/antagonistas & inhibidores , Colorantes Fluorescentes/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Triazenos/metabolismo , Triazenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Compuestos de Dansilo/química , Humanos , Ratones , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Transfección , Triazenos/química
11.
J Pharmacol Exp Ther ; 334(1): 9-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20348204

RESUMEN

Within the context of a new tumor-targeting strategy termed "combi-targeting," we designed RB24 to inhibit epidermal growth factor receptor (EGFR) or Her2 phosphorylation and to further degrade upon hydrolysis to 4-(3'-bromophenylamino)-6-aminoquinazoline (RB10; another EGFR/Her2 inhibitor) plus a strong DNA-alkylating species. 6-(3-Acetoxymethyl-3-methyltriazenyl)-4-(3'-bromophenylamino)quinazoline (RB24) showed significant antiproliferative activity against human breast cancer cells, and transfection of one such cell line, MDA-MB-435, with ErbB1 or ErbB2 (Her2) dramatically enhanced cell death by apoptosis. RB24 was capable of releasing 2- to 3-fold higher levels of RB10 in the transfectants than in their wild-type counterparts. More importantly, RB10 was abundantly distributed in the perinuclear region of the cells, and its elevated levels in the ErbB transfectants were concomitant with increased levels of DNA lesions in the latter cells. This selectivity could be abolished by coincubation of the cells with exogenous RB10, suggesting that the entire combi-molecule may bind primarily to its cognate perinuclear sites before degradation. This localization may exert a bystander effect, allowing the alkylating species to be abundantly propagated into the nucleus. Cell response to this novel targeting mechanism was mediated by 1) activation of c-Jun NH(2)-terminal kinase in response to DNA damage and 2) down-regulation of Bad through blockade of EGFR tyrosine kinase activity: two events that cooperatively converged into enhancement of apoptosis in the oncogene-transfected cells.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN , Receptores ErbB/antagonistas & inhibidores , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Triazenos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Receptores ErbB/biosíntesis , Citometría de Flujo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Microscopía Fluorescente , Estructura Molecular , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Quinazolinas/química , Quinazolinas/metabolismo , Receptor ErbB-2/biosíntesis , Transfección , Triazenos/química , Triazenos/metabolismo , Proteína Letal Asociada a bcl/metabolismo
12.
Anticancer Drugs ; 20(8): 659-67, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19581798

RESUMEN

ZRBA1 is a molecule termed 'combi-molecule' designed to induce DNA-alkylating lesions and to block epidermal growth factor receptor (EGFR) tyrosine kinase. Owing to its ability to downregulate the EGFR tyrosine kinase-mediated antiapoptotic signaling and DNA repair proteins, we inferred that it could significantly sensitize cells to ionizing radiation. Using the MDA-MB-468 human breast cancer cell line in which ZRBA1 has already been reported to induce significant EGFR/DNA-targeting potency, the results showed that: (i) concurrent administration of ZRBA1 and 4 Gy radiation led to a significant decrease in cell viability, (ii) the greater efficacy of the combination was sequential, being limited to conditions wherein the drug was administered concurrently with radiation or before radiation, and (iii) the efficacy enhancement of the combination was further confirmed by clonogenic assays from which a dose enhancement factor of 1.34 could be observed at survival fraction of 0.01. Flow cytometric analysis showed significant enhancement of cell cycle arrest in G2/M (P<0.046, irradiated cells vs. cells treated with ZRBA1 and radiation) and increased apoptosis when ZRBA1 was combined with radiation. Likewise, significant levels of double-strand breaks were observed for the combination, as determined by neutral comet assay (P<0.045, irradiated cells vs. cells treated with ZRBA1 and radiation). These results in toto suggest that the superior efficacy of the ZRBA1 plus radiation combination may be secondary to the ability of ZRBA1 to arrest the cells in G2/M, a cell cycle phase in which tumor cells are sensitive to radiation. Furthermore, the increased levels of DNA damage, combined with the concomitant downregulation of EGFR-mediated signaling by ZRBA1, may account for the significant levels of cell killing induced by the combination.


Asunto(s)
Quinazolinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Triazenos/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/efectos de la radiación , Receptores ErbB/antagonistas & inhibidores , Gefitinib , Humanos , Microscopía Fluorescente , Quinazolinas/metabolismo , Quinazolinas/farmacocinética , Quinazolinas/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/efectos de la radiación , Triazenos/metabolismo , Triazenos/farmacocinética , Triazenos/efectos de la radiación , Ensayo de Tumor de Célula Madre , Rayos X
13.
Eur J Med Chem ; 44(8): 3228-34, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19386398

RESUMEN

A range of triazene derivatives were synthesized and investigated as prodrug candidates for melanocyte-directed enzyme prodrug therapy (MDEPT). The prodrugs contained a tyramine or dopamine promoiety required for tyrosinase activation and this was joined via a urea functional group to the cytotoxic triazene. The stability of each of the prodrugs in phosphate buffer, human plasma and in mushroom tyrosinase is discussed. The identification of the main peak formed after the tyrosinase reaction was attempted by LC-MS and the conversion of prodrug to the quinone was confirmed.


Asunto(s)
Dopamina/análogos & derivados , Monofenol Monooxigenasa/metabolismo , Profármacos/química , Profármacos/metabolismo , Triazenos/química , Triazenos/metabolismo , Tiramina/análogos & derivados , Agaricales/enzimología , Alquilación , Diseño de Fármacos , Estabilidad de Medicamentos , Activación Enzimática , Humanos , Profármacos/síntesis química , Triazenos/sangre , Triazenos/síntesis química
14.
Drug Metab Lett ; 3(1): 1-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19356110

RESUMEN

Combi-molecules are novel agents designed to be hydrolyzed into two bioactive species: an epidermal growth factor receptor (EGFR) tyrosine kinase (TK) inhibitor + a DNA alkylating agent. With the purpose of enhancing the tumour concentration of the bioactive species, we synthesized and compared the activities of RB107, a quinazolinotriazene designed to generate the bioactive BJ2000 upon hydrolysis, ZRDM and RB107ZR that require metabolic activation to generate BJ2000. The results showed that RB107 released the highest level of BJ2000 and its degradation product FD105 in vivo and high levels of the DNA alkylating methyl diazonium ion in the brain, kidney, liver and the DU145 tumours as confirmed by (14)C-labeling. The results in toto suggest that RB107 was stable enough to deliver the bioactive species to the tumour site and for optimal tumour distribution of the bioactive species, combi-molecules of the triazene class must be designed to be primarily degraded by hydrolytic cleavage and not by metabolic activation.


Asunto(s)
Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Receptores ErbB/metabolismo , Triazenos/metabolismo , Triazenos/farmacocinética , Animales , Antineoplásicos Alquilantes/uso terapéutico , Biotransformación , Cromatografía Líquida de Alta Presión , Receptores ErbB/antagonistas & inhibidores , Humanos , Masculino , Espectrometría de Masas , Ratones , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacocinética , Espectrofotometría Ultravioleta , Distribución Tisular , Triazenos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Mol Neurosci ; 33(3): 268-77, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17952636

RESUMEN

Nerve growth factor (NGF)-deprivation or axotomy of dorsal root ganglion (DRG) neurons causes stress, which they cope by triggering various mechanisms. Among several molecular changes, in the present study, we demonstrate preprotachykinin-A-substance P (PPTA-SP) and activity-dependent neuroprotective protein-vasoactive intestinal peptide (ADNP-VIP) expression pattern using DRG neurons-Schwann cells coculture and axotomy model. In the presence of NGF, DRG cultures showed high levels of PPTA and ADNP mRNA expression, which were significantly suppressed in the absence of NGF and/or nitric oxide synthase (NOS) inhibition by NG-nitro-L-arginine methyl ester (L-NAME), suggesting that both NGF and nitric oxide (NO) can regulate PPTA and ADNP expression. However, treating coculture with NO donor, diethylenetriamine nitric oxide (DETA-NO) did not increase PPTA and ADNP expression in the presence or absence of NGF, although there was a marginal increase in ADNP expression in the absence of NGF. NGF-deprivation increases endogenous NO; thus, DETA-NO had no further effect on PPTA and ADNP expression. Alternatively, NGF produced from NO-stimulated Schwann cells influence gene expression. In addition, interestingly, DETA-NO treatment of Schwann cells alone suppresses both PPTA and ADNP, suggesting differential response of DRG neurons-Schwann cells coculture to DETA-NO. SP and ADNP immunostaining of axotomized DRGs revealed significant reduction in SP and ADNP compared to intact DRG, which was partially recovered in neuronal NOS blocker, 7-nitroindazole (7-NI)-treated DRGs, particularly intense ADNP staining in satellite glia. As ADNP is VIP-responsive gene, we further explored VIP expression in DRGs. Axotomy increased VIP in DRG neurons, but 7-NI treatment caused intense VIP staining in satellite glia. These observations suggest a complex interaction of NO-NGF with PPTA/SP and ADNP-VIP in neuron-glial communication when neurons are stressed.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico/metabolismo , Sistema Nervioso Periférico/fisiología , Precursores de Proteínas/metabolismo , Sustancia P/metabolismo , Taquicininas/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Axotomía , Células Cultivadas , Ganglios Espinales/citología , Proteínas de Homeodominio/genética , NG-Nitroarginina Metil Éster/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Precursores de Proteínas/genética , Ratas , Sustancia P/genética , Taquicininas/genética , Triazenos/metabolismo , Péptido Intestinal Vasoactivo/genética
16.
Free Radic Biol Med ; 40(6): 1028-33, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540398

RESUMEN

Nitric oxide (NO) is an important regulator of angiogenesis and neovascularization. The nature of endothelial cell motility responses to NO was examined using a Boyden chamber method. NO generated via decomposition of either DEA/NO or DETA/NO produced increases in human umbilical vein endothelial cell (HUVEC) chemotaxis, which were completely abrogated by ODQ, a soluble guanylyl cyclase inhibitor. Measurements of NO either directly by chemiluminescence or its chemistry with diaminofluorescein revealed that chemotaxis was driven by subtle NO gradients between the lower and the upper wells in this system. In addition to diffusion and volatilization from the upper chambers, the data showed that HUVEC consumption of NO contributed to these sustained gradients. Comparison of DEA/NO- and DETA/NO-mediated responses suggested that the persistence of spatial NO gradients is as significant as the absolute magnitude of NO exposure per unit time. The findings suggest that subnanomolar NO gradients are sufficient to mobilize endothelial cell migration into hypoxic tissue during neovascularization events, such as in wound healing and cancer.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Guanilato Ciclasa/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Difusión , Células Endoteliales/fisiología , Endotelio Vascular/citología , Humanos , Hidrazinas/metabolismo , Neovascularización Fisiológica , Óxido Nítrico/farmacología , Óxidos de Nitrógeno/metabolismo , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Triazenos/metabolismo , Volatilización
17.
J Biol Chem ; 281(2): 945-50, 2006 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-16263710

RESUMEN

Long chain acyl-CoA synthetase (ACSL) catalyzes the initial step in long chain fatty acid metabolism. Of the five mammalian ACSL isoforms cloned and characterized, ACSL5 is the only isoform found to be located, in part, on mitochondria and thus was hypothesized to be involved in fatty acid oxidation. To elucidate the specific roles of ACSL5 in fatty acid metabolism, we used adenoviral-mediated overexpression of ACSL5 (Ad-ACSL5) in rat hepatoma McArdle-RH7777 cells. Confocal microscopy revealed that Ad-ACSL5 colocalized to both mitochondria and endoplasmic reticulum. When compared with cells infected with Ad-GFP, Ad-ACSL5-infected cells at 24 h after infection had 2-fold higher acyl-CoA synthetase activities and 30% higher rates of fatty acid uptake when incubated with 500 microM [1-(14)C]oleic acid. Metabolism of [1-(14)C]oleic acid to cellular triacylglycerol (TAG) increased 42% in Ad-ACSL5-infected cells, but when compared with control cells, metabolism to acid-soluble metabolites, phospholipids, and medium TAG did not differ substantially. The incorporation of [1-(14)C]oleate and [1,2,3-(3)H]glycerol into TAG was similar in Ad-ACSL5-infected cells, thus indicating that Ad-ACSL5 increased TAG synthesis through both de novo and reacylation pathways. However, [1-(14)C]acetic acid incorporation into cellular lipids showed that, when compared with control cells, Ad-ACSL5-infected cells did not increase the metabolism of fatty acids that were derived from de novo synthesis. These results suggest that uptake of fatty acids into cells is regulated by metabolism and that overexpressed ACSL5 partitions exogenously derived fatty acids toward TAG synthesis and storage.


Asunto(s)
Coenzima A Ligasas/genética , Coenzima A Ligasas/fisiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Triglicéridos/metabolismo , Ácido Acético/química , Adenoviridae/genética , Animales , Carcinoma Hepatocelular/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Coenzima A Ligasas/antagonistas & inhibidores , Coenzima A Ligasas/química , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Lípidos/química , Hígado/metabolismo , Masculino , Microscopía Confocal , Ácido Oléico/química , Ácido Oléico/metabolismo , Fosfolípidos/química , Isoformas de Proteínas , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Triazenos/química , Triazenos/metabolismo , Triglicéridos/química
18.
Am J Physiol Cell Physiol ; 290(4): C1263-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16354758

RESUMEN

Recent data support the hypothesis that reactive oxygen species (ROS) play a central role in the initiation and progression of vascular diseases. An important vasoprotective function related to the regulation of ROS levels appears to be the antioxidant capacity of nitric oxide (NO). We previously reported that treatment with NO decreases phosphotyrosine levels of adapter protein p130(cas) by increasing protein tyrosine phosphatase-proline, glutamate, serine, and threonine sequence protein (PTP-PEST) activity, which leads to the suppression of agonist-induced H(2)O(2) elevation and motility in cultured rat aortic smooth muscle cells (SMCs). The present study was performed to investigate the hypotheses that 1) IGF-I increases the activity of the small GTPase Rac1 as well as H(2)O(2) levels and 2) NO suppresses IGF-I-induced H(2)O(2) elevation by decreasing Rac1 activity via increased PTP-PEST activity and dephosphorylation of p130(cas). We report that IGF-I induces phosphorylation of p130(cas) and activation of Rac1 and that NO attenuates these effects. The effects of NO are mimicked by the overexpression of PTP-PEST or dominant-negative (dn)-p130(cas) and antagonized by the expression of dn-PTP-PEST or p130(cas). We conclude that IGF-I induces rat aortic SMC motility by increasing phosphotyrosine levels of p130(cas) and activating Rac1 and that NO decreases motility by activating PTP-PEST, inducing dephosphorylating p130(cas), and decreasing Rac1 activity. Decreased Rac1 activity lowers intracellular H(2)O(2) levels, thus attenuating cell motility.


Asunto(s)
Proteína Sustrato Asociada a CrK/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Óxido Nítrico/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Aorta/citología , Movimiento Celular , Activación Enzimática , Femenino , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Donantes de Óxido Nítrico/metabolismo , Oxidantes/metabolismo , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12 , Ratas , Ratas Sprague-Dawley , Triazenos/metabolismo
19.
Mol Microbiol ; 58(1): 177-88, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16164557

RESUMEN

Nitric oxide (NO) is an important host defence molecule that varies its immune stimulatory effects depending on the concentrations at which it is produced, with low concentrations (< 1 microM) promoting an anti-inflammatory host response while higher concentrations (>1 microM) lead to inflammatory responses. Neisseria gonorrhoeae grows anaerobically by anaerobic respiration using nitrite reductase (Nir) to convert nitrite to NO and nitric oxide reductase (Nor) to convert NO to nitrous oxide. As N. gonorrhoeae can both produce and degrade NO, we have begun a study of NO metabolism in this bacterium to understand how gonococcal manipulation of NO concentration may influence the inflammatory response during infection. N. gonorrhoeae has an apparent Nir Km of 33 microM nitrite and an apparent Nor Km of 1.2 microM NO. The maximum specific activities for Nir and Nor were 135 nmoles nitrite reduced per minute per OD600 (pH 6.7) and 270 nmoles NO reduced per minute per OD600 (pH 7.5) respectively. N. gonorrhoeae established a steady-state concentration of NO after nitrite addition that was dependent on the nitrite concentration until saturation at 1 mM nitrite. The NO steady-state level decreased as pH increased, and the ratio of activities of Nir and Nor correlated to the NO steady-state level. When the NO donor DETA/NO was used to simulate host NO production, N. gonorrhoeae also established a NO steady-state level. The concentration of NO at steady state was found to be a function of the concentration of NO generated by DETA/NO, with N. gonorrhoeae reducing the NO from proinflammatory (>1 microM) to anti-inflammatory (approximately 100 nM) concentrations. The implications of the ability of N. gonorrhoeae to maintain an anti-inflammatory NO concentration is discussed in relation to asymptomatic infection in women.


Asunto(s)
Neisseria gonorrhoeae/metabolismo , Óxido Nítrico/metabolismo , Anaerobiosis , Femenino , Gonorrea/microbiología , Gonorrea/patología , Humanos , Concentración de Iones de Hidrógeno , Neisseria gonorrhoeae/genética , Óxido Nítrico/análisis , Nitrito Reductasas/metabolismo , Nitritos/metabolismo , Óxido Nitroso/metabolismo , Oxidorreductasas/metabolismo , Triazenos/metabolismo
20.
Leuk Res ; 29(6): 693-700, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15863211

RESUMEN

Recently, within the framework of a new strategy termed "combi-targeting," we designed ZRCM5 to contain a 2-phenylaminopyrimidopyridine moiety targeted to bcr-abl kinase and a triazene tail capable of generating a methyldiazonium species upon hydrolysis. The ability of ZRCM5 to block tyrosine kinase activity was tested in a short 10 min exposure ELISA involving isolated bcr-abl kinase and Western blotting assays. The results showed that: (a) ZRCM5 was hydrolyzed with a half-life of 27 min in cell culture media, (b) it blocked bcr-abl autophosphorylation in promyeloblastic leukemia K562 cells in a dose-dependent manner (IC(50)=14.01 microM) and (c) it induced dose-dependent levels of DNA strand breaks. In contrast, temozolomide (TEM), a clinical DNA damaging triazene capable of generating, like ZRCM5, a methyldiazonium species, could neither block bcr-abl tyrosine kinase activity in isolated enzyme nor in whole cell autophosphorylation assays. In cells expressing varied levels of bcr-abl, ZRCM5 was consistently more potent than TEM. The significant potency of ZRCM5 against the leukemia cells was attributed to its ability to simultaneously to block bcr-abl and related DNA repair activity while inducing significant DNA lesions in bcr-abl expressing leukemia cells. Further studies are ongoing to increase the affinity of ZRCM5 with the purpose of further enhancing its potency in bcr-abl expressing cells.


Asunto(s)
Dacarbazina/análogos & derivados , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Leucemia/enzimología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Triazenos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo/química , Daño del ADN , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Proteínas de Fusión bcr-abl , Humanos , Hidrólisis , Células K562 , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Proteínas Tirosina Quinasas/biosíntesis , Temozolomida , Factores de Tiempo , Triazenos/metabolismo , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...