Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2315018121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38625940

RESUMO

Heterotrimeric G proteins can be regulated by posttranslational modifications, including ubiquitylation. KCTD5, a pentameric substrate receptor protein consisting of an N-terminal BTB domain and a C-terminal domain, engages CUL3 to form the central scaffold of a cullin-RING E3 ligase complex (CRL3KCTD5) that ubiquitylates Gßγ and reduces Gßγ protein levels in cells. The cryo-EM structure of a 5:5:5 KCTD5/CUL3NTD/Gß1γ2 assembly reveals a highly dynamic complex with rotations of over 60° between the KCTD5BTB/CUL3NTD and KCTD5CTD/Gßγ moieties of the structure. CRL3KCTD5 engages the E3 ligase ARIH1 to ubiquitylate Gßγ in an E3-E3 superassembly, and extension of the structure to include full-length CUL3 with RBX1 and an ARIH1~ubiquitin conjugate reveals that some conformational states position the ARIH1~ubiquitin thioester bond to within 10 Å of lysine-23 of Gß and likely represent priming complexes. Most previously described CRL/substrate structures have consisted of monovalent complexes and have involved flexible peptide substrates. The structure of the KCTD5/CUL3NTD/Gßγ complex shows that the oligomerization of a substrate receptor can generate a polyvalent E3 ligase complex and that the internal dynamics of the substrate receptor can position a structured target for ubiquitylation in a CRL3 complex.


Assuntos
Proteínas de Transporte , Ubiquitina-Proteína Ligases , Ligação Proteica , Ubiquitinação , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Transporte/metabolismo , Ubiquitina/metabolismo , Proteínas Culina/genética , Proteínas Culina/metabolismo
2.
Nat Chem Biol ; 20(6): 732-741, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38321209

RESUMO

Glycosylation is a critical post-translational protein modification that affects folding, half-life and functionality. Glycosylation is a non-templated and heterogeneous process because of the promiscuity of the enzymes involved. We describe a platform for sequential glycosylation reactions for tailored sugar structures (SUGAR-TARGET) that allows bespoke, controlled N-linked glycosylation in vitro enabled by immobilized enzymes produced with a one-step immobilization/purification method. We reconstruct a reaction cascade mimicking a glycosylation pathway where promiscuity naturally exists to humanize a range of proteins derived from different cellular systems, yielding near-homogeneous glycoforms. Immobilized ß-1,4-galactosyltransferase is used to enhance the galactosylation profile of three IgGs, yielding 80.2-96.3% terminal galactosylation. Enzyme recycling is demonstrated for a reaction time greater than 80 h. The platform is easy to implement, modular and reusable and can therefore produce homogeneous glycan structures derived from various hosts for functional and clinical evaluation.


Assuntos
Enzimas Imobilizadas , Galactosiltransferases , Glicosilação , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Humanos , Galactosiltransferases/metabolismo , Galactosiltransferases/química , Polissacarídeos/metabolismo , Polissacarídeos/química , Processamento de Proteína Pós-Traducional
3.
Eur J Cell Biol ; 103(2): 151414, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38640594

RESUMO

The RAS isoforms (KRAS, HRAS and NRAS) have distinct cancer type-specific profiles. NRAS mutations are the second most prevalent RAS mutations in skin and hematological malignancies. Although RAS proteins were considered undruggable for decades, isoform and mutation-specific investigations have produced successful RAS inhibitors that are either specific to certain mutants, isoforms (pan-KRAS) or target all RAS proteins (pan-RAS). While extensive structural and biochemical investigations have focused mainly on K- and H-RAS mutations, NRAS mutations have received less attention, and the most prevalent NRAS mutations in human cancers, Q61K and Q61R, are rare in K- and H-RAS. This manuscript presents a crystal structure of the NRAS Q61K mutant in the GTP-bound form. Our structure reveals a previously unseen pocket near switch II induced by the binding of a ligand to the active form of the protein. This observation reveals a binding site that can potentially be exploited for development of inhibitors against mutant NRAS. Furthermore, the well-resolved catalytic site of this GTPase bound to native GTP provides insight into the stalled GTP hydrolysis observed for NRAS-Q61K.


Assuntos
GTP Fosfo-Hidrolases , Guanosina Trifosfato , Proteínas de Membrana , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/química , Humanos , Guanosina Trifosfato/metabolismo , Cristalografia por Raios X , Ligantes , Mutação , Modelos Moleculares
4.
ACS Med Chem Lett ; 14(2): 199-210, 2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36793435

RESUMO

B cell lymphoma 6 (BCL6), a highly regulated transcriptional repressor, is deregulated in several forms of non-Hodgkin lymphoma (NHL), most notably in diffuse large B-cell lymphoma (DLBCL). The activities of BCL6 are dependent on protein-protein interactions with transcriptional co-repressors. To find new therapeutic interventions addressing the needs of patients with DLBCL, we initiated a program to identify BCL6 inhibitors that interfere with co-repressor binding. A virtual screen hit with binding activity in the high micromolar range was optimized by structure-guided methods, resulting in a novel and highly potent inhibitor series. Further optimization resulted in the lead candidate 58 (OICR12694/JNJ-65234637), a BCL6 inhibitor with low nanomolar DLBCL cell growth inhibition and an excellent oral pharmacokinetic profile. Based on its overall favorable preclinical profile, OICR12694 is a highly potent, orally bioavailable candidate for testing BCL6 inhibition in DLBCL and other neoplasms, particularly in combination with other therapies.

5.
ACS Cent Sci ; 8(5): 571-580, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35647281

RESUMO

High-throughput experimentation (HTE) seeks to accelerate the exploration of materials space by uniting robotics, combinatorial methods, and parallel processing. HTE is particularly relevant to metal halide perovskites (MHPs), a diverse class of optoelectronic materials with a large chemical space. Here we develop an HTE workflow to synthesize and characterize light-emitting MHP single crystals, allowing us to generate the first reported data set of experimentally derived photoluminescence spectra for low-dimensional MHPs. We leverage the accelerated workflow to optimize the synthesis and emission of a new MHP, methoxy-phenethylammonium lead iodide ((4-MeO-PEAI)2-PbI2). We then synthesize 16 000 MHP single crystals and measure their photoluminescence to study the effects of synthesis parameters and compositional engineering on the emission intensity of 54 distinct MHPs: we achieve an acceleration factor of more than 100 times over previously reported HTE MHP synthesis and characterization methods. Using insights derived from this analysis, we screen an existing database for new, potentially emissive MHPs. On the basis of the Tanimoto similarity of the bright available emitters, we present our top candidates for future exploration. As a proof of concept, we use one of these (3,4-difluorophenylmethanamine) to synthesize an MHP which we find has a photoluminescence quantum yield of 10%.

6.
J Mol Biol ; 434(9): 167527, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35257782

RESUMO

Ral Guanine Nucleotide Dissociation Stimulator Like 1 (RGL1) is a RAS effector protein that activates Ral GTPase by stimulating nucleotide exchange. Most structures of RAS-effector complexes are for the HRAS isoform; relatively few KRAS-effector structures have been solved, even though KRAS mutations are more frequent in human cancers. We determined crystal structures of KRAS/RGL1-RAS-association (RA) domain complexes and characterized the interaction in solution using nuclear magnetic resonance spectroscopy, size-exclusion chromatography combined with multi-angle light scattering and biolayer interferometry. We report structures of wild-type KRAS and the oncogenic G12V mutant in complex with the RA domain of RGL1 at < 2 Å resolution. KRASWT/RGL1-RA crystallized as a 1:1 heterodimer, whilst KRASG12V/RGL1-RA crystallized as a heterotetrameric structure in which RGL1-RA dimerized via domain-swapping the C-terminal beta-strand. Solution data indicated that KRASWT and KRASG12V in complex with RGL1-RA both exist predominantly as 1:1 dimers, while tetramerization occurs through very slow association. Through detailed structural analyses, the distance and angle between RAS α1 helix and RBD/RA α1 helix were found to differ significantly among RAS and RBD/RA complexes. The KRAS/RGL1-RA structures possess some of the largest α1RAS/α1Effector distances (21.7-22.2 Å), whereas the corresponding distances in previously reported RAS/RAF complexes are significantly shorter (15.2-17.7 Å). Contact map analysis identified unique structural signatures involving contacts between the ß1-ß2 loop of RA and the α1 helix of RAS, clearly distinguishing the KRAS/RGL1-RA (and other RAS/RA complexes) from RAS/RBD complexes. These results demonstrate that RAS effectors employ an assortment of finely-tuned docking surfaces to achieve optimal interactions with RAS.


Assuntos
Fatores de Troca do Nucleotídeo Guanina , Proteínas Proto-Oncogênicas p21(ras) , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Mutação , Domínios Proteicos , Proteínas Proto-Oncogênicas p21(ras)/química , Proteínas Proto-Oncogênicas p21(ras)/genética
7.
Proc Natl Acad Sci U S A ; 105(28): 9570-5, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18599462

RESUMO

Golgi alpha-mannosidase II (GMII) is a key glycosyl hydrolase in the N-linked glycosylation pathway. It catalyzes the removal of two different mannosyl linkages of GlcNAcMan(5)GlcNAc(2), which is the committed step in complex N-glycan synthesis. Inhibition of this enzyme has shown promise in certain cancers in both laboratory and clinical settings. Here we present the high-resolution crystal structure of a nucleophile mutant of Drosophila melanogaster GMII (dGMII) bound to its natural oligosaccharide substrate and an oligosaccharide precursor as well as the structure of the unliganded mutant. These structures allow us to identify three sugar-binding subsites within the larger active site cleft. Our results allow for the formulation of the complete catalytic process of dGMII, which involves a specific order of bond cleavage, and a major substrate rearrangement in the active site. This process is likely conserved for all GMII enzymes-but not in the structurally related lysosomal mannosidase-and will form the basis for the design of specific inhibitors against GMII.


Assuntos
Proteínas de Drosophila/química , Manosidases/química , Animais , Carboidratos/química , Domínio Catalítico , Cristalografia por Raios X , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Manosidases/genética , Manosidases/metabolismo , Mutação , Ligação Proteica
8.
J Med Chem ; 64(2): 1139-1169, 2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33444025

RESUMO

The essential eukaryotic chaperone Hsp90 regulates the form and function of diverse client proteins, many of which govern thermotolerance, virulence, and drug resistance in fungal species. However, use of Hsp90 inhibitors as antifungal therapeutics has been precluded by human host toxicities and suppression of immune responses. We recently described resorcylate aminopyrazoles (RAPs) as the first class of Hsp90 inhibitors capable of discriminating between fungal (Cryptococcus neoformans, Candida albicans) and human isoforms of Hsp90 in biochemical assays. Here, we report an iterative structure-property optimization toward RAPs capable of inhibiting C. neoformans growth in culture. In addition, we report the first X-ray crystal structures of C. neoformans Hsp90 nucleotide binding domain (NBD), as the apoprotein and in complexes with the non-species-selective Hsp90 inhibitor NVP-AUY922 and three RAPs revealing unique ligand-induced conformational rearrangements, which reaffirm the hypothesis that intrinsic differences in protein flexibility can confer selective inhibition of fungal versus human Hsp90 isoforms.


Assuntos
Antifúngicos/farmacologia , Cryptococcus neoformans/efeitos dos fármacos , Fungos/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Pirazóis/farmacologia , Animais , Antifúngicos/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Microssomos Hepáticos/metabolismo , Ligação Proteica , Pirazóis/química , Especificidade da Espécie , Relação Estrutura-Atividade
9.
Chembiochem ; 11(5): 673-80, 2010 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-20209559

RESUMO

Golgi alpha-mannosidase II (GMII) is a key enzyme in the N-glycosylation pathway and is a potential target for cancer chemotherapy. The natural product swainsonine is a potent inhibitor of GMII. In this paper we characterize the binding of 5alpha-substituted swainsonine analogues to the soluble catalytic domain of Drosophila GMII by X-ray crystallography. These inhibitors enjoy an advantage over previously reported GMII inhibitors in that they did not significantly decrease the inhibitory potential of the swainsonine head-group. The phenyl groups of these analogues occupy a portion of the binding site not previously seen to be populated with either substrate analogues or other inhibitors and they form novel hydrophobic interactions. They displace a well-organized water cluster, but the presence of a C(10) carbonyl allows the reestablishment of important hydrogen bonds. Already approximately tenfold more active against the Golgi enzyme than the lysosomal enzyme, these inhibitors offer the potential of being extended into the N-acetylglucosamine binding site of GMII for the creation of even more potent and selective GMII inhibitors.


Assuntos
Antineoplásicos/química , Complexo de Golgi/enzimologia , Manosidases/antagonistas & inibidores , Swainsonina/análogos & derivados , Animais , Antineoplásicos/farmacologia , Sítios de Ligação , Domínio Catalítico , Drosophila melanogaster/enzimologia , Manosidases/metabolismo , Ligação Proteica , Swainsonina/farmacologia
10.
Anal Biochem ; 399(1): 7-12, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20026005

RESUMO

A simple and reliable continuous assay for measurement of alpha-mannosidase activity is described and demonstrated for analysis with two recombinant human enzymes using the new substrate resorufin alpha-d-mannopyranoside (Res-Man). The product of enzyme reaction, resorufin, exhibits fluorescence emission at 585 nm with excitation at 571 nm and has a pK(a) of 5.8, allowing continuous measurement of fluorescence turnover at or near physiological pH values for human lysosomal and Drosophila Golgi alpha-mannosidases. The assay performed using recombinant Drosophila Golgi alpha-mannosidase (dGMII) has been shown to give the kinetic parameters K(m) of 200 microM and V(max) of 11 nmol/min per nmol dGMII. Methods for performing the assay using several concentrations of the known alpha-mannosidase inhibitor swainsonine are also presented, demonstrating a potential for use of the assay as a simple method for high-throughput screening of inhibitors potentially useful in cancer treatment.


Assuntos
Corantes Fluorescentes/química , Fluorometria/métodos , Manosídeos/metabolismo , Oxazinas/metabolismo , alfa-Manosidase/metabolismo , Animais , Drosophila , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Complexo de Golgi/enzimologia , Ensaios de Triagem em Larga Escala , Humanos , Cinética , Lisossomos/enzimologia , Manosídeos/síntese química , Manosídeos/química , Oxazinas/síntese química , Oxazinas/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Swainsonina/química , Swainsonina/farmacologia , alfa-Manosidase/genética
11.
Bioorg Med Chem ; 18(8): 2829-35, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20363144

RESUMO

The syntheses of an isomer of kotalanol, a naturally occurring glucosidase inhibitor, and of kotalanol itself are described. The target compounds were synthesized by nucleophilic attack of PMB-protected 1,4-anhydro-4-thio-d-arabinitol at the least hindered carbon atom of two 1,3-cyclic sulfates, which were synthesized from d-mannose. Methoxymethyl ether and isopropylidene were chosen as protecting groups. The latter group was critical to ensure the facile deprotection of the coupled products in a one-step sequence to yield kotalanol and its isomer. The stereoisomer of kotalanol, with the opposite stereochemistry at the C-6' stereogenic centre, inhibited the N-terminal catalytic domain of intestinal human maltase glucoamylase (ntMGAM) with a K(i) value of 0.20+/-0.02microM; this compares to a K(i) value for kotalanol of 0.19+/-0.03microM. The results indicate that the configuration at C-6' is inconsequential for inhibitory activity against this enzyme.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores de Glicosídeo Hidrolases , Monossacarídeos/síntese química , Sulfatos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Isomerismo , Manose/química , Monossacarídeos/química , Monossacarídeos/farmacologia , Sulfatos/química , Sulfatos/farmacologia , alfa-Glucosidases/metabolismo
12.
Chembiochem ; 10(2): 268-77, 2009 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-19101978

RESUMO

Mannostatin A is a potent inhibitor of the mannose-trimming enzyme, Golgi alpha-mannosidase II (GMII), which acts late in the N-glycan processing pathway. Inhibition of this enzyme provides a route to blocking the transformation-associated changes in cancer cell surface oligosaccharide structures. Here, we report on the synthesis of new Mannostatin derivatives and analyze their binding in the active site of Drosophila GMII by X-ray crystallography. The results indicate that the interaction with the backbone carbonyl of Arg876 is crucial to the high potency of the inhibitor-an effect enhanced by the hydrophobic interaction between the thiomethyl group and an aromatic pocket vicinal to the cleavage site. The various structures indicate that differences in the hydration of protein-ligand complexes are also important determinants of plasticity as well as selectivity of inhibitor binding.


Assuntos
Ciclopentanos/farmacologia , Inibidores Enzimáticos/farmacologia , Manosidases/antagonistas & inibidores , Animais , Domínio Catalítico , Cristalografia por Raios X , Ciclopentanos/síntese química , Ciclopentanos/química , Ciclopentanos/metabolismo , Drosophila melanogaster/enzimologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Manosidases/química , Manosidases/metabolismo , Especificidade por Substrato , Enxofre/química
13.
Biochemistry ; 47(38): 10058-68, 2008 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-18759458

RESUMO

The N-glycosylation pathway is a target for pharmaceutical intervention in a number of pathological conditions including cancer. Golgi alpha-mannosidase II (GMII) is the final glycoside hydrolase in the pathway and has been the target for a number of synthetic efforts aimed at providing more selective and effective inhibitors. Drosophila GMII (dGMII) has been extensively studied due to the ease of obtaining high resolution structural data, allowing the observation of substrate distortion upon binding and after formation of a trapped covalent reaction intermediate. However, attempts to find new inhibitor leads by high-throughput screening of large commercial libraries or through in silico docking were unsuccessful. In this paper we provide a kinetic and structural analysis of five inhibitors derived from a small glycosidase-focused library. Surprisingly, four of these were known inhibitors of beta-glucosidases. X-ray crystallographic analysis of the dGMII:inhibitor complexes highlights the ability of the zinc-containing GMII active site to deform compounds, even ones designed as conformationally restricted transition-state mimics of beta-glucosidases, into binding entities that have inhibitory activity. Although these deformed conformations do not appear to be on the expected conformational itinerary of the enzyme, and are thus not transition-state mimics of GMII, they allow positioning of the three vicinal hydroxyls of the bound gluco-inhibitors into similar locations to those found with mannose-containing substrates, underlining the importance of these hydrogen bonds for binding. Further, these studies show the utility of targeting the acid-base catalyst using appropriately positioned positively charged nitrogen atoms, as well as the challenges associated with aglycon substitutions.


Assuntos
Inibidores Enzimáticos/química , Glicosídeo Hidrolases/antagonistas & inibidores , Glicosídeo Hidrolases/química , Complexo de Golgi/enzimologia , Manosidases/antagonistas & inibidores , Manosidases/química , Animais , Células Cultivadas , Cristalografia por Raios X , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/enzimologia , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Complexo de Golgi/efeitos dos fármacos , Estrutura Secundária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/fisiologia , Proteínas/antagonistas & inibidores , Proteínas/química
14.
Proteins ; 71(3): 1484-96, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18076078

RESUMO

Retaining glycosidases operate by a two-step catalytic mechanism in which the transition states are characterized by buildup of a partial positive charge at the anomeric center. Sulfonium-ion analogues of the naturally occurring glycosidase inhibitors, swainsonine and 8-epi-lentiginosine, in which the bridgehead nitrogen atom is replaced by a sulfonium-ion, were synthesized in order to test the hypothesis that a sulfonium salt carrying a permanent positive charge would be an effective glycosidase inhibitor. Initial prediction based on computational docking indicated three plausible binding modes to Drosophila Golgi alpha-mannosidase II (dGMII), the most likely being close to that of swainsonine. Observation of the binding of di-epi-thioswainsonine and 8-epi-thiolentiginosine to dGMII from crystallographic data, however, revealed an orientation different from swainsonine in the active site. Screening these two compounds against dGMII shows that they are inhibitors with IC(50) values of 2.0 and 0.014 mM, respectively. This dramatic difference in affinity between the two compounds, which differ by only one hydroxyl group, is rationalized in terms of bound water molecules and the water molecule substructure in the active site, as identified by comparison of high resolution X-ray crystal structures of several dGMII-inhibitor complexes.


Assuntos
Alcaloides/química , Proteínas de Drosophila/química , Manosidases/química , Compostos de Sulfônio/química , Swainsonina/análogos & derivados , Swainsonina/química , Água/química , Alcaloides/metabolismo , Animais , Sítios de Ligação , Cristalografia por Raios X , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Manosidases/antagonistas & inibidores , Manosidases/metabolismo , Ligação Proteica , Compostos de Sulfônio/metabolismo , Swainsonina/metabolismo , Água/fisiologia
15.
J Am Chem Soc ; 130(28): 8975-83, 2008 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-18558690

RESUMO

Inhibition of Golgi alpha-mannosidase II (GMII), which acts late in the N-glycan processing pathway, provides a route to blocking cancer-induced changes in cell surface oligosaccharide structures. To probe the substrate requirements of GMII, oligosaccharides were synthesized that contained an alpha(1,3)- or alpha(1,6)-linked 1-thiomannoside. Surprisingly, these oligosaccharides were not observed in X-ray crystal structures of native Drosophila GMII (dGMII). However, a mutant enzyme in which the catalytic nucleophilic aspartate was changed to alanine (D204A) allowed visualization of soaked oligosaccharides and led to the identification of the binding site for the alpha(1,3)-linked mannoside of the natural substrate. These studies also indicate that the conformational change of the bound mannoside to a high-energy B 2,5 conformation is facilitated by steric hindrance from, and the formation of strong hydrogen bonds to, Asp204. The observation that 1-thio-linked mannosides are not well tolerated by the catalytic site of dGMII led to the synthesis of a pentasaccharide containing the alpha(1,6)-linked Man of the natural substrate and the beta(1,2)-linked GlcNAc moiety proposed to be accommodated by the extended binding site of the enzyme. A cocrystal structure of this compound with the D204A enzyme revealed the molecular interactions with the beta(1,2)-linked GlcNAc. The structure is consistent with the approximately 80-fold preference of dGMII for the cleavage of substrates containing a nonreducing beta(1,2)-linked GlcNAc. By contrast, the lysosomal mannosidase lacks an equivalent GlcNAc binding site and kinetic analysis indicates oligomannoside substrates without non-reducing-terminal GlcNAc modifications are preferred, suggesting that selective inhibitors for GMII could exploit the additional binding specificity of the GlcNAc binding site.


Assuntos
Manosidases/metabolismo , Oligossacarídeos/metabolismo , Animais , Sítios de Ligação , Catálise , Cristalografia por Raios X , Drosophila/enzimologia , Drosophila/genética , Glicosilação , Manose/química , Manose/metabolismo , Manosidases/química , Manosidases/genética , Modelos Moleculares , Mutação , Oligossacarídeos/síntese química , Oligossacarídeos/química , Relação Estrutura-Atividade , Especificidade por Substrato , Tioglicosídeos/química , Tioglicosídeos/metabolismo
16.
Bioorg Med Chem ; 16(15): 7337-46, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18599296

RESUMO

Refining the chemical structure of functionalized pyrrolidine-based inhibitors of Golgi alpha-mannosidase II (GMII) to optimize binding affinity provided a lead molecule that demonstrated nanomolar competitive inhibition of alpha-mannosidases II and an optimal fit in the active site of Drosophila GMII by X-ray crystallography. Esters of this lead compound also inhibited the growth of human glioblastoma and brain-derived endothelial cells more than the growth of non-tumoral human fibroblasts, suggesting their potential for anti-cancer therapy.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Pirrolidinas/química , Pirrolidinas/farmacologia , alfa-Manosidase/antagonistas & inibidores , alfa-Manosidase/classificação , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Drosophila/enzimologia , Células Endoteliais/efeitos dos fármacos , Fabaceae/enzimologia , Glioblastoma , Humanos , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
17.
Proteins ; 69(1): 160-76, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17557336

RESUMO

Golgi alpha-mannosidase II (GMII), a zinc-dependent glycosyl hydrolase, is a promising target for drug development in anti-tumor therapies. Using X-ray crystallography, we have determined the structure of Drosophila melanogaster GMII (dGMII) complexed with three different inhibitors exhibiting IC50's ranging from 80 to 1000 microM. These structures, along with those of seven other available dGMII/inhibitor complexes, were then used as a basis for the evaluation of seven docking programs (GOLD, Glide, FlexX, AutoDock, eHiTS, LigandFit, and FITTED). We found that small inhibitors could be accurately docked by most of the software, while docking of larger compounds (i.e., those with extended aromatic cycles or long aliphatic chains) was more problematic. Overall, Glide provided the best docking results, with the most accurately predicted binding around the active site zinc atom. Further evaluation of Glide's performance revealed its ability to extract active compounds from a benchmark library of decoys.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Manosidases/antagonistas & inibidores , Manosidases/metabolismo , Algoritmos , Sítios de Ligação , Biologia Computacional , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Manosidases/química , Modelos Moleculares , Conformação Proteica , Software , Relação Estrutura-Atividade
18.
FEBS J ; 273(12): 2673-83, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16817895

RESUMO

Inhibitors targeting pancreatic alpha-amylase and intestinal alpha-glucosidases delay glucose production following digestion and are currently used in the treatment of Type II diabetes. Maltase-glucoamylase (MGA), a family 31 glycoside hydrolase, is an alpha-glucosidase anchored in the membrane of small intestinal epithelial cells responsible for the final step of mammalian starch digestion leading to the release of glucose. This paper reports the production and purification of active human recombinant MGA amino terminal catalytic domain (MGAnt) from two different eukaryotic cell culture systems. MGAnt overexpressed in Drosophila cells was of quality and quantity suitable for kinetic and inhibition studies as well as future structural studies. Inhibition of MGAnt was tested with a group of prospective alpha-glucosidase inhibitors modeled after salacinol, a naturally occurring alpha-glucosidase inhibitor, and acarbose, a currently prescribed antidiabetic agent. Four synthetic inhibitors that bind and inhibit MGAnt activity better than acarbose, and at comparable levels to salacinol, were found. The inhibitors are derivatives of salacinol that contain either a selenium atom in place of sulfur in the five-membered ring, or a longer polyhydroxylated, sulfated chain than salacinol. Six-membered ring derivatives of salacinol and compounds modeled after miglitol were much less effective as MGAnt inhibitors. These results provide information on the inhibitory profile of MGAnt that will guide the development of new compounds having antidiabetic activity.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores de Glicosídeo Hidrolases , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Álcoois Açúcares/química , Álcoois Açúcares/farmacologia , Sulfatos/química , Sulfatos/farmacologia , Acarbose/metabolismo , Acarbose/farmacologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Cinética , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Álcoois Açúcares/síntese química , Sulfatos/síntese química , Transfecção , alfa-Glucosidases/metabolismo
19.
PLoS One ; 7(8): e43437, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916261

RESUMO

BACKGROUND: The non-receptor tyrosine kinase JAK2 is implicated in a group of myeloproliferative neoplasms including polycythemia vera, essential thrombocythemia, and primary myelofibrosis. JAK2-selective inhibitors are currently being evaluated in clinical trials. Data from drug-resistant chronic myeloid leukemia patients demonstrate that treatment with a small-molecule inhibitor generates resistance via mutation or amplification of BCR-ABL. We hypothesize that treatment with small molecule inhibitors of JAK2 will similarly generate inhibitor-resistant mutants in JAK2. METHODOLOGY: In order to identify inhibitor-resistant JAK2 mutations a priori, we utilized TEL-JAK2 to conduct an in vitro random mutagenesis screen for JAK2 alleles resistant to JAK Inhibitor-I. Isolated mutations were evaluated for their ability to sustain cellular growth, stimulate downstream signaling pathways, and phosphorylate a novel JAK2 substrate in the presence of inhibitor. CONCLUSIONS: Mutations were found exclusively in the kinase domain of JAK2. The panel of mutations conferred resistance to high concentrations of inhibitor accompanied by sustained activation of the Stat5, Erk1/2, and Akt pathways. Using a JAK2 substrate, enhanced catalytic activity of the mutant JAK2 kinase was observed in inhibitor concentrations 200-fold higher than is inhibitory to the wild-type protein. When testing the panel of mutations in the context of the Jak2 V617F allele, we observed that a subset of mutations conferred resistance to inhibitor, validating the use of TEL-JAK2 in the initial screen. These results demonstrate that small-molecule inhibitors select for JAK2 inhibitor-resistant alleles, and the design of next-generation JAK2 inhibitors should consider the location of mutations arising in inhibitor-resistant screens.


Assuntos
Janus Quinase 2/química , Janus Quinase 2/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Linhagem Celular , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Humanos , Janus Quinase 2/genética , Mutagênese/genética , Mutagênese/fisiologia
20.
Structure ; 19(11): 1535-6, 2011 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22078550

RESUMO

Glycoside hydrolases are the tools that pathogenic bacteria use to cut through the defensive glycan structure on host cell surfaces. In this issue of Structure, Pluvinage et al. (2011) report how a bacterial polypeptide with more than one hydrolase module broadens the effective substrate specificity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA