Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Neurobiol ; 43(1): 265-281, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34988761

RESUMEN

Studies have suggested aminochrome as an endogenous neurotoxin responsible for the dopaminergic neuron degeneration in Parkinson's disease (PD). However, neuroinflammation, an important alteration in PD pathogenesis, has been strictly induced in vitro by aminochrome. The aim of this study was to characterize the neuroinflammation induced in vivo by aminochrome. Wistar rats (male, 250-270 g) received a unilateral single dose by stereotaxic injection of saline into three sites in the striatum in the negative control group, or 32 nmol 6-hydroxydopamine (6-OHDA) in the positive control, or 6 nmol aminochrome. After 14 days, histological and molecular analyses were performed. We observed by immunofluorescence that aminochrome, as well as 6-OHDA, induced an increase in the number of Iba-1+ cells and in the number of activated (Iba-1+/ CD68+) microglia. An increase in the number of S100b+ cells and in the GFAP expression were also evidenced in the striatum and the SNpc of animals from aminochrome and positive control group. Dopaminergic neuronal loss was marked by reduction of TH+ cells and confirmed with reduction in the number of Nissl-stained neurons in the SNpc of rats from aminochrome and positive control groups. In addition, we observed by qPCR that aminocrhome induced an increase in the levels of IL-1ß, TNF-α, NLRP3, CCL5 and CCR2 mRNA in the SNpc. This work provides the first evidence of microgliosis, astrogliosis and neuroinflammation induced by aminochrome in an in vivo model. Since aminochrome is an endogenous molecule derived from dopamine oxidation present in the targeted neurons in PD, these results reinforce the potential of aminochrome as a useful preclinical model to find anti-inflammatory and neuroprotective drugs for PD. Aminochrome induced dopaminergic neuronal loss, microglial activation, astroglial activation and neuroinflammation marked by an increase in NLRP3, IL1ß, TNF-α, CCL2, CCL5 and CCR2.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Ratas , Masculino , Animales , Enfermedad de Parkinson/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/metabolismo , Ratas Wistar , Oxidopamina , Enfermedades Neuroinflamatorias , Dopamina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas Dopaminérgicas/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/metabolismo , Modelos Animales de Enfermedad , Microglía/metabolismo
3.
Cell Mol Life Sci ; 73(18): 3583-97, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27001668

RESUMEN

L-Dopa continues to be the gold drug in Parkinson's disease (PD) treatment from 1967. The failure to translate successful results from preclinical to clinical studies can be explained by the use of preclinical models which do not reflect what happens in the disease since these induce a rapid and extensive degeneration; for example, MPTP induces a severe Parkinsonism in only 3 days in humans contrasting with the slow degeneration and progression of PD. This study presents a new anatomy and develops preclinical model based on aminochrome which induces a slow and progressive dysfunction of dopaminergic neurons. The unilateral injection of aminochrome into rat striatum resulted in (1) contralateral rotation when the animals are stimulated with apomorphine; (2) absence of significant loss of tyrosine hydroxylase-positive neuronal elements both in substantia nigra and striatum; (3) cell shrinkage; (4) significant reduction of dopamine release; (5) significant increase in GABA release; (6) significant decrease in the number of monoaminergic presynaptic vesicles; (7) significant increase of dopamine concentration inside of monoaminergic vesicles; (8) significant increase of damaged mitochondria; (9) significant decrease of ATP level in the striatum (10) significant decrease in basal and maximal mitochondrial respiration. These results suggest that aminochrome induces dysfunction of dopaminergic neurons where the contralateral behavior can be explained by aminochrome-induced ATP decrease required both for anterograde transport of synaptic vesicles and dopamine release. Aminochrome could be implemented as a new model neurotoxin to study Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Indolquinonas/farmacología , Enfermedad de Parkinson/patología , Adenosina Trifosfato/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/análisis , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Indolquinonas/síntesis química , Indolquinonas/química , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/veterinaria , Ratas , Ratas Sprague-Dawley , Sustancia Negra/metabolismo , Vesículas Sinápticas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Ácido gamma-Aminobutírico/análisis
4.
An Acad Bras Cienc ; 89(1): 247-261, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28423083

RESUMEN

Prosopis juliflora is a shrub that has been used to feed animals and humans. However, a synergistic action of piperidine alkaloids has been suggested to be responsible for neurotoxic damage observed in animals. We investigated the involvement of programmed cell death (PCD) and autophagy on the mechanism of cell death induced by a total extract (TAE) of alkaloids and fraction (F32) from P. juliflora leaves composed majoritary of juliprosopine in a model of neuron/glial cell co-culture. We saw that TAE (30 µg/mL) and F32 (7.5 µg/mL) induced reduction in ATP levels and changes in mitochondrial membrane potential at 12 h exposure. Moreover, TAE and F32 induced caspase-9 activation, nuclear condensation and neuronal death at 16 h exposure. After 4 h, they induced autophagy characterized by decreases of P62 protein level, increase of LC3II expression and increase in number of GFP-LC3 cells. Interestingly, we demonstrated that inhibition of autophagy by bafilomycin and vinblastine increased the cell death induced by TAE and autophagy induced by serum deprivation and rapamycin reduced cell death induced by F32 at 24 h. These results indicate that the mechanism neural cell death induced by these alkaloids involves PCD via caspase-9 activation and autophagy, which seems to be an important protective mechanism.


Asunto(s)
Alcaloides/toxicidad , Autofagia/fisiología , Neuroglía/efectos de los fármacos , Piperidinas/toxicidad , Prosopis/química , Adenosina Trifosfato/análisis , Alcaloides/aislamiento & purificación , Animales , Autofagia/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Neuroglía/fisiología , Piperidinas/aislamiento & purificación , Extractos Vegetales/toxicidad , Ratas , Ratas Wistar , Factores de Tiempo
5.
J Neurochem ; 129(6): 898-915, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24548101

RESUMEN

The molecular mechanisms causing the loss of dopaminergic neurons containing neuromelanin in the substantia nigra and responsible for motor symptoms of Parkinson's disease are still unknown. The discovery of genes associated with Parkinson's disease (such as alpha synuclein (SNCA), E3 ubiquitin protein ligase (parkin), DJ-1 (PARK7), ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL-1), serine/threonine-protein kinase (PINK-1), leucine-rich repeat kinase 2 (LRRK2), cation-transporting ATPase 13A1 (ATP13A), etc.) contributed enormously to basic research towards understanding the role of these proteins in the sporadic form of the disease. However, it is generally accepted by the scientific community that mitochondria dysfunction, alpha synuclein aggregation, dysfunction of protein degradation, oxidative stress and neuroinflammation are involved in neurodegeneration. Dopamine oxidation seems to be a complex pathway in which dopamine o-quinone, aminochrome and 5,6-indolequinone are formed. However, both dopamine o-quinone and 5,6-indolequinone are so unstable that is difficult to study and separate their roles in the degenerative process occurring in Parkinson's disease. Dopamine oxidation to dopamine o-quinone, aminochrome and 5,6-indolequinone seems to play an important role in the neurodegenerative processes of Parkinson's disease as aminochrome induces: (i) mitochondria dysfunction, (ii) formation and stabilization of neurotoxic protofibrils of alpha synuclein, (iii) protein degradation dysfunction of both proteasomal and lysosomal systems and (iv) oxidative stress. The neurotoxic effects of aminochrome in dopaminergic neurons can be inhibited by: (i) preventing dopamine oxidation of the transporter that takes up dopamine into monoaminergic vesicles with low pH and dopamine oxidative deamination catalyzed by monoamino oxidase (ii) dopamine o-quinone, aminochrome and 5,6-indolequinone polymerization to neuromelanin and (iii) two-electron reduction of aminochrome catalyzed by DT-diaphorase. Furthermore, dopamine conversion to NM seems to have a dual role, protective and toxic, depending mostly on the cellular context. Dopamine oxidation to dopamine o-quinone, aminochrome and 5,6-indolequinone plays an important role in neurodegeneration in Parkinson's disease since they induce mitochondria and protein degradation dysfunction; formation of neurotoxic alpha synuclein protofibrils and oxidative stress. However, the cells have a protective system against dopamine oxidation composed by dopamine uptake mediated by Vesicular monoaminergic transporter-2 (VMAT-2), neuromelanin formation, two-electron reduction and GSH-conjugation mediated by Glutathione S-transferase M2-2 (GSTM2).


Asunto(s)
Dopamina/toxicidad , Dopamina/uso terapéutico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/prevención & control , Animales , Dopamina/biosíntesis , Dopamina/metabolismo , Glutatión/metabolismo , Humanos , Indolquinonas/metabolismo , Melaninas/metabolismo , Melaninas/fisiología , Monoaminooxidasa/metabolismo , Quinonas/metabolismo
6.
Hum Mol Genet ; 21(10): 2245-62, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22337954

RESUMEN

Mutations leading to expansion of a poly-glutamine track in Huntingtin (Htt) cause Huntington's disease (HD). Signs of endoplasmic reticulum (ER) stress have been recently reported in animal models of HD, associated with the activation of the unfolded protein response (UPR). Here we have investigated the functional contribution of ER stress to HD by targeting the expression of two main UPR transcription factors, XBP1 and ATF4 (activating transcription factor 4), in full-length mutant Huntingtin (mHtt) transgenic mice. XBP1-deficient mice were more resistant to developing disease features, associated with improved neuronal survival and motor performance, and a drastic decrease in mHtt levels. The protective effects of XBP1 deficiency were associated with enhanced macroautophagy in both cellular and animal models of HD. In contrast, ATF4 deficiency did not alter mHtt levels. Although, XBP1 mRNA splicing was observed in the striatum of HD transgenic brains, no changes in the levels of classical ER stress markers were detected in symptomatic animals. At the mechanistic level, we observed that XBP1 deficiency led to augmented expression of Forkhead box O1 (FoxO1), a key transcription factor regulating autophagy in neurons. In agreement with this finding, ectopic expression of FoxO1 enhanced autophagy and mHtt clearance in vitro. Our results provide strong evidence supporting an involvement of XBP1 in HD pathogenesis probably due to an ER stress-independent mechanism involving the control of FoxO1 and autophagy levels.


Asunto(s)
Autofagia , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Animales , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/genética , Proteína 1 de Unión a la X-Box
7.
Biomolecules ; 14(6)2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38927076

RESUMEN

One of the biggest problems in the treatment of idiopathic Parkinson's disease is the lack of new drugs that slow its progression. L-Dopa remains the star drug in the treatment of this disease, although it induces severe side effects. The failure of clinical studies with new drugs depends on the use of preclinical models based on neurotoxins that do not represent what happens in the disease since they induce rapid and expansive neurodegeneration. We have recently proposed a single-neuron degeneration model for idiopathic Parkinson's disease that requires years to accumulate enough lost neurons for the onset of motor symptoms. This single-neuron degeneration model is based on the excessive formation of aminochrome during neuromelanin synthesis that surpass the neuroprotective action of the enzymes DT-diaphorase and glutathione transferase M2-2, which prevent the neurotoxic effects of aminochrome. Although the neurotoxic effects of aminochrome do not have an expansive effect, a stereotaxic injection of this endogenous neurotoxin cannot be used to generate a preclinical model in an animal. Therefore, the aim of this review is to evaluate the strategies for pharmacologically increasing the expression of DT diaphorase and GSTM2-2 and molecules that induce the expression of vesicular monoamine transporter 2, such as pramipexole.


Asunto(s)
Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Animales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Glutatión Transferasa/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Modelos Animales de Enfermedad , Antiparkinsonianos/farmacología , Antiparkinsonianos/uso terapéutico
8.
Biochim Biophys Acta ; 1822(7): 1125-36, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22483869

RESUMEN

We tested the hypothesis that both VMAT-2 and DT-diaphorase are an important cellular defense against aminochrome-dependent neurotoxicity during dopamine oxidation. A cell line with VMAT-2 and DT-diaphorase over-expressed was created. The transfection of RCSN-3 cells with a bicistronic plasmid coding for VMAT-2 fused with GFP-IRES-DT-diaphorase cDNA induced a significant increase in protein expression of VMAT-2 (7-fold; P<0.001) and DT-diaphorase (9-fold; P<0.001), accompanied by a 4- and 5.5-fold significant increase in transport and enzyme activity, respectively. Studies with synaptic vesicles from rat substantia nigra revealed that VMAT-2 uptake of ³H-aminochrome 6.3 ± 0.4nmol/min/mg was similar to dopamine uptake 6.2 ± 0.3nmol/min/mg that which were dependent on ATP. Interestingly, aminochrome uptake was inhibited by 2µM lobeline but not reserpine (1 and 10µM). Incubation of cells overexpressing VMAT-2 and DT-diaphorase with 20µM aminochrome resulted in (i) a significant decrease in cell death (6-fold, P<0.001); (ii) normal ultra structure determined by transmission electron microscopy contrasting with a significant increase of autophagosome and a dramatic remodeling of the mitochondrial inner membrane in wild type cells; (iii) normal level of ATP (256 ± 11µM) contrasting with a significant decrease in wild type cells (121±11µM, P<0.001); and (iv) a significant decrease in DNA laddering (21 ± 8pixels, P<0.001) cells in comparison with wild type cells treated with 20µM aminochrome (269 ± 9). These results support our hypothesis that VMAT-2 and DT-diaphorase are an important defense system against aminochrome formed during dopamine oxidation.


Asunto(s)
Dopamina/metabolismo , Indolquinonas/toxicidad , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Sustancia Negra/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Análisis de Varianza , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , ADN/efectos de los fármacos , ADN/metabolismo , Humanos , Indolquinonas/metabolismo , Lobelina/farmacología , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , NAD(P)H Deshidrogenasa (Quinona)/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Endogámicas F344 , Reserpina/farmacología , Sustancia Negra/citología , Transfección , Proteínas de Transporte Vesicular de Monoaminas/genética
9.
Antioxidants (Basel) ; 12(3)2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36978921

RESUMEN

Investigations of the effect of antioxidants on idiopathic Parkinson's disease have been unsuccessful because the preclinical models used to propose these clinical studies do not accurately represent the neurodegenerative process of the disease. Treatment with certain exogenous neurotoxins induces massive and extremely rapid degeneration; for example, MPTP causes severe Parkinsonism in just three days, while the degenerative process of idiopathic Parkinson´s disease proceeds over many years. The endogenous neurotoxin aminochrome seems to be a good alternative target since it is formed in the nigrostriatal system neurons where the degenerative process occurs. Aminochrome induces all the mechanisms reported to be involved in the degenerative processes of idiopathic Parkinson's disease. The presence of neuromelanin-containing dopaminergic neurons in the postmortem brain of healthy elderly people suggests that neuromelanin synthesis is a normal and harmless process despite the fact that it requires oxidation of dopamine to three ortho-quinones that are potentially toxic, especially aminochrome. The apparent contradiction that neuromelanin synthesis is harmless, despite its formation via neurotoxic ortho-quinones, can be explained by the protective roles of DT-diaphorase and glutathione transferase GSTM2-2 as well as the neuroprotective role of astrocytes secreting exosomes loaded with GSTM2-2. Increasing the expression of DT-diaphorase and GSTM2-2 may be a therapeutic goal to prevent the degeneration of new neuromelanin-containing dopaminergic neurons. Several phytochemicals that induce DT-diaphorase have been discovered and, therefore, an interesting question is whether these phytochemical KEAP1/NRF2 activators can inhibit or decrease aminochrome-induced neurotoxicity.

10.
Biomedicines ; 11(11)2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38002094

RESUMEN

The transition metal characteristics of iron allow it to play a fundamental role in several essential aspects of human life such as the transport of oxygen through hemoglobin or the transport of electrons in the mitochondrial respiratory chain coupled to the synthesis of ATP. However, an excess or deficiency of iron is related to certain pathologies. The maintenance of iron homeostasis is essential to avoid certain pathologies related to iron excess or deficiency. The existence of iron deposits in postmortem tissues of Parkinson's patients has been interpreted as evidence that iron plays a fundamental role in the degenerative process of the nigrostriatal system in this disease. The use of iron chelators has been successful in the treatment of diseases such as transfusion-dependent thalassemia and pantothenate kinase-associated neurodegeneration. However, a clinical study with the iron chelator deferiprone in patients with Parkinson's disease has not shown positive effects but rather worsened clinical symptoms. This suggests that iron may not play a role in the degenerative process of Parkinson's disease.

11.
Neurotox Res ; 41(3): 224-241, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36723781

RESUMEN

Causes of dopaminergic neuronal loss in Parkinson's disease (PD) are subject of investigation and the common use of models of acute neurodegeneration induced by neurotoxins 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 6-hydroxydopamine, and rotenone contributed to advances in the study of PD. However, the use of study models more similar to the pathophysiology of PD is required for advances in early diagnosis and translational pharmacology. Aminochrome (AMI), a compound derived from dopamine oxidation and a precursor of neuromelanin, is able to induce all the mechanisms associated with neurodegeneration. Previously, we showed AMI is cytotoxic in primary culture of mesencephalic cells (PCMC) and induces in vitro and in vivo neuroinflammation. On the other hand, the effect of rutin in central nervous system cells has revealed anti-inflammatory, antioxidative, and neuroprotective potential. However, there have been no data studies on the effect of rutin against aminochrome neurotoxicity. Here, we show that rutin prevents lysosomal dysfunction and aminochrome-induced cell death in SHSY-5Y cells, protects PCMC against aminochrome cytotoxicity, and prevents in vivo loss of dopaminergic neurons in substantia nigra pars compacta (SNPc), as well as microgliosis and astrogliosis. Additionally, we show that rutin decreases levels of interleukin-1ß (IL-1ß) mRNA and increases levels of glia-derived neurotrophic factor (GDNF) and nerve-derived neurotrophic factor (NGF) mRNA. We evidence for the first time the protective effect of rutin on PD aminochrome-induced models and suggest the potential role of the anti-inflammatory activity and upregulation of NGF and GDNF in the mechanism of rutin action against aminochrome neurotoxicity.


Asunto(s)
Fármacos Neuroprotectores , Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Animales , Ratones , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Flavonoides/farmacología , Rutina/farmacología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/prevención & control , Síndromes de Neurotoxicidad/metabolismo , Dopamina/metabolismo , Enfermedad de Parkinson/metabolismo , Neuronas Dopaminérgicas , Antiinflamatorios/farmacología , Fármacos Neuroprotectores/uso terapéutico , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología
13.
Biometals ; 25(4): 795-803, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22302610

RESUMEN

Hallmarks of idiopathic and some forms of familial Parkinson's disease are mitochondrial dysfunction, iron accumulation and oxidative stress in dopaminergic neurons of the substantia nigra. There seems to be a causal link between these three conditions, since mitochondrial dysfunction can give rise to increased electron leak and reactive oxygen species production. In turn, recent evidence indicates that diminished activity of mitochondrial complex I results in decreased Fe­S cluster synthesis and anomalous activation of Iron Regulatory Protein 1. Thus, mitochondrial dysfunction could be a founding event in the process that leads to neuronal death. Here, we present evidence showing that at low micromolar concentrations, the dopamine metabolite aminochrome inhibits complex I and ATP production in SH-SY5Y neuroblastoma cells differentiated into a dopaminergic phenotype. This effect is apparently direct, since it is replicated in isolated mitochondria. Additionally, overnight treatment with aminochrome increased the expression of the iron import transporter divalent metal transporter 1 and decreased the expression of the iron export transporter ferroportin 1. In accordance with these findings, cells treated with aminochrome presented increased iron uptake. These results suggest that aminochrome is an endogenous toxin that inhibits by oxidative modifications mitochondrial complex I and modifies the levels of iron transporters in a way that leads to iron accumulation.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Dopamina/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Indolquinonas/metabolismo , Indolquinonas/farmacología , Mitocondrias/metabolismo , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejo I de Transporte de Electrón/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Antioxidants (Basel) ; 11(2)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35204179

RESUMEN

Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.

16.
Neural Regen Res ; 17(9): 1861-1866, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35142659

RESUMEN

Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.

17.
Neurotox Res ; 40(6): 2135-2147, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35997936

RESUMEN

Studies showed that JM-20, a benzodiazepine-dihydropyridine hybrid molecule, protects against rotenone and 6-hydroxydopamine neurotoxicity. However, its protective effects against cytotoxicity induced by endogenous neurotoxins involved in Parkinson's disease (PD) pathogenesis have never been investigated. In this study, we evaluated the ability of JM-20 to inhibit alpha-synuclein (aSyn) aggregation. We also evaluated the interactions of JM-20 with aSyn by molecular docking and molecular dynamics and assessed the protective effect of JM-20 against aminochrome cytotoxicity. We demonstrated that JM-20 induced the formation of heterogeneous amyloid fibrils, which were innocuous to primary cultures of mesencephalic cells. Moreover, JM-20 reduced the average size of aSyn positive inclusions in H4 cells transfected with SynT wild-type and synphilin-1-V5, but not in HEK cells transfected with synphilin-1-GFP. In silico studies showed the interaction between JM-20 and the aSyn-binding site. Additionally, we showed that JM-20 protects SH-SY5Y cells against aminochrome cytotoxicity. These results reinforce the potential of JM-20 as a neuroprotective compound for PD and suggest aSyn as a molecular target for JM-20.


Asunto(s)
Dihidropiridinas , Neuroblastoma , Enfermedad de Parkinson , Humanos , alfa-Sinucleína , Benzodiazepinas , Simulación del Acoplamiento Molecular , Enfermedad de Parkinson/tratamiento farmacológico
18.
Stem Cell Res ; 51: 102193, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33517122

RESUMEN

KISCOi001-A is a healthy feeder-free and fully characterized human induced pluripotent stem (iPS) cell line cultured under xeno-free and defined conditions. The cell line is generated from normal human foreskin fibroblasts with non-integrating episomal plasmid vectors encoding OCT4, SOX2, KLF4, NANOG, LIN28, nontransforming L-MYC and dominant negative p53. The generated iPS cells are transgene-free and their pluripotency is confirmed by the expression of stem cell markers and capacity to differentiate into the cells of ectoderm, endoderm and mesoderm while their identity and karyotype stability is confirmed with Genomic assays.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Línea Celular , Fibroblastos , Prepucio , Humanos , Factor 4 Similar a Kruppel , Masculino
19.
Neurotox Res ; 39(2): 182-190, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33555546

RESUMEN

The enzyme glutathione transferase M2-2, expressed in human astrocytes, increases its expression in the presence of aminochrome and catalyzes the conjugation of aminochrome, preventing its toxic effects. Secretion of the enzyme glutathione transferase M2-2 from U373MG cells, used as a cellular model for astrocytes, has been reported, and the enzyme is taken up by neuroblastoma SYSH-S7 cells and provide protection against aminochrome. The present study provides evidence that glutathione transferase M2-2 is released in exosomes from U373MG cells, thereby providing a means for intercellular transport of the enzyme. With particular relevance to Parkinson disease and other degenerative conditions, we propose a new mechanism by which astrocytes may protect dopaminergic neurons against the endogenous neurotoxin aminochrome.


Asunto(s)
Astrocitos/metabolismo , Exosomas/metabolismo , Glutatión Transferasa/metabolismo , Transporte de Proteínas , Línea Celular Tumoral , Neuronas Dopaminérgicas/metabolismo , Humanos
20.
Chem Res Toxicol ; 23(9): 1492-6, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20849151

RESUMEN

DT-Diaphorase has been proposed to play a neuroprotective role in dopaminergic neurons by preventing aminochrome neurotoxicity. There are several studies supporting this idea, but in all studies, we used dicoumarol, an inhibitor of DT-diaphorase. We have designed and developed two siRNA to silence the expression of DT-diaphorase to study its role in aminochrome metabolism. We transduced RCSN-3 cells with retroviral particles containing a pRetroSuper plasmid coding a siRNA for DT-diaphorase. The cells selected in the presence of puromycin generated a stable cell line RCSN-3Nq6 and RCSN-3Nq7 with low expression of DT-diaphorase (27% and 33% of wild type, respectively). A significant cell death was observed in RCSN-3 cells expressing siRNA Nq6 and Nq7 for DT-diaphorase when were incubated with 100 µM aminochrome during 48 (4- and 3.5-fold, respectively; P < 0.01). These results support the protective role of DT-diaphorase against aminochrome neurotoxicity in dopaminergic neurons containing neuromelanin and show that Nq6 and Nq7 siRNA are very useful tools to study the role of DT-diaphorase in aminochrome metabolism.


Asunto(s)
NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Fármacos Neuroprotectores/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Apoptosis , Línea Celular , Indolquinonas/metabolismo , Indolquinonas/toxicidad , Melaninas/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Interferencia de ARN , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA