Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Fitoterapia ; 172: 105716, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37926399

RESUMEN

Four previously undescribed angucyclinones umezawaones A-D (1-4) were isolated from the liquid cultures of Umezawaea beigongshangensis. Their structures were determined by spectroscopic analyses, single crystal X-ray diffraction, quantum chemical 13C NMR and electronic circular dichroism calculations. All compounds displayed strong inhibitory activities against indoleamine 2,3-dioxygenase and tryptophan-2,3-dioxygenase in enzymatic assay, especially compound 2.


Asunto(s)
Actinobacteria , Triptófano Oxigenasa , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo , Anguciciclinas y Anguciclinonas , Actinomyces/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa , Estructura Molecular
2.
J Biol Chem ; 299(6): 104753, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37116709

RESUMEN

Indoleamine-2, 3-dioxygenase (IDO1) and Tryptophan-2, 3-dioxygenase (TDO) catalyze the conversion of L-tryptophan to N-formyl-kynurenine and thus play primary roles in metabolism, inflammation, and tumor immune surveillance. Because their activities depend on their heme contents, which vary in biological settings and go up or down in a dynamic manner, we studied how their heme levels may be impacted by nitric oxide (NO) in mammalian cells. We utilized cells expressing TDO or IDO1 either naturally or via transfection and determined their activities, heme contents, and expression levels as a function of NO exposure. We found NO has a bimodal effect: a narrow range of low NO exposure promoted cells to allocate heme into the heme-free TDO and IDO1 populations and consequently boosted their heme contents and activities 4- to 6-fold, while beyond this range the NO exposure transitioned to have a negative impact on their heme contents and activities. NO did not alter dioxygenase protein expression levels, and its bimodal impact was observed when NO was released by a chemical donor or was generated naturally by immune-stimulated macrophage cells. NO-driven heme allocations to IDO1 and TDO required participation of a GAPDH-heme complex and for IDO1 required chaperone Hsp90 activity. Thus, cells can up- or downregulate their IDO1 and TDO activities through a bimodal control of heme allocation by NO. This mechanism has important biomedical implications and helps explain why the IDO1 and TDO activities in animals go up and down in response to immune stimulation.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa , Óxido Nítrico , Triptófano Oxigenasa , Animales , Hemo/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Mamíferos/metabolismo , Triptófano/metabolismo , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34667125

RESUMEN

Two histidine-ligated heme-dependent monooxygenase proteins, TyrH and SfmD, have recently been found to resemble enzymes from the dioxygenase superfamily currently named after tryptophan 2,3-dioxygenase (TDO), that is, the TDO superfamily. These latest findings prompted us to revisit the structure and function of the superfamily. The enzymes in this superfamily share a similar core architecture and a histidine-ligated heme. Their primary functions are to promote O-atom transfer to an aromatic metabolite. TDO and indoleamine 2,3-dioxygenase (IDO), the founding members, promote dioxygenation through a two-step monooxygenation pathway. However, the new members of the superfamily, including PrnB, SfmD, TyrH, and MarE, expand its boundaries and mediate monooxygenation on a broader set of aromatic substrates. We found that the enlarged superfamily contains eight clades of proteins. Overall, this protein group is a more sizeable, structure-based, histidine-ligated heme-dependent, and functionally diverse superfamily for aromatics oxidation. The concept of TDO superfamily or heme-dependent dioxygenase superfamily is no longer appropriate for defining this growing superfamily. Hence, there is a pressing need to redefine it as a heme-dependent aromatic oxygenase (HDAO) superfamily. The revised concept puts HDAO in the context of thiol-ligated heme-based enzymes alongside cytochrome P450 and peroxygenase. It will update what we understand about the choice of heme axial ligand. Hemoproteins may not be as stringent about the type of axial ligand for oxygenation, although thiolate-ligated hemes (P450s and peroxygenases) more frequently catalyze oxygenation reactions. Histidine-ligated hemes found in HDAO enzymes can likewise mediate oxygenation when confronted with a proper substrate.


Asunto(s)
Hemoproteínas/química , Oxigenasas/química , Aminoácidos Aromáticos/metabolismo , Biocatálisis , Hemo/metabolismo , Hemo Oxigenasa (Desciclizante)/química , Hemo Oxigenasa (Desciclizante)/clasificación , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemoproteínas/clasificación , Hemoproteínas/metabolismo , Humanos , Ligandos , Redes y Vías Metabólicas , Modelos Moleculares , Oxidación-Reducción , Oxigenasas/clasificación , Oxigenasas/metabolismo , Filogenia , Conformación Proteica , Triptófano Oxigenasa/química , Triptófano Oxigenasa/clasificación , Triptófano Oxigenasa/metabolismo
4.
J Inorg Biochem ; 225: 111604, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34571402

RESUMEN

The kynurenine pathway is the major route of tryptophan metabolism. The first step of this pathway is catalysed by one of two heme-dependent dioxygenase enzymes - tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO) - leading initially to the formation of N-formylkynurenine (NFK). In this paper, we present a crystal structure of a bacterial TDO from X. campestris in complex with l-kynurenine, the hydrolysed product of NFK. l-kynurenine is bound at the active site in a similar location to the substrate (l-Trp). Hydrogen bonding interactions with Arg117 and the heme 7-propionate anchor the l-kynurenine molecule into the pocket. A mechanism for the hydrolysis of NFK in the active site is presented.


Asunto(s)
Quinurenina/metabolismo , Triptófano Oxigenasa/metabolismo , Enlace de Hidrógeno , Hierro/química , Quinurenina/química , Oxidación-Reducción , Unión Proteica , Estereoisomerismo , Triptófano/química , Triptófano Oxigenasa/química , Xanthomonas campestris/enzimología
5.
Biochem J ; 478(10): 1999-2017, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33960368

RESUMEN

Human hepatic tryptophan 2,3-dioxygenase (hTDO) is a homotetrameric hemoprotein. It is one of the most rapidly degraded liver proteins with a half-life (t1/2) of ∼2.3 h, relative to an average t1/2 of ∼2-3 days for total liver protein. The molecular mechanism underlying the poor longevity of hTDO remains elusive. Previously, we showed that hTDO could be recognized and ubiquitinated by two E3 ubiquitin (Ub) ligases, gp78/AMFR and CHIP, and subsequently degraded via Ub-dependent proteasomal degradation pathway. Additionally, we identified 15 ubiquitination K-sites and demonstrated that Trp-binding to an exosite impeded its proteolytic degradation. Here, we further established autophagic-lysosomal degradation as an alternative back-up pathway for cellular hTDO degradation. In addition, with protein kinases A and C, we identified 13 phosphorylated Ser/Thr (pS/pT) sites. Mapping these pS/pT sites on the hTDO surface revealed their propinquity to acidic Asp/Glu (D/E) residues engendering negatively charged DEpSpT clusters vicinal to the ubiquitination K-sites over the entire protein surface. Through site-directed mutagenesis of positively charged patches of gp78, previously documented to interact with the DEpSpT clusters in other target proteins, we uncovered the likely role of the DEpSpT clusters in the molecular recognition of hTDO by gp78 and plausibly other E3 Ub-ligases. Furthermore, cycloheximide-chase analyses revealed the critical structural relevance of the disordered N- and C-termini not only in the Ub-ligase recognition, but also in the proteasome engagement. Together, the surface DEpSpT clusters and the N- and C-termini constitute an intrinsic bipartite degron for hTDO physiological turnover.


Asunto(s)
Autofagia , Lisosomas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Triptófano Oxigenasa/metabolismo , Triptófano/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Células Hep G2 , Humanos , Mutación , Fosforilación , Proteolisis , Triptófano Oxigenasa/química , Triptófano Oxigenasa/genética
6.
Biochemistry ; 59(30): 2813-2822, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32659080

RESUMEN

The first step of the kynurenine pathway for l-tryptophan (l-Trp) degradation is catalyzed by heme-dependent dioxygenases, tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase. In this work, we employed stopped-flow optical absorption spectroscopy to study the kinetic behavior of the Michaelis complex of Cupriavidus metallidurans TDO (cmTDO) to improve our understanding of oxygen activation and initial oxidation of l-Trp. On the basis of the stopped-flow results, rapid freeze-quench (RFQ) experiments were performed to capture and characterize this intermediate by Mössbauer spectroscopy. By incorporating the chlorite dismutase-chlorite system to produce high concentrations of solubilized O2, we were able to capture the Michaelis complex of cmTDO in a nearly quantitative yield. The RFQ-Mössbauer results confirmed the identity of the Michaelis complex as an O2-bound ferrous species. They revealed remarkable similarities between the electronic properties of the Michaelis complex and those of the O2 adduct of myoglobin. We also found that the decay of this reactive intermediate is the rate-limiting step of the catalytic reaction. An inverse α-secondary substrate kinetic isotope effect was observed with a kH/kD of 0.87 ± 0.03 when (indole-d5)-l-Trp was employed as the substrate. This work provides an important piece of spectroscopic evidence of the chemical identity of the Michaelis complex of bacterial TDO.


Asunto(s)
Biocatálisis , Triptófano Oxigenasa/química , Cupriavidus/enzimología , Isótopos , Cinética , Espectrofotometría Ultravioleta , Espectroscopía de Mossbauer , Análisis Espectral , Factores de Tiempo , Triptófano/metabolismo
7.
Chem Commun (Camb) ; 56(20): 3089-3092, 2020 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-32052805

RESUMEN

Reaction of FeIII(O2˙-)(TPP) with 2,3-dimethylindole at -40 °C gives the ring-opened, dioxygenated N-(2-acetyl-phenyl)-acetamide product. The reaction was monitored in situ by low-temperature UV-vis and 1H NMR spectroscopies. This work demonstrates that a discrete iron(iii)(superoxo) porphyrin is competent to carry out indole oxidation, as proposed for the tryptophan and indoleamine 2,3-dioxygenases.


Asunto(s)
Compuestos Férricos/química , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indoles/química , Metaloporfirinas/química , Superóxidos/química , Triptófano Oxigenasa/química , Compuestos Férricos/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indoles/metabolismo , Metaloporfirinas/metabolismo , Estructura Molecular , Oxidación-Reducción , Superóxidos/metabolismo , Triptófano Oxigenasa/metabolismo
8.
Biochim Biophys Acta Proteins Proteom ; 1868(1): 140247, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31276825

RESUMEN

Tryptophan 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO) have an independent origin; however, they have distinctly evolved to catalyze the same reaction. In general, TDO is a single-copy gene in each metazoan species, and TDO enzymes demonstrate similar enzyme activity regardless of their biological origin. In contrast, multiple IDO paralogues are observed in many species, and they display various enzymatic properties. Similar to vertebrate IDO2, invertebrate IDOs generally show low affinity/catalytic efficiency for L-Trp. Meanwhile, two IDO isoforms from scallop (IDO-I and -III) and sponge IDOs show high L-Trp catalytic activity, which is comparable to vertebrate IDO1. Site-directed mutagenesis experiments have revealed that primarily two residues, Tyr located at the 2nd residue on the F-helix (F2nd) and His located at the 9th residue on the G-helix (G9th), are crucial for the high affinity/catalytic efficiency of these 'high performance' invertebrate IDOs. Conversely, those two amino acid substitutions (F2nd/Tyr and G9th/His) resulted in high affinity and catalytic activity in other molluscan 'low performance' IDOs. In human IDO1, G9th is Ser167, whereas the counterpart residue of G9th in human TDO is His76. Previous studies have shown that Ser167 could not be substituted by His because the human IDO1 Ser167His variant showed significantly low catalytic activity. However, this may be specific for human IDO1 because G9th/His was demonstrated to be very effective in increasing the L-Trp affinity even in vertebrate IDOs. Therefore, these findings indicate that the active sites of TDO and IDO are more similar to each other than previously expected.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/química , Invertebrados/enzimología , Triptófano Oxigenasa/química , Triptófano/química , Animales , Dominio Catalítico , Evolución Molecular , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Filogenia , Triptófano Oxigenasa/genética
9.
Angew Chem Int Ed Engl ; 59(8): 3043-3047, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31828916

RESUMEN

Herein, we report an engineered enzyme that can monooxygenate unprotected tryptophan into the corresponding 3a-hydroxyhexahydropyrrolo[2,3-b]indole-2-carboxylic acid (HPIC) in a single, scalable step with excellent turnover number and diastereoselectivity. Taking advantage of directed evolution, we analyzed the stepwise oxygen-insertion mechanism of tryptophan 2,3-dioxygenases, and transformed tryptophan 2,3-dioxygenase from Xanthomonas campestris into a monooxygenase for oxidative cyclization of tryptophans. It was revealed that residue F51 is vital in determining the product ratio of HPIC to N'-formylkynurenine. Our reactions and purification procedures use no organic solvents, resulting in an eco-friendly method to prepare HPICs for further applications.


Asunto(s)
Oxigenasas de Función Mixta/química , Triptófano Oxigenasa/química , Triptófano/química , Humanos , Oxidación-Reducción
10.
Sci Rep ; 9(1): 18455, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804586

RESUMEN

The improvement of body's own immune system is considered one of the safest approaches to fight against cancer and several other diseases. Excessive catabolism of the essential amino acid, L-tryptophan (L-Trp) assists the cancer cells to escape normal immune obliteration. The formation of disproportionate kynurenine and other downstream metabolites suppress the T cell functions. Blocking of this immunosuppressive mechanism is considered as a promising approach against cancer, neurological disorders, autoimmunity, and other immune-mediated diseases. Overexpression of indoleamine 2,3-dioxygenase 1 (IDO1) enzyme is directly related to the induction of immunosuppressive mechanisms and represents an important therapeutic target. Several classes of small molecule-based IDO1 inhibitors have been already reported, but only few compounds are currently being evaluated in various stages of clinical trials as adjuvants or in combination with chemo- and radiotherapies. In the quest for novel structural class(s) of IDO1 inhibitors, we developed a series of 4,5-disubstituted 1,2,3-triazole derivatives. The optimization of 4,5-disubstituted 1,2,3-triazole scaffold and comprehensive biochemical and biophysical studies led to the identification of compounds, 3i, 4i, and 4k as potent and selective inhibitors of IDO1 enzyme with IC50 values at a low nanomolar level. These potent compounds also showed strong IDO1 inhibitory activities in MDA-MB-231 cells with no/negligible level of cytotoxicity. The T cell activity studies revealed that controlled regulation of IDO1 enzyme activity in the presence of these potent compounds could induce immune response against breast cancer cells. The compounds also showed excellent in vivo antitumor efficacy (of tumor growth inhibition = 79-96%) in the female Swiss albino mice. As a consequence, this study describes the first example of 4,5-disubstituted 1,2,3-triazole based IDO1 inhibitors with potential applications for immunotherapeutic studies.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Linfocitos T Citotóxicos/efectos de los fármacos , Triazoles/farmacología , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinoma de Ehrlich/tratamiento farmacológico , Carcinoma de Ehrlich/inmunología , Carcinoma de Ehrlich/patología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Pruebas de Enzimas , Femenino , Células HEK293 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Concentración 50 Inhibidora , Quinurenina/inmunología , Quinurenina/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/inmunología , Ratones , Simulación del Acoplamiento Molecular , Cultivo Primario de Células , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Linfocitos T Citotóxicos/inmunología , Triazoles/química , Triazoles/uso terapéutico , Triptófano/inmunología , Triptófano/metabolismo , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo , Escape del Tumor/efectos de los fármacos
11.
Molecules ; 24(23)2019 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-31795096

RESUMEN

Cancers express tryptophan catabolising enzymes indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO2) to produce immunosuppressive tryptophan metabolites that undermine patients' immune systems, leading to poor disease outcomes. Both enzymes are validated targets for cancer immunotherapy but there is a paucity of potent TDO2 and dual IDO1/TDO2 inhibitors. To identify novel dual IDO1/TDO2 scaffolds, 3D shape similarity and pharmacophore in silico screening was conducted using TDO2 as a model for both systems. The obtained hits were tested in cancer cell lines expressing mainly IDO1 (SKOV3-ovarian), predominantly TDO2 (A172-brain), and both IDO1 and TDO2 (BT549-breast). Three virtual screening hits were confirmed as inhibitors (TD12, TD18 and TD34). Dose response experiments showed that TD34 is the most potent inhibitor capable of blocking both IDO1 and TDO2 activity, with the IC50 value for BT549 at 3.42 µM. This work identified new scaffolds able to inhibit both IDO1 and TDO2, thus enriching the collection of dual IDO1/TDO2 inhibitors and providing chemical matter for potential development into future anticancer drugs.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/química , Antineoplásicos/química , Antineoplásicos/farmacología , Desarrollo de Medicamentos , Descubrimiento de Drogas/métodos , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
12.
J Am Chem Soc ; 141(47): 18771-18779, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31682426

RESUMEN

Indoleamine 2,3-dioxygenase 1 (hIDO1) and tryptophan dioxygenase (hTDO) are two of the only three heme-based dioxygenases in humans. They have recently been identified as key cancer immunotherapeutic drug targets. While structures of hIDO1 in complex with inhibitors have been documented, so far there are no structures of hTDO-inhibitor complexes available. Here we use PF-06840003 (IPD), a hIDO1-selective inhibitor in clinical trials, as a structural probe to elucidate inhibitor-selectivity in hIDO1 versus hTDO. Spectroscopic studies show that IPD exhibits 400-fold higher inhibition activity toward hIDO1 with respect to hTDO. Crystallographic structures reveal that the binding pocket of IPD in the active site in hIDO1 is much more flexible as compared to that in hTDO, which offers a molecular explanation for the superior inhibition activity of IPD in hIDO1 with respect to hTDO. In addition to the IPD bound in the active site, a second IPD molecule was identified in an inhibitory site on the proximal side of the heme in hIDO1 and in an exosite that is ∼40 Šaway from the active site in hTDO. Taken together the data provide new insights into structure-based design of mono and dual inhibitors targeting hIDO1 and/or hTDO.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/antagonistas & inhibidores , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Hemo/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Modelos Moleculares , Dominios Proteicos , Especificidad por Sustrato , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo
13.
Angew Chem Int Ed Engl ; 58(46): 16480-16484, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31584750

RESUMEN

Electrochemical sensors are essential for point-of-care testing (POCT) and wearable sensing devices. Establishing an efficient electron transfer route between redox enzymes and electrodes is key for converting enzyme-catalyzed reactions into electrochemical signals, and for the development of robust, sensitive, and selective biosensors. We demonstrate that the site-specific incorporation of a novel synthetic amino acid (2-amino-3-(4-mercaptophenyl)propanoic acid) into redox enzymes, followed by an S-click reaction to wire the enzyme to the electrode, facilitates electron transfer. The fabricated biosensor demonstrated real-time and selective monitoring of tryptophan (Trp) in blood and sweat samples, with a linear range of 0.02-0.8 mm. Further developments along this route may result in dramatic expansion of portable electrochemical sensors for diverse health-determination molecules.


Asunto(s)
Oxidorreductasas/metabolismo , Técnicas Biosensibles/métodos , Técnicas Electroquímicas , Electrodos , Transporte de Electrón , Células HeLa , Humanos , Oxidorreductasas/química , Sistemas de Atención de Punto , Sudor/metabolismo , Triptófano/análisis , Triptófano/sangre , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo , Dispositivos Electrónicos Vestibles
14.
J Biochem ; 164(5): 359-367, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30053101

RESUMEN

l-Tryptophan oxidase, VioA from Chromobacterium violaceum, which has a high substrate specificity for tryptophan, is useful for quantitative assay of tryptophan. However, stability of wild type VioA is not enough for its application in clinical or industrial use. To improve the thermal stability of the enzyme, we developed a VioA (C395A) mutant, with higher stability than wild type VioA. The VioA (C395A) exhibited similar specificity and kinetic parameter for tryptophan to wild type. Conventionally, the quantity of tryptophan is determined by instrumental methods, such as high-performance liquid chromatography (HPLC) after pre-column-derivatization. Using the mutant enzyme, we succeeded in the tryptophan quantification in human plasma samples, to an accuracy of <2.9% when compared to the instrumental method, and to a precision of CV <3.2%. To analyse the improvement in storage stability and substrate specificity, we further determined the crystal structures of VioA (C395A) complexed with FAD, and with FAD and tryptophan at 1.8 Å resolution.


Asunto(s)
Ingeniería de Proteínas , Temperatura , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo , Cromatografía Líquida de Alta Presión , Chromobacterium/enzimología , Estabilidad de Enzimas , Conformación Proteica , Triptófano Oxigenasa/genética
15.
J Am Chem Soc ; 140(27): 8518-8525, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29897749

RESUMEN

Human indoleamine 2,3-dioxygenase 1 (hIDO1) and tryptophan dioxygenase (hTDO) catalyze the same dioxygenation reaction of Trp to generate N-formyl kynurenine (NFK). They share high structural similarity, especially in the active site. However, hIDO1 possesses a unique inhibitory substrate binding site (Si) that is absent in hTDO. In addition, in hIDO1, the indoleamine group of the substrate Trp is H-bonded to S167 through a bridging water, while that in hTDO is directly H-bonded to H76. Here we show that Trp binding to the Si site or the mutation of S167 to histidine in hIDO1 retards its turnover activity and that the inhibited activity can be rescued by an effector, 3-indole ethanol (IDE). Kinetic studies reveal that the inhibited activity introduced by Trp binding to the Si site is a result of retarded recombination of the ferryl moiety with Trp epoxide to form NFK and that IDE reverses the effect by preventing Trp from binding to the Si site. In contrast, the abolished activity induced by the S167H mutation is primarily a result of ∼5000-fold reduction in the O2 binding rate constant, possibly due to the blockage of a ligand delivery tunnel, and that IDE binding to the Si site reverses the effect by reopening the tunnel. The data offer new insights into structure-based design of hIDO1-selective inhibitors.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/análogos & derivados , Triptófano/metabolismo , Sitios de Unión , Dominio Catalítico , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Cinética , Quinurenina/metabolismo , Modelos Moleculares , Unión Proteica , Especificidad por Sustrato , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo
16.
J Am Chem Soc ; 140(12): 4372-4379, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29506384

RESUMEN

Heme-based tryptophan dioxygenases are established immunosuppressive metalloproteins with significant biomedical interest. Here, we synthesized two mechanistic probes to specifically test if the α-amino group of the substrate directly participates in a critical step of the O atom transfer during catalysis in human tryptophan 2,3-dioxygenase (TDO). Substitution of the nitrogen atom of the substrate to a carbon (probe 1) or oxygen (probe 2) slowed the catalytic step following the first O atom transfer such that transferring the second O atom becomes less likely to occur, although the dioxygenated products were observed with both probes. A monooxygenated product was also produced from probe 2 in a significant quantity. Analysis of this new product by HPLC coupled UV-vis spectroscopy, high-resolution mass spectrometry, 1H NMR, 13C NMR, HSQC, HMBC, and infrared (IR) spectroscopies concluded that this monooxygenated product is a furoindoline compound derived from an unstable epoxyindole intermediate. These results prove that small molecules can manipulate the stepwise O atom transfer reaction of TDO and provide a showcase for a tunable mechanism by synthetic compounds. The product analysis results corroborate the presence of a substrate-based epoxyindole intermediate during catalysis and provide the first substantial experimental evidence for the involvement of the substrate α-amino group in the epoxide ring-opening step during catalysis. This combined synthetic, biochemical, and biophysical study establishes the catalytic role of the α-amino group of the substrate during the O atom transfer reactions and thus represents a substantial advance to the mechanistic comprehension of the heme-based tryptophan dioxygenases.


Asunto(s)
Compuestos de Amonio/metabolismo , Compuestos Epoxi/metabolismo , Oxígeno/metabolismo , Triptófano Oxigenasa/metabolismo , Compuestos de Amonio/química , Compuestos Epoxi/química , Humanos , Conformación Molecular , Oxígeno/química , Especificidad por Sustrato , Triptófano Oxigenasa/química
17.
J Insect Physiol ; 107: 104-109, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29551569

RESUMEN

The Tribolium castaneum vermilion gene encodes tryptophan 2,3-dioxygenase, a pivotal enzyme in the ommochrome pathway that is required for proper pigmentation of the eye. A white-eyed mutant strain of T. castaneum, vermilionwhite (vw), lacks eye pigmentation due to a deletion of unknown size that removes all but the 3'-end of the vermilion gene. To create a more defined mutation in vermilion, the CRISPR/Cas9-nuclease system was used to target wild type vermilion in preblastoderm T. castaneum embryos. As adults, all injected beetles had wild type (black) eye pigmentation; however, when outcrossed to vw mates, one cross produced 19% white-eyed offspring. When the vermilion locus of these offspring was analyzed by target-site sequencing, it was determined that white-eyed individuals had a 2 bp deletion that resulted in a frame-shift mutation, presumably producing a nonfunctional enzyme. Interestingly, some of their black-eyed siblings also had a small deletion of 6 bp, but the resultant loss of two amino acids had no apparent impact on enzyme function. To establish a mutant strain homozygous for the CRISPR-induced knock-out allele, a CRISPR positive G0 male was crossed to wild type females. Their progeny were self-crossed, and white-eyed progeny were used to establish the new strain. This mutant strain is herein named vermilionICE and will be used in future work in addition to or in place of vw.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes , Proteínas de Insectos/genética , Pigmentación/genética , Tribolium/fisiología , Triptófano Oxigenasa/genética , Secuencia de Aminoácidos , Animales , Femenino , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/fisiología , Masculino , Redes y Vías Metabólicas , Fenómenos Fisiológicos Oculares , Filogenia , Alineación de Secuencia , Tribolium/genética , Tribolium/crecimiento & desarrollo , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo
18.
J Phys Chem B ; 121(31): 7412-7420, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28715185

RESUMEN

The human heme enzyme tryptophan 2,3-dioxygenase (hTDO) catalyzes the insertion of dioxygen into its cognate substrate, l-tryptophan (l-Trp). Its active site structure is highly dynamic, and the mechanism of enzyme-substrate-ligand complex formation and the ensuing enzymatic reaction is not yet understood. Here we have studied complex formation in hTDO by using time-resolved optical and infrared spectroscopy with carbon monoxide (CO) as a ligand. We have observed that both substrate-free and substrate-bound hTDO coexist in two discrete conformations with greatly different ligand binding rates. In the fast rebinding hTDO conformation, there is facile ligand access to the heme iron, but it is greatly hindered in the slowly rebinding conformation. Spectroscopic evidence implicates active site solvation as playing a crucial role for the observed kinetic differences. Substrate binding shifts the conformational equilibrium markedly toward the fast species and thus primes the active site for subsequent ligand binding, ensuring that formation of the ternary complex occurs predominantly by first binding l-Trp and then the ligand. Consequently, the efficiency of catalysis is enhanced because O2 binding prior to substrate binding, resulting in nonproductive oxidation of the heme iron, is greatly suppressed.


Asunto(s)
Ligandos , Triptófano Oxigenasa/metabolismo , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Dominio Catalítico , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Fotólisis , Unión Proteica , Espectroscopía Infrarroja por Transformada de Fourier , Especificidad por Sustrato , Temperatura , Triptófano Oxigenasa/química , Triptófano Oxigenasa/genética
19.
Metallomics ; 9(9): 1230-1240, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28650043

RESUMEN

Iron-containing enzymes such as heme enzymes play crucial roles in biological systems. Three distinct heme-containing dioxygenase enzymes, tryptophan 2,3-dioxygenase (TDO), indoleamine 2,3-dioxygenase 1 (IDO1) and indoleamine 2,3-dioxygenase 2 (IDO2) catalyze the initial and rate-limiting step of l-tryptophan catabolism through the kynurenine pathway in mammals. Overexpression of these enzymes causes depletion of tryptophan and the accumulation of metabolic products, which contributes to tumor immune tolerance and immune dysregulation in a variety of disease pathologies. In the past few decades, IDO1 has garnered the most attention as a therapeutic target with great potential in cancer immunotherapy. Many potential inhibitors of IDO1 have been designed, synthesized and evaluated, among which indoximod (d-1-MT), INCB024360, GDC-0919 (formerly NLG-919), and an IDO1 peptide-based vaccine have advanced to the clinical trial stage. However, recently, the roles of TDO and IDO2 have been elucidated in immune suppression. In this review, the current drug discovery landscape for targeting TDO, IDO1 and IDO2 is highlighted, with particular attention to the recent use of drugs in clinical trials. Moreover, the crystal structures of these enzymes, in complex with inhibitors, and the mechanisms of Trp catabolism in the first step, are summarized to provide information for facilitating the discovery of new enzyme inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Enzimas/metabolismo , Hemoproteínas/antagonistas & inhibidores , Hemoproteínas/metabolismo , Triptófano/metabolismo , Animales , Inhibidores Enzimáticos/química , Enzimas/química , Hemoproteínas/química , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Estructura Molecular , Conformación Proteica , Triptófano/química , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/química , Triptófano Oxigenasa/metabolismo
20.
Mol Genet Metab ; 120(4): 317-324, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28285122

RESUMEN

In this report we describe the first human case of hypertryptophanemia confirmed to be due to tryptophan 2,3-dioxygenase deficiency. The underlying etiology was established by sequencing the TDO2 gene, in which there was compound heterozygosity for two rare variants: c.324G>C, p.Met108Ile and c.491dup, p.Ile165Aspfs*12. The pathogenicity of these variants was confirmed by molecular-level studies, which showed that c.491dup does not produce soluble protein and c.324G>C results in a catalytically less efficient Met108Ile enzyme that is prone to proteolytic degradation. The biochemical phenotype of hypertryptophanemia and hyperserotoninemia does not appear to have significant clinical consequences.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Mutación , Triptófano Oxigenasa/genética , Dominio Catalítico , Femenino , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Recién Nacido , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN , Triptófano Oxigenasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...