Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Anaesth ; 131(4): 745-763, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37567808

RESUMEN

BACKGROUND: Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS: In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS: Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS: These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.


Asunto(s)
Agonismo Inverso de Drogas , Neuralgia , Ratas , Animales , Calidad de Vida , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/uso terapéutico , Neuralgia/tratamiento farmacológico , Fenómenos Electrofisiológicos
2.
Proc Natl Acad Sci U S A ; 113(7): 1778-83, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26831078

RESUMEN

Cancer cells reprogram their metabolism to promote growth and proliferation. The genetic evidence pointing to the importance of the amino acid serine in tumorigenesis is striking. The gene encoding the enzyme 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the first committed step of serine biosynthesis, is overexpressed in tumors and cancer cell lines via focal amplification and nuclear factor erythroid-2-related factor 2 (NRF2)-mediated up-regulation. PHGDH-overexpressing cells are exquisitely sensitive to genetic ablation of the pathway. Here, we report the discovery of a selective small molecule inhibitor of PHGDH, CBR-5884, identified by screening a library of 800,000 drug-like compounds. CBR-5884 inhibited de novo serine synthesis in cancer cells and was selectively toxic to cancer cell lines with high serine biosynthetic activity. Biochemical characterization of the inhibitor revealed that it was a noncompetitive inhibitor that showed a time-dependent onset of inhibition and disrupted the oligomerization state of PHGDH. The identification of a small molecule inhibitor of PHGDH not only enables thorough preclinical evaluation of PHGDH as a target in cancers, but also provides a tool with which to study serine metabolism.


Asunto(s)
Neoplasias/metabolismo , Fosfoglicerato-Deshidrogenasa/antagonistas & inhibidores , Serina/biosíntesis , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias/patología
3.
Molecules ; 24(8)2019 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-31022852

RESUMEN

Determining chemokine receptor CXCR4 expression is significant in multiple diseases due to its role in promoting inflammation, cell migration and tumorigenesis. [68Ga]Pentixafor is a promising ligand for imaging CXCR4 expression in multiple tumor types, but its utility is limited by the physical properties of 68Ga. We screened a library of >200 fluorine-containing structural derivatives of AMD-3465 to identify promising candidates for in vivo imaging of CXCR4 expression by positron emission tomography (PET). Compounds containing fluoroethyltriazoles consistently achieved higher docking scores. Six of these higher scoring compounds were radiolabeled by click chemistry and evaluated in PC3-CXCR4 cells and BALB/c mice bearing bilateral PC3-WT and PC3-CXCR4 xenograft tumors. The apparent CXCR4 affinity of the ligands was relatively low, but tumor uptake was CXCR4-specific. The tumor uptake of [18F]RPS-534 (7.2 ± 0.3 %ID/g) and [18F]RPS-547 (3.1 ± 0.5 %ID/g) at 1 h p.i. was highest, leading to high tumor-to-blood, tumor-to-muscle, and tumor-to-lung ratios. Total cell-associated activity better predicted in vivo tumor uptake than did the docking score or apparent CXCR4 affinity. By this metric, and on the basis of their high yielding radiosynthesis, high tumor uptake, and good contrast to background, [18F]RPS-547, and especially [18F]RPS-534, are promising 18F-labeled candidates for imaging CXCR4 expression.


Asunto(s)
Complejos de Coordinación/administración & dosificación , Imagen Molecular , Péptidos Cíclicos/administración & dosificación , Radiofármacos/administración & dosificación , Receptores CXCR4/genética , Animales , Línea Celular Tumoral , Complejos de Coordinación/química , Radioisótopos de Flúor/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Ratones , Péptidos Cíclicos/química , Tomografía de Emisión de Positrones , Radiofármacos/química , Receptores CXCR4/química , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Proc Natl Acad Sci U S A ; 111(14): E1402-8, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706818

RESUMEN

Accumulation of lipofuscin bisretinoids (LBs) in the retinal pigment epithelium (RPE) is the alleged cause of retinal degeneration in genetic blinding diseases (e.g., Stargardt) and a possible etiological agent for age-related macular degeneration. Currently, there are no approved treatments for these diseases; hence, agents that efficiently remove LBs from RPE would be valuable therapeutic candidates. Here, we show that beta cyclodextrins (ß-CDs) bind LBs and protect them against oxidation. Computer modeling and biochemical data are consistent with the encapsulation of the retinoid arms of LBs within the hydrophobic cavity of ß-CD. Importantly, ß-CD treatment reduced by 73% and 48% the LB content of RPE cell cultures and of eyecups obtained from Abca4-Rdh8 double knock-out (DKO) mice, respectively. Furthermore, intravitreal administration of ß-CDs reduced significantly the content of bisretinoids in the RPE of DKO animals. Thus, our results demonstrate the effectiveness of ß-CDs to complex and remove LB deposits from RPE cells and provide crucial data to develop novel prophylactic approaches for retinal disorders elicited by LBs.


Asunto(s)
Lipofuscina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Retinoides/metabolismo , beta-Ciclodextrinas/metabolismo , Animales , Sitios de Unión , Cromatografía Líquida de Alta Presión , Simulación por Computador , Fluorescencia , Técnicas In Vitro , Lipofuscina/aislamiento & purificación , Ratones , Ratones Noqueados , Oxidación-Reducción , Retinoides/aislamiento & purificación
5.
J Bacteriol ; 197(6): 1040-50, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25561717

RESUMEN

Phenolic glycolipids (PGLs) are polyketide synthase-derived glycolipids unique to pathogenic mycobacteria. PGLs are found in several clinically relevant species, including various Mycobacterium tuberculosis strains, Mycobacterium leprae, and several nontuberculous mycobacterial pathogens, such as M. marinum. Multiple lines of investigation implicate PGLs in virulence, thus underscoring the relevance of a deep understanding of PGL biosynthesis. We report mutational and biochemical studies that interrogate the mechanism by which PGL biosynthetic intermediates (p-hydroxyphenylalkanoates) synthesized by the iterative polyketide synthase Pks15/1 are transferred to the noniterative polyketide synthase PpsA for acyl chain extension in M. marinum. Our findings support a model in which the transfer of the intermediates is dependent on a p-hydroxyphenylalkanoyl-AMP ligase (FadD29) acting as an intermediary between the iterative and the noniterative synthase systems. Our results also establish the p-hydroxyphenylalkanoate extension ability of PpsA, the first-acting enzyme of a multisubunit noniterative polyketide synthase system. Notably, this noniterative system is also loaded with fatty acids by a specific fatty acyl-AMP ligase (FadD26) for biosynthesis of phthiocerol dimycocerosates (PDIMs), which are nonglycosylated lipids structurally related to PGLs. To our knowledge, the partially overlapping PGL and PDIM biosynthetic pathways provide the first example of two distinct, pathway-dedicated acyl-AMP ligases loading the same type I polyketide synthase system with two alternate starter units to produce two structurally different families of metabolites. The studies reported here advance our understanding of the biosynthesis of an important group of mycobacterial glycolipids.


Asunto(s)
Membrana Celular/metabolismo , Glucolípidos/biosíntesis , Mycobacterium marinum/metabolismo , Fenoles/metabolismo , Proteína Transportadora de Acilo/genética , Proteína Transportadora de Acilo/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Membrana Celular/química , Regulación Bacteriana de la Expresión Génica/fisiología , Glucolípidos/química , Glucolípidos/metabolismo , Estructura Molecular , Mutación , Mycobacterium marinum/genética , Fenoles/química , Estructura Terciaria de Proteína , Especificidad de la Especie
6.
Antimicrob Agents Chemother ; 59(10): 6521-38, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239979

RESUMEN

The search for drugs that can kill replicating and nonreplicating Mycobacterium tuberculosis faces practical bottlenecks. Measurement of CFU and discrimination of bacteriostatic from bactericidal activity are costly in compounds, supplies, labor, and time. Testing compounds against M. tuberculosis under conditions that prevent the replication of M. tuberculosis often involves a second phase of the test in which conditions are altered to permit the replication of bacteria that survived the first phase. False-positive determinations of activity against nonreplicating M. tuberculosis may arise from carryover of compounds from the nonreplicating stage of the assay that act in the replicating stage. We mitigate these problems by carrying out a 96-well microplate liquid MIC assay and then transferring an aliquot of each well to a second set of plates in which each well contains agar supplemented with activated charcoal. After 7 to 10 days-about 2 weeks sooner than required to count CFU-fluorometry reveals whether M. tuberculosis bacilli in each well have replicated extensively enough to reduce a resazurin dye added for the final hour. This charcoal agar resazurin assay (CARA) distinguishes between bacterial biomasses in any two wells that differ by 2 to 3 log10 CFU. The CARA thus serves as a pretest and semiquantitative surrogate for longer, more laborious, and expensive CFU-based assays, helps distinguish bactericidal from bacteriostatic activity, and identifies compounds that are active under replicating conditions, nonreplicating conditions, or both. Results for 14 antimycobacterial compounds, including tuberculosis (TB) drugs, revealed that PA-824 (pretomanid) and TMC207 (bedaquiline) are largely bacteriostatic.


Asunto(s)
Antituberculosos/farmacología , Bioensayo , Recuento de Colonia Microbiana/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Agar , Antituberculosos/clasificación , Carbón Orgánico/química , Recuento de Colonia Microbiana/instrumentación , Colorantes/química , Diarilquinolinas/farmacología , Fluorometría , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Nitroimidazoles/farmacología , Oxazinas/química , Xantenos/química
7.
Nature ; 461(7264): 621-6, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19759536

RESUMEN

Many anti-infectives inhibit the synthesis of bacterial proteins, but none selectively inhibits their degradation. Most anti-infectives kill replicating pathogens, but few preferentially kill pathogens that have been forced into a non-replicating state by conditions in the host. To explore these alternative approaches we sought selective inhibitors of the proteasome of Mycobacterium tuberculosis. Given that the proteasome structure is extensively conserved, it is not surprising that inhibitors of all chemical classes tested have blocked both eukaryotic and prokaryotic proteasomes, and no inhibitor has proved substantially more potent on proteasomes of pathogens than of their hosts. Here we show that certain oxathiazol-2-one compounds kill non-replicating M. tuberculosis and act as selective suicide-substrate inhibitors of the M. tuberculosis proteasome by cyclocarbonylating its active site threonine. Major conformational changes protect the inhibitor-enzyme intermediate from hydrolysis, allowing formation of an oxazolidin-2-one and preventing regeneration of active protease. Residues outside the active site whose hydrogen bonds stabilize the critical loop before and after it moves are extensively non-conserved. This may account for the ability of oxathiazol-2-one compounds to inhibit the mycobacterial proteasome potently and irreversibly while largely sparing the human homologue.


Asunto(s)
Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Dominio Catalítico/efectos de los fármacos , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Mycobacterium tuberculosis/crecimiento & desarrollo , Oxazolidinonas/metabolismo , Oxazolidinonas/farmacología , Inhibidores de Proteasas/química , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Carbonilación Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína , Especificidad por Sustrato , Tiazoles/farmacología , Treonina/metabolismo
8.
Proc Natl Acad Sci U S A ; 109(40): 16004-11, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23012453

RESUMEN

Existing drugs are slow to eradicate Mycobacterium tuberculosis (Mtb) in patients and have failed to control tuberculosis globally. One reason may be that host conditions impair Mtb's replication, reducing its sensitivity to most antiinfectives. We devised a high-throughput screen for compounds that kill Mtb when its replication has been halted by reactive nitrogen intermediates (RNIs), acid, hypoxia, and a fatty acid carbon source. At concentrations routinely achieved in human blood, oxyphenbutazone (OPB), an inexpensive anti-inflammatory drug, was selectively mycobactericidal to nonreplicating (NR) Mtb. Its cidal activity depended on mild acid and was augmented by RNIs and fatty acid. Acid and RNIs fostered OPB's 4-hydroxylation. The resultant 4-butyl-4-hydroxy-1-(4-hydroxyphenyl)-2-phenylpyrazolidine-3,5-dione (4-OH-OPB) killed both replicating and NR Mtb, including Mtb resistant to standard drugs. 4-OH-OPB depleted flavins and formed covalent adducts with N-acetyl-cysteine and mycothiol. 4-OH-OPB killed Mtb synergistically with oxidants and several antituberculosis drugs. Thus, conditions that block Mtb's replication modify OPB and enhance its cidal action. Modified OPB kills both replicating and NR Mtb and sensitizes both to host-derived and medicinal antimycobacterial agents.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Oxifenilbutazona/farmacología , Animales , Cromatografía Líquida de Alta Presión , Farmacorresistencia Microbiana/fisiología , Ácidos Grasos/metabolismo , Femenino , Hidroxilación , Espectroscopía de Resonancia Magnética , Ratones , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/fisiología , Oxifenilbutazona/metabolismo , Oxifenilbutazona/farmacocinética , Especies de Nitrógeno Reactivo/metabolismo
9.
Nat Methods ; 9(1): 68-71, 2011 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-22081126

RESUMEN

Fluorescence applications requiring high photostability often depend on the use of solution additives to enhance fluorophore performance. Here we demonstrate that the direct or proximal conjugation of cyclooctatetraene (COT), 4-nitrobenzyl alcohol (NBA) or Trolox to the cyanine fluorophore Cy5 dramatically enhanced fluorophore photostability without otherwise affecting its native spectral characteristics. Such conjugation is a powerful means of improving the robustness of fluorescence-based applications demanding long-lived, nonblinking fluorescence emission.


Asunto(s)
Carbocianinas/química , Colorantes Fluorescentes/química , Animales , Alcoholes Bencílicos/química , Células CHO , Cromanos/química , Cricetinae , Ciclooctanos/química , ADN/química , Estabilidad de Medicamentos , Fluorescencia , Humanos , Cinética , Simulación de Dinámica Molecular , Fotoblanqueo/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Oxígeno Singlete/análisis
10.
J Am Soc Nephrol ; 24(8): 1250-61, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23813215

RESUMEN

Ischemia causes AKI as a result of ATP depletion, and rapid recovery of ATP on reperfusion is important to minimize tissue damage. ATP recovery is often delayed, however, because ischemia destroys the mitochondrial cristae membranes required for mitochondrial ATP synthesis. The mitochondria-targeted compound SS-31 accelerates ATP recovery after ischemia and reduces AKI, but its mechanism of action remains unclear. Here, we used a polarity-sensitive fluorescent analog of SS-31 to demonstrate that SS-31 binds with high affinity to cardiolipin, an anionic phospholipid expressed on the inner mitochondrial membrane that is required for cristae formation. In addition, the SS-31/cardiolipin complex inhibited cytochrome c peroxidase activity, which catalyzes cardiolipin peroxidation and results in mitochondrial damage during ischemia, by protecting its heme iron. Pretreatment of rats with SS-31 protected cristae membranes during renal ischemia and prevented mitochondrial swelling. Prompt recovery of ATP on reperfusion led to rapid repair of ATP-dependent processes, such as restoration of the actin cytoskeleton and cell polarity. Rapid recovery of ATP also inhibited apoptosis, protected tubular barrier function, and mitigated renal dysfunction. In conclusion, SS-31, which is currently in clinical trials for ischemia-reperfusion injury, protects mitochondrial cristae by interacting with cardiolipin on the inner mitochondrial membrane.


Asunto(s)
Cardiolipinas/metabolismo , Citocromo-c Peroxidasa/metabolismo , Isquemia/metabolismo , Mitocondrias/efectos de los fármacos , Oligopéptidos/farmacología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Oligopéptidos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo
11.
J Med Chem ; 67(7): 5473-5501, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38554135

RESUMEN

Proteolysis-Targeting Chimeras (PROTACs) are bifunctional molecules that bring a target protein and an E3 ubiquitin ligase into proximity to append ubiquitin, thus directing target degradation. Although numerous PROTACs have entered clinical trials, their development remains challenging, and their large size can produce poor drug-like properties. To overcome these limitations, we have modified our Coferon platform to generate Combinatorial Ubiquitination REal-time PROteolysis (CURE-PROs). CURE-PROs are small molecule degraders designed to self-assemble through reversible bio-orthogonal linkers to form covalent heterodimers. By modifying known ligands for Cereblon, MDM2, VHL, and BRD with complementary phenylboronic acid and diol/catechol linkers, we have successfully created CURE-PROs that direct degradation of BRD4 both in vitro and in vivo. The combinatorial nature of our platform significantly reduces synthesis time and effort to identify the optimal linker length and E3 ligase partner to each target and is readily amenable to screening for new targets.


Asunto(s)
Proteínas Nucleares , Factores de Transcripción , Proteolisis , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo , Ligandos
12.
Antimicrob Agents Chemother ; 57(10): 5138-40, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23856770

RESUMEN

Mycobactin biosynthesis in Mycobacterium tuberculosis facilitates iron acquisition, which is required for growth and virulence. The mycobactin biosynthesis inhibitor salicyl-AMS [5'-O-(N-salicylsulfamoyl)adenosine] inhibits M. tuberculosis growth in vitro under iron-limited conditions. Here, we conducted a single-dose pharmacokinetic study and a monotherapy study of salicyl-AMS with mice. Intraperitoneal injection yielded much better pharmacokinetic parameter values than oral administration did. Monotherapy of salicyl-AMS at 5.6 or 16.7 mg/kg significantly inhibited M. tuberculosis growth in the mouse lung, providing the first in vivo proof of concept for this novel antibacterial strategy.


Asunto(s)
Antibacterianos/farmacología , Pulmón/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Oxazoles/metabolismo , Animales , Femenino , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria
13.
Proc Natl Acad Sci U S A ; 107(12): 5477-82, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20212119

RESUMEN

Niemann-Pick type C disease (NPC) is a lysosomal storage disorder causing accumulation of unesterified cholesterol in lysosomal storage organelles. Recent studies have shown that hydroxypropyl-beta-cyclodextrin injections in npc1(-/-) mice are partially effective in treating this disease. Using cultured fibroblasts, we have investigated the cellular mechanisms responsible for reduction of cholesterol accumulation. We show that decreased levels of cholesterol accumulation are maintained for several days after removal of cyclodextrin from the culture medium. This suggests that endocytosed cyclodextrin can reduce the cholesterol storage by acting from inside endocytic organelles rather than by removing cholesterol from the plasma membrane. To test this further, we incubated both NPC1 and NPC2 mutant cells with cholesterol-loaded cyclodextrin for 1 h, followed by chase in serum-containing medium. Although the cholesterol content of the treated cells increased after the 1-h incubation, the cholesterol levels in the storage organelles were later reduced significantly. We covalently coupled cyclodextrin to fluorescent dextran polymers. These cyclodextrin-dextran conjugates were delivered to cholesterol-enriched lysosomal storage organelles and were effective at reducing the cholesterol accumulation. We demonstrate that methyl-beta-cyclodextrin is more potent than hydroxypropyl-beta-cyclodextrin in reducing both cholesterol and bis(monoacylglycerol) phosphate accumulation in NPC mutant fibroblasts. Brief treatment of cells with cyclodextrins causes an increase in cholesterol esterification by acyl CoA:cholesterol acyl transferase, indicating increased cholesterol delivery to the endoplasmic reticulum. These findings suggest that cyclodextrin-mediated enhanced cholesterol transport from the endocytic system can reduce cholesterol accumulation in cells with defects in either NPC1 or NPC2.


Asunto(s)
Colesterol/metabolismo , Mutación , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , beta-Ciclodextrinas/metabolismo , Androstenos/farmacología , Animales , Transporte Biológico Activo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Ésteres del Colesterol/metabolismo , Endocitosis , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Proteínas de Transporte Vesicular , beta-Ciclodextrinas/farmacología
14.
Arch Biochem Biophys ; 509(1): 90-9, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21303655

RESUMEN

Succinic semialdehyde dehydrogenases (SSADHs) are ubiquitous enzymes that catalyze the NAD(P)+-coupled oxidation of succinic semialdehyde (SSA) to succinate, the last step of the γ-aminobutyrate shunt. Mycobacterium tuberculosis encodes two paralogous SSADHs (gabD1 and gabD2). Here, we describe the first mechanistic characterization of GabD1, using steady-state kinetics, pH-rate profiles, ¹H NMR, and kinetic isotope effects. Our results confirmed SSA and NADP+ as substrates and demonstrated that a divalent metal, such as Mg²+, linearizes the time course. pH-rate studies failed to identify any ionizable groups with pK(a) between 5.5 and 10 involved in substrate binding or rate-limiting chemistry. Primary deuterium, solvent and multiple kinetic isotope effects revealed that nucleophilic addition to SSA is very fast, followed by a modestly rate-limiting hydride transfer and fast thioester hydrolysis. Proton inventory studies revealed that a single proton is associated with the solvent-sensitive rate-limiting step. Together, these results suggest that product dissociation and/or conformational changes linked to it are rate-limiting. Using structural information for the human homolog enzyme and ¹H NMR, we further established that nucleophilic attack takes place at the Si face of SSA, generating a thiohemiacetal with S stereochemistry. Deuteride transfer to the Pro-R position in NADP+ generates the thioester intermediate and [4A-²H, 4B-¹H] NADPH. A chemical mechanism based on these data and the structural information available is proposed.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Succionato-Semialdehído Deshidrogenasa/metabolismo , Secuencia de Aminoácidos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Mycobacterium tuberculosis/química , NADP/metabolismo , Resonancia Magnética Nuclear Biomolecular , Alineación de Secuencia , Estereoisomerismo , Especificidad por Sustrato , Succionato-Semialdehído Deshidrogenasa/química , Ácido gamma-Aminobutírico/análogos & derivados , Ácido gamma-Aminobutírico/metabolismo
15.
Curr Cancer Drug Targets ; 21(4): 306-325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33535953

RESUMEN

Epigenetic modulation of gene expression is essential for tissue-specific development and maintenance in mammalian cells. Disruption of epigenetic processes, and the subsequent alteration of gene functions, can result in inappropriate activation or inhibition of various cellular signaling pathways, leading to cancer. Recent advancements in the understanding of the role of epigenetics in cancer initiation and progression have uncovered functions for DNA methylation, histone modifications, nucleosome positioning, and non-coding RNAs. Epigenetic therapies have shown some promise for hematological malignancies, and a wide range of epigenetic-based drugs are undergoing clinical trials. However, in a dynamic survival strategy, cancer cells exploit their heterogeneous population which frequently results in the rapid acquisition of therapy resistance. Here, we describe novel approaches in drug discovery targeting the epigenome, highlighting recent advances the selective degradation of target proteins using Proteolysis Targeting Chimera (PROTAC) to address drug resistance.


Asunto(s)
Resistencia a Antineoplásicos , Terapia Molecular Dirigida , Neoplasias , Proteolisis , Descubrimiento de Drogas/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Epigenoma/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Neoplasias/tratamiento farmacológico , Neoplasias/genética
16.
NPJ Breast Cancer ; 7(1): 108, 2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34426581

RESUMEN

Tetrathiomolybdate (TM) is a novel, copper-depleting compound associated with promising survival in a phase II study of patients with high-risk and triple-negative breast cancer. We sought to elucidate the mechanism of TM by exploring its effects on collagen processing and immune function in the tumor microenvironment (TME). Using an exploratory cohort, we identified markers of collagen processing (LOXL2, PRO-C3, C6M, and C1M) that differed between those with breast cancer versus controls. We measured these collagen biomarkers in TM-treated patients on the phase II study and detected evidence of decreased collagen cross-linking and increased degradation over formation in those without disease compared to those who experienced disease progression. Preclinical studies revealed decreased collagen deposition, lower levels of myeloid-derived suppressor cells, and higher CD4+ T-cell infiltration in TM-treated mice compared with controls. This study reveals novel mechanisms of TM targeting the TME and immune response with potential applications across cancer types.

17.
Biochemistry ; 49(8): 1616-27, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20078138

RESUMEN

Mycobacterium tuberculosis (Mtb) remains the leading single cause of death from bacterial infection. Here we explored the possibility of species-selective inhibition of lipoamide dehydrogenase (Lpd), an enzyme central to Mtb's intermediary metabolism and antioxidant defense. High-throughput screening of combinatorial chemical libraries identified triazaspirodimethoxybenzoyls as high-nanomolar inhibitors of Mtb's Lpd that were noncompetitive versus NADH, NAD(+), and lipoamide and >100-fold selective compared to human Lpd. Efficacy required the dimethoxy and dichlorophenyl groups. The structure of an Lpd-inhibitor complex was resolved to 2.42 A by X-ray crystallography, revealing that the inhibitor occupied a pocket adjacent to the Lpd NADH/NAD(+) binding site. The inhibitor did not overlap with the adenosine moiety of NADH/NAD(+) but did overlap with positions predicted to bind the nicotinamide rings in NADH and NAD(+) complexes. The dimethoxy ring occupied a deep pocket adjacent to the FAD flavin ring where it would block coordination of the NADH nicotinamide ring, while the dichlorophenyl group occupied a more exposed pocket predicted to coordinate the NAD(+) nicotinamide. Several residues that are not conserved between the bacterial enzyme and its human homologue were predicted to contribute both to inhibitor binding and to species selectivity, as confirmed for three residues by analysis of the corresponding mutant Mtb Lpd proteins. Thus, nonconservation of residues lining the electron-transfer tunnel in Mtb Lpd can be exploited for development of species-selective Lpd inhibitors.


Asunto(s)
Antituberculosos/química , Antituberculosos/metabolismo , Dihidrolipoamida Deshidrogenasa/antagonistas & inhibidores , Dihidrolipoamida Deshidrogenasa/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Mycobacterium tuberculosis/enzimología , Animales , Antituberculosos/efectos adversos , Antituberculosos/farmacología , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cristalografía por Rayos X , Dihidrolipoamida Deshidrogenasa/química , Dihidrolipoamida Deshidrogenasa/genética , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacología , Macrófagos/citología , Ratones , Modelos Biológicos , Estructura Molecular , Mutagénesis Sitio-Dirigida , NAD/química , NAD/metabolismo , Relación Estructura-Actividad , Ácido Tióctico/análogos & derivados , Ácido Tióctico/química , Ácido Tióctico/metabolismo
19.
J Med Chem ; 63(21): 13103-13123, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33095579

RESUMEN

The immunoproteasome (i-20S) has emerged as a therapeutic target for autoimmune and inflammatory disorders and hematological malignancies. Inhibition of the chymotryptic ß5i subunit of i-20S inhibits T cell activation, B cell proliferation, and dendritic cell differentiation in vitro and suppresses immune responses in animal models of autoimmune disorders and allograft rejection. However, cytotoxicity to immune cells has accompanied the use of covalently reactive ß5i inhibitors, whose activity against the constitutive proteasome (c-20S) is cumulative with the time of exposure. Herein, we report a structure-activity relationship study of a class of noncovalent proteasome inhibitors with picomolar potencies and 1000-fold selectivity for i-20S over c-20S. Furthermore, these inhibitors are specific for ß5i over the other five active subunits of i-20S and c-20S, providing useful tools to study the functions of ß5i in immune responses. The potency of these compounds in inhibiting human T cell activation suggests that they may have therapeutic potential.


Asunto(s)
Dipéptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Dipéptidos/metabolismo , Dipéptidos/farmacología , Células HeLa , Humanos , Concentración 50 Inhibidora , Cinética , Activación de Linfocitos/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/farmacología , Unión Proteica , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
20.
J Am Chem Soc ; 131(46): 16744-50, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19799378

RESUMEN

Several Mycobacterium tuberculosis strains, Mycobacterium leprae, and other mycobacterial pathogens produce a group of small-molecule virulence factors called phenolic glycolipids (PGLs). PGLs play key roles in pathogenicity and host-pathogen interaction. Thus, elucidation of the PGL biosynthetic pathway will not only expand our understanding of natural product biosynthesis, but may also illuminate routes to novel therapeutics to afford alternative lines of defense against mycobacterial infections. In this study, we report an investigation of the enzymatic requirements for the production of long-chain p-hydroxyphenylalkanoate intermediates of PGL biosynthesis. We demonstrate a functional cooperation between a coenzyme A-independent stand-alone didomain initiation module (FadD22) and a 6-domain reducing iterative type I polyketide synthase (Pks15/1) for production of p-hydroxyphenylalkanoate intermediates in in vitro and in vivo FadD22-Pks15/1 reconstituted systems. Our results suggest that Pks15/1 is an iterative type I polyketide synthase with a relaxed control of catalytic cycle iterations, a mechanistic property that explains the origin of a characteristic alkyl chain length variability seen in mycobacterial PGLs. The FadD22-Pks15/1 reconstituted systems lay an initial foundation for future efforts to unveil the mechanism of iterative catalysis control by which the structures of the final products of Pks15/1 are defined, and to scrutinize the functional partnerships of the FadD22-Pks15/1 system with downstream enzymes of the PGL biosynthetic pathway.


Asunto(s)
Coenzima A/metabolismo , Ácidos Grasos/biosíntesis , Glucolípidos/biosíntesis , Mycobacterium marinum/enzimología , Fenoles/metabolismo , Sintasas Poliquetidas/metabolismo , Glucolípidos/química , Fenoles/química , Sintasas Poliquetidas/química , Sintasas Poliquetidas/genética , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA