Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
1.
Acta Chim Slov ; 68(3): 667-682, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34897536

RESUMEN

The activation of caspases is central to apoptotic process in living systems. Defects in apoptosis have been implicated with carcinogenesis. Need to develop smart agents capable of inducing apoptosis in tumor cells is obvious. With this motive, diversity oriented synthesis of 1-benzylpyrrolidin-3-ol analogues was envisaged. The multi component Ugi reaction synthesized library of electronically diverse analogues was explored for cytotoxic propensity towards a panel of human cancer cell lines at 10 µM. The lead compounds exhibit a selective cytotoxicity towards HL-60 cells as compared to cell lines derived from solid tumors. Besides, their milder cytotoxic effect on non-cancerous cell lines reaffirm their selective action towards cancer cells only. The lead molecules were tested for their ability to target caspase-3, as a vital protease triggering apoptosis. The lead compounds were observed to induce apoptosis in HL-60 cells around 10 µM concentration. The lead compounds exhibited various non-covalent supra type interactions with caspase-3 key residues around the active site. The binding ability of lead compounds with caspase-3 was studied via molecular docking and molecular dynamic (MD) simulations. MD simulations indicated the stability of compound-caspase-3 complex throughout the 50 ns simulation run. The stability and bio-availability of the lead compounds under physiological conditions was assessed by their interaction with Bovine Serum Albumin (BSA) as model protein. BSA interactions of lead compounds were studied by various bio-physical methods and further substantiated with in silico MD simulations.


Asunto(s)
Antineoplásicos/farmacología , Caspasa 3/metabolismo , Activadores de Enzimas/farmacología , Pirrolidinas/farmacología , Animales , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Bovinos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Activadores de Enzimas/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Pirrolidinas/metabolismo , Albúmina Sérica Bovina/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
2.
ACS Chem Biol ; 16(7): 1159-1163, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34165961

RESUMEN

The nuclear receptor-related 1 protein, Nurr1, is a transcription factor critical for the development and maintenance of dopamine-producing neurons in the substantia nigra pars compacta, a cell population that progressively loses the ability to make dopamine and degenerates in Parkinson's disease. Recently, we demonstrated that Nurr1 binds directly to and is regulated by the endogenous dopamine metabolite 5,6-dihydroxyindole (DHI). Unfortunately, DHI is an unstable compound, and thus a poor tool for studying Nurr1 function. Here, we report that 5-chloroindole, an unreactive analog of DHI, binds directly to the Nurr1 ligand binding domain with micromolar affinity and stimulates the activity of Nurr1, including the transcription of genes governing the synthesis and packaging of dopamine.


Asunto(s)
Activadores de Enzimas/farmacología , Indoles/farmacología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/agonistas , Animales , Línea Celular , Activadores de Enzimas/metabolismo , Activadores de Enzimas/toxicidad , Indoles/metabolismo , Indoles/toxicidad , Ratones , Mutación , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Unión Proteica , Dominios Proteicos/genética
3.
J Med Chem ; 64(9): 5323-5344, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33872507

RESUMEN

Herein we describe the discovery, mode of action, and preclinical characterization of the soluble guanylate cyclase (sGC) activator runcaciguat. The sGC enzyme, via the formation of cyclic guanosine monophoshphate, is a key regulator of body and tissue homeostasis. sGC activators with their unique mode of action are activating the oxidized and heme-free and therefore NO-unresponsive form of sGC, which is formed under oxidative stress. The first generation of sGC activators like cinaciguat or ataciguat exhibited limitations and were discontinued. We overcame limitations of first-generation sGC activators and identified a new chemical class via high-throughput screening. The investigation of the structure-activity relationship allowed to improve potency and multiple solubility, permeability, metabolism, and drug-drug interactions parameters. This program resulted in the discovery of the oral sGC activator runcaciguat (compound 45, BAY 1101042). Runcaciguat is currently investigated in clinical phase 2 studies for the treatment of patients with chronic kidney disease and nonproliferative diabetic retinopathy.


Asunto(s)
Diseño de Fármacos , Activadores de Enzimas/química , Guanilil Ciclasa Soluble/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/metabolismo , Perros , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Activadores de Enzimas/uso terapéutico , Semivida , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Simulación de Dinámica Molecular , Ratas , Ratas Endogámicas SHR , Solubilidad , Guanilil Ciclasa Soluble/metabolismo , Relación Estructura-Actividad
4.
Biochem Soc Trans ; 49(1): 131-144, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33367567

RESUMEN

Enzyme catalysis is omnipresent in the cell. The mechanisms by which highly evolved protein folds enable rapid and specific chemical transformation of substrates belong to the marvels of structural biology. Targeting of enzymes with inhibitors has immediate application in drug discovery, from chemotherapeutics over antibiotics to antivirals. NMR spectroscopy combines multiple assets for the investigation of enzyme function. The non-invasive technique can probe enzyme structure and dynamics and map interactions with substrates, cofactors and inhibitors at the atomic level. With experiments performed at close to native conditions, catalytic transformations can be monitored in real time, giving access to kinetic parameters. The power of NMR in the solid state, in contrast with solution, lies in the absence of fundamental size limitations, which is crucial for enzymes that are either membrane-embedded or assemble into large soluble complexes exceeding hundreds of kilodaltons in molecular weight. Here we review recent progress in solid-state NMR methodology, which has taken big leaps in the past years due to steady improvements in hardware design, notably magic angle spinning, and connect it to parallel biochemical advances that enable isotope labelling of increasingly complex enzymes. We first discuss general concepts and requirements of the method and then highlight the state-of-the-art in sample preparation, structure determination, dynamics and interaction studies. We focus on examples where solid-state NMR has been instrumental in elucidating enzyme mechanism, alone or in integrative studies.


Asunto(s)
Enzimas , Espectroscopía de Resonancia Magnética/métodos , Complejos Multiproteicos/química , Animales , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Enzimas/química , Enzimas/metabolismo , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/análisis , Complejos Multiproteicos/metabolismo , Especificidad por Sustrato
5.
Angew Chem Int Ed Engl ; 60(10): 5436-5442, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33238058

RESUMEN

Genetic, preclinical and clinical data link Parkinson's disease and Gaucher's disease and provide a rational entry point to disease modification therapy via enhancement of ß-Glucocerebrosidase (GCase) activity. We discovered a new class of pyrrolo[2,3-b]pyrazine activators effecting both Vmax and Km. They bind to human GCase and increase substrate metabolism in the lysosome in a cellular assay. We obtained the first crystal structure for an activator and identified a novel non-inhibitory binding mode at the interface of a dimer, rationalizing the observed structure-activity relationship (SAR). The compound binds GCase inducing formation of a dimeric state at both endoplasmic reticulum (ER) and lysosomal pHs, as confirmed by analytical ultracentrifugation. Importantly, the pyrrolo[2,3-b]pyrazines have central nervous system (CNS) drug-like properties. Our findings are important for future drug discovery efforts in the field of GCase activation and provide a deeper mechanistic understanding of the requirements for enzymatic activation, pointing to the relevance of dimerization.


Asunto(s)
Activadores de Enzimas/metabolismo , Glucosilceramidasa/metabolismo , Multimerización de Proteína/efectos de los fármacos , Pirazinas/metabolismo , Pirroles/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Activadores de Enzimas/química , Glucosilceramidasa/química , Humanos , Cinética , Estructura Molecular , Unión Proteica , Pirazinas/química , Pirroles/química , Relación Estructura-Actividad
6.
J Med Chem ; 63(18): 10474-10495, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32787077

RESUMEN

SIRT6 activation is thought to be a promising target for the treatment of many diseases, particularly cancer. Herein, we report the discovery of a series of new small-molecule SIRT6 activators. Structure-activity relationship analyses led to the identification of the most potent compound, 2-(1-benzofuran-2-yl)-N-(diphenylmethyl) quinoline-4-carboxamide (12q), which showed an EC1.5 value of 0.58 ± 0.12 µM and an EC50 value of 5.35 ± 0.69 µM against SIRT6-dependent peptide deacetylation in FLUOR DE LYS assay. It exhibited weak or no activity against other HDAC family members as well as 415 kinases, indicating good selectivity for SIRT6. 12q significantly inhibited the proliferation and migration of pancreatic ductal adenocarcinoma (PDAC) cells in vitro. It also markedly suppressed the tumor growth in a PDAC tumor xenograft model. This compound showed attractive pharmacokinetic properties. Overall, 12q could be a good lead compound for the treatment of PDAC, and it is worthy of further study.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Activadores de Enzimas/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Quinolinas/uso terapéutico , Sirtuinas/metabolismo , Adenocarcinoma/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activadores de Enzimas/síntesis química , Activadores de Enzimas/metabolismo , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Quinolinas/síntesis química , Quinolinas/metabolismo , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ChemMedChem ; 15(24): 2521-2529, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32762073

RESUMEN

AMP-activated protein kinase (AMPK) is currently the subject of intensive study and active discussions. AMPK performs its functions both at the cellular level, providing the switch between energy-consuming and energy-producing processes, and at the whole body level, particularly, regulating certain aspects of higher nervous activity and behavior. Control of such a 'main switch' compensates dysfunctions and associated diseases. In the present paper, we studied the binding of 3-benzylidene oxindoles to the kinase domain of the AMPK α-subunit, which is thought to prevent its interaction with the autoinhibitory domain and thus result in the AMPK activation. For this purpose, we developed the cellular test system based on the AMPKAR plasmid, which implements the FRET effect, synthesized a number of 3-benzylidene oxindole compounds and simulated their binding to various sites of the kinase domain. The most probable binding site for the studied compounds was established by the correlation of calculated and experimental data. The obtained results allow to analyze various classes of AMPK activators using virtual and high-content screening.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Compuestos de Bencilideno/farmacología , Activadores de Enzimas/farmacología , Oxindoles/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Quinasas Activadas por AMP/química , Secuencia de Aminoácidos , Compuestos de Bencilideno/síntesis química , Compuestos de Bencilideno/metabolismo , Sitios de Unión , Línea Celular Tumoral , Activadores de Enzimas/síntesis química , Activadores de Enzimas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Oxindoles/síntesis química , Oxindoles/metabolismo , Unión Proteica , Dominios Proteicos , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/metabolismo
8.
Eur J Pharmacol ; 881: 173203, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32446711

RESUMEN

Soluble guanylyl cyclase (sGC), the major receptor for nitric oxide (NO), is a heterodimer consisting of two subunits, the α and the ß subunit. The NO/sGC/cGMP signaling pathway is protective in different disease pathomechanisms including angina pectoris, pulmonary hypertension and fibrotic diseases. The natural ligand heme has two carboxylic acids which interact in the ß1 heme nitric oxide oxygen binding (HNOX) domain with the amino acids of the highly conserved Y-x-S-x-R motif. The Y-x-S-x-R motif is also involved in binding of the dicarboxylic activators cinaciguat and BAY 60-2770 as indicated by crystallization studies of sGC activator and bacterial HNOX homologs. To what extent the Y-x-S-x-R motif hydrogen bond network contributes to binding of monocarboxylic acids has not been examined so far. In the current paper, the chemical structural formula of the novel monocarboxylic drug BAY-543 is described for the first time. Using this novel drug, we evaluate the importance of the amino acids Y135 and R139 for thermostabilization and activation in comparison to the dicarboxylic acid BAY 60-2770. Measurements with point mutated sGC variants demonstrate tyrosine 135 as exclusive binding site of the monocarboxylic acid BAY-543 but not the dicarboxylic BAY 60-2770.


Asunto(s)
Activadores de Enzimas/farmacología , Guanilil Ciclasa Soluble/metabolismo , Secuencias de Aminoácidos , Animales , Benzoatos/metabolismo , Benzoatos/farmacología , Sitios de Unión , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/farmacología , Activación Enzimática , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Células HEK293 , Humanos , Hidrocarburos Fluorados/metabolismo , Hidrocarburos Fluorados/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacología , Mutación Puntual , Unión Proteica , Conformación Proteica , Subunidades de Proteína , Células Sf9 , Guanilil Ciclasa Soluble/química , Guanilil Ciclasa Soluble/genética , Relación Estructura-Actividad , Tirosina
9.
Cell ; 181(3): 688-701.e16, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32315618

RESUMEN

Impairment of protein phosphatases, including the family of serine/threonine phosphatases designated PP2A, is essential for the pathogenesis of many diseases, including cancer. The ability of PP2A to dephosphorylate hundreds of proteins is regulated by over 40 specificity-determining regulatory "B" subunits that compete for assembly and activation of heterogeneous PP2A heterotrimers. Here, we reveal how a small molecule, DT-061, specifically stabilizes the B56α-PP2A holoenzyme in a fully assembled, active state to dephosphorylate selective substrates, such as its well-known oncogenic target, c-Myc. Our 3.6 Å structure identifies molecular interactions between DT-061 and all three PP2A subunits that prevent dissociation of the active enzyme and highlight inherent mechanisms of PP2A complex assembly. Thus, our findings provide fundamental insights into PP2A complex assembly and regulation, identify a unique interfacial stabilizing mode of action for therapeutic targeting, and aid in the development of phosphatase-based therapeutics tailored against disease specific phospho-protein targets.


Asunto(s)
Proteína Fosfatasa 2/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Activadores de Enzimas/metabolismo , Células HEK293 , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Proteína Fosfatasa 2/química , Subunidades de Proteína
10.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32315619

RESUMEN

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Asunto(s)
Proteína Fosfatasa 2/metabolismo , Apoptosis , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Activadores de Enzimas/metabolismo , Fase G1 , Humanos , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Fenotiazinas/farmacología , Fosforilación , Proteína Fosfatasa 2/fisiología , Subunidades de Proteína/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
11.
J Med Chem ; 63(10): 5324-5340, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32329617

RESUMEN

Janus kinases (JAKs) are non-receptor tyrosine kinases that are essential components of the JAK-STAT signaling pathway. Associated aberrant signaling is responsible for many forms of cancer and disorders of the immune system. The present focus is on the discovery of molecules that may regulate the activity of JAK2 by selective binding to the JAK2 pseudokinase domain, JH2. Specifically, the Val617Phe mutation in JH2 stimulates the activity of the adjacent kinase domain (JH1) resulting in myeloproliferative disorders. Starting from a non-selective screening hit, we have achieved the goal of discovering molecules that preferentially bind to the ATP binding site in JH2 instead of JH1. We report the design and synthesis of the compounds and binding results for the JH1, JH2, and JH2 V617F domains, as well as five crystal structures for JH2 complexes. Testing with a selective and non-selective JH2 binder on the autophosphorylation of wild-type and V617F JAK2 is also contrasted.


Asunto(s)
Amitrol (Herbicida)/química , Amitrol (Herbicida)/metabolismo , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Janus Quinasa 2/química , Janus Quinasa 2/metabolismo , Animales , Células HEK293 , Humanos , Ligandos , Unión Proteica/fisiología , Células Sf9 , Difracción de Rayos X/métodos
12.
Arch Biochem Biophys ; 687: 108285, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32074500

RESUMEN

Polyphyllin I (PPI), a bioactive constituent extracted from the rhizomes of Paris polyphylla, is cytotoxic to several cancer types. This study was designed to explore whether PPI prevents non-small-cell lung cancer (NSCLC) growth and to investigate the molecular mechanism. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. In cultured human NSCLC cell lines, PPI induces autophagy by activating AMPK and then inhibiting mTOR signaling in a concentration-dependent manner. Furthermore, the activation of autophagy induced by PPI was reversed by the AMPK inhibitor compound C. Computational docking showed that PPI directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. Microscale thermophoresis and Drug Affinity Responsive Targeting Stability (DARTS) assay further confirmed the high affinity between PPI and AMPK. In vivo studies indicated that PPI suppressed the growth of NSCLC and increased the levels of LC3-II and phosphorylated AMPK in tumors isolated from a xenograft model of NSCLC in mice. Moreover, PPI exhibited favorable pharmacokinetics in rats. In summary, PPI conclusively acts as a direct AMPK activator to induce cell autophagy which inhibits the growth of NSCLC cells. In the future, PPI therapy should be applied to treat patients with NSCLC.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Diosgenina/análogos & derivados , Activadores de Enzimas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/química , Sitio Alostérico , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Diosgenina/metabolismo , Diosgenina/farmacocinética , Diosgenina/uso terapéutico , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacocinética , Femenino , Humanos , Masculino , Ratones Desnudos , Simulación del Acoplamiento Molecular , Unión Proteica , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Med Chem ; 63(6): 3348-3358, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32109056

RESUMEN

ER aminopeptidase 1 (ERAP1) is an intracellular enzyme that generates antigenic peptides and is an emerging target for cancer immunotherapy and the control of autoimmunity. ERAP1 inhibitors described previously target the active site and are limited in selectivity, minimizing their clinical potential. To address this, we targeted the regulatory site of ERAP1 using a high-throughput screen and discovered a small molecule hit that is highly selective for ERAP1. (4aR,5S,6R,8S,8aR)-5-(2-(Furan-3-yl)ethyl)-8-hydroxy-5,6,8a-trimethyl-3,4,4a,5,6,7,8,8a-octahydronaphthalene-1-carboxylic acid is a natural product found in Dodonaea viscosa that constitutes a submicromolar, highly selective, and cell-active modulator of ERAP1. Although the compound activates hydrolysis of small model substrates, it is a competitive inhibitor for physiologically relevant longer peptides. Crystallographic analysis confirmed that the compound targets the regulatory site of the enzyme that normally binds the C-terminus of the peptide substrate. Our findings constitute a novel starting point for the development of selective ERAP1 modulators that have potential for further clinical development.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Presentación de Antígeno/efectos de los fármacos , Diterpenos de Tipo Clerodano/farmacología , Epítopos/metabolismo , Péptidos/metabolismo , Inhibidores de Proteasas/farmacología , Sitio Alostérico , Aminopeptidasas/química , Aminopeptidasas/metabolismo , Animales , Dominio Catalítico , Cristalografía por Rayos X , Diterpenos de Tipo Clerodano/química , Diterpenos de Tipo Clerodano/metabolismo , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Epítopos/química , Células HeLa , Humanos , Ratones , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/metabolismo , Péptidos/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Unión Proteica , Proteolisis/efectos de los fármacos
14.
Biochem Biophys Res Commun ; 524(1): 123-128, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-31980172

RESUMEN

Circadian rhythms are the endogenous oscillation of biological reactions and behaviors in most organisms on Earth. Circadian clocks are the pacemakers regulating circadian rhythms, and the transcription-translation dependent feedback loop (TTFL) model was supposed to be the sole model of circadian clocks. However, recent years have witnessed rapid progresses in the study of non-TTFL circadian clocks. The cyanobacterial circadian clock consists of three proteins (KaiA, KaiB, and KaiC), and is extensively studied as a non-TTFL circadian clock model. Although containing only three proteins, the molecular mechanism of the KaiABC circadian clock remains elusive. We recently noticed that KaiA has an auto-inhibition conformation during the oscillation, but how this auto-inhibition is regulated is unclear. Here, we started from the design of light modulated KaiAs to investigate this mechanism. We designed different KaiA constructs fused with the light modulable LOV2 protein, and used light-modulated KaiAs to regulate the phosphorylation and dephosphorylation of KaiC. Our data indicated that the N-terminal domain of KaiA is important for KaiA's reversible off/on switching during the unidirectional oscillation of the KaiABC system. This work provides an updated model to explain the molecular mechanism of the KaiABC circadian clock.


Asunto(s)
Proteínas Bacterianas/metabolismo , Relojes Circadianos/fisiología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Cianobacterias/fisiología , Activadores de Enzimas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Ritmo Circadiano/fisiología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Fosforilación , Unión Proteica , Conformación Proteica , Multimerización de Proteína , ARN Bacteriano
15.
Bioorg Med Chem ; 28(1): 115232, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31818630

RESUMEN

Glucose flux through glucokinase (GK) controls insulin release from the pancreas in response to high levels of glucose. Flux through GK is also responsible for reducing hepatic glucose output. Since many individuals with type 2 diabetes appear to have an inadequacy or defect in one or both of these processes, identifying compounds that can activate GK could provide a therapeutic benefit. Herein we report the further structure activity studies of a novel series of glucokinase activators (GKA). These studies led to the identification of pyridine 72 as a potent GKA that lowered post-prandial glucose in normal C57BL/6J mice, and after 14d dosing in ob/ob mice.


Asunto(s)
Activadores de Enzimas/química , Glucoquinasa/química , Hipoglucemiantes/química , Animales , Sitios de Unión , Glucemia/análisis , Cristalografía por Rayos X , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Activadores de Enzimas/metabolismo , Activadores de Enzimas/uso terapéutico , Glucoquinasa/metabolismo , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Cinética , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Relación Estructura-Actividad , Tiadiazoles/química , Tiadiazoles/metabolismo
16.
Bioorg Med Chem Lett ; 30(2): 126790, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31744674

RESUMEN

Adenosine monophosphate-activated protein kinase (AMPK) has been considered as a promising drug target for its regulation in both glucose and lipid metabolism. Mogrol was originally identified from high throughput screening as a small molecule activator of AMPK subtype α2ß1γ1. In order to enhance its potency on AMPK and summarize the structure-activity relationships, a series of mogrol derivatives were designed, synthesized and evaluated in pharmacological AMPK activation assays. The results showed that the amine derivatives at the 24-position can improve the potency. Among them, compounds 3 and 4 exhibited the best potency (EC50: 0.15 and 0.14 µM) which was 20 times more potent than mogrol (EC50: 3.0 µM).


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diseño de Fármacos , Activadores de Enzimas/síntesis química , Triterpenos/metabolismo , Proteínas Quinasas Activadas por AMP/química , Regulación Alostérica/efectos de los fármacos , Cucurbitaceae/química , Cucurbitaceae/metabolismo , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Humanos , Relación Estructura-Actividad , Triterpenos/farmacología
17.
ACS Infect Dis ; 5(11): 1915-1925, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31588734

RESUMEN

Acyldepsipeptides are a unique class of antibiotics that act via allosterically dysregulated activation of the bacterial caseinolytic protease (ClpP). The ability of ClpP activators to kill nongrowing bacteria represents a new opportunity to combat deep-seated biofilm infections. However, the acyldepsipeptide scaffold is subject to rapid metabolism. Herein, we explore alteration of the potentially metabolically reactive α,ß unsaturated acyl chain. Through targeted synthesis, a new class of phenyl urea substituted depsipeptide ClpP activators with improved metabolic stability is described. The ureadepsipeptides are potent activators of Staphylococcus aureus ClpP and show activity against Gram-positive bacteria, including S. aureus biofilms. These studies demonstrate that a phenyl urea motif can successfully mimic the double bond, maintaining potency equivalent to acyldepsipeptides but with decreased metabolic liability. Although removal of the double bond from acyldepsipeptides generally has a significant negative impact on potency, structural studies revealed that the phenyl ureadepsipeptides can retain potency through the formation of a third hydrogen bond between the urea and the key Tyr63 residue in the ClpP activation domain. Ureadepsipeptides represent a new class of ClpP activators with improved drug-like properties, potent antibacterial activity, and the tractability to be further optimized.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Depsipéptidos/química , Endopeptidasa Clp/metabolismo , Activadores de Enzimas/química , Staphylococcus aureus/enzimología , Antibacterianos/química , Antibacterianos/metabolismo , Proteínas Bacterianas/agonistas , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Depsipéptidos/metabolismo , Depsipéptidos/farmacología , Endopeptidasa Clp/química , Endopeptidasa Clp/genética , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Dominios Proteicos , Staphylococcus aureus/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Urea/química , Urea/metabolismo
18.
Expert Opin Drug Metab Toxicol ; 15(11): 975-984, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31619082

RESUMEN

Objectives: Riociguat is a soluble guanylate cyclase stimulator licensed for the treatment of pulmonary arterial hypertension (PAH), a potentially fatal complication of human immunodeficiency virus infection. This study investigated the inhibitory potency of selected antiretroviral regimens on the metabolic clearance of riociguat.Methods: The inhibitory potential of the components of six antiretroviral combinations (ATRIPLA® (efavirenz/emtricitabine/tenofovir disoproxil), COMPLERA® (rilpivirine/emtricitabine/tenofovir disoproxil), STRIBILD® (elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil), TRIUMEQ® (abacavir/dolutegravir/lamivudine), and two ritonavir-boosted regimens) on riociguat metabolism were evaluated in recombinant human CYP1A1 and CYP3A4 as well as in human hepatocytes exhibiting both CYP1A1 and CYP3A4 activity. In vitro-in vivo correlation was performed between calculated and observed increases in riociguat exposure in vivo.Results: Using both in vitro systems, the predicted increase in exposure of riociguat was highest with components of TRIUMEQ® followed by COMPLERA®, ATRIPLA®, STRIBILD®, and the ritonavir-boosted regimens. Further experiments in human hepatocytes confirmed CYP1A1 to be the predominant enzyme in the metabolic clearance of riociguat.Conclusion: Antiretroviral treatment containing the potent CYP1A1 inhibitor abacavir had the greatest impact on riociguat metabolic clearance. The impact of comedications containing only strong CYP3A4 inhibitors e.g. ritonavir was less pronounced, suggesting a benefit of riociguat over PAH-targeting medications with contraindications for use with strong CYP3A4 inhibitors.


Asunto(s)
Fármacos Anti-VIH/farmacología , Citocromo P-450 CYP1A1/metabolismo , Activadores de Enzimas/metabolismo , Pirazoles/metabolismo , Pirimidinas/metabolismo , Fármacos Anti-VIH/administración & dosificación , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Citocromo P-450 CYP3A/efectos de los fármacos , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/administración & dosificación , Inhibidores del Citocromo P-450 CYP3A/farmacología , Interacciones Farmacológicas , Activadores de Enzimas/administración & dosificación , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación
19.
Genes Dev ; 33(17-18): 1280-1292, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371438

RESUMEN

All cells use proteases to adjust protein amounts. Proteases maintain protein homeostasis by degrading nonfunctional toxic proteins and play regulatory roles by targeting particular substrates in response to specific signals. Here we address how cells tune protease specificity to nutritional signals. We report that Salmonella enterica increases the specificity of the broadly conserved proteases Lon and ClpSAP by transforming the Lon activator and substrate HspQ into an inhibitor of the N-degron recognin ClpS, the adaptor of the ClpAP protease. We establish that upon acetylation, HspQ stops being a Lon activator and substrate and that the accumulated HspQ binds to ClpS, hindering degradation of ClpSAP substrates. Growth on glucose promotes HspQ acetylation by increasing acetyl-CoA amounts, thereby linking metabolism to proteolysis. By altering protease specificities but continuing to degrade junk proteins, cells modify the abundance of particular proteins while preserving the quality of their proteomes. This rapid response mechanism linking protease specificity to nutritional signals is broadly conserved.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fenómenos Fisiológicos de la Nutrición , Salmonella enterica/enzimología , Acetilación , Activadores de Enzimas/metabolismo , Inhibidores Enzimáticos/metabolismo , Glucosa/metabolismo , Proteínas de Choque Térmico , Proteasa La/metabolismo , Unión Proteica , Proteolisis , Salmonella enterica/crecimiento & desarrollo , Especificidad por Sustrato
20.
Mini Rev Med Chem ; 19(18): 1544-1557, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31362687

RESUMEN

Soluble Guanylate Cyclase (sGC) is the intracellular receptor of Nitric Oxide (NO). The activation of sGC results in the conversion of Guanosine Triphosphate (GTP) to the secondary messenger cyclic Guanosine Monophosphate (cGMP). cGMP modulates a series of downstream cascades through activating a variety of effectors, such as Phosphodiesterase (PDE), Protein Kinase G (PKG) and Cyclic Nucleotide-Gated Ion Channels (CNG). NO-sGC-cGMP pathway plays significant roles in various physiological processes, including platelet aggregation, smooth muscle relaxation and neurotransmitter delivery. With the approval of an sGC stimulator Riociguat for the treatment of Pulmonary Arterial Hypertension (PAH), the enthusiasm in the discovery of sGC modulators continues for broad clinical applications. Notably, through activating the NO-sGC-cGMP pathway, sGC stimulator and activator potentiate for the treatment of various diseases, such as PAH, Heart Failure (HF), Diabetic Nephropathy (DN), Systemic Sclerosis (SS), fibrosis as well as other diseases including Sickle Cell Disease (SCD) and Central Nervous System (CNS) disease. Here, we review the preclinical and clinical studies of sGC stimulator and activator in recent years and prospect for the development of sGC modulators in the near future.


Asunto(s)
Activadores de Enzimas/química , Guanilato Ciclasa/metabolismo , Derivados del Benceno/química , Derivados del Benceno/metabolismo , Derivados del Benceno/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/patología , GMP Cíclico/metabolismo , Activadores de Enzimas/metabolismo , Activadores de Enzimas/uso terapéutico , Guanilato Ciclasa/química , Humanos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/patología , Óxido Nítrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA