Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 695: 149400, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38160530

RESUMEN

SETD2 (SET-domain containing protein 2) is a histone methyltransferase (HMT) of the SET family responsible for the trimethylation of K36 of histone H3, thus producing the epigenetic mark H3K36me3. Recent studies have shown that certain SET family HMTs, such as SMYD2, SMYD3 or SETDB1 can also methylate protein kinases and therefore be involved in signaling pathways. Here we provide structural and enzymatic evidence showing that SETD2 methylates the protein tyrosine kinase ACK1 in vitro. ACK1 is recognized as a major integrator of signaling from various receptor tyrosine kinases. Using ACK1 peptides and recombinant proteins, we show that SETD2 methylates the K514 residue of ACK1 generating K514 mono, di or tri-methylation. Interestingly, K514 is found in a "H3K36-like" motif of ACK1 which is known to be post-translationally modified and to be involved in protein-protein interaction. The crystal structure of SETD2 catalytic domain in complex with an ACK1 peptide further provides the structural basis for the methylation of ACK1 K514 by SETD2. Our work therefore strongly suggests that ACK1 could be a novel non-histone substrate of SETD2 and further supports that SET HMTs, such as SETD2, could be involved in both epigenetic regulations and cell signaling.


Asunto(s)
Histonas , Proteínas Tirosina Quinasas , Proteínas Tirosina Quinasas/metabolismo , Histonas/metabolismo , Metilación , N-Metiltransferasa de Histona-Lisina/genética , Procesamiento Proteico-Postraduccional
2.
Nucleic Acids Res ; 50(14): 7938-7958, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35871293

RESUMEN

Although originally described as transcriptional activator, SPI1/PU.1, a major player in haematopoiesis whose alterations are associated with haematological malignancies, has the ability to repress transcription. Here, we investigated the mechanisms underlying gene repression in the erythroid lineage, in which SPI1 exerts an oncogenic function by blocking differentiation. We show that SPI1 represses genes by binding active enhancers that are located in intergenic or gene body regions. HDAC1 acts as a cooperative mediator of SPI1-induced transcriptional repression by deacetylating SPI1-bound enhancers in a subset of genes, including those involved in erythroid differentiation. Enhancer deacetylation impacts on promoter acetylation, chromatin accessibility and RNA pol II occupancy. In addition to the activities of HDAC1, polycomb repressive complex 2 (PRC2) reinforces gene repression by depositing H3K27me3 at promoter sequences when SPI1 is located at enhancer sequences. Moreover, our study identified a synergistic relationship between PRC2 and HDAC1 complexes in mediating the transcriptional repression activity of SPI1, ultimately inducing synergistic adverse effects on leukaemic cell survival. Our results highlight the importance of the mechanism underlying transcriptional repression in leukemic cells, involving complex functional connections between SPI1 and the epigenetic regulators PRC2 and HDAC1.


Asunto(s)
Histona Desacetilasa 1 , Leucemia Eritroblástica Aguda , Complejo Represivo Polycomb 2 , Proteínas Proto-Oncogénicas , Transactivadores , Acetilación , Animales , Cromatina/genética , Histona Desacetilasa 1/genética , Leucemia Eritroblástica Aguda/genética , Ratones , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética
3.
Clin Genet ; 101(5-6): 552-558, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35132614

RESUMEN

Variants in aminoacyl-tRNA synthetases (ARSs) genes are associated to a broad spectrum of human inherited diseases. Patients with defective PheRS, encoded by FARSA and FARSB, display brain abnormalities, interstitial lung disease and facial dysmorphism. We investigated four children from two unrelated consanguineous families carrying two missense homozygous variants in FARSA with significantly reduced PheRS-mediated aminoacylation activity. In addition to the core ARS-phenotype, all patients showed an inflammatory profile associated with autoimmunity and interferon score, a clinical feature not ascribed to PheRS-deficient patients to date. JAK inhibition improved lung disease in one patient. Our findings expand the genetic and clinical spectrum of FARSA-related disease.


Asunto(s)
Aminoacil-ARNt Sintetasas , Enfermedad de Charcot-Marie-Tooth , Enfermedades Pulmonares Intersticiales , Aminoacil-ARNt Sintetasas/genética , Enfermedad de Charcot-Marie-Tooth/genética , Consanguinidad , Humanos , Enfermedades Pulmonares Intersticiales/genética , Fenotipo , Síndrome
4.
Int J Mol Sci ; 23(13)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35806030

RESUMEN

Phosphorylation is an essential process in biological events and is considered critical for biological functions. In tissues, protein phosphorylation mainly occurs on tyrosine (Tyr), serine (Ser) and threonine (Thr) residues. The balance between phosphorylation and dephosphorylation is under the control of two super enzyme families, protein kinases (PKs) and protein phosphatases (PPs), respectively. Although there are many selective and effective drugs targeting phosphokinases, developing drugs targeting phosphatases is challenging. PTP1B, one of the most central protein tyrosine phosphatases (PTPs), is a key player in several human diseases and disorders, such as diabetes, obesity, and hematopoietic malignancies, through modulation of different signaling pathways. However, due to high conservation among PTPs, most PTP1B inhibitors lack specificity, raising the need to develop new strategies targeting this enzyme. In this mini-review, we summarize three classes of PTP1B inhibitors with different mechanisms: (1) targeting multiple aryl-phosphorylation sites including the catalytic site of PTP1B; (2) targeting allosteric sites of PTP1B; (3) targeting specific mRNA sequence of PTP1B. All three types of PTP1B inhibitors present good specificity over other PTPs and are promising for the development of efficient small molecules targeting this enzyme.


Asunto(s)
Inhibidores Enzimáticos , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Sitio Alostérico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Transducción de Señal
5.
Int J Mol Sci ; 23(13)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35806064

RESUMEN

Human protein tyrosine phosphatase 1B (PTP1B) is a ubiquitous non-receptor tyrosine phosphatase that serves as a major negative regulator of tyrosine phosphorylation cascades of metabolic and oncogenic importance such as the insulin, epidermal growth factor receptor (EGFR), and JAK/STAT pathways. Increasing evidence point to a key role of PTP1B-dependent signaling in cancer. Interestingly, genetic defects in PTP1B have been found in different human malignancies. Notably, recurrent somatic mutations and splice variants of PTP1B were identified in human B cell and Hodgkin lymphomas. In this work, we analyzed the molecular and functional levels of three PTP1B mutations identified in primary mediastinal B cell lymphoma (PMBCL) patients and located in the WPD-loop (V184D), P-loop (R221G), and Q-loop (G259V). Using biochemical, enzymatic, and molecular dynamics approaches, we show that these mutations lead to PTP1B mutants with extremely low intrinsic tyrosine phosphatase activity that display alterations in overall protein stability and in the flexibility of the active site loops of the enzyme. This is in agreement with the key role of the active site loop regions, which are preorganized to interact with the substrate and to enable catalysis. Our study provides molecular and enzymatic evidence for the loss of protein tyrosine phosphatase activity of PTP1B active-site loop mutants identified in human lymphoma.


Asunto(s)
Linfoma de Células B , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Dominio Catalítico , Humanos , Linfoma de Células B/genética , Mutación , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Tirosina/metabolismo
6.
Mol Pharmacol ; 100(3): 283-294, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34266924

RESUMEN

Human SETD2 is the unique histone methyltransferase that generates H3K36 trimethylation (H3K36me3), an epigenetic mark that plays a key role in normal hematopoiesis. Interestingly, recurrent inactivating mutations of SETD2 and aberrant H3K36me3 are increasingly reported to be involved in hematopoietic malignancies. Benzene (BZ) is a ubiquitous environmental pollutant and carcinogen that causes leukemia. The leukemogenic properties of BZ depend on its biotransformation in the bone marrow into oxidative metabolites, in particular 1,4-benzoquinone (BQ). This hematotoxic metabolite can form DNA and protein adducts that result in the damage and the alteration of cellular processes. Recent studies suggest that BZ-dependent leukemogenesis could depend on epigenetic perturbations, notably aberrant histone methylation. We investigated whether H3K36 trimethylation by SETD2 could be impacted by BZ and its hematotoxic metabolites. Herein, we show that BQ, the major leukemogenic metabolite of BZ, inhibits irreversibly the human histone methyltransferase SETD2, resulting in decreased H3K36me3. Our mechanistic studies further indicate that the BQ-dependent inactivation of SETD2 is due to covalent binding of BQ to reactive Zn-finger cysteines within the catalytic domain of the enzyme. The formation of these quinoprotein adducts results in loss of enzyme activity and protein crosslinks/oligomers. Experiments conducted in hematopoietic cells confirm that exposure to BQ results in the formation of SETD2 crosslinks/oligomers and concomitant loss of H3K36me3 in cells. Taken together, our data indicate that BQ, a major hematotoxic metabolite of BZ, could contribute to BZ-dependent leukemogenesis by perturbing the functions of SETD2, a histone lysine methyltransferase of hematopoietic relevance. SIGNIFICANCE STATEMENT: Benzoquinone is a major leukemogenic metabolite of benzene. Dysregulation of histone methyltransferase is involved in hematopoietic malignancies. This study found that benzoquinone irreversibly impairs SET domain containing 2, a histone H3K36 methyltransferase that plays a key role in hematopoiesis. Benzoquinone forms covalent adducts on Zn-finger cysteines within the catalytic site, leading to loss of activity, protein crosslinks/oligomers, and concomitant decrease of H3K36me3 histone mark. These data provide evidence that a leukemogenic metabolite of benzene can impair a key epigenetic enzyme.


Asunto(s)
Benceno/metabolismo , Benceno/toxicidad , Benzoquinonas/toxicidad , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Benceno/química , Benzoquinonas/química , Línea Celular , Cisteína/química , Cisteína/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Humanos , Leucemia/inducido químicamente , Leucemia/genética , Leucemia/metabolismo , Metilación , Cultivo Primario de Células , Dedos de Zinc/efectos de los fármacos
7.
Haematologica ; 106(9): 2478-2488, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32855277

RESUMEN

Vaso-occlusive crises are the hallmark of sickle cell disease (SCD). They are believed to occur in two steps, starting with adhesion of deformable low-dense red blood cells (RBCs), or other blood cells such as neutrophils, to the wall of post-capillary venules, followed by trapping of the denser RBCs or leukocytes in the areas of adhesion because of reduced effective lumen-diameter. In SCD, RBCs are heterogeneous in terms of density, shape, deformability and surface proteins, which accounts for the differences observed in their adhesion and resistance to shear stress. Sickle RBCs exhibit abnormal adhesion to laminin mediated by Lu/BCAM protein at their surface. This adhesion is triggered by Lu/BCAM phosphorylation in reticulocytes but such phosphorylation does not occur in mature dense RBCs despite firm adhesion to laminin. In this study, we investigated the adhesive properties of sickle RBC subpopulations and addressed the molecular mechanism responsible for the increased adhesion of dense RBCs to laminin in the absence of Lu/BCAM phosphorylation. We provide evidence for the implication of oxidative stress in post-translational modifications of Lu/BCAM that impact its distribution and cis-interaction with glycophorin C at the cell surface activating its adhesive function in sickle dense RBCs.


Asunto(s)
Anemia de Células Falciformes , Laminina , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Eritrocitos/metabolismo , Humanos , Laminina/metabolismo , Sistema del Grupo Sanguíneo Lutheran/metabolismo , Estrés Oxidativo
8.
J Biol Chem ; 294(33): 12483-12494, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31248982

RESUMEN

Protein tyrosine phosphatase, nonreceptor type 2 (PTPN2) is mainly expressed in hematopoietic cells, where it negatively regulates growth factor and cytokine signaling. PTPN2 is an important regulator of hematopoiesis and immune/inflammatory responses, as evidenced by loss-of-function mutations of PTPN2 in leukemia and lymphoma and knockout mice studies. Benzene is an environmental chemical that causes hematological malignancies, and its hematotoxicity arises from its bioactivation in the bone marrow to electrophilic metabolites, notably 1,4-benzoquinone, a major hematotoxic benzene metabolite. Although the molecular bases for benzene-induced leukemia are not well-understood, it has been suggested that benzene metabolites alter topoisomerases II function and thereby significantly contribute to leukemogenesis. However, several studies indicate that benzene and its hematotoxic metabolites may also promote the leukemogenic process by reacting with other targets and pathways. Interestingly, alterations of cell-signaling pathways, such as Janus kinase (JAK)/signal transducer and activator of transcription (STAT), have been proposed to contribute to benzene-induced malignant blood diseases. We show here that 1,4-benzoquinone directly impairs PTPN2 activity. Mechanistic and kinetic experiments with purified human PTPN2 indicated that this impairment results from the irreversible formation (kinact = 645 m-1·s-1) of a covalent 1,4-benzoquinone adduct at the catalytic cysteine residue of the enzyme. Accordingly, cell experiments revealed that 1,4-benzoquinone exposure irreversibly inhibits cellular PTPN2 and concomitantly increases tyrosine phosphorylation of STAT1 and expression of STAT1-regulated genes. Our results provide molecular and cellular evidence that 1,4-benzoquinone covalently modifies key signaling enzymes, implicating it in benzene-induced malignant blood diseases.


Asunto(s)
Benceno , Benzoquinonas/metabolismo , Leucemia , Proteínas de Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Factor de Transcripción STAT1 , Transducción de Señal/efectos de los fármacos , Benceno/farmacocinética , Benceno/farmacología , Células HEK293 , Humanos , Células Jurkat , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/genética
9.
Biochem Biophys Res Commun ; 525(2): 308-312, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32089267

RESUMEN

Transglutaminases (TG) and arylamine N-acetyltransferases (NAT) are important family of enzymes. Although they catalyze different reactions and have distinct structures, these two families of enzymes share a spatially conserved catalytic triad (Cys, His, Asp residues). In active TGs, a conserved Trp residue located close to the triad cysteine is crucial for catalysis through stabilization of transition states. Here, we show that in addition to sharing a similar catalytic triad with TGs, functional NAT enzymes also possess in their active site an aromatic residue (Phe, Tyr or Trp) occupying a structural position similar to the Trp residue of active TGs. More importantly, as observed in active TGs, our data indicates that in functional NAT enzymes this conserved aromatic residue is also involved in stabilization of transition states. These results thus indicate that in addition to the three triad residues, these two families of enzymes also share a spatially conserved aromatic amino acid position important for catalysis. Identification of residues involved in the stabilization of transition states is important to develop potent inhibitors. Interestingly, NAT enzymes have been shown as potential targets of clinical interest.


Asunto(s)
Secuencia de Aminoácidos , Arilamina N-Acetiltransferasa/química , Secuencia Conservada , Transglutaminasas/química , Aminoácidos Aromáticos , Animales , Biocatálisis , Dominio Catalítico , Humanos , Transglutaminasas/genética
10.
Mol Pharmacol ; 96(2): 297-306, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31221825

RESUMEN

Etoposide is a widely prescribed anticancer drug that is, however, associated with an increased risk of secondary leukemia. Although the molecular basis underlying the development of these leukemias remains poorly understood, increasing evidence implicates the interaction of etoposide metabolites [i.e., etoposide quinone (EQ)] with topoisomerase II enzymes. However, effects of etoposide quinone on other cellular targets could also be at play. We investigated whether T-cell protein tyrosine phosphatase (TCPTP), a protein tyrosine phosphatase that plays a key role in normal and malignant hematopoiesis through regulation of Janus kinase/signal transducer and activator of transcription signaling, could be a target of EQ. We report here that EQ is an irreversible inhibitor of TCPTP phosphatase (IC50 = ∼7 µM, second-order rate inhibition constant of ∼810 M-1⋅min-1). No inhibition was observed with the parent drug. The inhibition by EQ was found to be due to the formation of a covalent adduct at the catalytic cysteine residue in the active site of TCPTP. Exposure of human hematopoietic cells (HL60 and Jurkat) to EQ led to inhibition of endogenous TCPTP and concomitant increase in STAT1 tyrosine phosphorylation. Our results suggest that in addition to alteration of topoisomerase II functions, EQ could also contribute to etoposide-dependent leukemogenesis through impairment of key hematopoietic signaling enzymes, such as TCPTP.


Asunto(s)
Etopósido/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Quinonas/farmacología , Sitios de Unión , Dominio Catalítico , Cisteína/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Células Jurkat , Fosforilación/efectos de los fármacos , Quinonas/química , Factor de Transcripción STAT1/metabolismo
11.
Blood ; 129(19): 2645-2656, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28288979

RESUMEN

CREBBP is targeted by inactivating mutations in follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL). Here, we provide evidence from transgenic mouse models that Crebbp deletion results in deficits in B-cell development and can cooperate with Bcl2 overexpression to promote B-cell lymphoma. Through transcriptional and epigenetic profiling of these B cells, we found that Crebbp inactivation was associated with broad transcriptional alterations, but no changes in the patterns of histone acetylation at the proximal regulatory regions of these genes. However, B cells with Crebbp inactivation showed high expression of Myc and patterns of altered histone acetylation that were localized to intragenic regions, enriched for Myc DNA binding motifs, and showed Myc binding. Through the analysis of CREBBP mutations from a large cohort of primary human FL and DLBCL, we show a significant difference in the spectrum of CREBBP mutations in these 2 diseases, with higher frequencies of nonsense/frameshift mutations in DLBCL compared with FL. Together, our data therefore provide important links between Crebbp inactivation and Bcl2 dependence and show a role for Crebbp inactivation in the induction of Myc expression. We suggest this may parallel the role of CREBBP frameshift/nonsense mutations in DLBCL that result in loss of the protein, but may contrast the role of missense mutations in the lysine acetyltransferase domain that are more frequently observed in FL and yield an inactive protein.


Asunto(s)
Linfocitos B/patología , Proteína de Unión a CREB/genética , Regulación Neoplásica de la Expresión Génica , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Epigénesis Genética , Eliminación de Gen , Humanos , Linfoma Folicular/genética , Ratones , Ratones Transgénicos , Mutación
12.
J Biol Chem ; 292(5): 1603-1612, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27965358

RESUMEN

Dithiocarbamates (DTCs) are important industrial chemicals used extensively as pesticides and in a variety of therapeutic applications. However, they have also been associated with neurotoxic effects and in particular with the development of Parkinson-like neuropathy. Although different pathways and enzymes (such as ubiquitin ligases or the proteasome) have been identified as potential targets of DTCs in the brain, the molecular mechanisms underlying their neurotoxicity remain poorly understood. There is increasing evidence that alteration of glycogen metabolism in the brain contributes to neurodegenerative processes. Interestingly, recent studies with N,N-diethyldithiocarbamate suggest that brain glycogen phosphorylase (bGP) and glycogen metabolism could be altered by DTCs. Here, we provide molecular and mechanistic evidence that bGP is a target of DTCs. To examine this system, we first tested thiram, a DTC pesticide known to display neurotoxic effects, observing that it can react rapidly with bGP and readily inhibits its glycogenolytic activity (kinact = 1.4 × 105 m-1 s-1). Using cysteine chemical labeling, mass spectrometry, and site-directed mutagenesis approaches, we show that thiram (and certain of its metabolites) alters the activity of bGP through the formation of an intramolecular disulfide bond (Cys318-Cys326), known to act as a redox switch that precludes the allosteric activation of bGP by AMP. Given the key role of glycogen metabolism in brain functions and neurodegeneration, impairment of the glycogenolytic activity of bGP by DTCs such as thiram may be a new mechanism by which certain DTCs exert their neurotoxic effects.


Asunto(s)
Glucógeno Fosforilasa de Forma Encefálica/química , Neurotoxinas/química , Tiocarbamatos/química , Glucógeno/metabolismo , Glucógeno Fosforilasa de Forma Encefálica/genética , Glucógeno Fosforilasa de Forma Encefálica/metabolismo , Humanos , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Neurotoxinas/toxicidad , Tiocarbamatos/toxicidad
13.
Mol Pharmacol ; 92(3): 358-365, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28674152

RESUMEN

Thiram (tetramethylthiuram disulfide) is a representative dithiocarbamate (DTC) pesticide used in both the field and as a seed protectant. The widespread use of Thiram and other DTC pesticides has raised concerns for health, because these compounds can exert neuropathic, endocrine disruptive, and carcinogenic effects. These toxic effects are thought to rely, at least in part, on the reaction of Thiram (and certain of its metabolites) with cellular protein thiols with subsequent loss of protein function. So far, a limited number of molecular targets of Thiram have been reported, including few enzymes such as dopamine ß-hydroxylase, 11ß-hydroxysteroid dehydrogenase, and brain glycogen phosphorylase. We provide evidence that Thiram is an inhibitor (KI = 23 µM; kinact = 0.085 second-1; kinact/KI = 3691 M-1⋅s-1) of human arylamine N-acetyltransferase 1 (NAT1), a phase II xenobiotic-metabolizing enzyme that plays a key role in the biotransformation of aromatic amine xenobiotics. Thiram was found to act as an irreversible inhibitor through the modification of NAT1 catalytic cysteine residue as also reported for other enzymes targeted by this pesticide. We also showed using purified NAT1 and human keratinocytes that Thiram impaired the N-acetylation of 3,4-dichloroaniline (3,4-DCA), a major toxic metabolite of aromatic amine pesticides (such as Diuron or Propanil). As coexposure to different classes of pesticides is common, our data suggest that pharmacokinetic drug-drug interactions between DTC pesticides such as Thiram and aromatic amine pesticides may occur through alteration of NAT1 enzymes functions.


Asunto(s)
Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Fungicidas Industriales/farmacología , Isoenzimas/antagonistas & inhibidores , Tiram/farmacología , Acetilación , Compuestos de Anilina/metabolismo , Células Cultivadas , Ditiotreitol/farmacología , Humanos
14.
J Biol Chem ; 291(46): 23842-23853, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27660393

RESUMEN

Brain glycogen and its metabolism are increasingly recognized as major players in brain functions. Moreover, alteration of glycogen metabolism in the brain contributes to neurodegenerative processes. In the brain, both muscle and brain glycogen phosphorylase isozymes regulate glycogen mobilization. However, given their distinct regulatory features, these two isozymes could confer distinct metabolic functions of glycogen in brain. Interestingly, recent proteomics studies have identified isozyme-specific reactive cysteine residues in brain glycogen phosphorylase (bGP). In this study, we show that the activity of human bGP is redox-regulated through the formation of a disulfide bond involving a highly reactive cysteine unique to the bGP isozyme. We found that this disulfide bond acts as a redox switch that precludes the allosteric activation of the enzyme by AMP without affecting its activation by phosphorylation. This unique regulatory feature of bGP sheds new light on the isoform-specific regulation of glycogen phosphorylase and glycogen metabolism.


Asunto(s)
Disulfuros/química , Glucógeno Fosforilasa de Forma Encefálica/química , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Regulación Alostérica/fisiología , Animales , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Disulfuros/metabolismo , Glucógeno/química , Glucógeno/metabolismo , Glucógeno Fosforilasa de Forma Encefálica/genética , Glucógeno Fosforilasa de Forma Encefálica/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Oxidación-Reducción , Fosforilación/fisiología , Conejos , Ratas
15.
J Biol Chem ; 291(35): 18072-83, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27402852

RESUMEN

Brain glycogen metabolism plays a critical role in major brain functions such as learning or memory consolidation. However, alteration of glycogen metabolism and glycogen accumulation in the brain contributes to neurodegeneration as observed in Lafora disease. Glycogen phosphorylase (GP), a key enzyme in glycogen metabolism, catalyzes the rate-limiting step of glycogen mobilization. Moreover, the allosteric regulation of the three GP isozymes (muscle, liver, and brain) by metabolites and phosphorylation, in response to hormonal signaling, fine-tunes glycogenolysis to fulfill energetic and metabolic requirements. Whereas the structures of muscle and liver GPs have been known for decades, the structure of brain GP (bGP) has remained elusive despite its critical role in brain glycogen metabolism. Here, we report the crystal structure of human bGP in complex with PEG 400 (2.5 Å) and in complex with its allosteric activator AMP (3.4 Å). These structures demonstrate that bGP has a closer structural relationship with muscle GP, which is also activated by AMP, contrary to liver GP, which is not. Importantly, despite the structural similarities between human bGP and the two other mammalian isozymes, the bGP structures reveal molecular features unique to the brain isozyme that provide a deeper understanding of the differences in the activation properties of these allosteric enzymes by the allosteric effector AMP. Overall, our study further supports that the distinct structural and regulatory properties of GP isozymes contribute to the different functions of muscle, liver, and brain glycogen.


Asunto(s)
Adenosina Monofosfato/química , Glucógeno Fosforilasa de Forma Encefálica/química , Proteínas del Tejido Nervioso/química , Adenosina Monofosfato/genética , Adenosina Monofosfato/metabolismo , Regulación Alostérica , Cristalografía por Rayos X , Glucógeno Fosforilasa de Forma Encefálica/genética , Glucógeno Fosforilasa de Forma Encefálica/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Enfermedad de Lafora/genética , Enfermedad de Lafora/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Dominios Proteicos
16.
Mol Cell ; 34(2): 223-33, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19394299

RESUMEN

Rho family GTPases are important cellular switches and control a number of physiological functions. Understanding the molecular basis of interaction of these GTPases with their effectors is crucial in understanding their functions in the cell. Here we present the crystal structure of the complex of Rac2 bound to the split pleckstrin homology (spPH) domain of phospholipase C-gamma(2) (PLCgamma(2)). Based on this structure, we illustrate distinct requirements for PLCgamma(2) activation by Rac and EGF and generate Rac effector mutants that specifically block activation of PLCgamma(2), but not the related PLCbeta(2) isoform. Furthermore, in addition to the complex, we report the crystal structures of free spPH and Rac2 bound to GDP and GTPgammaS. These structures illustrate a mechanism of conformational switches that accompany formation of signaling active complexes and highlight the role of effector binding as a common feature of Rac and Cdc42 interactions with a variety of effectors.


Asunto(s)
Fosfolipasa C gamma/química , Proteínas de Unión al GTP rac/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Activación Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Fosfolipasa C gamma/metabolismo , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Alineación de Secuencia , Especificidad por Sustrato , Termodinámica , Proteínas de Unión al GTP rac/metabolismo , Proteína RCA2 de Unión a GTP
18.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 2): 266-73, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25664736

RESUMEN

Arylamine N-acetyltransferases (NATs) are xenobiotic metabolizing enzymes that catalyze the acetyl-CoA-dependent acetylation of arylamines. To better understand the mode of binding of the cofactor by this family of enzymes, the structure of Mesorhizobium loti NAT1 [(RHILO)NAT1] was determined in complex with CoA. The F42W mutant of (RHILO)NAT1 was used as it is well expressed in Escherichia coli and displays enzymatic properties similar to those of the wild type. The apo and holo structures of (RHILO)NAT1 F42W were solved at 1.8 and 2 Šresolution, respectively. As observed in the Mycobacterium marinum NAT1-CoA complex, in (RHILO)NAT1 CoA binding induces slight structural rearrangements that are mostly confined to certain residues of its `P-loop'. Importantly, it was found that the mode of binding of CoA is highly similar to that of M. marinum NAT1 but different from the modes reported for Bacillus anthracis NAT1 and Homo sapiens NAT2. Therefore, in contrast to previous data, this study shows that different orthologous NATs can bind their cofactors in a similar way, suggesting that the mode of binding CoA in this family of enzymes is less diverse than previously thought. Moreover, it supports the notion that the presence of the `mammalian/eukaryotic insertion loop' in certain NAT enzymes impacts the mode of binding CoA by imposing structural constraints.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Coenzima A/metabolismo , Mesorhizobium/enzimología , Secuencia de Aminoácidos , Arilamina N-Acetiltransferasa/química , Arilamina N-Acetiltransferasa/genética , Sitios de Unión , Coenzima A/química , Cristalografía por Rayos X , Mesorhizobium/química , Mesorhizobium/genética , Mesorhizobium/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Puntual , Conformación Proteica , Alineación de Secuencia
19.
Fungal Genet Biol ; 81: 82-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26068093

RESUMEN

Arylamines are frequent pollutants in soils. Fungi have proven to be efficient in detoxifying these chemicals by acetylating them using arylamine N-acetyl transferase enzymes. Here, we selected from natural soils fungi highly resistant to 3,4-dichloroaniline (DCA). Fusarium species were the most frequently isolated species, especially Fusarium solani. The sequenced strain of F. solani contains five NAT genes, as did all the DCA-resistant isolates. RT-PCR analysis showed that the five genes were expressed in F. solani. Expression of the F. solani genes in Podospora anserina and analysis of acetylation directly in F. solani showed that only the NhNAT2B gene conferred significant resistance to DCA and that F. solani likely uses pathways different from acetylation to resist high doses of DCA, as observed previously for Trichoderma.


Asunto(s)
Compuestos de Anilina/toxicidad , Fusarium/aislamiento & purificación , Microbiología del Suelo , Contaminantes del Suelo/toxicidad , Acetiltransferasas , Secuencia de Aminoácidos , Fusarium/enzimología , Perfilación de la Expresión Génica , Inactivación Metabólica , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
20.
Anal Biochem ; 486: 35-7, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26099937

RESUMEN

CREB-binding protein (CBP) is a lysine acetyltransferase that regulates transcription by acetylating histone and non-histone substrates. Defects in CBP activity are associated with hematologic malignancies, neurodisorders, and congenital malformations. Sensitive and quantitative enzymatic assays are essential to better characterize the pathophysiological features of CBP. We describe a sensitive nonradioactive method to measure purified and immunopurified cellular CBP enzymatic activity through rapid reverse phase-ultra-fast liquid chromatography (RP-UFLC) analysis of fluorescent histone H3 peptide substrates. The applicability and biological relevance of the assay are supported by kinetic, inhibition, and immunoprecipitation studies. More broadly, this approach could be easily adapted to assay other lysine acetyltransferases or methyltransferases.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Pruebas de Enzimas/métodos , Colorantes Fluorescentes/metabolismo , Histonas/química , Fragmentos de Péptidos/metabolismo , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Colorantes Fluorescentes/química , Humanos , Cinética , Datos de Secuencia Molecular , Fragmentos de Péptidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA