Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 2): 185-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25664730

RESUMEN

Lactate dehydrogenase (LDH) is an essential metabolic enzyme that catalyzes the interconversion of pyruvate and lactate using NADH/NAD(+) as a co-substrate. Many cancer cells exhibit a glycolytic phenotype known as the Warburg effect, in which elevated LDH levels enhance the conversion of glucose to lactate, making LDH an attractive therapeutic target for oncology. Two known inhibitors of the human muscle LDH isoform, LDHA, designated 1 and 2, were selected, and their IC50 values were determined to be 14.4 ± 3.77 and 2.20 ± 0.15 µM, respectively. The X-ray crystal structures of LDHA in complex with each inhibitor were determined; both inhibitors bind to a site overlapping with the NADH-binding site. Further, an apo LDHA crystal structure solved in a new space group is reported, as well as a complex with both NADH and the substrate analogue oxalate bound in seven of the eight molecules and an oxalate only bound in the eighth molecule in the asymmetric unit. In this latter structure, a kanamycin molecule is located in the inhibitor-binding site, thereby blocking NADH binding. These structures provide insights into LDHA enzyme mechanism and inhibition and a framework for structure-assisted drug design that may contribute to new cancer therapies.


Asunto(s)
L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/química , Neoplasias/enzimología , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Simulación del Acoplamiento Molecular , NAD/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Ácido Oxálico/metabolismo , Conformación Proteica
2.
Bioorg Med Chem ; 21(16): 4839-45, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23816041

RESUMEN

Certain bacterial pathogens possess a repertoire of carbohydrate processing enzymes that process host N-linked glycans and many of these enzymes are required for full virulence of harmful human pathogens such as Clostridium perfringens and Streptococcus pneumoniae. One bacterial carbohydrate processing enzyme that has been studied is the pneumococcal virulence factor SpGH125 from S. pneumoniae and its homologue, CpGH125, from C. perfringens. These exo-α-1,6-mannosidases from glycoside hydrolase family 125 show poor activity toward aryl α-mannopyranosides. To circumvent this problem, we describe a convenient synthesis of the fluorogenic disaccharide substrate 4-methylumbelliferone α-d-mannopyranosyl-(1→6)-ß-d-mannopyranoside. We show this substrate can be used in a coupled fluorescent assay by using ß-mannosidases from either Cellulomonas fimi or Helix pomatia as the coupling enzyme. We find that this disaccharide substrate is processed much more efficiently than aryl α-mannopyranosides by CpGH125, most likely because inclusion of the second mannose residue makes this substrate more like the natural host glycan substrates of this enzyme, which enables it to bind better. Using this sensitive coupled assay, the detailed characterization of these metal-independent exo-α-mannosidases GH125 enzymes should be possible, as should screening chemical libraries for inhibitors of these virulence factors.


Asunto(s)
Disacáridos/síntesis química , Umbeliferonas/síntesis química , alfa-Manosidasa/metabolismo , Clostridium perfringens/enzimología , Disacáridos/química , Disacáridos/metabolismo , Pruebas de Enzimas , Colorantes Fluorescentes/química , Cinética , Streptococcus pneumoniae/enzimología , Especificidad por Sustrato , Umbeliferonas/química , Umbeliferonas/metabolismo , alfa-Manosidasa/química
3.
Bioorg Med Chem Lett ; 21(23): 6950-4, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22033460

RESUMEN

A series of CCR5 antagonists were optimized for potent inhibition of R5 HIV-1 replication in peripheral blood mononuclear cells. Compounds that met acceptable ADME criteria, selectivity, human plasma protein binding, potency shift in the presence of α-glycoprotein were evaluated in rat and dog pharmacokinetics.


Asunto(s)
Amidas/síntesis química , Fármacos Anti-VIH/síntesis química , Antagonistas de los Receptores CCR5 , Diseño de Fármacos , VIH-1 , Leucocitos Mononucleares , Amidas/química , Amidas/farmacología , Animales , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Perros , Humanos , Concentración 50 Inhibidora , Leucocitos Mononucleares/efectos de los fármacos , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Piperidinas/farmacología , Piridinas/síntesis química , Piridinas/química , Piridinas/farmacología , Ratas , Replicación Viral/efectos de los fármacos
4.
J Med Chem ; 49(17): 5262-72, 2006 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-16913715

RESUMEN

The synthesis of four new oxorhenium(V) complexes containing the "3 + 1" mixed-ligand donor set, ReO(SYS)X (where Y = S, py; X = Cl, Br), is described. All of the complexes tested exhibited selectivity for cathepsin B over K. Most notably, compound 6, ReO(SSS-2,2')Br (IC50(cathepsin B) = 1.0 nM), was 260 times more potent against cathepsin B. It was also discovered that complexes containing the same tridentate (SSS) ligand were more potent when the leaving group was bromide versus chloride (e.g., IC50(cathepsin B): ReO(SSS-2,2')Cl (4), 8.8 nM; ReO(SSS-2,2')Br (6), 1.0 nM). Mechanistic studies with cathepsin B showed that both compounds 2 (ReO(SpyS)(SPhOMe-p)) and 4 were active-site-directed. Compound 2 was determined to be a tight-binding, reversible inhibitor, while compound 4 was a time-dependent, slowly reversible inhibitor. The results described in this paper show that the oxorhenium(V) "3 + 1" complexes are potent, selective inhibitors of cathepsin B and have potential for the treatment of cancer.


Asunto(s)
Catepsina B/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/farmacología , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/farmacología , Renio/química , Sitios de Unión , Catepsina B/química , Catepsina K , Catepsinas/antagonistas & inhibidores , Catepsinas/química , Inhibidores de Cisteína Proteinasa/química , Humanos , Ligandos , Estructura Molecular , Compuestos Organometálicos/química , Estereoisomerismo , Relación Estructura-Actividad , Factores de Tiempo
5.
Biochem Pharmacol ; 72(5): 588-96, 2006 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-16815309

RESUMEN

The chemokine receptor CXCR4 is widely expressed on different cell types, is involved in leukocyte chemotaxis, and is a co-receptor for HIV. AMD3100 has been shown to be a CXCR4 receptor antagonist, and to block HIV infection of T-tropic, X4-using, virus in vitro and in vivo. AMD3100 is an effective mobilizer of hematopoietic stem cells and is being investigated in clinical trials in multiple myeloma and non-Hodgkins lymphoma patients. Using the CCRF-CEM T-cell line that constitutively expresses CXCR4 we confirmed that AMD3100 was an antagonist of SDF-1/CXCL12 ligand binding (IC50=651+/-37 nM). We have also shown that AMD3100 inhibits SDF-1 mediated GTP-binding (IC50=27+/-2.2 nM), SDF-1 mediated calcium flux (IC50=572+/-190 nM), and SDF-1 stimulated chemotaxis (IC50=51+/-17 nM). AMD3100 did not inhibit calcium flux against cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7 when stimulated with their cognate ligands, nor did it inhibit receptor binding of LTB4. AMD3100 did not, on its own, induce a calcium flux in the CCRF-CEM cells, which express multiple GPCRs including CXCR4, CCR4 and CCR7. Furthermore, AMD3100 neither stimulated GTP-binding, an assay for GPCR activation, in CEM cell membranes; nor chemotaxis of CCRF-CEM cells. These data therefore demonstrate that AMD3100 is a specific antagonist of CXCR4, is not cross-reactive with other chemokine receptors, and is not an agonist of CXCR4.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Bencilaminas , Calcio/metabolismo , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiotaxis/efectos de los fármacos , Ciclamas , Humanos , Unión Proteica , Transducción de Señal/efectos de los fármacos
6.
Assay Drug Dev Technol ; 3(6): 637-48, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16438659

RESUMEN

Chemokine receptors have been implicated in several disease processes such as acute and chronic inflammation, cancer, and allograft rejection and are therefore targets for drug development. The chemokine receptors CCR5 and CXCR4 are of particular interest as they serve as entry cofactors for human immunodeficiency virus. These receptors are members of the G protein-coupled receptor (GPCR) family. In this respect, assessing GPCR activation by GTP binding is an important tool to study the early stage of signal transduction. The assay normally utilizes the non-hydrolysable GTP analogue guanosine 5'-gamma-[35S]thiotriphosphate. In order to avoid the problems involved in working with radioactivity, a new non-radioactive version of the assay was developed using a europium-labeled GTP analogue in which europium-GTP binding can be assayed using time-resolved fluorescence. The assay was optimized for CXCR4 and CCR5 and validated for screening of chemokine antagonists using the small molecule CXCR4 antagonist AMD3100 and CCR5 antagonists.


Asunto(s)
Antagonistas de los Receptores CCR5 , Europio , Guanosina Trifosfato/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Fármacos Anti-VIH/farmacología , Bencilaminas , Calcio/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Quimiocinas/antagonistas & inhibidores , Quimiocinas/metabolismo , Ciclamas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Fluorescencia , Guanosina Difosfato , Guanosina Trifosfato/análogos & derivados , Compuestos Heterocíclicos/farmacología , Humanos , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Reproducibilidad de los Resultados , Saponinas , Transducción de Señal , Cloruro de Sodio , Temperatura , Factores de Tiempo
7.
J Med Chem ; 56(20): 8049-65, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24090135

RESUMEN

The redesign of the previously reported thiophene-3-yl-methyl urea series, as a result of potential cardiotoxicity, was successfully accomplished, resulting in the identification of a novel potent series of CCR5 antagonists containing the imidazolidinylpiperidinyl scaffold. The main redesign criteria were to reduce the number of rotatable bonds and to maintain an acceptable lipophilicity to mitigate hERG inhibition. The structure-activity relationship (SAR) that was developed was used to identify compounds with the best pharmacological profile to inhibit HIV-1. As a result, five advanced compounds, 6d, 6e, 6i, 6h, and 6k, were further evaluated for receptor selectivity, antiviral activity against CCR5 using (R5) HIV-1 clinical isolates, and in vitro and in vivo safety. On the basis of these results, 6d and 6h were selected for further development.


Asunto(s)
Fármacos Anti-VIH/farmacología , Benzoatos/farmacología , Antagonistas de los Receptores CCR5 , Replicación Viral/efectos de los fármacos , Animales , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Benzoatos/síntesis química , Benzoatos/química , Células CHO , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Diseño de Fármacos , Células HEK293 , VIH-1/efectos de los fármacos , VIH-1/fisiología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Imidazoles/síntesis química , Imidazoles/química , Imidazoles/farmacología , Imidazolidinas/química , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Modelos Químicos , Estructura Molecular , Piperidinas/química , Receptores CCR5/genética , Receptores CCR5/metabolismo , Relación Estructura-Actividad
8.
ACS Med Chem Lett ; 3(3): 216-21, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-24900457

RESUMEN

A series of CCR5 antagonists representing the thiophene-3-yl-methyl ureas were designed that met the pharmacological criteria for HIV-1 inhibition and mitigated a human ether-a-go-go related gene (hERG) inhibition liability. Reducing lipophilicity was the main design criteria used to identify compounds that did not inhibit the hERG channel, but subtle structural modifications were also important. Interestingly, within this series, compounds with low hERG inhibition prolonged the action potential duration (APD) in dog Purkinje fibers, suggesting a mixed effect on cardiac ion channels.

9.
Biochem Pharmacol ; 83(4): 472-9, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22146583

RESUMEN

In order to enter and infect human cells HIV must bind to CD4 in addition to either the CXCR4 or the CCR5 chemokine receptor. AMD11070 was the first orally available small molecule antagonist of CXCR4 to enter the clinic. Herein we report the molecular pharmacology of AMD11070 which is a potent inhibitor of X4 HIV-1 replication and the gp120/CXCR4 interaction. Using the CCRF-CEM T cell line that endogenously expresses CXCR4 we have demonstrated that AMD11070 is an antagonist of SDF-1α ligand binding (IC50 = 12.5 ± 1.3 nM), inhibits SDF-1 mediated calcium flux (IC50 = 9.0 ± 2.0 nM) and SDF-1α mediated activation of the CXCR4 receptor as measured by a Eu-GTP binding assay (IC50 =39.8 ± 2.5 nM) or a [(35)S]-GTPγS binding assay (IC50 =19.0 ± 4.1 nM), and inhibits SDF-1α stimulated chemotaxis (IC50 =19.0 ± 4.0 nM). AMD11070 does not inhibit calcium flux of cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7, or ligand binding to CXCR7 and BLT1, demonstrating selectivity for CXCR4. In addition AMD11070 is able to inhibit the SDF-1ß isoform interactions with CXCR4; and N-terminal truncated variants of CXCR4 with equal potency to wild type receptor. Further mechanistic studies indicate that AMD11070 is an allosteric inhibitor of CXCR4.


Asunto(s)
Aminoquinolinas/farmacología , Aminoquinolinas/farmacocinética , Bencimidazoles/farmacología , Bencimidazoles/farmacocinética , VIH-1/efectos de los fármacos , Receptores CXCR4/metabolismo , Internalización del Virus/efectos de los fármacos , Administración Oral , Aminoquinolinas/administración & dosificación , Animales , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/farmacología , Bencimidazoles/administración & dosificación , Disponibilidad Biológica , Butilaminas , Línea Celular , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Perros , Regulación de la Expresión Génica/efectos de los fármacos , VIH-1/fisiología , Compuestos Heterocíclicos con 1 Anillo , Humanos , Estructura Molecular , Unión Proteica , Receptores CXCR4/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
10.
J Inorg Biochem ; 105(5): 754-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21481817

RESUMEN

Gold(III) compounds have been examined for potential anti-cancer activity. It is proposed that the molecular targets of these compounds are thiol-containing biological molecules such as the cathepsin cysteine proteases. These enzymes have been implicated in many diseases including cancer. The catalytic mechanism of the cathepsin cysteine proteases is dependent upon a cysteine at the active site which is accessible to the interaction of thiophilic metals such as gold. The synthesis and biological activity of square-planar six-membered cycloaurated Au(III) compounds with a pyridinyl-phenyl linked backbone and two monodentate or one bidentate leaving group is described. Gold(III) cycloaurated compounds were able to inhibit both cathepsins B and K. Structure/activity was investigated by modifications to the pyridinyl-phenyl backbone, and leaving groups. Optimal activity was seen with substitution at the 6 position of the pyridine ring. The reversibility of inhibition was tested by reactivation in the presence of cysteine with a bidentate thiosalicylate compound being an irreversible inhibitor. Five compounds were evaluated for in vitro cytotoxicity against a panel of human tumor cell lines. The thiosalicylate compound was tested in vivo against the HT29 human colon tumor xenograft model. A modest decrease in tumor growth was observed compared with the untreated control tumor.


Asunto(s)
Antineoplásicos/química , Catepsina B/antagonistas & inhibidores , Catepsina K/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/química , Oro/química , Animales , Antineoplásicos/farmacología , Catepsina B/química , Catepsina B/metabolismo , Catepsina K/química , Catepsina K/metabolismo , Línea Celular Tumoral , Proteasas de Cisteína/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Masculino , Ratones , Ratones SCID
12.
Biochem Pharmacol ; 78(8): 993-1000, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19540208

RESUMEN

CXCR4 is widely expressed in multiple cell types, and is involved in neonatal development, hematopoiesis, and lymphocyte trafficking and homing. Disruption of the CXCL12/CXCR4 interaction has been implicated in stem cell mobilization. Additionally CXCR4 is a co-receptor for HIV. Selective small molecule antagonists of CXCR4 therefore have therapeutic potential. AMD3465 is an N-pyridinylmethylene monocyclam CXCR4 antagonist which can block infection of T-tropic, CXCR4-using HIV. Using the CCRF-CEM T-cell line which expresses CXCR4 we have demonstrated that AMD3465 is an antagonist of SDF-1 ligand binding (K(i) of 41.7+/-1.2nM), and inhibits SDF-1 mediated signaling as shown by inhibition of GTP binding, calcium flux, and inhibition of chemotaxis. AMD3465 is selective for CXCR4 and does not inhibit chemokine-stimulated calcium flux in cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7, nor does it inhibit binding of LTB(4) to its receptor, BLT1. The pharmacokinetics of AMD3465 was investigated in mice and dogs. Absorption was rapid following subcutaneous administration. AMD3465 was cleared from dog plasma in a biphasic manner with a terminal half-life of 1.56-4.63h. Comparison of exposure to the intravenous and subcutaneous doses indicated 100% bioavailability following subcutaneous administration. AMD3465 caused leukocytosis when administered subcutaneously in mice and dogs, with peak mobilization occurring between 0.5 and 1.5h following subcutaneous dosing in mice and with maximum peak plasma concentration of compound preceding peak mobilization in dogs, indicating that AMD3465 has the potential to mobilize hematopoietic stem cells. These data demonstrate the therapeutic potential for the CXCR4 antagonist AMD3465.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Piridinas/farmacología , Piridinas/farmacocinética , Receptores CXCR4/antagonistas & inhibidores , Absorción , Animales , Área Bajo la Curva , Células CHO , Calcio/análisis , Calcio/metabolismo , Línea Celular , Quimiocina CXCL12/antagonistas & inhibidores , Quimiotaxis/efectos de los fármacos , Cricetinae , Cricetulus , Perros , Relación Dosis-Respuesta a Droga , Fluoresceínas/metabolismo , Colorantes Fluorescentes/metabolismo , Semivida , Humanos , Concentración 50 Inhibidora , Riñón/citología , Leucocitosis/etiología , Masculino , Dosis Máxima Tolerada , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Estructura Molecular , Unión Proteica , Piridinas/efectos adversos , Piridinas/sangre , Piridinas/química , Transfección
13.
J Inorg Biochem ; 102(10): 1839-45, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18684510

RESUMEN

The cysteine proteases of the trypanosomatid parasitic protozoa have been validated as targets for chemotherapy of Chagas' disease and leishmaniasis. Metal complexes of gold, platinum, iridium, palladium, rhodium and osmium have been reported to have activity against a variety of trypanosomatids, but the molecular target of these compounds has not been defined. The activity of gold(III) and palladium(II) cyclometallated complexes, and oxorhenium(V) complexes against mammalian and parasitic cysteine proteases was investigated. All gold(III) complexes (1-6) inhibited cathepsin B with IC(50) values in the range of 0.2-1.4 microM. Of the six palladium compounds, aceto[2,6-bis[(butylthio-kappa S)methyl]phenyl-kappa C]-, (SP-4-3)-palladium(II) (11) was the most potent inhibitor of cathepsin B with an IC(50) of 0.4 microM. A clear structure-activity relationship was observed with the oxorhenium(V) complexes with chloro[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium(V) (16) being the most potent inhibitor of cathepsin B with an IC(50) of 0.009 microM. Six complexes were further tested against the parasite cysteine proteases, cruzain from T. cruzi, and cpB from L. major; the most potent inhibitors were the two rhenium complexes (2(1H)-pyridinethionato-kappa S(2))[2,6-bis[(mercapto-kappa S)methyl]pyridine-kappa N(1)] oxorhenium(V) (15) and chloro[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium(V) (16). The compounds were also evaluated in assays for parasite growth. Two oxorhenium(V) compounds ((p-methoxyphenylthiolato-S)[2,6-bis[(mercapto-kappa S)methyl]pyridine-kappa N(1)] oxorhenium(V) (14) and (methanethiolato)[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium (V) (18)) and the palladium compound 11 inhibited T. cruzi intracellular growth, and compound 11 inhibited promastigote growth in three Leishmania species. In conclusion this preliminary data indicates that metal complexes targeted at parasite cysteine proteases show promise for the treatment of both Chagas' disease and leishmaniasis.


Asunto(s)
Catepsina B/metabolismo , Enfermedad de Chagas/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/farmacología , Leishmania/efectos de los fármacos , Leishmaniasis/tratamiento farmacológico , Metales/farmacología , Tripanocidas/farmacología , Trypanosoma/efectos de los fármacos , Animales , Catepsina B/antagonistas & inhibidores , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Humanos , Compuestos Inorgánicos/química , Compuestos Inorgánicos/farmacología , Metales/uso terapéutico , Tripanocidas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA