Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros













Intervalo de año de publicación
1.
J Biol Chem ; 299(3): 102941, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36702251

RESUMEN

Glutamine synthetase (GS), which catalyzes the ATP-dependent synthesis of L-glutamine from L-glutamate and ammonia, is a ubiquitous and conserved enzyme that plays a pivotal role in nitrogen metabolism across all life domains. In vertebrates, GS is highly expressed in astrocytes, where its activity sustains the glutamate-glutamine cycle at glutamatergic synapses and is thus essential for maintaining brain homeostasis. In fact, decreased GS levels or activity have been associated with neurodegenerative diseases, with these alterations attributed to oxidative post-translational modifications of the protein, in particular tyrosine nitration. In this study, we expressed and purified human GS (HsGS) and performed an in-depth analysis of its oxidative inactivation by peroxynitrite (ONOO-) in vitro. We found that ONOO- exposure led to a dose-dependent loss of HsGS activity, the oxidation of cysteine, methionine, and tyrosine residues and also the nitration of tryptophan and tyrosine residues. Peptide mapping by LC-MS/MS through combined H216O/H218O trypsin digestion identified up to 10 tyrosine nitration sites and five types of dityrosine cross-links; these modifications were further scrutinized by structural analysis. Tyrosine residues 171, 185, 269, 283, and 336 were the main nitration targets; however, tyrosine-to-phenylalanine HsGS mutants revealed that their sole nitration was not responsible for enzyme inactivation. In addition, we observed that ONOO- induced HsGS aggregation and activity loss. Thiol oxidation was a key modification to elicit aggregation, as it was also induced by hydrogen peroxide treatment. Taken together, our results indicate that multiple oxidative events at various sites are responsible for the inactivation and aggregation of human GS.


Asunto(s)
Glutamato-Amoníaco Ligasa , Ácido Peroxinitroso , Procesamiento Proteico-Postraduccional , Humanos , Cromatografía Liquida , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/metabolismo , Ácido Peroxinitroso/química , Ácido Peroxinitroso/farmacología , Espectrometría de Masas en Tándem , Tirosina/metabolismo , Activación Enzimática/efectos de los fármacos , Oxidación-Reducción , Mutación , Agregación Patológica de Proteínas/inducido químicamente
2.
PLoS One ; 16(11): e0260519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34847196

RESUMEN

Endoplasmic reticulum (ER) stress is associated with acute kidney injury (AKI) caused by various mechanisms, including antibiotics, non-steroidal anti-inflammatory drugs, cisplatin, and radiocontrast. Tunicamycin (TM) is a nucleoside antibiotic that induces ER stress and is a commonly used model of AKI. 4-phenylbutyrate (4-PBA) is a chemical chaperone and histone deacetylase (HDAC) inhibitor and has been shown to protect the kidney from ER stress, apoptosis, and structural damage in a tunicamycin model of AKI. The renal protection provided by 4-PBA is attributed to its ability to prevent misfolded protein aggregation and inhibit ER stress; however, the HDAC inhibitor effects of 4-PBA have not been examined in the TM-induced model of AKI. As such, the main objective of this study was to determine if histone hyperacetylation provides any protective effects against TM-mediated AKI. The FDA-approved HDAC inhibitor vorinostat was used, as it has no ER stress inhibitory effects and therefore the histone hyperacetylation properties alone could be investigated. In vitro work demonstrated that vorinostat inhibited histone deacetylation in cultured proximal tubular cells but did not prevent ER stress or protein aggregation induced by TM. Vorinostat induced a significant increase in cell death, and exacerbated TM-mediated total cell death and apoptotic cell death. Wild type male mice were treated with TM (0.5 mg/kg, intraperitoneal injection), with or without vorinostat (50 mg/kg/day) or 4-PBA (1 g/kg/day). Mice treated with 4-PBA or vorinostat exhibited similar levels of histone hyperacetylation. Expression of the pro-apoptotic protein CHOP was induced with TM, and not inhibited by vorinostat. Further, vorinostat did not prevent any renal damage or decline in renal function caused by tunicamycin. These data suggest that the protective mechanisms found by 4-PBA are primarily due to its molecular chaperone properties, and the HDAC inhibitors used did not provide any protection against renal injury caused by ER stress.


Asunto(s)
Lesión Renal Aguda , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Tunicamicina/efectos adversos , Vorinostat/farmacología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/prevención & control , Animales , Línea Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/prevención & control , Tunicamicina/farmacología
3.
J Ethnopharmacol ; 268: 113640, 2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33307058

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Terminalia chebula Retz. (T.chebula) is an important medicinal plant in Tibetan medicine and Ayurveda. T.chebula is known as the "King of Tibetan Medicine", due to its widespread clinical pharmacological activity such as anti-inflammatory, antioxidative, antidiabetic as well as anticancer in lots of in vivo and in vitro models. In this study, we use transgenic and/or RNAi Caenorhabditis elegans (C.elegans) model to simulation the AD pathological features induced by Aß, to detect the effect of TWE on improving Aß-induced toxicity and the corresponding molecular mechanism. AIM OF STUDY: The study aimed to tested the activities and its possible mechanism of T.chebula to against Aß1-42 induced toxicity and Aß1-42 aggregation. MATERIALS AND METHODS: Using transgenic C.elegans strain CL2006 and CL4176 as models respond to paralytic induced by Aß toxicity. The transcription factors DAF-16 and SKN-1 were analyzed used a fluorescence microscope in transgenic strains (DAF-16:GFP, SKN-1:GFP). The function of DAF-16 and SKN-1 was further investigated using loss-of-function strains by feeding RNA interference (RNAi) bacteria. To evaluate the aggregation level of Aß in the transgenic C.elegans, Thioflavin S (ThS) staining and WB visualized the levels of Aß monomers and oligomers. RESULTS: TWE treatment can significantly improve the paralysis of transgenic C.elegans caused by Aß aggregation (up to 14%). The Aß aggregates in transgenic C.elegans are significantly inhibited under TWE exposure (up to 70%). TWE increases the nuclear localization of the key transcription factor DAF-16 and HSF-1, which in turn leads to the expression of downstream Hsp-16.2 protein and exerts its inhibitory effect on Aß aggregation. Meanwhile, paralysis improved has not observed in SKN-1 mutation and/or RNAi C.elegans. CONCLUSION: Our results indicate that TWE can protect C.elegans against the Aß1-42-induced toxicity, inhibition Aß1-42 aggregation and delaying Aß-induced paralysis. The neuroprotective effect of TWE involves the activation of DAF-16/HSF-1/Hsp-16.2 pathway.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Fragmentos de Péptidos/toxicidad , Extractos Vegetales/uso terapéutico , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/prevención & control , Terminalia , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Relación Dosis-Respuesta a Droga , Humanos , Fragmentos de Péptidos/antagonistas & inhibidores , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Agregación Patológica de Proteínas/patología
4.
Int J Biol Macromol ; 163: 128-134, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32615214

RESUMEN

The protein misfolded structure called amyloids is related with extensive range of pathologies like local amyloidosis and neurodegenerative diseases. Several studies have reported the potential of insulin to generate local amyloidosis under certain state. Reports also showed that fibrils of insulin generated local amyloid mass due to continuous subcutaneous injection in mouse as well as rat. The present study was designed to examine the consequence of insulin fibril injections in rats, as well as the ability of enzymes, Lumbrokinase (LK) and Serratiopeptidase (SP) in diminishing this amyloid mass progression. The results showed that insulin fibrils generated amyloid masses in rats after subcutaneous injection for two weeks which was significantly condensed in size for the groups injected with insulin fibrils combined with LK or SP. At higher doses of LK and SP, the absence of amyloid structure was observed in histopathological studies. Light microscopy, polarized microscopy as well as Lumia live in vivo imaging system was used to analyze the results. In conclusion, the overall outcome of this study showed the anti-amyloid potential of enzyme LK and SP in the attenuation of local amyloidosis.


Asunto(s)
Endopeptidasas/farmacología , Insulina/efectos adversos , Péptido Hidrolasas/farmacología , Placa Amiloide/metabolismo , Placa Amiloide/prevención & control , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/prevención & control , Animales , Benzotiazoles/química , Rojo Congo/química , Modelos Animales de Enfermedad , Reacción en el Punto de Inyección/patología , Reacción en el Punto de Inyección/prevención & control , Inyecciones Subcutáneas , Insulina/administración & dosificación , Masculino , Microscopía Fluorescente , Microscopía de Polarización , Placa Amiloide/inducido químicamente , Placa Amiloide/patología , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/patología , Ratas , Ratas Wistar , Tejido Subcutáneo/efectos de los fármacos , Tejido Subcutáneo/patología
5.
Neurobiol Dis ; 141: 104947, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32422283

RESUMEN

Human and animal studies have shown that exposure to the organochlorine pesticide dieldrin is associated with increased risk of Parkinson's disease (PD). Previous work showed that developmental dieldrin exposure increased neuronal susceptibility to MPTP toxicity in male C57BL/6 mice, possibly via changes in dopamine (DA) packaging and turnover. However, the relevance of the MPTP model to PD pathophysiology has been questioned. We therefore studied dieldrin-induced neurotoxicity in the α-synuclein (α-syn)-preformed fibril (PFF) model, which better reflects the α-syn pathology and toxicity observed in PD pathogenesis. Specifically, we used a "two-hit" model to determine whether developmental dieldrin exposure increases susceptibility to α-syn PFF-induced synucleinopathy. Dams were fed either dieldrin (0.3 mg/kg, every 3-4 days) or vehicle corn oil starting 1 month prior to breeding and continuing through weaning of pups at postnatal day 22. At 12 weeks of age, male and female offspring received intrastriatal α-syn PFF or control saline injections. Consistent with the male-specific increased susceptibility to MPTP, our results demonstrate that developmental dieldrin exposure exacerbates PFF-induced toxicity in male mice only. Specifically, in male offspring, dieldrin exacerbated PFF-induced motor deficits on the challenging beam and increased DA turnover in the striatum 6 months after PFF injection. However, male offspring showed neither exacerbation of phosphorylated α-syn aggregation (pSyn) in the substantia nigra (SN) at 1 or 2 months post-PFF injection, nor exacerbation of PFF-induced TH and NeuN loss in the SN 6 months post-PFF injection. Collectively, these data indicate that developmental dieldrin exposure produces a male-specific exacerbation of synucleinopathy-induced behavioral and biochemical deficits. This sex-specific result is consistent with both previous work in the MPTP model, our previously reported sex-specific effects of this exposure paradigm on the male and female epigenome, and the higher prevalence and more severe course of PD in males. The novel two-hit environmental toxicant/PFF exposure paradigm established in this project can be used to explore the mechanisms by which other PD-related exposures alter neuronal vulnerability to synucleinopathy in sporadic PD.


Asunto(s)
Dieldrín/toxicidad , Actividad Motora/efectos de los fármacos , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/patología , Plaguicidas/toxicidad , Agregación Patológica de Proteínas , alfa-Sinucleína/toxicidad , Animales , Dopamina/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo , Factores Sexuales , Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/administración & dosificación
6.
Anal Bioanal Chem ; 412(24): 6485-6497, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32322953

RESUMEN

Extracellular accumulation of amyloid beta peptide (Aß) is believed to be one of the main factors responsible for neurodegeneration in Alzheimer's disease (AD). Metals could induce Aß aggregation, by their redox activity or binding properties to amyloid ß fibrils, leading to their accumulation and deposition outside neurons. For this reason, metal chelation may have an acknowledged part to play in AD prevention and treatment. In the current work, the role of different selenium species, including selenium nanoparticles, in Aß aggregation, was studied by evaluating their metal-chelating properties and their ability both to inhibit metal-induced Aß1-42 aggregation fibrils and to disaggregate them once formed. Transition biometals such as Fe(II), Cu(II), and Zn(II) at 50 µM were selected to establish the in vitro models. The DPPH assay was used to determine the antioxidant capacity of the evaluated selenium species. Selenium nanoparticles stabilized with chitosan (Ch-SeNPs) and with both chitosan and chlorogenic acid polyphenol (CGA@ChSeNPs) showed the highest antioxidant properties with EC50 of 0.9 and 0.07 mM, respectively. UV-Vis and d1(UV-Vis) spectra also revealed that selenium species, in particular selenomethionine (SeMet), were able to interact with metals. Regarding Aß1-42 incubation experiments, Fe(II), Cu(II), and Zn(II) induced Aß aggregation, in a similar way to most of the evaluated selenium species. However, Ch-SeNPs produced a high inhibition of metal-induced Aß aggregation, as well as a high disaggregation capacity of Aß fibrils in both the presence and absence of biometals, in addition to reducing the length and width (20% of reduction in the presence of Zn(II)) of the generated Aß fibrils. Graphical abstract.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Antioxidantes/farmacología , Metales/efectos adversos , Agregado de Proteínas/efectos de los fármacos , Selenio/farmacología , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Antioxidantes/química , Cobre/efectos adversos , Humanos , Hierro/efectos adversos , Nanopartículas/química , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/metabolismo , Selenio/química , Zinc/efectos adversos
7.
Biometals ; 33(2-3): 97-106, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32170541

RESUMEN

Alterations in metal ion homeostasis appear coupled to neurodegenerative disorders but mechanisms are unknown. Amyloid formation of the protein α-synuclein in brain cells is a hallmark of Parkinson's disease. α-Synuclein can bind several metal ions in vitro and such interactions may affect the assembly process. Here we used biophysical methods to study the effects of micromolar concentrations of Cu2+ and Fe3+ ions on amyloid formation of selected α-synuclein variants (wild-type and A53T α-synuclein, in normal and N-terminally acetylated forms). As shown previously, Cu2+ speeds up aggregation of normal wild-type α-synuclein, but not the acetylated form. However, Cu2+ has a minimal effect on (the faster) aggregation of normal A53T α-synuclein, despite that Cu2+ binds to this variant. Like Cu2+, Fe3+ speeds up aggregation of non-acetylated wild-type α-synuclein, but with acetylation, Fe3+ instead slows down aggregation. In contrast, for A53T α-synuclein, regardless of acetylation, Fe3+ slows down aggregation with the effect being most dramatic for acetylated A53T α-synuclein. The results presented here suggest a correlation between metal-ion modulation effect and intrinsic aggregation speed of the various α-synuclein variants.


Asunto(s)
Amiloide/metabolismo , Cobre/farmacología , Compuestos Férricos/farmacología , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/biosíntesis , Cobre/química , Relación Dosis-Respuesta a Droga , Compuestos Férricos/química , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/inducido químicamente , Conformación Proteica , alfa-Sinucleína/genética
8.
Cells ; 9(2)2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085572

RESUMEN

Research on repurposing the old alcohol-aversion drug disulfiram (DSF) for cancer treatment has identified inhibition of NPL4, an adaptor of the p97/VCP segregase essential for turnover of proteins involved in multiple pathways, as an unsuspected cancer cell vulnerability. While we reported that NPL4 is targeted by the anticancer metabolite of DSF, the bis-diethyldithiocarbamate-copper complex (CuET), the exact, apparently multifaceted mechanism(s) through which the CuET-induced aggregation of NPL4 kills cancer cells remains to be fully elucidated. Given the pronounced sensitivity to CuET in tumor cell lines lacking the genome integrity caretaker proteins BRCA1 and BRCA2, here we investigated the impact of NPL4 targeting by CuET on DNA replication dynamics and DNA damage response pathways in human cancer cell models. Our results show that CuET treatment interferes with DNA replication, slows down replication fork progression and causes accumulation of single-stranded DNA (ssDNA). Such a replication stress (RS) scenario is associated with DNA damage, preferentially in the S phase, and activates the homologous recombination (HR) DNA repair pathway. At the same time, we find that cellular responses to the CuET-triggered RS are seriously impaired due to concomitant malfunction of the ATRIP-ATR-CHK1 signaling pathway that reflects an unorthodox checkpoint silencing mode through ATR (Ataxia telangiectasia and Rad3 related) kinase sequestration within the CuET-evoked NPL4 protein aggregates.


Asunto(s)
Disuasivos de Alcohol/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Disulfiram/farmacología , Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Neoplasias/patología , Proteínas Nucleares/metabolismo , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/inducido químicamente , Transducción de Señal/efectos de los fármacos , Proteína que Contiene Valosina/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-30870786

RESUMEN

Phenothiazine molecules are effective and commonly used antipsychotic drugs, especially in the treatment of schizophrenia. However, they produce strong extrapyramidal side-effects manifested by drug-induced parkinsonism. Because Parkinson's disease as a neurodegenerative illness is associated with the formation of amyloid fibrils in neuronal cells, it is postulated that the development of phenothiazine-induced parkinsonism may be related to the phenothiazine-induced formation of fibrillar aggregates. The effect of phenothiazine compounds (fluphenazine (FPh), chlorpromazine (ChP) and propionylpromazine (PP)) on the fibrillogenesis of poly-l-lysine (PLL) was studied using Fourier-transform infrared (FTIR) spectroscopy supported by principal component analysis (PCA), vibrational circular dichroism (VCD), transmission electron microscopy (TEM) and Congo red binding assay. The fibrillogenesis of PLL is accompanied by fibril formation with charged or uncharged polypeptides with PPII (polyproline-like extended helix), α-helix or ß-sheet conformations. All of the phenothiazine molecules investigated effectively reduced the temperature required to induce the formation of ß-sheet-rich fibrils from α-helix-rich fibrils of PLL.


Asunto(s)
Amiloide/metabolismo , Antipsicóticos/efectos adversos , Enfermedad de Parkinson Secundaria/etiología , Fenotiazinas/efectos adversos , Polilisina/metabolismo , Agregación Patológica de Proteínas/inducido químicamente , Estructura Secundaria de Proteína/efectos de los fármacos , Amiloide/química , Amiloide/ultraestructura , Antipsicóticos/química , Dicroismo Circular , Humanos , Modelos Moleculares , Enfermedad de Parkinson Secundaria/metabolismo , Fenotiazinas/química , Polilisina/química , Agregación Patológica de Proteínas/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier
10.
Sci Signal ; 12(572)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862700

RESUMEN

The aggregation of α-synuclein (αSyn) is considered a key pathophysiological feature of certain neurodegenerative disorders, collectively termed synucleinopathies. Given that a prion-like, cell-to-cell transfer of misfolded αSyn has been recognized in the spreading of αSyn pathology in synucleinopathies, we investigated the biological mechanisms underlying the propagation of the disease with respect to environmental neurotoxic stress. Considering the potential role of the divalent metal manganese (Mn2+) in protein aggregation, we characterized its effect on αSyn misfolding and transmission in experimental models of Parkinson's disease. In cultured dopaminergic neuronal cells stably expressing wild-type human αSyn, misfolded αSyn was secreted through exosomes into the extracellular medium upon Mn2+ exposure. These exosomes were endocytosed through caveolae into primary microglial cells, thereby mounting neuroinflammatory responses. Furthermore, Mn2+-elicited exosomes exerted a neurotoxic effect in a human dopaminergic neuronal model (LUHMES cells). Moreover, bimolecular fluorescence complementation (BiFC) analysis revealed that Mn2+ accelerated the cell-to-cell transmission of αSyn, resulting in dopaminergic neurotoxicity in a mouse model of Mn2+ exposure. Welders exposed to Mn2+ had increased misfolded αSyn content in their serum exosomes. Stereotaxically delivering αSyn-containing exosomes, isolated from Mn2+-treated αSyn-expressing cells, into the striatum initiated Parkinsonian-like pathological features in mice. Together, these results indicate that Mn2+ exposure promotes αSyn secretion in exosomal vesicles, which subsequently evokes proinflammatory and neurodegenerative responses in both cell culture and animal models.


Asunto(s)
Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Exosomas/metabolismo , Manganeso/toxicidad , Enfermedad de Parkinson Secundaria/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Exosomas/patología , Manganeso/farmacología , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Priones/metabolismo , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/patología
11.
Neurochem Res ; 43(12): 2333-2342, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30302613

RESUMEN

Trans-cinnamaldehyde (CNM) has recently drawn attention due to its potent anti-inflammatory and antioxidant properties. The current study explored the memory enhancing effects of CNM against lipopolysaccharide (LPS)-induced neuroinflammation in mice. CNM and curcumin (a reference antioxidant) were administered at a dose of 50 mg/kg i.p. 3 h after a single LPS injection (0.8 mg/kg, i.p.) and continued daily for 7 days. Our results displayed that CNM and curcumin significantly ameliorated the LPS-induced impairment of learning and memory, neuroinflammation, oxidative stress and neuronal apoptosis. Memory functions and locomotor activity were assessed by Morris water maze, object recognition test and open field test. Both CNM and curcumin activated the nuclear factor erythroid 2 related factor 2 (Nrf2) and restored levels of downstream antioxidant enzymes superoxide dismutase and glutathione-S-transferase (GST) in the hippocampus. They also attenuated LPS-induced increase in hippocampal contents of interleukin-1ß (IL-1ß), malondialdehyde and caspase-3. Immunohistochemistry results showed that both CNM and curcumin reduced Aß1-42 protein accumulation in brain of mice. Remarkably CNM's effect on IL-1ß was less pronounced than curcumin; however it showed higher GST activity and more potent anti-apoptotic and anti-amylodogenic effect. We conclude that, CNM produces its memory enhancing effects through modulation of Nrf2 antioxidant defense in hippocampus, inhibition of neuroinflammation, apoptosis and amyloid protein burden.


Asunto(s)
Acroleína/análogos & derivados , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Lipopolisacáridos/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Agregación Patológica de Proteínas/tratamiento farmacológico , Acroleína/farmacología , Acroleína/uso terapéutico , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo , Distribución Aleatoria
12.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3595-3604, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30251679

RESUMEN

Cysteine (Cys) residues are major causes of crystallin disulfide formation and aggregation in aging and cataractous human lenses. We recently found that disulfide linkages are highly and partly conserved in ß- and γ-crystallins, respectively, in human age-related nuclear cataract and glutathione depleted LEGSKO mouse lenses, and could be mimicked by in vitro oxidation. Here we determined which Cys residues are involved in disulfide-mediated crosslinking of recombinant human γD-crystallin (hγD). In vitro diamide oxidation revealed dimer formation by SDS-PAGE and LC-MS analysis with Cys 111-111 and C111-C19 as intermolecular disulfides and Cys 111-109 as intramolecular sites. Mutation of Cys111 to alanine completely abolished dimerization. Addition of αB-crystallin was unable to protect Cys 111 from dimerization. However, Cu2+-induced hγD-crystallin aggregation was suppressed up to 50% and 80% by mutants C109A and C111A, respectively, as well as by total glutathionylation. In contrast to our recently published results using ICAT-labeling method, manual mining of the same database confirmed the specific involvement of Cys111 in disulfides with no free Cys111 detectable in γD-crystallin from old and cataractous human lenses. Surface accessibility studies show that Cys111 in hγD is the most exposed Cys residue (29%), explaining thereby its high propensity toward oxidation and polymerization in the aging lens.


Asunto(s)
Catarata/patología , Cisteína/metabolismo , Agregación Patológica de Proteínas/patología , Multimerización de Proteína/genética , gamma-Cristalinas/metabolismo , Adolescente , Factores de Edad , Anciano , Catarata/genética , Cationes Bivalentes/toxicidad , Niño , Preescolar , Cobre/toxicidad , Disulfuros/metabolismo , Glutatión/farmacología , Humanos , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Cristalino/patología , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Estrés Oxidativo/efectos de los fármacos , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , gamma-Cristalinas/química , gamma-Cristalinas/genética
13.
Artículo en Inglés | MEDLINE | ID: mdl-29698685

RESUMEN

Cadmium is a highly toxic environmental pollutant that can cause many adverse effects including cancer, neurological disease and kidney damage. Aquatic amphibians are particularly susceptible to this toxicant as it was shown to cause developmental abnormalities and genotoxic effects. In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. In the present study, CdCl2 treatment of A6 kidney epithelial cells, derived from the frog, Xenopus laevis, induced the accumulation of HO-1, heat shock protein 70 (HSP70) and HSP30 as well as an increase in the production of aggregated protein and aggresome-like structures. Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. Treatment of cells with hemin and baicalein, which were previously shown to provide cytoprotection against various stresses, induced HO-1 accumulation in a concentration-dependent manner. Also, treatment of cells with hemin and baicalein suppressed CdCl2-induced actin dysregulation and the accumulation of aggregated protein and aggresome-like structures. This cytoprotective effect was inhibited by SnPP. These results suggest that HO-1-mediated protection against CdCl2 toxicity includes the maintenance of actin cytoskeletal and microtubular structure and the suppression of aggregated protein and aggresome-like structures.


Asunto(s)
Cadmio/toxicidad , Contaminantes Ambientales/toxicidad , Proteínas del Choque Térmico HSP30/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Hemo-Oxigenasa 1/metabolismo , Riñón/efectos de los fármacos , Agregación Patológica de Proteínas/inducido químicamente , Animales , Antioxidantes/química , Antioxidantes/metabolismo , Línea Celular , Suplementos Dietéticos , Inhibidores Enzimáticos/farmacología , Flavanonas/antagonistas & inhibidores , Flavanonas/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/química , Hemina/antagonistas & inhibidores , Hemina/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Metaloporfirinas/farmacología , Microscopía Confocal , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/prevención & control , Protoporfirinas/farmacología , Proteínas de Xenopus/agonistas , Proteínas de Xenopus/antagonistas & inhibidores , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis
14.
Sci Rep ; 8(1): 5275, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588456

RESUMEN

Anesthetic agents have been implicated in the causation of neurological and cognitive deficits after surgery, the exacerbation of chronic neurodegenerative disease, and were recently reported to promote the onset of the neurologic respiratory disease Congenital Central Hypoventilation Syndrome (CCHS), related to misfolding of the transcription factor Phox2B. To study how anesthetic agents could affect neuronal function through alterations to protein folding, we created neuronal cell models emulating the graded disease severity of CCHS. We found that the gas anesthetic isoflurane and the opiate morphine potentiated aggregation and mislocalization of Phox2B variants, similar to that seen in CCHS, and observed transcript and protein level changes consistent with activation of the endoplasmic reticulum (ER) unfolded protein response. Attenuation of ER stress pathways did not result in a correction of Phox2B misfolding, indicating a primary effect of isoflurane on protein structure. We also observed that isoflurane hindered the folding and activity of proteins that rely heavily on ER function, like the CFTR channel. Our results show how anesthetic drugs can alter protein folding and induce ER stress, indicating a mechanism by which these agents may affect neuronal function after surgery.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Proteínas de Homeodominio/metabolismo , Hipoventilación/congénito , Isoflurano/efectos adversos , Morfina/efectos adversos , Agregación Patológica de Proteínas/inducido químicamente , Apnea Central del Sueño/inducido químicamente , Factores de Transcripción/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/análisis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Homeodominio/análisis , Humanos , Hipoventilación/inducido químicamente , Hipoventilación/metabolismo , Hipoventilación/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Pliegue de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Apnea Central del Sueño/metabolismo , Apnea Central del Sueño/patología , Factores de Transcripción/análisis , Respuesta de Proteína Desplegada/efectos de los fármacos
15.
J Mol Recognit ; 31(6): e2697, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29318655

RESUMEN

Dysfunctional accumulation of amyloid ß-protein (Aß) mediated by Cu2+ exhibits higher neurotoxicity and accelerates the progress of Alzheimer's disease, so inhibition of Cu2+ -mediated Aß aggregation and cytotoxicity has been considered as a therapeutic strategy for the disease. Herein, a nonapeptide was designed by linking HH to the C-terminus of a peptide inhibitor of Aß aggregation, LVFFARK (LK7). We found that the nonapeptide, LK7-HH, possessed dual functionality, including enhanced inhibition capability on Aß aggregation as compared to LK7, and chelating Cu2+ with a dissociation constant of 5.50 µM. This enabled LK7-HH to arrest the generation of reactive oxygen species catalyzed by Cu2+ or Cu2+ -Aß complex, and to inhibit Cu2+ -induced Aß aggregation. Moreover, in contrast with the cytotoxicity of LK7 aggregates, LK7-HH was biocompatible because HH conjugation made its aggregation behavior different from LK7. Thus, LK7-HH efficiently suppressed Cu2+ -mediated Aß aggregation and cytotoxicity. An equimolar concentration of LK7-HH increased cell viability from 50% to 90% when treating Aß40 -Cu2+ complexes. The results provided insights into the roles of HH in enhancing the inhibition of Aß and Cu2+ -induced Aß aggregations, in eliminating Cu2+ -induced cytotoxicities by arresting generation of reactive oxygen species, and in making the peptide biocompatible. Therefore, this work would contribute to the design of potent peptide-based inhibitors of Cu2+ -mediated Aß aggregation and cytotoxicity.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Quelantes/farmacología , Péptidos/química , Agregación Patológica de Proteínas/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quelantes/química , Cobre/toxicidad , Humanos , Péptidos/genética , Péptidos/farmacología , Agregado de Proteínas/genética , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/patología , Especies Reactivas de Oxígeno/química
16.
Nutr Neurosci ; 21(2): 92-96, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27697018

RESUMEN

Multiple sclerosis (MS) is a disease which manifests demyelination of neuronal cells in the brain. Despite extensive research on the mechanisms of disease development and progression, the exact mechanism is not elucidated yet, which has hampered drug development and subsequent treatment of the disease. We have recently shown that the serum levels of arsenic and malondialdehyde, a lipid peroxidation marker, are high in MS patients. In this article, we would like to formulate the hypothesis that arsenic may cause MS by induction of inflammation, degeneration, and apoptosis in neuronal cells. The induction of ROS generation in cells upon exposure to arsenic as a heavy metal may be involved in the pathogenesis of MS. Tau protein, a member of the family of microtubule-associated proteins, is mainly expressed in neurons and contribute to the assembly of neuronal microtubules network. Arsenic may affect the hyperphosphorylation and aggregation of tau proteins and may be involved in the cascade leading to deregulation of tau function associated with neurodegeneration. For validation of this hypothesis, studies might be conducted to evaluate the association of arsenic levels and tau protein levels in MS patients. Further studies might also focus on the trafficking along microtubules in neurons of MS patient with regard to hyperphosphorylation of tau protein. This hypothesis may add a new dimension to the understanding of MS etiology and help to design novel therapeutic agents against potential targets that might be discovered. If this hypothesis proves to be true, tau phosphorylation inhibitors can be potential candidates for MS drug development.


Asunto(s)
Arsénico/toxicidad , Inflamación/diagnóstico , Esclerosis Múltiple/diagnóstico , Agregación Patológica de Proteínas/diagnóstico , Proteínas tau/metabolismo , Animales , Apoptosis , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Progresión de la Enfermedad , Humanos , Inflamación/inducido químicamente , Peroxidación de Lípido , Malondialdehído/sangre , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Esclerosis Múltiple/inducido químicamente , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Agregación Patológica de Proteínas/inducido químicamente , Proteínas tau/genética
17.
Mol Neurobiol ; 55(1): 138-144, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28822080

RESUMEN

Soluble epoxide hydrolase (sEH) is widely expressed in the mammalian brain and possesses dual enzymatic activities, including C-terminal epoxide hydrolase (C-EH) which degrades epoxyeicosatrienoic acid (EET), a beneficial arachidonic acid metabolite. In the present study, the neuroprotective effect of sEH inhibition on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurodegeneration of nigrostriatal dopaminergic system was investigated using genetic and pharmacological approaches. MPTP (15 mg/kg) was intraperitoneally injected in sEH knockout (KO) mice and C57BL/6J mice as wild-type (WT) mice. Compared with the MPTP-treated WT mice, MPTP-induced reductions in striatal dopamine content and nigral tyrosine hydroxylase level (TH, a biomarker of dopaminergic neurons) were less significant in the treated sEH mice. Furthermore, MPTP-induced HO-1 elevation (a redox-regulated protein), α-synuclein aggregation, and caspase 12 activation (a hallmark of ER stress) were less prominent in sEH KO mice than in WT mice. These data indicate that sEH KO mice are more resistant to MPTP-induced neurotoxicity. The pharmacological effect of N-[1-(1-oxopropyl)-4-piperidinyl]-N0-[4-(trifluoromethoxy)phenyl)-urea (TPPU, an sEH inhibitor) on MPTP-induced neurotoxicity was investigated in WT mice. TPPU (1 mg/kg, i.p.) attenuated MPTP-induced reduction in striatal dopamine content, TH-positive cell numbers, TH, and pro-caspase 9 protein levels (an initiator caspase of apoptosis) in mouse SN. Moreover, TPPU reduced MPTP-induced HO-1 elevation, α-synuclein aggregation and caspase 12 activation, indicating that TPPU is effective in attenuating MPTP-induced oxidative stress, apoptosis, protein aggregation, and ER stress. In conclusion, our study suggests that sEH is a potential target for developing therapies for parkinsonism. Furthermore, sEH inhibitors may be of clinical significance for treating CNS neurodegenerative diseases.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Epóxido Hidrolasas/antagonistas & inhibidores , Intoxicación por MPTP/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Piperidinas/uso terapéutico , Agregación Patológica de Proteínas/tratamiento farmacológico , alfa-Sinucleína/antagonistas & inhibidores , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Epóxido Hidrolasas/metabolismo , Intoxicación por MPTP/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Compuestos de Fenilurea/farmacología , Piperidinas/farmacología , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , alfa-Sinucleína/metabolismo
18.
Toxicology ; 394: 11-18, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29196190

RESUMEN

Cigarette smoking is a significant risk factor for cataract. However, the mechanism by which cigarette smoke (CS) causes cataract remains poorly understood. We had earlier shown that in CS-exposed guinea pig, p-benzoquinone (p-BQ) derived from CS in the lungs is carried by the circulatory system to distant organs and induces various smoke-related pathogeneses. Here, we observed that CS exposure caused accumulation of the p-BQ-protein adduct in the eye lens of guinea pigs. We also observed accumulation of the p-BQ-protein adduct in resected lens from human smokers with cataract. No such accumulation was observed in the lens of never smokers. p-BQ is a strong arylating agent that forms Michael adducts with serum albumin and haemoglobin resulting in alterations of structure and function. A major protein in the mammalian eye lens is αA-crystallin, which is a potent molecular chaperone. αA-crystallin plays a key role in maintaining the integrity and transparency of the lens. SDS-PAGE indicated that p-BQ induced aggregation of αA-crystallin. Various biophysical techniques including UV-vis spectroscopy, fluorescence spectroscopy, FT-IR, bis-ANS titration suggested a perturbation of structure and chaperone function of αA-crystallin upon p-BQ modification. Our results indicate that p-BQ is a causative agent involved in the modification of αA-crystallin and pathogenesis of CS-induced cataract. Our findings would educate public about the impacts of smoking on eye health and help to discourage them from smoking. The study might also help scientists to develop new drugs for the intervention of CS-induced cataract at an early stage.


Asunto(s)
Benzoquinonas/toxicidad , Catarata/etiología , Catarata/metabolismo , Fumar Cigarrillos/efectos adversos , alfa-Cristalinas/metabolismo , Anciano , Animales , Benzoquinonas/química , Benzoquinonas/farmacocinética , Benzoquinonas/envenenamiento , Catarata/inducido químicamente , Catarata/patología , Fumar Cigarrillos/metabolismo , Fumar Cigarrillos/patología , Escherichia coli/genética , Escherichia coli/metabolismo , Cobayas , Humanos , Cápsula del Cristalino/efectos de los fármacos , Cápsula del Cristalino/metabolismo , Cápsula del Cristalino/patología , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/metabolismo , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , alfa-Cristalinas/biosíntesis , alfa-Cristalinas/química , alfa-Cristalinas/genética
19.
Neuroscience ; 367: 47-59, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29080716

RESUMEN

Ubiquitin-proteasome system (UPS) has emerged as major molecular mechanism which modulates synaptic plasticity. However, very little is known about what happens if this system fails during postnatal brain development. In the present study, MG132 was administered intracerebroventricularly in BALB/c mice pups at postnatal day one (P1), a very crucial period for synaptogenesis. Both 20S proteasome and calpain activities were found to be reduced in the mid brain of MG132-administered pups after 24 h. Mice (P40) which received MG132 on P1 were subjected to Morris water maze (MWM) training. Analysis showed spatial learning and memory of MG132 mice was significantly impaired when compared to age-matched controls. Hematoxylin and eosin as well as Cresyl Violet staining revealed substantial loss of cellular connections, distorted architecture and increased pyknosis in hippocampal CA1 and CA3 regions of MG132 mice. Immunohistochemical analysis of MG132 mice showed increased accumulation of intracellular amyloid-ß in hippocampal cells when compared to control. Moreover, double immunostaining revealed increased expression of amyloid precursor protein C-terminal fragments (APP-CTFß) without affecting ß-secretase expression in MG132 mice. Real-Time PCR analyses showed significant increase in hippocampal expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit glutamate A1 (GluA1), but no change in the brain-derived neurotrophic factor (Bdnf) expression in MG132 mice. Western blot analyses showed decreased levels of pThr286-CaMKIIα:CaMKIIα and pSer133-CREB:CREB ratio but increased pro:mature BDNF ratio in the hippocampus of MG132 mice. Taken together, postnatal proteasome inhibition could lead to accumulation of intracellular amyloid-ß protein aggregates, which mediate hippocampus-dependent spatial memory impairments in adult mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Aprendizaje Espacial/fisiología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Recién Nacidos , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Discapacidades para el Aprendizaje/inducido químicamente , Leupeptinas/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Inhibidores de Proteasoma/toxicidad , Agregación Patológica de Proteínas/inducido químicamente , Receptores AMPA/genética , Receptores AMPA/metabolismo , Aprendizaje Espacial/efectos de los fármacos
20.
BMC Neurosci ; 18(1): 51, 2017 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-28651647

RESUMEN

BACKGROUND: In the recent decade, iron oxide nanoparticles (IONPs) have been proposed for several applications in the central nervous system (CNS), including targeting amyloid beta (Aß) in the arteries, inhibiting the microglial cells, delivering drugs, and increasing contrast in magnetic resonance imaging. Conversely, a notable number of studies have reported the role of iron in neurodegenerative diseases. Therefore, this study has reviewed the recent studies to determine whether IONPs iron can threaten the cellular viability same as iron. RESULTS: Iron contributes in Fenton's reaction and produces reactive oxygen species (ROS). ROS cause to damage the macromolecules and organelles of the cell via oxidative stress. Iron accumulation and oxidative stress are able to aggregate some proteins, including Aß and α-synuclein, which play a critical role in Alzheimer's and Parkinson's diseases, respectively. Iron accumulation, oxidative stress, and protein aggregation make a positive feedback loop, which can be toxic for the cell. The release of iron ions from IONPs may result in iron accumulation in the targeted tissue, and thus, activate the positive feedback loop. However, the levels of IONPs induced toxicity depend on the size, concentration, surface charge, and the type of coating and functional groups of IONPs. CONCLUSION: IONPs depending on their properties can lead to iron accumulation, oxidative stress and protein aggregation in the neural cells. Therefore, in order to apply IONPs in the CNS, the consideration of IONPs properties is crucial.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Citotoxinas/farmacología , Compuestos Férricos/toxicidad , Nanopartículas del Metal/toxicidad , Animales , Humanos , Hierro/metabolismo , Estrés Oxidativo , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA