Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Oncol ; 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38506049

RESUMEN

An immunosuppressive tumor microenvironment promotes tumor growth and is one of the main factors limiting the response to cancer immunotherapy. We have previously reported that inhibition of vacuolar protein sorting 34 (VPS34), a crucial lipid kinase in the autophagy/endosomal trafficking pathway, decreases tumor growth in several cancer models, increases infiltration of immune cells and sensitizes tumors to anti-programmed cell death protein 1/programmed cell death 1 ligand 1 therapy by upregulation of C-C motif chemokine 5 (CCL5) and C-X-C motif chemokine 10 (CXCL10) chemokines. The purpose of this study was to investigate the signaling mechanism leading to the VPS34-dependent chemokine increase. NanoString gene expression analysis was applied to tumors from mice treated with the VPS34 inhibitor SB02024 to identify key pathways involved in the anti-tumor response. We showed that VPS34 inhibitors increased the secretion of T-cell-recruitment chemokines in a cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes protein (STING)-dependent manner in cancer cells. Both pharmacological and small interfering RNA (siRNA)-mediated VPS34 inhibition increased cGAS/STING-mediated expression and secretion of CCL5 and CXCL10. The combination of VPS34 inhibitor and STING agonist further induced cytokine release in both human and murine cancer cells as well as monocytic or dendritic innate immune cells. Finally, the VPS34 inhibitor SB02024 sensitized B16-F10 tumor-bearing mice to STING agonist treatment and significantly improved mice survival. These results show that VPS34 inhibition augments the cGAS/STING pathway, leading to greater tumor control through immune-mediated mechanisms. We propose that pharmacological VPS34 inhibition may synergize with emerging therapies targeting the cGAS/STING pathway.

3.
Sci Transl Med ; 10(428)2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29444979

RESUMEN

Cancer cells adapt to their inherently increased oxidative stress through activation of the glutathione (GSH) and thioredoxin (TXN) systems. Inhibition of both of these systems effectively kills cancer cells, but such broad inhibition of antioxidant activity also kills normal cells, which is highly unwanted in a clinical setting. We therefore evaluated targeting of the TXN pathway alone and, more specifically, selective inhibition of the cytosolic selenocysteine-containing enzyme TXN reductase 1 (TXNRD1). TXNRD1 inhibitors were discovered in a large screening effort and displayed increased specificity compared to pan-TXNRD inhibitors, such as auranofin, that also inhibit the mitochondrial enzyme TXNRD2 and additional targets. For our lead compounds, TXNRD1 inhibition correlated with cancer cell cytotoxicity, and inhibitor-triggered conversion of TXNRD1 from an antioxidant to a pro-oxidant enzyme correlated with corresponding increases in cellular production of H2O2 In mice, the most specific TXNRD1 inhibitor, here described as TXNRD1 inhibitor 1 (TRi-1), impaired growth and viability of human tumor xenografts and syngeneic mouse tumors while having little mitochondrial toxicity and being better tolerated than auranofin. These results display the therapeutic anticancer potential of irreversibly targeting cytosolic TXNRD1 using small molecules and present potent and selective TXNRD1 inhibitors. Given the pronounced up-regulation of TXNRD1 in several metastatic malignancies, it seems worthwhile to further explore the potential benefit of specific irreversible TXNRD1 inhibitors for anticancer therapy.


Asunto(s)
Citosol/enzimología , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Tiorredoxina Reductasa 1/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones SCID , Oxidación-Reducción , Relación Estructura-Actividad , Tiorredoxina Reductasa 1/química , Tiorredoxina Reductasa 1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Med Chem ; 61(6): 2533-2551, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29485874

RESUMEN

Recent literature has both suggested and questioned MTH1 as a novel cancer target. BAY-707 was just published as a target validation small molecule probe for assessing the effects of pharmacological inhibition of MTH1 on tumor cell survival, both in vitro and in vivo. (1) In this report, we describe the medicinal chemistry program creating BAY-707, where fragment-based methods were used to develop a series of highly potent and selective MTH1 inhibitors. Using structure-based drug design and rational medicinal chemistry approaches, the potency was increased over 10,000 times from the fragment starting point while maintaining high ligand efficiency and drug-like properties.


Asunto(s)
Antineoplásicos/farmacología , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Morfolinas/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Células CACO-2 , Permeabilidad de la Membrana Celular , Diseño de Fármacos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Hepatocitos/metabolismo , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Estructura Molecular , Morfolinas/química , Morfolinas/farmacocinética , Ratas , Ratas Wistar , Relación Estructura-Actividad
5.
ACS Chem Biol ; 12(8): 1986-1992, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28679043

RESUMEN

MTH1 is a hydrolase responsible for sanitization of oxidized purine nucleoside triphosphates to prevent their incorporation into replicating DNA. Early tool compounds published in the literature inhibited the enzymatic activity of MTH1 and subsequently induced cancer cell death; however recent studies have questioned the reported link between these two events. Therefore, it is important to validate MTH1 as a cancer dependency with high quality chemical probes. Here, we present BAY-707, a substrate-competitive, highly potent and selective inhibitor of MTH1, chemically distinct compared to those previously published. Despite superior cellular target engagement and pharmacokinetic properties, inhibition of MTH1 with BAY-707 resulted in a clear lack of in vitro or in vivo anticancer efficacy either in mono- or in combination therapies. Therefore, we conclude that MTH1 is dispensable for cancer cell survival.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Sistemas de Liberación de Medicamentos , Morfolinas/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Células CACO-2 , Células Cultivadas , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Células HeLa , Hepatocitos/efectos de los fármacos , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Microsomas Hepáticos/efectos de los fármacos , Modelos Moleculares , Morfolinas/química , Neoplasias/fisiopatología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Pirimidinas/química , Pirimidinas/farmacología , Ratas
6.
J Med Chem ; 60(4): 1262-1271, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28001384

RESUMEN

Selective inhibitors could help unveil the mechanisms by which inhibition of poly(ADP-ribose) polymerases (PARPs) elicits clinical benefits in cancer therapy. We profiled 10 clinical PARP inhibitors and commonly used research tools for their inhibition of multiple PARP enzymes. We also determined crystal structures of these compounds bound to PARP1 or PARP2. Veliparib and niraparib are selective inhibitors of PARP1 and PARP2; olaparib, rucaparib, and talazoparib are more potent inhibitors of PARP1 but are less selective. PJ34 and UPF1069 are broad PARP inhibitors; PJ34 inserts a flexible moiety into hydrophobic subpockets in various ADP-ribosyltransferases. XAV939 is a promiscuous tankyrase inhibitor and a potent inhibitor of PARP1 in vitro and in cells, whereas IWR1 and AZ-6102 are tankyrase selective. Our biochemical and structural analysis of PARP inhibitor potencies establishes a molecular basis for either selectivity or promiscuity and provides a benchmark for experimental design in assessment of PARP inhibitor effects.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Tanquirasas/antagonistas & inhibidores , Animales , Bencimidazoles/química , Bencimidazoles/farmacología , Células HEK293 , Humanos , Indazoles/química , Indazoles/farmacología , Modelos Moleculares , Fenantrenos/química , Fenantrenos/farmacología , Ftalazinas/química , Ftalazinas/farmacología , Piperazinas/química , Piperazinas/farmacología , Piperidinas/química , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Tanquirasas/metabolismo
7.
Biochemistry ; 55(9): 1384-97, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26854536

RESUMEN

The inositol polyphosphate 5-phosphatase INPP5B hydrolyzes the 5-phosphate group from water- and lipid-soluble signaling messengers. Two synthetic benzene and biphenyl polyphosphates (BzP/BiPhPs), simplified surrogates of inositol phosphates and phospholipid headgroups, were identified by thermodynamic studies as potent INPP5B ligands. The X-ray structure of the complex between INPP5B and biphenyl 3,3',4,4',5,5'-hexakisphosphate [BiPh(3,3',4,4',5,5')P6, IC50 5.5 µM] was determined at 2.89 Å resolution. One inhibitor pole locates in the phospholipid headgroup binding site and the second solvent-exposed ring binds to the His-Tag of another INPP5B molecule, while a molecule of inorganic phosphate is also present in the active site. Benzene 1,2,3-trisphosphate [Bz(1,2,3)P3] [one ring of BiPh(3,3',4,4',5,5')P6] inhibits INPP5B ca. 6-fold less potently. Co-crystallization with benzene 1,2,4,5-tetrakisphosphate [Bz(1,2,4,5)P4, IC50 = 6.3 µM] yielded a structure refined at 2.9 Å resolution. Conserved residues among the 5-phosphatase family mediate interactions with Bz(1,2,4,5)P4 and BiPh(3,3',4,4',5,5')P6 similar to those with the polar groups present in positions 1, 4, 5, and 6 on the inositol ring of the substrate. 5-Phosphatase specificity most likely resides in the variable zone located close to the 2- and 3-positions of the inositol ring, offering insights to inhibitor design. We propose that the inorganic phosphate present in the INPP5B-BiPh(3,3',4,4',5,5')P6 complex mimics the postcleavage substrate 5-phosphate released by INPP5B in the catalytic site, allowing elucidation of two new key features in the catalytic mechanism proposed for the family of phosphoinositide 5-phosphatases: first, the involvement of the conserved Arg-451 in the interaction with the 5-phosphate and second, identification of the water molecule that initiates 5-phosphate hydrolysis. Our model also has implications for the proposed "moving metal" mechanism.


Asunto(s)
Fosfatos de Inositol/química , Fosfatos de Inositol/metabolismo , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Sitios de Unión/fisiología , Cristalografía por Rayos X , Estructura Secundaria de Proteína
8.
FEBS J ; 283(6): 1107-23, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26783088

RESUMEN

UNLABELLED: Human sphingomyelinase phosphodiesterase like 3a (SMPDL3a) is a secreted enzyme that shares a conserved catalytic domain with human acid sphingomyelinase (aSMase), the enzyme carrying mutations causative of Niemann-Pick disease. We have solved the structure of SMPDL3a revealing a calcineurin-like fold. A dimetal site, glycosylation pattern and a disulfide bond network are likely to be conserved also in human aSMase. We show that the binuclear site of SMPDL3a is occupied by two Zn(2+) ions and that excess Zn(2+) leads to inhibition of enzyme activity through binding to additional sites. As an extension of recent biochemical work we uncovered that SMPDL3a catalyses the hydrolysis of several modified nucleotides that include cytidine 5'-diphosphocholine, cytidine diphosphate ethanolamine and ADP-ribose, but not the aSMase substrate, sphingomyelin. We subsequently determined the structure of SMPDL3a in complex with the product 5'-cytidine monophosphate (CMP), a structure that is consistent with several distinct coordination modes of the substrate/product in the active site during the reaction cycle. Based on the structure of CMP complexes, we propose a phosphoryl transfer mechanism for SMPDL3a. Finally, a homology model of human aSMase was constructed to allow for the mapping of selected Niemann-Pick disease mutations on a three-dimensional framework to guide further characterization of their effects on aSMase function. DATABASE: Structural data are available in the PDB database under the accession numbers 5EBB and 5EBE.


Asunto(s)
Esfingomielina Fosfodiesterasa/química , Esfingomielina Fosfodiesterasa/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , Citidina Monofosfato/metabolismo , Disulfuros/química , Glicosilación , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Enfermedades de Niemann-Pick/enzimología , Enfermedades de Niemann-Pick/genética , Filogenia , Mutación Puntual , Conformación Proteica , Homología de Secuencia de Aminoácido , Esfingomielina Fosfodiesterasa/genética , Electricidad Estática , Especificidad por Sustrato , Zinc/metabolismo
9.
PLoS One ; 10(6): e0131507, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121039

RESUMEN

Human NUDT16 is a member of the NUDIX hydrolase superfamily. After having been initially described as an mRNA decapping enzyme, recent studies conferred it a role as an "housecleaning" enzyme specialized in the removal of hazardous (deoxy)inosine diphosphate from the nucleotide pool. Here we present the crystal structure of human NUDT16 both in its apo-form and in complex with its product inosine monophosphate (IMP). NUDT16 appears as a dimer whose formation generates a positively charged trench to accommodate substrate-binding. Complementation of the structural data with detailed enzymatic and biophysical studies revealed the determinants of substrate recognition and particularly the importance of the substituents in position 2 and 6 on the purine ring. The affinity for the IMP product, harboring a carbonyl in position 6 on the base, compared to purine monophosphates lacking a H-bond acceptor in this position, implies a catalytic cycle whose rate is primarily regulated by the product-release step. Finally, we have also characterized a phenomenon of inhibition by the product of the reaction, IMP, which might exclude non-deleterious nucleotides from NUDT16-mediated hydrolysis regardless of their cellular concentration. Taken together, this study details structural and regulatory mechanisms explaining how substrates are selected for hydrolysis by human NUDT16.


Asunto(s)
Inosina Monofosfato/metabolismo , Pirofosfatasas/química , Pirofosfatasas/metabolismo , Adenosina Difosfato/metabolismo , Biocatálisis/efectos de los fármacos , Calorimetría , Secuencia Conservada , Cristalografía por Rayos X , Humanos , Inosina Monofosfato/farmacología , Cinética , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos
10.
Structure ; 22(5): 744-55, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24704254

RESUMEN

SHIP2, OCRL, and INPP5B belong to inositol polyphosphate 5-phophatase subfamilies involved in insulin regulation and Lowes syndrome. The structural basis for membrane recognition, substrate specificity, and regulation of inositol polyphosphate 5-phophatases is still poorly understood. We determined the crystal structures of human SHIP2, OCRL, and INPP5B, the latter in complex with phosphoinositide substrate analogs, which revealed a membrane interaction patch likely to assist in sequestering substrates from the lipid bilayer. Residues recognizing the 1-phosphate of the substrates are highly conserved among human family members, suggesting similar substrate binding modes. However, 3- and 4-phosphate recognition varies and determines individual substrate specificity profiles. The high conservation of the environment of the scissile 5-phosphate suggests a common reaction geometry for all members of the human 5-phosphatase family.


Asunto(s)
Membrana Celular/metabolismo , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Fosfatos de Inositol/química , Fosfatos de Inositol/metabolismo , Modelos Moleculares , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Fosfatidilinositoles/química , Fosfatidilinositoles/metabolismo , Especificidad por Sustrato
11.
J Mol Biol ; 425(22): 4323-33, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23816837

RESUMEN

Guanine monophosphate (GMP) synthetase is a bifunctional two-domain enzyme. The N-terminal glutaminase domain generates ammonia from glutamine and the C-terminal synthetase domain aminates xanthine monophosphate (XMP) to form GMP. Mammalian GMP synthetases (GMPSs) contain a 130-residue-long insert in the synthetase domain in comparison to bacterial proteins. We report here the structure of a eukaryotic GMPS. Substrate XMP was bound in the crystal structure of the human GMPS enzyme. XMP is bound to the synthetase domain and covered by a LID motif. The enzyme forms a dimer in the crystal structure with subunit orientations entirely different from the bacterial counterparts. The inserted sub-domain is shown to be involved in substrate binding and dimerization. Furthermore, the structural basis for XMP recognition is revealed as well as a potential allosteric site. Enzymes in the nucleotide metabolism typically display an increased activity in proliferating cells due to the increased need for nucleotides. Many drugs used as immunosuppressants and for treatment of cancer and viral diseases are indeed nucleobase- and nucleoside-based compounds, which are acting on or are activated by enzymes in this pathway. The information obtained from the crystal structure of human GMPS might therefore aid in understanding interactions of nucleoside-based drugs with GMPS and in structure-based design of GMPS-specific inhibitors.


Asunto(s)
Ligasas de Carbono-Nitrógeno/química , Ligasas de Carbono-Nitrógeno/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Escherichia coli/química , Escherichia coli/enzimología , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Especificidad por Sustrato
12.
EMBO Rep ; 14(9): 804-10, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23867627

RESUMEN

Short-chain peptides are transported across membranes through promiscuous proton-dependent oligopeptide transporters (POTs)--a subfamily of the major facilitator superfamily (MFS). The human POTs, PEPT1 and PEPT2, are also involved in the absorption of various drugs in the gut as well as transport to target cells. Here, we present a structure of an oligomeric POT transporter from Shewanella oneidensis (PepTSo2), which was crystallized in the inward open conformation in complex with the peptidomimetic alafosfalin. All ligand-binding residues are highly conserved and the structural insights presented here are therefore likely to also apply to human POTs.


Asunto(s)
Proteínas Bacterianas/química , Shewanella/química , Simportadores/química , Alanina/análogos & derivados , Alanina/farmacología , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Unión Proteica , Shewanella/metabolismo , Simportadores/metabolismo
13.
Nat Struct Mol Biol ; 20(6): 766-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624861

RESUMEN

Here we present two structures of the major facilitator (MFS) xylose transporter XylE from Escherichia coli in inward open and partially occluded inward open conformations. These structures provide key information about the transport cycle of XylE and the closely related human GLUT transporters. This is, to our knowledge, the first MFS transporter structure determined in more than one conformational state, which may establish XylE as an important MFS model protein.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Simportadores/química , Simportadores/metabolismo , Cristalografía por Rayos X , Escherichia coli/química , Escherichia coli/metabolismo , Modelos Moleculares , Conformación Proteica
14.
J Lipid Res ; 54(6): 1630-1643, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526831

RESUMEN

Lipocalin prostaglandin D synthase (L-PGDS) regulates synthesis of an important inflammatory and signaling mediator, prostaglandin D2 (PGD2). Here, we used structural, biophysical, and biochemical approaches to address the mechanistic aspects of substrate entry, catalysis, and product exit of this enzyme. Structure of human L-PGDS was solved in a complex with a substrate analog (SA) and in ligand-free form. Its catalytic Cys 65 thiol group was found in two different conformations, each making a distinct hydrogen bond network to neighboring residues. These help in elucidating the mechanism of the cysteine nucleophile activation. Electron density for ligand observed in the active site defined the substrate binding regions, but did not allow unambiguous fitting of the SA. To further understand ligand binding, we used NMR spectroscopy to map the binding sites and to show the dynamics of protein-substrate and protein-product interactions. A model for ligand binding at the catalytic site is proposed, showing a second binding site involved in ligand exit and entry. NMR chemical shift perturbations and NMR resonance line-width alterations (observed as changes of intensity in two-dimensional cross-peaks in [¹H,¹5N]-transfer relaxation optimization spectroscopy) for residues at the Ω loop (A-B loop), E-F loop, and G-H loop besides the catalytic sites indicate involvement of these residues in ligand entry/egress.


Asunto(s)
Oxidorreductasas Intramoleculares/química , Lipocalinas/química , Simulación de Dinámica Molecular , Catálisis , Dominio Catalítico , Humanos , Resonancia Magnética Nuclear Biomolecular/métodos , Unión Proteica , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Especificidad por Sustrato
15.
ACS Chem Biol ; 7(5): 822-8, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22330088

RESUMEN

Phosphoinositides regulate many cellular processes, and cellular levels are controlled by kinases and phosphatases. SHIP2 (SH2 (Src homology 2)-domain-containing inositol-phosphatase-2) plays a critical role in phosphoinositide signaling, cleaving the 5-phosphate from phosphatidylinositol 3,4,5-trisphosphate. SHIP2 is thought to be involved in type-2 diabetes and obesity, conditions that could therefore be open to pharmacological modulation of the enzyme. However, rational design of SHIP2 inhibitors has been limited by the absence of a high-resolution structure. Here, we present a 2.1 Å resolution crystal structure of the phosphatase domain of SHIP2 bound to the synthetic ligand biphenyl 2,3',4,5',6-pentakisphosphate (BiPh(2,3',4,5',6)P(5)). BiPh(2,3',4,5',6)P(5) is not a SHIP2 substrate but inhibits Ins(1,3,4,5)P(4) hydrolysis with an IC(50) of 24.8 ± 3.0 µM, (K(m) for Ins(1,3,4,5)P(4) is 215 ± 28 µM). Molecular dynamics simulations suggest that when BiPh(2,3',4,5',6)P(5) binds to SHIP2, a flexible loop folds over and encloses the ligand. Compounds targeting such a closed conformation might therefore deliver SHIP2-specific drugs.


Asunto(s)
Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Diseño de Fármacos , Fosfatos/química , Fosfatos/farmacología , Fosfatos de Fosfatidilinositol/química , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/química , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Fosfatos de Fosfatidilinositol/farmacología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/metabolismo , Dominios Homologos src
16.
FEBS J ; 277(23): 4920-30, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21054786

RESUMEN

Human hypoxanthine-guanine phosphoribosyltransferase (HPRT) (EC 2.4.2.8) catalyzes the conversion of hypoxanthine and guanine to their respective nucleoside monophosphates. Human HPRT deficiency as a result of genetic mutations is linked to both Lesch-Nyhan disease and gout. In the present study, we have characterized phosphoribosyltransferase domain containing protein 1 (PRTFDC1), a human HPRT homolog of unknown function. The PRTFDC1 structure has been determined at 1.7 Å resolution with bound GMP. The overall structure and GMP binding mode are very similar to that observed for HPRT. Using a thermal-melt assay, a nucleotide metabolome library was screened against PRTFDC1 and revealed that hypoxanthine and guanine specifically interacted with the enzyme. It was subsequently confirmed that PRTFDC1 could convert these two bases into their corresponding nucleoside monophosphate. However, the catalytic efficiency (k(cat)/K(m)) of PRTFDC1 towards hypoxanthine and guanine was only 0.26% and 0.09%, respectively, of that of HPRT. This low activity could be explained by the fact that PRTFDC1 has a Gly in the position of the proposed catalytic Asp of HPRT. In PRTFDC1, a water molecule at the position of the aspartic acid side chain position in HPRT might be responsible for the low activity observed by acting as a weak base. The data obtained in the present study indicate that PRTFDC1 does not have a direct catalytic role in the nucleotide salvage pathway.


Asunto(s)
Hipoxantina Fosforribosiltransferasa/química , Hipoxantina Fosforribosiltransferasa/metabolismo , Secuencia de Aminoácidos , Clonación Molecular , Cristalografía por Rayos X , Guanina/metabolismo , Guanosina Monofosfato/metabolismo , Humanos , Hipoxantina/metabolismo , Hipoxantina Fosforribosiltransferasa/genética , Técnicas In Vitro , Cinética , Metaboloma , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Especificidad por Sustrato
17.
PLoS One ; 5(9): e12907, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20957027

RESUMEN

UNLABELLED: The human SnoN is an oncoprotein that interacts with several transcription-regulatory proteins such as the histone-deacetylase, N-CoR containing co-repressor complex and Smad proteins. This study presents the crystal structure of the Dachshund homology domain of human SnoN. The structure reveals a groove composed of conserved residues with characteristic properties of a protein-interaction surface. A comparison of the 12 monomers in the asymmetric unit reveals the presence of two major conformations: an open conformation with a well accessible groove and a tight conformation with a less accessible groove. The variability in the backbone between the open and the tight conformations matches the differences seen in previously determined structures of individual Dachshund homology domains, suggesting a general plasticity within this fold family. The flexibility observed in the putative protein binding groove may enable SnoN to recognize multiple interaction partners. ENHANCED VERSION: This article can also be viewed as an enhanced version in which the text of the article is integrated with interactive 3D representations and animated transitions. Please note that a web plugin is required to access this enhanced functionality. Instructions for the installation and use of the web plugin are available in Text S1.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Proto-Oncogénicas/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Alineación de Secuencia
18.
Nucleic Acids Res ; 38(20): 7308-19, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20631005

RESUMEN

Human purine de novo synthesis pathway contains several multi-functional enzymes, one of which, tri-functional GART, contains three enzymatic activities in a single polypeptide chain. We have solved structures of two domains bearing separate catalytic functions: glycinamide ribonucleotide synthetase and aminoimidazole ribonucleotide synthetase. Structures are compared with those of homologous enzymes from prokaryotes and analyzed in terms of the catalytic mechanism. We also report small angle X-ray scattering models for the full-length protein. These models are consistent with the enzyme forming a dimer through the middle domain. The protein has an approximate seesaw geometry where terminal enzyme units display high mobility owing to flexible linker segments. This resilient seesaw shape may facilitate internal substrate/product transfer or forwarding to other enzymes in the pathway.


Asunto(s)
Ligasas de Carbono-Nitrógeno/química , Fosforribosilglicinamida-Formiltransferasa/química , Adenosina Trifosfato/química , Sitios de Unión , Dominio Catalítico , Cristalografía , Glicina/química , Humanos , Modelos Moleculares , Estructura Cuaternaria de Proteína , Ribosamonofosfatos/química , Dispersión del Ángulo Pequeño , Difracción de Rayos X
19.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 65(Pt 11): 1091-6, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19923725

RESUMEN

The inhibitor of apoptosis (IAP) family of proteins contains key modulators of apoptosis and inflammation that interact with caspases through baculovirus IAP-repeat (BIR) domains. Overexpression of IAP proteins frequently occurs in cancer cells, thus counteracting the activated apoptotic program. The IAP proteins have therefore emerged as promising targets for cancer therapy. In this work, X-ray crystallography was used to determine the first structures of BIR domains from human NAIP and cIAP2. Both structures harbour an N-terminal tetrapeptide in the conserved peptide-binding groove. The structures reveal that these two proteins bind the tetrapeptides in a similar mode as do other BIR domains. Detailed interactions are described for the P1'-P4' side chains of the peptide, providing a structural basis for peptide-specific recognition. An arginine side chain in the P3' position reveals favourable interactions with its hydrophobic moiety in the binding pocket, while hydrophobic residues in the P2' and P4' pockets make similar interactions to those seen in other BIR domain-peptide complexes. The structures also reveal how a serine in the P1' position is accommodated in the binding pockets of NAIP and cIAP2. In addition to shedding light on the specificity determinants of these two proteins, the structures should now also provide a framework for future structure-based work targeting these proteins.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/química , Proteína Inhibidora de la Apoptosis Neuronal/química , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Cristalografía por Rayos X , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Modelos Moleculares , Datos de Secuencia Molecular , Proteína Inhibidora de la Apoptosis Neuronal/genética , Estructura Terciaria de Proteína/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Ubiquitina-Proteína Ligasas
20.
FEBS Lett ; 582(25-26): 3590-4, 2008 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-18812174

RESUMEN

Evasion of apoptosis is recognized as a characteristic of malignant growth. Anti-apoptotic B-cell lymphoma-2 (Bcl-2) family members have therefore emerged as potential therapeutic targets due to their critical role in proliferating cancer cells. Here, we present the crystal structure of Bfl-1, the last anti-apoptotic Bcl-2 family member to be structurally characterized, in complex with a peptide corresponding to the BH3 region of the pro-apoptotic protein Bim. The structure reveals distinct features at the peptide-binding site, likely to define the binding specificity for pro-apoptotic proteins. Superposition of the Bfl-1:Bim complex with that of Mcl-1:Bim reveals a significant local plasticity of hydrophobic interactions contributed by the Bim peptide, likely to be the basis for the multi specificity of Bim for anti-apoptotic proteins.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas de la Membrana/química , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas/química , Secuencia de Aminoácidos , Apoptosis , Proteína 11 Similar a Bcl2 , Cristalografía por Rayos X , Humanos , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Estructura Secundaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA