Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Microb Cell Fact ; 23(1): 178, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879464

RESUMEN

BACKGROUND: Computational mining of useful enzymes and biosynthesis pathways is a powerful strategy for metabolic engineering. Through systematic exploration of all conceivable combinations of enzyme reactions, including both known compounds and those inferred from the chemical structures of established reactions, we can uncover previously undiscovered enzymatic processes. The application of the novel alternative pathways enables us to improve microbial bioproduction by bypassing or reinforcing metabolic bottlenecks. Benzylisoquinoline alkaloids (BIAs) are a diverse group of plant-derived compounds with important pharmaceutical properties. BIA biosynthesis has developed into a prime example of metabolic engineering and microbial bioproduction. The early bottleneck of BIA production in Escherichia coli consists of 3,4-dihydroxyphenylacetaldehyde (DHPAA) production and conversion to tetrahydropapaveroline (THP). Previous studies have selected monoamine oxidase (MAO) and DHPAA synthase (DHPAAS) to produce DHPAA from dopamine and oxygen; however, both of these enzymes produce toxic hydrogen peroxide as a byproduct. RESULTS: In the current study, in silico pathway design is applied to relieve the bottleneck of DHPAA production in the synthetic BIA pathway. Specifically, the cytochrome P450 enzyme, tyrosine N-monooxygenase (CYP79), is identified to bypass the established MAO- and DHPAAS-mediated pathways in an alternative arylacetaldoxime route to DHPAA with a peroxide-independent mechanism. The application of this pathway is proposed to result in less formation of toxic byproducts, leading to improved production of reticuline (up to 60 mg/L at the flask scale) when compared with that from the conventional MAO pathway. CONCLUSIONS: This study showed improved reticuline production using the bypass pathway predicted by the M-path computational platform. Reticuline production in E. coli exceeded that of the conventional MAO-mediated pathway. The study provides a clear example of the integration of pathway mining and enzyme design in creating artificial metabolic pathways and suggests further potential applications of this strategy in metabolic engineering.


Asunto(s)
Bencilisoquinolinas , Escherichia coli , Ingeniería Metabólica , Ingeniería Metabólica/métodos , Bencilisoquinolinas/metabolismo , Escherichia coli/metabolismo , Escherichia coli/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Vías Biosintéticas , Simulación por Computador , Tetrahidropapaverolina/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Ácido 3,4-Dihidroxifenilacético/análogos & derivados
2.
Sci Rep ; 14(1): 11410, 2024 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762658

RESUMEN

A series of novel Schiff base derivatives (1-28) of 3,4-dihydroxyphenylacetic acid were synthesized in a multi-step reaction. All the synthesized Schiff bases were obtained in high yields and their structures were determined by 1HNMR, 13CNMR, and HR-ESI-MS spectroscopy. Except for compounds 22, 26, 27, and 28, all derivatives show excellent to moderate α-glucosidase inhibition. Compounds 5 (IC50 = 12.84 ± 0.52 µM), 4 (IC50 = 13.64 ± 0.58 µM), 12 (IC50 = 15.73 ± 0.71 µM), 13 (IC50 = 16.62 ± 0.47 µM), 15 (IC50 = 17.40 ± 0.74 µM), 3 (IC50 = 18.45 ± 1.21 µM), 7 (IC50 = 19.68 ± 0.82 µM), and 2 (IC50 = 20.35 ± 1.27 µM) shows outstanding inhibition as compared to standard acarbose (IC50 = 873.34 ± 1.67 µM). Furthermore, a docking study was performed to find out the interaction between the enzyme and the most active compounds. With this research work, 3,4-dihydroxyphenylacetic acid Schiff base derivatives have been introduced as a potential class of α-glucosidase inhibitors that have remained elusive till now.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético , Diseño de Fármacos , Inhibidores de Glicósido Hidrolasas , Simulación del Acoplamiento Molecular , Bases de Schiff , alfa-Glucosidasas , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/farmacología , Inhibidores de Glicósido Hidrolasas/síntesis química , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/química , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/química , Ácido 3,4-Dihidroxifenilacético/metabolismo , Ácido 3,4-Dihidroxifenilacético/farmacología , Bases de Schiff/química , Bases de Schiff/farmacología , Hidrazonas/química , Hidrazonas/farmacología , Hidrazonas/síntesis química , Relación Estructura-Actividad
3.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34769179

RESUMEN

Neurogenerative diseases, such as Parkinson's disease, are associated, not only with the selective loss of dopamine (DA), but also with the accumulation of reactive catechol-aldehyde, 3,4-dihydroxyphenylacetaldehyde (DOPAL), which is formed as the immediate oxidation product of cytoplasmic DA by monoamine oxidase. DOPAL is well known to exhibit toxic effects on neuronal cells. Both catecholic and aldehyde groups seem to be associated with the neurotoxicity of DOPAL. However, the exact cause of toxicity caused by this compound remains unknown. Since the reactivity of DOPAL could be attributed to its immediate oxidation product, DOPAL-quinone, we examined the potential reactions of this toxic metabolite. The oxidation of DOPAL by mushroom tyrosinase at pH 5.3 produced conventional DOPAL-quinone, but oxidation at pH 7.4 produced the tautomeric quinone-methide, which gave rise to 3,4-dihydroxyphenylglycolaldehyde and 3,4-dihydroxybenzaldehyde as products through a series of reactions. When the oxidation reaction was performed in the presence of ascorbic acid, two additional products were detected, which were tentatively identified as the cyclized products, 5,6-dihydroxybenzofuran and 3,5,6-trihydroxybenzofuran. Physiological concentrations of Cu(II) ions could also cause the oxidation of DOPAL to DOPAL-quinone. DOPAL-quinone exhibited reactivity towards the cysteine residues of serum albumin. DOPAL-oligomer, the oxidation product of DOPAL, exhibited pro-oxidant activity oxidizing GSH to GSSG and producing hydrogen peroxide. These results indicate that DOPAL-quinone generates several toxic compounds that could augment the neurotoxicity of DOPAL.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Dopamina/química , Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Ácido 3,4-Dihidroxifenilacético/química , Animales , Oxidación-Reducción
4.
Chem Res Toxicol ; 34(10): 2184-2193, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34506109

RESUMEN

Monoamine oxidase (MAO) catalyzes the oxidative deamination of dopamine and norepinephrine to produce 3,4-dihydroxyphenylacetaldehyde (DOPAL) and 3,4-dihydroxyphenylglycolaldehyde (DOPEGAL), respectively. Both of these aldehydes are potently cytotoxic and have been implicated in pathogenesis of neurodegenerative and cardiometabolic disorders. Previous work has demonstrated that both the catechol and aldehyde moieties of DOPAL are reactive and cytotoxic via their propensity to cause macromolecular cross-linking. With certain amines, DOPAL likely reacts via a Schiff base before oxidative activation of the catechol and rearrangement to a stable indole product. Our current work expands on this reactivity and includes the less-studied DOPEGAL. Although we confirmed that antioxidants mediated DOPAL's reactivity with carnosine and N-acetyl-l-lysine, antioxidants had no effect on reactivity with l-cysteine. Therefore, we propose a non-oxidative mechanism where, following Schiff base formation, the thiol of l-cysteine reacts to form a thiazolidine. Similarly, we demonstrate that DOPEGAL forms a putative thiazolidine conjugate with l-cysteine. We identified and characterized both l-cysteine conjugates via HPLC-MS and additionally identified a DOPEGAL adduct with carnosine, which is likely an Amadori product. Furthermore, we were able to demonstrate that these conjugates are produced in biological systems via MAO after treatment of the cell lysate with norepinephrine or dopamine along with the corresponding nucleophiles (i.e., l-cysteine and carnosine). As it has been established that metabolic and oxidative stress leads to increased MAO activity and accumulation of DOPAL and DOPEGAL, it is conceivable that conjugation of these aldehydes to carnosine or l-cysteine is a newly identified detoxification pathway. Furthermore, the ability to characterize these adducts via analytical techniques reveals their potential for use as biomarkers of dopamine or norepinephrine metabolic disruption.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Carnosina/metabolismo , Catecoles/metabolismo , Cisteína/metabolismo , Monoaminooxidasa/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular
5.
Int J Mol Sci ; 22(11)2021 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-34199427

RESUMEN

The interplay between α-synuclein and dopamine derivatives is associated with oxidative stress-dependent neurodegeneration in Parkinson's disease (PD). The formation in the dopaminergic neurons of intraneuronal inclusions containing aggregates of α-synuclein is a typical hallmark of PD. Even though the biochemical events underlying the aberrant aggregation of α-synuclein are not completely understood, strong evidence correlates this process with the levels of dopamine metabolites. In vitro, 3,4-dihydroxyphenylacetaldehyde (DOPAL) and the other two metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and 3,4-dihydroxyphenylethanol (DOPET), share the property to inhibit the growth of mature amyloid fibrils of α-synuclein. Although this effect occurs with the formation of differently toxic products, the molecular basis of this inhibition is still unclear. Here, we provide information on the effect of DOPAC on the aggregation properties of α-synuclein and its ability to interact with membranes. DOPAC inhibits α-synuclein aggregation, stabilizing monomer and inducing the formation of dimers and trimers. DOPAC-induced oligomers did not undergo conformational transition in the presence of membranes, and penetrated the cell, where they triggered autophagic processes. Cellular assays showed that DOPAC reduced cytotoxicity and ROS production induced by α-synuclein aggregates. Our findings show that the early radicals resulting from DOPAC autoxidation produced covalent modifications of the protein, which were not by themselves a primary cause of either fibrillation or membrane binding inhibition. These findings are discussed in the light of the potential mechanism of DOPAC protection against the toxicity of α-synuclein aggregates to better understand protein and catecholamine biology and to eventually suggest a scaffold that can help in the design of candidate molecules able to interfere in α-synuclein aggregation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Enfermedad de Parkinson/genética , Agregación Patológica de Proteínas/genética , alfa-Sinucleína/genética , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/farmacología , Amiloide/efectos de los fármacos , Amiloide/genética , Dopamina/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Multimerización de Proteína/genética , alfa-Sinucleína/antagonistas & inhibidores
6.
Neurotoxicology ; 86: 85-93, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34314733

RESUMEN

Parkinson's disease is characterized by dopamine dyshomeostasis and oxidative stress. The aldehyde metabolite of dopamine, 3,4-dihydroxyphenylacetaldehyde (DOPAL), has been reported to be cytotoxic and capable of protein modification. Protein modification by DOPAL has been implicated in the pathogenesis of Parkinson's disease, but the complete pathology is unknown. Our findings show that DOPAL modifies glutathione S-transferase (GST), an important enzyme in the antioxidant defense system. DOPAL, dopamine, and the metabolite 3,4-dihydroxyphenylacetic acid (DOPAC), inhibited the activity of GST isolated from N27 dopaminergic cells at an IC50 of 31.46 µM, 82.32 µM, and 260.0 µM, respectively. DOPAL, dopamine, and DOPAC inhibited commercially available equine liver GST at an IC50 of 23.72 µM, 32.17 µM, and 73.70 µM, respectively. This inhibition was time dependent and irreversible. 1 mM ʟ-cysteine or glutathione fully protected GST activity from DOPAL, DA, and DOPAC inhibition. 1 mM carnosine partially protected GST activity from DA inhibition. Furthermore, ʟ-cysteine was found to protect GST by forming a putative thiazolidine conjugate with DOPAL. We conclude that GST inactivation may be a part of the broader etiopathology of Parkinson's disease.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/farmacología , Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Glutatión Transferasa/antagonistas & inhibidores , Animales , Línea Celular , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Relación Dosis-Respuesta a Droga , Glutatión Transferasa/metabolismo , Caballos , Ratas
7.
J Neurochem ; 158(4): 960-979, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33991113

RESUMEN

In Parkinson's disease, dopamine-containing nigrostriatal neurons undergo profound degeneration. Tyrosine hydroxylase (TH) is the rate-limiting enzyme in dopamine biosynthesis. TH increases in vitro formation of reactive oxygen species, and previous animal studies have reported links between cytosolic dopamine build-up and oxidative stress. To examine effects of increased TH activity in catecholaminergic neurons in vivo, we generated TH-over-expressing mice (TH-HI) using a BAC-transgenic approach that results in over-expression of TH with endogenous patterns of expression. The transgenic mice were characterized by western blot, qPCR, and immunohistochemistry. Tissue contents of dopamine, its metabolites, and markers of oxidative stress were evaluated. TH-HI mice had a 3-fold increase in total and phosphorylated TH levels and an increased rate of dopamine synthesis. Coincident with elevated dopamine turnover, TH-HI mice showed increased striatal production of H2 O2 and reduced glutathione levels. In addition, TH-HI mice had elevated striatal levels of the neurotoxic dopamine metabolites 3,4-dihydroxyphenylacetaldehyde and 5-S-cysteinyl-dopamine and were more susceptible than wild-type mice to the effects of amphetamine and methamphetamine. These results demonstrate that increased TH alone is sufficient to produce oxidative stress in vivo, build up autotoxic dopamine metabolites, and augment toxicity.


Asunto(s)
Anfetamina/farmacología , Catecolaminas/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Estrés Oxidativo , Tirosina 3-Monooxigenasa/metabolismo , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Dopamina/análogos & derivados , Dopamina/metabolismo , Femenino , Dosificación de Gen , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Tirosina 3-Monooxigenasa/genética
8.
CNS Neurosci Ther ; 27(5): 540-551, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33475253

RESUMEN

AIM: To understand why autonomic failures, a common non-motor symptom of Parkinson's disease (PD), occur earlier than typical motor disorders. METHODS: Vagal application of DOPAL (3,4-dihydroxyphenylacetaldehyde) to simulate PD-like autonomic dysfunction and understand the connection between PD and cardiovascular dysfunction. Molecular and morphological approaches were employed to test the time-dependent alternation of α-synuclein aggregation and the ultrastructure changes in the heart and nodose (NG)/nucleus tractus solitarius (NTS). RESULTS: Blood pressure (BP) and baroreflex sensitivity of DOPAL-treated rats were significantly reduced accompanied with a time-dependent change in orthostatic BP, consistent with altered echocardiography and cardiomyocyte mitochondrial ultrastructure. Notably, time-dependent and collaborated changes in Mon-/Tri-α-synuclein were paralleled with morphological alternation in the NG and NTS. CONCLUSION: These all demonstrate that early autonomic dysfunction mediated by vagal application of DOPAL highly suggests the plausible etiology of PD initiated from peripheral, rather than central site. It will provide a scientific basis for the prevention and early diagnosis of PD.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Enfermedades del Sistema Nervioso Autónomo/patología , Enfermedad de Parkinson Secundaria/patología , Nervio Vago , Ácido 3,4-Dihidroxifenilacético/farmacología , Animales , Enfermedades del Sistema Nervioso Autónomo/etiología , Barorreflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Electrocardiografía , Hipotensión Ortostática/fisiopatología , Masculino , Mitocondrias Cardíacas/patología , Miocardio/patología , Miocitos Cardíacos/patología , Ganglio Nudoso/patología , Enfermedad de Parkinson Secundaria/complicaciones , Ratas , Ratas Sprague-Dawley , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/genética
9.
J Pharm Biomed Anal ; 195: 113822, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33358301

RESUMEN

3,4-dihydroxyphenylacetaldehyde (DOPAL), a toxic intermediary metabolite of dopamine (DA), causes catecholaminergic neurodegeneration via covalent binding with functional proteins or other biomolecules. Accurate quantification of DOPAL is essential to investigate the etiological factors associated with DOPAL and the pathogenetic role of DOPAL in Parkinson's disease (PD). However, no validated quantitative methods are available. Quantification of DOPAL in biosample is challenging since it is a reactive endogenous aldehyde with poor ionization efficiency and chromatographic behavior in the LC-MS system. Here, a sensitive, simple, and robust UPLC-MS/MS method has been established and validated for the determination of DOPAL in rat brain tissue specimens. DOPAL was found to be unstable in biosample due to reactive aldehyde whereas it was stable in acidic condition. The analyte was stabilized by pH and temperature control during the sample preparation and derivatization. Then, a chemical derivatization method that can be readily performed in acidic conditions and at low temperature was employed using 2-hydrazino-4-(trifluoromethyl)-pyrimidine (HTP) to block the reactive aldehyde and improve the detection sensitivity (about 100-fold increase) and chromatographic retention. Bovine serum albumin was used as a surrogate matrix, which was validated by the parallelism assay and post-column infusion experiment. This method was fully validated and the lower limit of quantification (LLOQ) was 0.5 ng/mL. With the method, a significant increase of DOPAL level was found in striatum region of rats received 6-hydroxydopamine (6-OHDA) injection for 12 h, indicating DOPAL may play a pathogenic role in 6-OHDA-induced PD model.


Asunto(s)
Cuerpo Estriado , Espectrometría de Masas en Tándem , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Animales , Cromatografía Liquida , Ratas
10.
Semin Neurol ; 40(5): 502-514, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32906170

RESUMEN

The catecholamines dopamine and norepinephrine are key central neurotransmitters that participate in many neurobehavioral processes and disease states. Norepinephrine is also the main neurotransmitter mediating regulation of the circulation by the sympathetic nervous system. Several neurodegenerative disorders feature catecholamine deficiency. The most common is Parkinson's disease (PD), in which putamen dopamine content is drastically reduced. PD also entails severely decreased myocardial norepinephrine content, a feature that characterizes two other Lewy body diseases-pure autonomic failure and dementia with Lewy bodies. It is widely presumed that tissue catecholamine depletion in these conditions results directly from loss of catecholaminergic neurons; however, as highlighted in this review, there are also important functional abnormalities in extant residual catecholaminergic neurons. We refer to this as the "sick-but-not-dead" phenomenon. The malfunctions include diminished dopamine biosynthesis via tyrosine hydroxylase (TH) and L-aromatic-amino-acid decarboxylase (LAAAD), inefficient vesicular sequestration of cytoplasmic catecholamines, and attenuated neuronal reuptake via cell membrane catecholamine transporters. A unifying explanation for catecholaminergic neurodegeneration is autotoxicity exerted by 3,4-dihydroxyphenylacetaldehyde (DOPAL), an obligate intermediate in cytoplasmic dopamine metabolism. In PD, putamen DOPAL is built up with respect to dopamine, associated with a vesicular storage defect and decreased aldehyde dehydrogenase activity. Probably via spontaneous oxidation, DOPAL potently oligomerizes and forms quinone-protein adducts with ("quinonizes") α-synuclein (AS), a major constituent in Lewy bodies, and DOPAL-induced AS oligomers impede vesicular storage. DOPAL also quinonizes numerous intracellular proteins and inhibits enzymatic activities of TH and LAAAD. Treatments targeting DOPAL formation and oxidation therefore might rescue sick-but-not-dead catecholaminergic neurons in Lewy body diseases.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Enfermedades del Sistema Nervioso Autónomo/metabolismo , Dopamina/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Norepinefrina/metabolismo , Enfermedad de Parkinson/metabolismo , Sistema Nervioso Simpático/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Humanos
11.
PLoS Genet ; 16(6): e1008868, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32579581

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder featuring progressive loss of midbrain dopaminergic (DA) neurons that leads to motor symptoms. The etiology and pathogenesis of PD are not clear. We found that expression of COUP-TFII, an orphan nuclear receptor, in DA neurons is upregulated in PD patients through the analysis of public datasets. We show here that through epigenetic regulation, COUP-TFII contributes to oxidative stress, suggesting that COUP-TFII may play a role in PD pathogenesis. Elevated COUP-TFII expression specifically in DA neurons evokes DA neuronal loss in mice and accelerates the progression of phenotypes in a PD mouse model, MitoPark. Compared to control mice, those with elevated COUP-TFII expression displayed reduced cristae in mitochondria and enhanced cellular electron-dense vacuoles in the substantia nigra pars compacta. Mechanistically, we found that overexpression of COUP-TFII disturbs mitochondrial pathways, resulting in mitochondrial dysfunction. In particular, there is repressed expression of genes encoding cytosolic aldehyde dehydrogenases, which could enhance oxidative stress and interfere with mitochondrial function via 3,4-dihydroxyphenylacetaldehyde (DOPAL) buildup in DA neurons. Importantly, under-expression of COUP-TFII in DA neurons slowed the deterioration in motor functions of MitoPark mice. Taken together, our results suggest that COUP-TFII may be an important contributor to PD development and a potential therapeutic target.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Neuronas Dopaminérgicas/patología , Mitocondrias/patología , Enfermedad de Parkinson/genética , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Aldehído Deshidrogenasa , Animales , Encéfalo/citología , Encéfalo/patología , Línea Celular , Línea Celular Tumoral , Estudios de Cohortes , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neuronas Dopaminérgicas/citología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , RNA-Seq , Ratas , Regulación hacia Arriba
12.
J Neural Transm (Vienna) ; 127(2): 169-177, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31807952

RESUMEN

Monoamine oxidase (MAO) plays a central role in the metabolism of the neurotransmitters dopamine, norepinephrine, and serotonin. This brief review focuses on 3,4-dihydroxyphenylacetaldehyde (DOPAL), which is the immediate product of MAO acting on cytoplasmic dopamine. DOPAL is toxic; however, normally DOPAL is converted via aldehyde dehydrogenase (ALDH) to 3,4-dihydroxyphenylacetic acid (DOPAC), which rapidly exits the neurons. In addition to vesicular uptake of dopamine via the vesicular monoamine transporter (VMAT), the two-enzyme sequence of MAO and ALDH keeps cytoplasmic dopamine levels low. Dopamine oxidizes readily to form toxic products that could threaten neuronal homeostasis. The catecholaldehyde hypothesis posits that diseases featuring catecholaminergic neurodegeneration result from harmful interactions between DOPAL and the protein alpha-synuclein, a major component of Lewy bodies in diseases such as Parkinson disease, dementia with Lewy bodies, and pure autonomic failure. DOPAL potently oligomerizes alpha-synuclein, and alpha-synuclein oligomers impede vesicular functions, shifting the fate of cytoplasmic dopamine toward MAO-catalyzed formation of DOPAL-a vicious cycle. When MAO deaminates dopamine to form DOPAL, hydrogen peroxide is generated; and DOPAL, hydrogen peroxide, and divalent metal cations react to form hydroxyl radicals, which peroxidate lipid membranes. Lipid peroxidation products in turn inhibit ALDH, causing DOPAL to accumulate-another vicious cycle. MAO inhibition decreases DOPAL formation but concurrently increases the spontaneous oxidation of dopamine, potentially trading off one form of toxicity for another. These considerations rationalize a neuroprotection strategy based on concurrent treatment with an MAO inhibitor and an anti-oxidant.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Aldehído Deshidrogenasa/metabolismo , Dopamina/metabolismo , Monoaminooxidasa/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Animales , Humanos
13.
J Pharmacol Exp Ther ; 372(2): 157-165, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31744850

RESUMEN

Lewy body diseases such as Parkinson's disease involve intraneuronal deposition of the protein α-synuclein (AS) and depletion of nigrostriatal dopamine (DA). Interactions of AS with DA oxidation products may link these neurohistopathologic and neurochemical abnormalities via two potential pathways: spontaneous oxidation of DA to dopamine-quinone and enzymatic oxidation of DA catalyzed by monoamine oxidase to form 3,4-dihydroxyphenylacetaldehyde (DOPAL), which is then oxidized to DOPAL-Q. We compared these two pathways in terms of the ability of DA and DOPAL to modify AS. DOPAL was far more potent than DA both in oligomerizing and forming quinone-protein adducts with (quinonizing) AS. The DOPAL-induced protein modifications were enhanced similarly by pro-oxidation with Cu(II) or tyrosinase and inhibited similarly by antioxidation with N-acetylcysteine. Dopamine oxidation evoked by Cu(II) or tyrosinase did not quinonize AS. In cultured MO3.13 human oligodendrocytes DOPAL resulted in the formation of numerous intracellular quinoproteins that were visualized by near-infrared spectroscopy. We conclude that of the two routes by which oxidation of DA modifies AS and other proteins the route via DOPAL is more prominent. The results support developing experimental therapeutic strategies that might mitigate deleterious modifications of proteins such as AS in Lewy body diseases by targeting DOPAL formation and oxidation. SIGNIFICANCE STATEMENT: Interactions of the protein α-synuclein with products of dopamine oxidation in the neuronal cytoplasm may link two hallmark abnormalities of Parkinson disease: Lewy bodies (which contain abundant AS) and nigrostriatal DA depletion (which produces the characteristic movement disorder). Of the two potential routes by which DA oxidation may alter AS and other proteins, the route via the autotoxic catecholaldehyde 3,4-dihydroxyphenylacetaldehyde is more prominent; the results support experimental therapeutic strategies targeting DOPAL formation and DOPAL-induced protein modifications.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Dopamina/análogos & derivados , Dopamina/química , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/química , Ácido 3,4-Dihidroxifenilacético/efectos adversos , Ácido 3,4-Dihidroxifenilacético/química , Ácido 3,4-Dihidroxifenilacético/metabolismo , Acetilcisteína/química , Antioxidantes/química , Línea Celular , Cobre/química , Cobre/metabolismo , Dopamina/efectos adversos , Dopamina/metabolismo , Humanos , Monoaminooxidasa/metabolismo , Monofenol Monooxigenasa/química , Monofenol Monooxigenasa/metabolismo , Oligodendroglía/citología , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Tolcapona/metabolismo , alfa-Sinucleína/metabolismo
14.
Nat Commun ; 10(1): 2015, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043610

RESUMEN

Previous studies have utilized monoamine oxidase (MAO) and L-3,4-dihydroxyphenylalanine decarboxylase (DDC) for microbe-based production of tetrahydropapaveroline (THP), a benzylisoquinoline alkaloid (BIA) precursor to opioid analgesics. In the current study, a phylogenetically distinct Bombyx mori 3,4-dihydroxyphenylacetaldehyde synthase (DHPAAS) is identified to bypass MAO and DDC for direct production of 3,4-dihydroxyphenylacetaldehyde (DHPAA) from L-3,4-dihydroxyphenylalanine (L-DOPA). Structure-based enzyme engineering of DHPAAS results in bifunctional switching between aldehyde synthase and decarboxylase activities. Output of dopamine and DHPAA products is fine-tuned by engineered DHPAAS variants with Phe79Tyr, Tyr80Phe and Asn192His catalytic substitutions. Balance of dopamine and DHPAA products enables improved THP biosynthesis via a symmetrical pathway in Escherichia coli. Rationally engineered insect DHPAAS produces (R,S)-THP in a single enzyme system directly from L-DOPA both in vitro and in vivo, at higher yields than that of the wild-type enzyme. However, DHPAAS-mediated downstream BIA production requires further improvement.


Asunto(s)
Descarboxilasas de Aminoácido-L-Aromático/metabolismo , Escherichia coli/metabolismo , Proteínas de Insectos/metabolismo , Ingeniería Metabólica/métodos , Tetrahidropapaverolina/metabolismo , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Secuencias de Aminoácidos/genética , Animales , Descarboxilasas de Aminoácido-L-Aromático/química , Descarboxilasas de Aminoácido-L-Aromático/genética , Descarboxilasas de Aminoácido-L-Aromático/aislamiento & purificación , Bombyx , Dopamina/metabolismo , Proteínas de Insectos/química , Proteínas de Insectos/genética , Proteínas de Insectos/aislamiento & purificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
15.
Brain Res ; 1702: 74-84, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29030055

RESUMEN

This review provides an update about cardiac sympathetic denervation in Lewy body diseases. The family of Lewy body diseases includes Parkinson's disease (PD), pure autonomic failure (PAF), and dementia with Lewy bodies (DLB). All three feature intra-neuronal cytoplasmic deposits of the protein, alpha-synuclein. Multiple system atrophy (MSA), the parkinsonian form of which can be difficult to distinguish from PD with orthostatic hypotension, involves glial cytoplasmic inclusions that contain alpha-synuclein. By now there is compelling neuroimaging, neuropathologic, and neurochemical evidence for cardiac sympathetic denervation in Lewy body diseases. In addition to denervation, there is decreased storage of catecholamines in the residual terminals. The degeneration develops in a centripetal, retrograde, "dying back" sequence. Across synucleinopathies the putamen and cardiac catecholaminergic lesions seem to occur independently of each other, whereas non-motor aspects of PD (e.g., anosmia, dementia, REM behavior disorder, OH) are associated with each other and with cardiac sympathetic denervation. Cardiac sympathetic denervation can be caused by synucleinopathy in inherited PD. According to the catecholaldehyde hypothesis, 3,4-dihydroxyphenylacetaldehyde (DOPAL), an intermediary metabolite of dopamine, causes or contributes to the death of catecholamine neurons, especially by interacting with proteins such as alpha-synuclein. DOPAL oxidizes spontaneously to DOPAL-quinone, which probably converts alpha-synuclein to its toxic oligomeric form. Decreasing DOPAL production and oxidation might slow the neurodegenerative process. Tracking cardiac sympathetic innervation over time could be the basis for a proof of principle experimental therapeutics trial targeting DOPAL.


Asunto(s)
Corazón/inervación , Miocardio/metabolismo , Enfermedad de Parkinson/metabolismo , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Catecolaminas/metabolismo , Dopamina/metabolismo , Humanos , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/patología , Atrofia de Múltiples Sistemas/patología , Neuroimagen , Neuronas/metabolismo , Sistema Nervioso Simpático , alfa-Sinucleína/metabolismo
16.
ACS Chem Neurosci ; 10(1): 690-703, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30352158

RESUMEN

Identifying the mechanisms by which the presynaptic protein α-synuclein (aSyn) is associated with neurodegeneration of dopamine neurons is a major priority in the Parkinson's disease (PD) field. Studies indicate that DOPAL (3,4-dihydroxyphenylacetaldehyde), an aldehyde generated from the enzymatic oxidation of dopamine, may convert aSyn monomer into a neurotoxin via formation of covalently stabilized toxic oligomers. Herein we investigated the role of N-terminal acetylation and familial aSyn mutations (A30P, A53T, E46K, G51D, and H50Q) on DOPAL-induced oligomerization of the protein. Our results indicate that the wild-type (WT) N-terminally acetylated aSyn (Ac-aSyn) is less prone to form oligomers upon incubation with DOPAL than the non-N-terminally acetylated protein. On the other hand, familial mutants from Ac-aSyn, particularly A53T, E46K, and H50Q increased the formation of DOPAL-derived aSyn oligomers, especially large oligomers. Binding of aSyn to synaptic-like small unilamellar vesicles (SUVs) protected distinctive aSyn variants against the effects of DOPAL. While N-terminal acetylation increased the protective action of SUVs against DOPAL-induced aSyn oligomerization, A53T, A30P, and H50Q mutations in Ac-aSyn had an opposite effect. This means that PD-linked mutations may not only perturb the affinity of aSyn for membranes but also influence the formation of DOPAL-mediated oligomers. Overall, our findings provide important evidence for the existence of a connection between familial mutations of aSyn, their distinct affinity to lipid membranes, and the formation of potentially toxic oligomers of the protein mediated by DOPAL.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Neuronas Dopaminérgicas/efectos de los fármacos , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Ácido 3,4-Dihidroxifenilacético/farmacología , Acetilación/efectos de los fármacos , Dopamina/metabolismo , Humanos , Lípidos de la Membrana/metabolismo , Mutación/genética , Oxidación-Reducción/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Multimerización de Proteína/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , alfa-Sinucleína/metabolismo
17.
Amino Acids ; 51(1): 97-102, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30191330

RESUMEN

Oxidative deamination of norepinephrine (NE) and dopamine (DA) by monoamine oxidase (MAO) generates the catecholaldehydes 3,4-dihydroxyphenylglycolaldehyde (DOPEGAL) and 3,4-dihydroxyphenylacetaldehyde (DOPAL), respectively, and H2O2. Catecholaldehydes are highly reactive electrophiles that have been implicated as causal factors in the etiology of neurodegenerative diseases and cardiac injury from ischemia and diabetes. The reactivity of both catechol and aldehyde groups enables the catecholaldehdyes to cross-link proteins and other biological molecules. Carnosine is a ß-alanyl-histidine dipeptide found in millimolar concentrations in brain and myocardium. It is well known to detoxify aldehydes formed from oxidized lipids and sugars, yet the reactivity of carnosine with catecholaldehydes has never been reported. Here, we investigated the ability of carnosine to form conjugates with DOPAL and DOPEGAL. Both catecholaldehydes were highly reactive towards L-cysteine (L-Cys), as well as carnosine; however, glutathione (GSH) showed essentially no reactivity towards DOPAL. In contrast, GSH readily reacted with the lipid peroxidation product 4-hydroxy-2-nonenal (4HNE), while carnosine showed low reactivity to 4HNE by comparison. To determine whether carnosine mitigates catecholaldehyde toxicity, samples of atrial myocardium were collected from patients undergoing elective cardiac surgery. Using permeabilized myofibers prepared from this tissue, mitochondrial respiration analysis revealed a concentration-dependent decrease in ADP-stimulated respiration with DOPAL. Pre-incubation with carnosine, but not GSH or L-Cys, significantly reduced this effect (p < 0.05). Carnosine was also able to block formation of catecholaldehyde protein adducts in isolated human cardiac mitochondria treated with NE. These findings demonstrate the unique reactivity of carnosine towards catecholaldehydes and, therefore, suggest a novel and distinct biological role for histidine dipeptides in this detoxification reaction. The therapeutic potential of carnosine in diseases associated with catecholamine-related toxicity is worthy of further examination.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Aldehídos/metabolismo , Carnosina/farmacología , Mitocondrias/metabolismo , Miocardio/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Anciano , Catecoles , Cisteína/farmacología , Glutatión/farmacología , Humanos , Persona de Mediana Edad , Oxidación-Reducción
18.
Eur J Pharmacol ; 845: 65-73, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30579934

RESUMEN

3,4-Dihydroxyphenylacetaldehyde (DOPAL), the monoamine oxidase (MAO) metabolite of dopamine, plays a role in pathogenesis of Parkinson disease, inducing α-synuclein aggregation. DOPAL generates discrete α-synuclein aggregates. Inhibiting this aggregation could provide therapy for slowing Parkinson disease progression. Primary and secondary amines form adducts with aldehydes. Rasagiline and aminoindan contain these amine groups. DOPAL-induced α-synuclein aggregates were resolved in the presence and absence of rasagiline or aminoindan using quantitative Western blotting. DOPAL levels in incubation mixtures, containing increased rasagiline or aminoindan concentrations, were determined by high pressure liquid chromatography (HPLC). Schiff base adducts between DOPAL and rasagiline or aminoindan were determined using mass spectrometry. A neuroprotective effect of rasagiline and aminoindan against DOPAL-induced toxicity was demonstrated using PC-12 cells. Rasagiline and aminoindan significantly reduced aggregation of α-synuclein of all sizes in test tube and PC-12 cells experiments. Dimethylaminoindan did not reduce aggregation. DOPAL levels in incubation mixtures were reduced with increasing rasagiline or aminoindan concentrations but not with dimethylaminoindan. Schiff base adducts between DOPAL and either rasagiline or aminoindan were demonstrated by mass spectrometry. A neuroprotective effect against DOPAL-induced toxicity in PC-12 cells was demonstrated for both rasagiline and aminoindan. Inhibiting DOPAL-induced α-synuclein aggregation through amine adducts provides a therapeutic approach for slowing Parkinson disease progression.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Aldehídos/farmacología , Indanos/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Ácido 3,4-Dihidroxifenilacético/toxicidad , Aldehídos/uso terapéutico , Animales , Indanos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Células PC12 , Ratas
19.
Biochem Biophys Res Commun ; 509(2): 367-372, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30591215

RESUMEN

The formation of neurotoxic oligomers of the presynaptic protein α-Synuclein (aSyn) is suggested to be associated with Parkinson's disease neurodegeneration. In this respect, it was demonstrated that the aldehyde 3,4-dihydroxyphenylacetaldehyde (DOPAL), a product from the enzymatic oxidation of dopamine, is capable of stabilizing potentially toxic aSyn oligomers via formation of covalent adducts with Lys residues of the protein. In addition, DOPAL-induced production of reactive oxygen species (ROS) leads to the oxidation of aSyn's Met residues to Met-sulfoxide. Recently, our group pointed out that the pre-oxidation of all-four Met residues of aSyn, upon treatment with H2O2, decreases the formation of large aSyn-DOPAL oligomers, which are suggested to be more toxic to neurons than the corresponding small oligomers (Carmo-Gonçalves et al., Biochem. Biophys. Res. Comm. 505, 295-301. 2018). By using a series of Met to Val mutants of aSyn, we demonstrated that the ability of aSyn to scavenge ROS/H2O2 generated from DOPAL oxidation is primarily dependent on Met residues located at the C-terminal domain of the protein, which contrasts with the reactivity of aSyn against H2O2 itself in which N-terminal Met residues (notably Met5) were more readily oxidized. Interestingly, the substitution of C-terminal Met residues (particularly Met127) by Val increased the formation of DOPAL-induced large oligomers in comparison with the wild-type protein. In this context, we demonstrated that the hydrophobicity of aSyn monomer, which is affected distinctively by the oxidation of N- versus C-terminal methionines, is correlated with the formation of large (but not small) oligomers of aSyn mediated by DOPAL.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Peróxido de Hidrógeno/química , Metionina/química , Valina/química , alfa-Sinucleína/química , Ácido 3,4-Dihidroxifenilacético/química , Ácido 3,4-Dihidroxifenilacético/metabolismo , Sustitución de Aminoácidos , Naftalenosulfonatos de Anilina/química , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Metionina/metabolismo , Mutación , Oxidación-Reducción , Dominios Proteicos , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia , Valina/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
20.
Biochem Biophys Res Commun ; 505(1): 295-301, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30249394

RESUMEN

The dopamine metabolite 3,4-dihydroxyphenylacetaldehyde (DOPAL) is believed to play a central role in Parkinson's disease neurodegeneration by stabilizing potentially toxic oligomers of the presynaptic protein α-Synuclein (aSyn). Besides the formation of covalent DOPAL-Lys adducts, DOPAL promotes the oxidation of Met residues of aSyn, which is also a common oxidative post-translational modification found in the protein in vivo. Herein we set out to address the role of Met residues on the oligomerization and neurotoxic properties of DOPAL-modified aSyn. Our data indicate that DOPAL promotes the formation of two distinct types of aSyn oligomers: large and small (dimer and trimers) oligomers, which seem to be generated by independent mechanisms and cannot be interconverted by using denaturing agents. Interestingly, H2O2-treated aSyn monomer, which exhibits all-four Met residues oxidized to Met-sulfoxide, exhibited a reduced ability to form large oligomers upon treatment with DOPAL, with no effect on the population of small oligomers. In this context, triple Met-Val mutant M5V/M116V/M127V exhibited an increased population of large aSyn-DOPAL oligomers in comparison with the wild-type protein. Interestingly, the stabilization of large rather than small oligomers seems to be associated with an enhanced toxicity of DOPAL-aSyn adducts. Collectively, these findings indicate that Met residues may play an important role in modulating both the oligomerization and the neurotoxic properties of DOPAL-derived aSyn species.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/farmacología , Metionina/química , Neuronas/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/toxicidad , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Peróxido de Hidrógeno/farmacología , Metionina/genética , Ratones , Mutación , Neuronas/citología , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , alfa-Sinucleína/química , alfa-Sinucleína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA