Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Biochem ; 123(6): 1064-1076, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35434822

RESUMEN

Vestigial-like family member 3 (VGLL3) is a member of the VGLL family that serves as cofactors for TEA-domain transcription factors. Although VGLL3 is involved in the proliferation of cancer cells, the molecular mechanisms underlying VGLL3-mediated cell proliferation remain largely unknown. In this study, we found that stable expression of VGLL3 in human lung cancer A549 cells affects glutamine metabolism and increases their dependency on de novo nucleotide synthesis for proliferation. Mechanistically, VGLL3 was found to induce the expression of GART, which encodes a trifunctional enzyme that catalyzes de novo purine synthesis from glutamine. GART knockdown and the glycinamide ribonucleotide synthase, aminoimidazole ribonucleotide synthase, and glycinamide ribonucleotide formyltransferase trifunctional protein (GART) inhibitor lometrexol repressed the proliferation and survival of A549 cells stably expressing VGLL3. Mesenchymal breast cancer BT549 cells and MDA-MB-231 cells showed high expression of VGLL3, and VGLL3 knockdown was found to reduce GART expression. Lometrexol also repressed the proliferation of these breast cancer cells, whereas addition of inosine monophosphate, an important metabolite downstream of GART, rescued this repression. Taken together, these results suggest that VGLL3 induces GART expression and thereby confers de novo nucleotide-dependent cell proliferation in cancer cells.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Neoplasias/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Línea Celular Tumoral , Glutamina , Humanos , Neoplasias/patología , Nucleótidos/biosíntesis , Factores de Transcripción
2.
Bioorg Med Chem ; 37: 116093, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33773393

RESUMEN

We discovered 6-substituted thieno[2,3-d]pyrimidine compounds (3-9) with 3-4 bridge carbons and side-chain thiophene or furan rings for dual targeting one-carbon (C1) metabolism in folate receptor- (FR) expressing cancers. Synthesis involved nine steps starting from the bromo-aryl carboxylate. From patterns of growth inhibition toward Chinese hamster ovary cells expressing FRα or FRß, the proton-coupled folate transporter or reduced folate carrier, specificity for uptake by FRs was confirmed. Anti-proliferative activities were demonstrated toward FRα-expressing KB tumor cells and NCI-IGROV1 ovarian cancer cells. Inhibition of de novo purine biosynthesis at both 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and glycinamide ribonucleotide formyltransferase (GARFTase) was confirmed by metabolite rescue, metabolomics and enzyme assays. X-ray crystallographic structures were obtained with compounds 3-5 and human GARFTase. Our studies identify first-in-class C1 inhibitors with selective uptake by FRs and dual inhibition of enzyme targets in de novo purine biosynthesis, resulting in anti-tumor activity. This series affords an exciting new platform for selective multi-targeted anti-tumor agents.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Pirimidinas/farmacología , Tiofenos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetulus , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Receptores de Folato Anclados a GPI/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Unión Proteica , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/metabolismo
3.
Bioorg Med Chem ; 28(12): 115544, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32503687

RESUMEN

Tumor-targeted 6-substituted pyrrolo[2,3-d]pyrimidine benzoyl compounds based on 2 were isosterically modified at the 4-carbon bridge by replacing the vicinal (C11) carbon by heteroatoms N (4), O (5) or S (6), or with an N-substituted formyl (7), trifluoroacetyl (8) or acetyl (9). Replacement with sulfur (6) afforded the most potent KB tumor cell inhibitor, ~6-fold better than the parent 2. In addition, 6 retained tumor transport selectivity via folate receptor (FR) α and -ß over the ubiquitous reduced folate carrier (RFC). FRα-mediated cell inhibition for 6 was generally equivalent to 2, while the FRß-mediated activity was improved by 16-fold over 2. N (4) and O (5) substitutions afforded similar tumor cell inhibitions as 2, with selectivity for FRα and -ß over RFC. The N-substituted analogs 7-9 also preserved transport selectivity for FRα and -ß over RFC. For FRα-expressing CHO cells, potencies were in the order of 8 > 7 > 9. Whereas 8 and 9 showed similar results with FRß-expressing CHO cells, 7 was ~16-fold more active than 2. By nucleoside rescue experiments, all the compounds inhibited de novo purine biosynthesis, likely at the step catalyzed by glycinamide ribonucleotide formyltransferase. Thus, heteroatom replacements of the CH2 in the bridge of 2 afford analogs with increased tumor cell inhibition that could provide advantages over 2, as well as tumor transport selectivity over clinically used antifolates including methotrexate and pemetrexed.


Asunto(s)
Antineoplásicos/síntesis química , Diseño de Fármacos , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Ácido Fólico/metabolismo , Pirimidinas/química , Pirroles/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión , Células CHO , Dominio Catalítico , Línea Celular Tumoral , Cricetinae , Cricetulus , Receptor 1 de Folato/química , Receptor 1 de Folato/genética , Receptor 2 de Folato/química , Receptor 2 de Folato/genética , Ácido Fólico/química , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Humanos , Simulación del Acoplamiento Molecular , Fosforribosilglicinamida-Formiltransferasa/química , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Pirroles/metabolismo , Pirroles/farmacología , Relación Estructura-Actividad
4.
J Proteome Res ; 18(5): 2078-2087, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-30964683

RESUMEN

Purines represent a class of essential metabolites produced by the cell to maintain cellular homeostasis and facilitate cell proliferation. In times of high purine demand, the de novo purine biosynthetic pathway is activated; however, the mechanisms that facilitate this process are largely unknown. One plausible mechanism is through intracellular signaling, which results in enzymes within the pathway becoming post-translationally modified to enhance their individual enzyme activities and the overall pathway metabolic flux. Here, we employ a proteomic strategy to investigate the extent to which de novo purine biosynthetic pathway enzymes are post-translationally modified in 293T cells. We identified 7 post-translational modifications on 135 residues across the 6 human pathway enzymes. We further asked whether there were differences in the post-translational modification state of each pathway enzyme isolated from cells cultured in the presence or absence of purines. Of the 174 assigned modifications, 67% of them were only detected in one experimental growth condition in which a significant number of serine and threonine phosphorylations were noted. A survey of the most-probable kinases responsible for these phosphorylation events uncovered a likely AKT phosphorylation site at residue Thr397 of PPAT, which was only detected in cells under purine-supplemented growth conditions. These data suggest that this modification might alter enzyme activity or modulate its interaction(s) with downstream pathway enzymes. Together, these findings propose a role for post-translational modifications in pathway regulation and activation to meet intracellular purine demand.


Asunto(s)
Amidofosforribosiltransferasa/metabolismo , Mapeo Peptídico/métodos , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/metabolismo , Acetilación , Adenilosuccinato Liasa/genética , Adenilosuccinato Liasa/metabolismo , Amidofosforribosiltransferasa/genética , Secuencia de Aminoácidos , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Péptidos/síntesis química , Péptidos/metabolismo , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Transducción de Señal , Treonina/metabolismo , Ubiquitinación
5.
Biochemistry ; 55(32): 4574-82, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27439469

RESUMEN

Pemetrexed and methotrexate are antifolates used for cancer chemotherapy and inflammatory diseases. These agents have toxic side effects resulting, in part, from nonspecific cellular transport by the reduced folate carrier (RFC), a ubiquitously expressed facilitative transporter. We previously described 2-amino-4-oxo-6-substituted pyrrolo[2,3-d]pyrimidine antifolates with modifications of the side chain linker and aromatic ring that are poor substrates for RFC but are efficiently transported via folate receptors (FRs) and the proton-coupled folate transporter (PCFT). These targeted antifolates are cytotoxic in vitro toward FR- and PCFT-expressing tumor cells and in vivo with human tumor xenografts in immune-compromised mice, reflecting selective cellular uptake. Antitumor efficacy is due to inhibition of glycinamide ribonucleotide (GAR) formyltransferase (GARFTase) activity in de novo synthesis of purine nucleotides. This study used purified human GARFTase (formyltransferase domain) to assess in vitro inhibition by eight novel thieno- and pyrrolo[2,3-d]pyrimidine antifolates. Seven analogues (AGF23, AGF71, AGF94, AGF117, AGF118, AGF145, and AGF147) inhibited GARFTase with Ki values in the low- to mid-nanomolar concentration range, whereas AGF50 inhibited GARFTase with micromolar potency similar to that of PMX. On the basis of crystal structures of ternary complexes with GARFTase, ß-GAR, and the monoglutamyl antifolates, differences in inhibitory potencies correlated well with antifolate binding and the positions of the terminal carboxylates. Our data provide a mechanistic basis for differences in inhibitory potencies between these novel antifolates and a framework for future structure-based drug design. These analogues could be more efficacious than clinically used antifolates, reflecting their selective cellular uptake by FRs and PCFT and potent GARFTase inhibition.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Animales , Humanos , Células KB , Ratones , Modelos Moleculares , Fosforribosilglicinamida-Formiltransferasa/química , Conformación Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Apoptosis ; 21(12): 1386-1397, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27718035

RESUMEN

Glycinamide ribonucleotide formyltransferase (GART) has been established as a pivotal enzyme in de novo purine synthesis, and mediates cellular apoptosis in many diseases. We aimed to investigate the role of GART in the pathogenesis of Crohn's disease (CD). In our study, we demonstrated for the first time that GART expression is up-regulated in patients with active CD and in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced acute colitis model. Moreover, the inhibition of GART induced cellular apoptosis and suppressed the migration of IECs through the activation of the MEKK3-MKK3-p38 mitogen-activated protein kinase (MAPK) pathway, following with the dys-regulation of p53 and p53 up-regulated modulator of apoptosis (PUMA). Taken together, GART plays a critical role in the protection of cellular apoptosis and migration of intestinal epithelial cells to maintain the integrity of the epithelial barrier, thus providing a new potential approach in designing a novel therapy for CD.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Colitis/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Ligasas de Carbono-Nitrógeno/genética , Proliferación Celular , Colitis/enzimología , Colitis/genética , Colitis/fisiopatología , Células Epiteliales/citología , Células Epiteliales/enzimología , Humanos , Intestinos/citología , Intestinos/enzimología , Sistema de Señalización de MAP Quinasas , Fosforribosilglicinamida-Formiltransferasa/genética , Proteínas Proto-Oncogénicas/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
7.
Biochemistry ; 52(30): 5133-44, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23869564

RESUMEN

Glycinamide ribonucleotide transformylase (GAR Tfase) is a folate-dependent enzyme in the de novo purine biosynthesis pathway, which has long been considered a potential target for development of anti-neoplastic therapeutics. Here we report the biological and X-ray crystallographic evaluations of both independent C10 diastereomers, 10S- and 10R-methylthio-DDACTHF, bound to human GAR Tfase, including the highest-resolution apo GAR Tfase structure to date (1.52 Å). Both diastereomers are potent inhibitors (Ki = 210 nM for 10R, and Ki = 180 nM for 10S) of GAR Tfase and exhibit effective inhibition of human leukemia cell growth (IC50 = 80 and 50 nM, respectively). Their inhibitory activity was surprisingly high, and these lipophilic C10-substituted analogues show distinct advantages over their hydrophilic counterparts, most strikingly in retaining potency in mutant human leukemia cell lines that lack reduced folate carrier protein activity (IC50 = 70 and 60 nM, respectively). Structural characterization reveals a new binding mode for these diastereoisomers, in which the lipophilic thiomethyl groups penetrate deeper into a hydrophobic pocket within the folate-binding site. In silico docking simulations of three other sulfur-containing folate analogues also indicate that this hydrophobic cleft represents a favorable region for binding lipophilic substituents. Overall, these results suggest sulfur and its substitutions play an important role in not only the binding of anti-folates to GAR Tfase but also the selectivity and cellular activity (growth inhibition), thereby presenting new possibilities for the future design of potent and selective anti-folate drugs that target GAR Tfase.


Asunto(s)
Antimetabolitos Antineoplásicos/química , Ligasas de Carbono-Nitrógeno/química , Inhibidores Enzimáticos/química , Modelos Moleculares , Fosforribosilglicinamida-Formiltransferasa/química , Tetrahidrofolatos/química , Antimetabolitos Antineoplásicos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Apoproteínas/antagonistas & inhibidores , Apoproteínas/química , Apoproteínas/metabolismo , Sitios de Unión , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Conformación Molecular , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/química , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/genética , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/metabolismo , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Tetrahidrofolatos/metabolismo , Tetrahidrofolatos/farmacología
8.
Adv Sci (Weinh) ; 10(25): e2301264, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37439412

RESUMEN

Tumor stemness is associated with the recurrence and incurability of colorectal cancer (CRC), which lacks effective therapeutic targets and drugs. Glycinamide ribonucleotide transformylase (GART) fulfills an important role in numerous types of malignancies. The present study aims to identify the underlying mechanism through which GART may promote CRC stemness, as to developing novel therapeutic methods. An elevated level of GART is associated with poor outcomes in CRC patients and promotes the proliferation and migration of CRC cells. CD133+ cells with increased GART expression possess higher tumorigenic and proliferative capabilities both in vitro and in vivo. GART is identified to have a novel methyltransferase function, whose enzymatic activity center is located at the E948 site. GART also enhances the stability of RuvB-like AAA ATPase 1 (RUVBL1) through methylating its K7 site, which consequently aberrantly activates the Wnt/ß-catenin signaling pathway to induce tumor stemness. Pemetrexed (PEM), a compound targeting GART, combined with other chemotherapy drugs greatly suppresses tumor growth both in a PDX model and in CRC patients. The present study demonstrates a novel methyltransferase function of GART and the role of the GART/RUVBL1/ß-catenin signaling axis in promoting CRC stemness. PEM may be a promising therapeutic agent for the treatment of CRC.


Asunto(s)
Ligasas de Carbono-Nitrógeno , Neoplasias Colorrectales , Humanos , Línea Celular Tumoral , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Metiltransferasas/metabolismo , beta Catenina/metabolismo , Neoplasias Colorrectales/patología , Vía de Señalización Wnt , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas Portadoras/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/farmacología , Ligasas de Carbono-Nitrógeno/metabolismo
9.
Front Endocrinol (Lausanne) ; 14: 1129162, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37143728

RESUMEN

Targeting tumor cell metabolism is a new frontier in cancer management. Thus, metabolic pathway inhibitors could be used as anti-estrogen receptor α (ERα) breast cancer (BC) drugs. Here, the interplay among metabolic enzyme(s), the ERα levels and cell proliferation was studied. siRNA-based screen directed against different metabolic proteins in MCF10a, MCF-7 and MCF-7 cells genetically resistant to endocrine therapy (ET) drugs and metabolomic analyses in numerous BC cell lines unveil that the inhibition of GART, a key enzyme in the purine de novo biosynthetic pathway, induces ERα degradation and prevent BC cell proliferation. We report here that a reduced GART expression correlates with a longer relapse-free-survival (RFS) in women with ERα-positive BCs. ERα-expressing luminal A invasive ductal carcinomas (IDCs) are sensitive to GART inhibition and GART expression is increased in receptor-positive IDCs of high grade and stage and plays a role in the development of ET resistance. Accordingly, GART inhibition reduces ERα stability and cell proliferation in IDC luminal A cells where it deregulates 17ß-estradiol (E2):ERα signaling to cell proliferation. Moreover, the GART inhibitor lometrexol (LMX) and drugs approved for clinical treatment of primary and metastatic BC (4OH-tamoxifen and the CDK4/CDK6 inhibitors) exert synergic antiproliferative effects in BC cells. In conclusion, GART inhibition by LMX or other inhibitors of the de novo purine biosynthetic pathway could be a novel effective strategy for the treatment of primary and metastatic BCs.


Asunto(s)
Neoplasias de la Mama , Ligasas de Carbono-Nitrógeno , Carcinoma Ductal de Mama , Femenino , Humanos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Vías Biosintéticas , Recurrencia Local de Neoplasia , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Purinas , Ligasas de Carbono-Nitrógeno/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo
10.
Ann Oncol ; 23(7): 1723-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22186609

RESUMEN

BACKGROUND: Clinical results of a randomized phase III trial comparing pemetrexed-carboplatin (PC) with etoposide-carboplatin (EC) in chemonaive patients with extensive-stage disease small-cell lung cancer (ED-SCLC) resulted in trial closure for futility; biomarker analyses using immunohistochemistry (IHC) and single-nucleotide polymorphisms (SNPs) are described herein. PATIENTS AND METHODS: Thymidylate synthase (TS), excision repair cross complementing-1 (ERCC1), glycinamide ribonucleotide formyltransferase (GARFT), and folylpolyglutamate synthetase (FPGS) were investigated using IHC (n=395). SNPs were genotyped for TS, FPGS, γ-glutamyl hydrolase (GGH), methylenetetrahydrofolate reductase (MTHFR), folate receptor-α FR-α, and solute carrier 19A1 (SLC19A1; n=611). RESULTS: None of the IHC biomarkers (folate pathway or ERCC1) were found to be predictive or prognostic in this setting. rs2838952 (adjacent to SLC19A1) had significant treatment-independent association with overall survival (OS; hazard ratio 0.590, P=0.01). Nine GGH-associated SNPs interacted with rs3788205 (SLC19A1) for OS on the PC arm. rs12379987 (FPGS) interacted with treatment for OS (interaction P=0.036). CONCLUSION: Potential ERCC1 and folate pathway IHC biomarkers failed to predict outcome in either study arm in ED-SCLC. SNPs in regions including FPGS and SLC19A1 and interacting SNPs in GGH and SLC19A1 were associated with differences in OS; however, none of these SNPs predicted for greater survival with PC over EC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/metabolismo , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Biomarcadores de Tumor/genética , Carboplatino/administración & dosificación , Ensayos Clínicos Fase III como Asunto , Colágeno Tipo XVIII/genética , Proteínas de Unión al ADN/metabolismo , Supervivencia sin Enfermedad , Endonucleasas/metabolismo , Etopósido/administración & dosificación , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Estimación de Kaplan-Meier , Modelos Logísticos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Pemetrexed , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Polimorfismo de Nucleótido Simple , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteína Portadora de Folato Reducido/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Timidilato Sintasa/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-21301108

RESUMEN

Phosphoribosylglycinamide formyltransferase (PurN) from Streptococcus mutans was recombinantly expressed in Escherichia coli. An effective purification protocol was established. The purified protein, which had a purity of >95%, was identified by SDS-PAGE and MALDI-TOF MS. The protein was crystallized using the vapour-diffusion method in hanging-drop mode with PEG 3350 as the primary precipitant. X-ray diffraction data were collected to 2.1 Šresolution. Preliminary X-ray analysis indicated that the crystal belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 52.25, b = 63.29, c = 131.81 Å.


Asunto(s)
Proteínas Bacterianas/química , Fosforribosilglicinamida-Formiltransferasa/química , Streptococcus mutans/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Tampones (Química) , Cristalización , Difusión , Electroforesis en Gel de Poliacrilamida , Escherichia coli/química , Escherichia coli/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/aislamiento & purificación , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Difracción de Rayos X , Rayos X
12.
FEMS Yeast Res ; 10(2): 199-208, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20082641

RESUMEN

Candida albicans is the principal human fungal pathogen that leads to life-threatening mycoses worldwide. To study its pathobiology, we characterized genes for two enzymes involved in the de novo purine biosynthesis pathway: ADE8 (encoding phosphoribosylglycinamide formyl-transferase) and GUA1 (GMP synthase). Heterozygous and homozygous disruption strains were constructed for both genes. We found that ADE8 and GUA1 are conditionally essential; i.e. can be bypassed in the presence of exogenous adenine and guanine, respectively, and that ADE8 plays an additional role in the C1-folate pool. Furthermore, the heterozygotes of ADE8/ade8 and GUA1/gua1 were hypersensitive to methotrexate (an inhibitor of de novo synthesis of tetrahydrofolate) and 6-azauracil (a known inhibitor of the IMP dehydrogenase involved in GMP biosynthesis), respectively. In a murine model of systemic candidiasis, the virulence of both heterozygous strains was marginally attenuated, while the ade8/ade8 and gua1/gua1 strains were completely avirulent. Our results and those of others indicate that many conditional essential genes involved in different biosynthesis pathways are required for systemic candidiasis, likely due to the host nutritional constraints imposed on the pathogen.


Asunto(s)
Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Candidiasis/microbiología , Ligasas de Carbono-Nitrógeno , Fosforribosilglicinamida-Formiltransferasa , Purinas/biosíntesis , Animales , Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/genética , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Humanos , Masculino , Metotrexato/farmacología , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Uracilo/análogos & derivados , Uracilo/farmacología , Virulencia
13.
Mol Biotechnol ; 42(3): 306-19, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19301155

RESUMEN

Human GARS-AIRS-GART encodes a fused tri-functional enzyme protein involved in de novo purine biosynthesis, aberrant function being implicated in Down syndrome and Leukemia. We performed phylogenetic analysis to discern evolutionary relationships and in silico characterization to identify elements potentially important for gene regulation. We report that murine, bovine and chimpanzee sequences are the nearest neighbors of human GARS-AIRS-GART and that endo-duplication of the AIRS protein is restricted to insect orthologs. Convergent evolution of mono-functional bacterial orthologs to bi-functional, partly fused, yeast orthologs is observed from the rooted-NJ tree topology that bears bootstrap values exceeding 9000 in majority of the nodes. Sequence alignments reveal that introns 11-15 of human GARS-AIRS-GART are conserved among vertebrates. An inverse correlation is observed between intron size and intron density without bias for intron position. The generation time of organisms is independent of intron density. Human, bovine and murine sequences possess similar GC content with CpG islands in promoter regions. The long isoforms of cow and chicken transcripts and short isoforms of human, bovine and murine mRNA form energetically stable stem-like structures in the 3'-UTR and may regulate translational stability of GARS-AIRS-GART transcripts. Glycine-rich loops important for enzyme structure and ATP-, folate-binding residues are partially conserved.


Asunto(s)
Ligasas de Carbono-Nitrógeno/genética , Fosforribosilglicinamida-Formiltransferasa/genética , Secuencia de Aminoácidos , Animales , Composición de Base , Ligasas de Carbono-Nitrógeno/metabolismo , Análisis por Conglomerados , Simulación por Computador , Islas de CpG , Regulación de la Expresión Génica , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Filogenia , Alineación de Secuencia , Estadísticas no Paramétricas , Regiones no Traducidas
14.
Cancer Res ; 66(7): 3836-44, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16585211

RESUMEN

Transport is required before reduced folates and anticancer antifolates [e.g., methotrexate (MTX)] exert their physiologic functions or cytotoxic effects. The folate/antifolate transporter with the widest tissue distribution and greatest activity is the reduced folate carrier (RFC). There is little evidence that RFC-mediated influx is posttranscriptionally regulated. We show that [(3)H]MTX influx in CCRF-CEM human childhood T-leukemia cells is potentiated up to 6-fold by exogenous 5-amino-4-imidazolecarboxamide riboside (AICAr) in a AICAr and MTX concentration-dependent manner. Metabolism to more biologically active polyglutamate forms is also potentiated for MTX and other antifolates. That potentiation of influx by AICAr is mediated by effects on the RFC is supported by analyses +/-AICAr showing (a) similarity and magnitude of kinetic constants for [(3)H]MTX influx; (b) similarity of inhibitory potency of known RFC substrates; (c) lack of potentiation in a CCRF-CEM subline that does not express the RFC; and (d) similarity of time and temperature dependence. Potentiation occurs rapidly and does not require new protein synthesis. Effects of specific inhibitors of folate metabolism and the time and sequence of AICAr incubation with cells suggest that both dihydrofolate reductase inhibition and metabolism of AICAr are essential for potentiation. Acute folate deficiency or incubation of CCRF-CEM with AICAr-related metabolites (e.g., adenosine) does not initiate potentiation. AICAr increases growth inhibitory potency of MTX and aminopterin against CCRF-CEM cells when both AICAr and antifolate are present for the first 24 hours of a 120-hour growth period. AICAr is the first small molecule that regulates RFC activity.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Leucemia de Células T/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Ribonucleótidos/farmacología , Aminoimidazol Carboxamida/farmacología , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Niño , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacocinética , Humanos , Cinética , Leucemia de Células T/tratamiento farmacológico , Metotrexato/análogos & derivados , Metotrexato/metabolismo , Metotrexato/farmacocinética , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Ácido Poliglutámico/análogos & derivados , Ácido Poliglutámico/biosíntesis , Ácido Poliglutámico/farmacocinética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteína Portadora de Folato Reducido , Fracciones Subcelulares , Temperatura , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/metabolismo , Tritio
15.
Methods Mol Biol ; 1764: 279-289, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29605921

RESUMEN

A long-standing hypothesis in the de novo purine biosynthetic pathway is that there must be highly coordinated processes to allow for enhanced metabolic flux when a cell demands purines. One mechanism by which the pathway meets its cellular demand is through the spatial organization of pathway enzymes into multienzyme complexes called purinosomes. Cellular conditions known to impact the activity of enzymes in the pathway or overall pathway flux have been reflected in a change in the number of purinosome-positive cells or the density of purinosomes in a given cell. The following general protocols outline the steps needed for purinosome detection through transient expression of fluorescent protein chimeras or through immunofluorescence in purine-depleted HeLa cells using confocal laser scanning microscopy. These protocols define a purinosome as a colocalization of FGAMS with one additional pathway enzyme, such as PPAT or GART, and provide insights into the proper identification of a purinosome from other reported cellular bodies.


Asunto(s)
Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Metaboloma , Microscopía Fluorescente/métodos , Nucleotidiltransferasas/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Purinas/metabolismo , Vías Biosintéticas , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos
16.
Mol Cancer Ther ; 5(9): 2211-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16985054

RESUMEN

Because of its ability to mimic a low energy status of the cell, the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside was proposed as an antineoplastic agent switching off major energy-consuming processes associated with the malignant phenotype (lipid production, DNA synthesis, cell proliferation, cell migration, etc.). Key to the antineoplastic action of AICA riboside is its conversion to ZMP, an AMP mimetic that at high concentrations activates the AMP-activated protein kinase (AMPK). Here, in an attempt to increase the efficacy of AICA riboside, we pretreated cancer cells with methotrexate, an antimetabolite blocking the metabolism of ZMP. Methotrexate enhanced the AICA riboside-induced accumulation of ZMP and led to a decrease in the levels of ATP, which functions as an intrasteric inhibitor of AMPK. Consequently, methotrexate markedly sensitized AMPK for activation by AICA riboside and potentiated the inhibitory effects of AICA riboside on tumor-associated processes. As cotreatment elicited antiproliferative effects already at concentrations of compounds that were only marginally effective when used alone, our findings on the cooperation between methotrexate and AICA riboside provide new opportunities both for the application of classic antimetabolic chemotherapeutics, such as methotrexate, and for the exploitation of the energy-sensing machinery as a target for cancer intervention.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Metotrexato/farmacología , Ribonucleósidos/farmacología , Proteínas Quinasas Activadas por AMP , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/antagonistas & inhibidores , Aminoimidazol Carboxamida/metabolismo , Aminoimidazol Carboxamida/farmacocinética , Aminoimidazol Carboxamida/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/biosíntesis , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Humanos , Lípidos/biosíntesis , Complejos Multienzimáticos/metabolismo , Nucleótido Desaminasas/antagonistas & inhibidores , Nucleótido Desaminasas/genética , Nucleótido Desaminasas/metabolismo , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/genética , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/metabolismo , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Purinas/antagonistas & inhibidores , Purinas/biosíntesis , Interferencia de ARN , Ribonucleósidos/farmacocinética , Ribonucleótidos/antagonistas & inhibidores , Ribonucleótidos/metabolismo
17.
Eur J Med Chem ; 139: 531-541, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-28830032

RESUMEN

A novel series of 6-substituted benzoyl and non-benzoyl straight chain pyrrolo[2,3-d]pyrimidines were designed and synthesized as potential antitumor agents targeting both thymidylate and purine nucleotide biosynthesis. Starting from the key intermediate 2-amino-4-oxo-pyrrolo[2,3-d]pyrimidin-6-yl-acetic acid, target compounds 1-6 were successfully obtained through two sequential condensation and saponification reactions in decent yield. The newly synthesized compounds showed antiproliferative potencies against a panel of tumor cell lines including KB, SW620 and MCF7. In particular, most compounds of this series exhibited nanomolar to subnanomolar inhibitory activities toward KB tumor cells, significantly more potent than the positive control methotrexate (MTX) and pemetrexed (PMX). Along with the results of nucleoside protection assays, molecular modeling studies suggested that the antitumor activity of compound 6 could be attributed to multitargeted inhibition of folate-dependent enzymes thymidylate synthase (TS), glycinamide ribonucleotide formyltransferase (GARFTase) and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFTase). Growth inhibition by compound 6 also induced distinct early apoptosis and cell cycle arrest at S-phase, which resulted in cell death.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Pirimidinas/farmacología , Pirroles/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/metabolismo , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Pirimidinas/síntesis química , Pirimidinas/química , Pirroles/síntesis química , Pirroles/química , Relación Estructura-Actividad , Timidilato Sintasa/metabolismo
18.
Cell Rep ; 21(5): 1331-1346, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29091770

RESUMEN

Mechanistic (or mammalian) target of rapamycin complex 1 (mTORC1) integrates signals from growth factors and nutrients to control biosynthetic processes, including protein, lipid, and nucleic acid synthesis. We find that the mTORC1 pathway is responsive to changes in purine nucleotides in a manner analogous to its sensing of amino acids. Depletion of cellular purines, but not pyrimidines, inhibits mTORC1, and restoration of intracellular adenine nucleotides via addition of exogenous purine nucleobases or nucleosides acutely reactivates mTORC1. Adenylate sensing by mTORC1 is dependent on the tuberous sclerosis complex (TSC) protein complex and its regulation of Rheb upstream of mTORC1, but independent of energy stress and AMP-activated protein kinase (AMPK). Even though mTORC1 signaling is not acutely sensitive to changes in intracellular guanylates, long-term depletion of guanylates decreases Rheb protein levels. Our findings suggest that nucleotide sensing, like amino acid sensing, enables mTORC1 to tightly coordinate nutrient availability with the synthesis of macromolecules, such as protein and nucleic acids, produced from those nutrients.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Nucleótidos de Purina/metabolismo , Células A549 , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Dihidroorotato Deshidrogenasa , Inhibidores Enzimáticos/farmacología , Fluorouracilo/farmacología , Células HeLa , Humanos , Mercaptopurina/farmacología , Metotrexato/farmacología , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Interferencia de ARN , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
19.
BMC Med Genet ; 7: 24, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16539728

RESUMEN

BACKGROUND: Down syndrome (DS) is caused by trisomy 21 (+21), but the aberrations in gene expression resulting from this chromosomal aneuploidy are not yet completely understood. METHODS: We used oligonucleotide microarrays to survey mRNA expression in early- and late-passage control and +21 fibroblasts and mid-gestation fetal hearts. We supplemented this analysis with northern blotting, western blotting, real-time RT-PCR, and immunohistochemistry. RESULTS: We found chromosome 21 genes consistently over-represented among the genes over-expressed in the +21 samples. However, these sets of over-expressed genes differed across the three cell/tissue types. The chromosome 21 gene MX1 was strongly over-expressed (mean 16-fold) in senescent +21 fibroblasts, a result verified by northern and western blotting. MX1 is an interferon target gene, and its mRNA was induced by interferons present in +21 fibroblast conditioned medium, suggesting an autocrine loop for its over-expression. By immunohistochemistry the p78MX1 protein was induced in lesional tissue of alopecia areata, an autoimmune disorder associated with DS. We found strong over-expression of the purine biosynthesis gene GART (mean 3-fold) in fetal hearts with +21 and verified this result by northern blotting and real-time RT-PCR. CONCLUSION: Different subsets of chromosome 21 genes are over-expressed in different cell types with +21, and for some genes this over-expression is non-linear (>1.5X). Hyperactive interferon signaling is a candidate pathway for cell senescence and autoimmune disorders in DS, and abnormal purine metabolism should be investigated for a potential role in cardiac defects.


Asunto(s)
Cromosomas Humanos Par 21 , Síndrome de Down/genética , Corazón Fetal/metabolismo , Fibroblastos/metabolismo , Alopecia Areata/genética , Alopecia Areata/metabolismo , Alopecia Areata/patología , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Células Cultivadas , Síndrome de Down/embriología , Síndrome de Down/metabolismo , Proteínas de Unión al GTP/biosíntesis , Proteínas de Unión al GTP/genética , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Proteínas de Resistencia a Mixovirus , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo
20.
Plant Physiol Biochem ; 44(10): 551-5, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17064924

RESUMEN

Levels of ATP and other nucleotides increased in wounded potato tuber slices, maintained on moist paper for 24 h after preparation. The relative expression intensity of genes encoding adenosine kinase (AK) and adenine phosphoribosyltransferase (APRT) in wounded slices was greater than the intensity of genes of the de novo pathway, glycineamide ribonucleotide formyltransferase (GART) and 5-aminoimidazole ribonucleotide synthetase (AIRS). In vitro activities of adenosine kinase (ATP:adenosine 5'-phosphotransferase; EC 2.7.1.20) and adenine phosphoribosyltransferase (AMP:pyrophosphate phospho-d-ribosyltransferase; EC 2.4.2.7) increased during wounding. Adenosine nucleosidase (adenosine ribohydrolase; EC 3.2.2.7) activity was negligible in freshly prepared slices, but its activity is dramatically enhanced in wounded slices. In situ adenosine salvage activity, estimated from the incorporation of radioactivity from exogenously supplied [8-(14)C]adenosine into nucleotides and RNA, increased more than five times in the wounded slices. These results strongly suggest that greater expression of the genes encoding enzymes of adenosine salvage during wounding is closely related to the increased supply of adenine nucleotides in the wounded slices.


Asunto(s)
Adenosina Monofosfato/biosíntesis , Adenosina/metabolismo , Tubérculos de la Planta/metabolismo , Solanum tuberosum/metabolismo , Adenina Fosforribosiltransferasa/metabolismo , Adenosina Quinasa/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Fosforribosilglicinamida-Formiltransferasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA