Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 47(8): 2373-2382, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35589915

RESUMEN

Potentially druggable mechanisms underlying synaptic deficits seen in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are under intense interrogations. In addition to defective synaptic vesicle trafficking, cytoskeletal disruption, autophagic perturbation, and neuroinflammation, hyperphosphorylation of microtubule-associated protein collapsin response mediator protein 2 (CRMP2, also known as DPYSL2) is newly determined to correlate with synaptic deficits in human DLB. The small molecule experimental therapeutic, lanthionine ketimine-5-ethyl ester (LKE), appears to interact with CRMP2 in a host of neurodegenerative mouse models, normalizing its phosphorylation level while promoting healthful autophagy in cell culture models and suppressing the proinflammatory phenotype of activated microglia. Accordingly, this study examined the effect of LKE on α-synuclein A53T transgenic (Tg) mice which were employed as a DLB model. We found that chronic administration of LKE to A53T mice suppressed (1) the accumulation of LBs, (2) neuroinflammatory activation of microglia, (3) impairment of contextual fear memory, and (4) CRMP2 phosphorylation at Thr509 in A53T Tg mice. These results suggest that CRMP2 phosphorylation by GSK3ß in the hippocampus is related to pathology and memory impairment in DLB, and LKE may have clinical implications in the treatment of α-synucleinopathy.


Asunto(s)
Aminoácidos Sulfúricos , Sinucleinopatías , Aminoácidos Sulfúricos/farmacología , Aminoácidos Sulfúricos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Ésteres , Humanos , Ratones , Ratones Transgénicos , alfa-Sinucleína
2.
Genes Cells ; 24(1): 31-40, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30375127

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by slow and progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc). Levodopa (l-Dopa), the current main treatment for PD, supplies dopamine, but it does not prevent neurodegeneration. There is thus no promising remedy for PD. Recent in vitro study showed the increase in the phosphorylation levels of Collapsin Response Mediator Protein 2 (CRMP2) is involved in dopaminergic axon degeneration. In the present study, we report elevation of CRMP2 phosphorylation in dopaminergic neurons in SNc after challenge with the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a common model for PD. Genetic suppression of CRMP2 phosphorylation by mutation of the obligatory Cyclin-dependent kinase 5 (Cdk5)-targeted serine-522 site prevented axonal degradation in the nigrostriatal pathway of transgenic mice. As a result, the degree of MPTP-induced motor impairment in the rotarod test was suppressed. These results suggest that suppression of CRMP2 phosphorylation may be a novel therapeutic target for PD.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/genética , Supresión Genética , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Actividad Motora , Neostriado/patología , Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Fosforilación , Sustancia Negra/patología
3.
J Biol Chem ; 293(47): 18242-18269, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30282635

RESUMEN

Oxidative stress triggers and exacerbates neurodegeneration in Alzheimer's disease (AD). Various antioxidants reduce oxidative stress, but these agents have little efficacy due to poor blood-brain barrier (BBB) permeability. Additionally, single-modal antioxidants are easily overwhelmed by global oxidative stress. Activating nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) and its downstream antioxidant system are considered very effective for reducing global oxidative stress. Thus far, only a few BBB-permeable agents activate the Nrf2-dependent antioxidant system. Here, we discovered a BBB-bypassing Nrf2-activating polysaccharide that may attenuate AD pathogenesis. Mini-GAGR, a 0.7-kDa cleavage product of low-acyl gellan gum, increased the levels and activities of Nrf2-dependent antioxidant enzymes, decreased reactive oxygen species (ROS) under oxidative stress in mouse cortical neurons, and robustly protected mitochondria from oxidative insults. Moreover, mini-GAGR increased the nuclear localization and transcriptional activity of Nrf2 similarly to known Nrf2 activators. Mechanistically, mini-GAGR increased the dissociation of Nrf2 from its inhibitor, Kelch-like ECH-associated protein 1 (Keap1), and induced phosphorylation and nuclear translocation of Nrf2 in a protein kinase C (PKC)- and fibroblast growth factor receptor (FGFR1)-dependent manner. Finally, 20-day intranasal treatment of 3xTg-AD mice with 100 nmol of mini-GAGR increased nuclear p-Nrf2 and growth-associated protein 43 (GAP43) levels in hippocampal neurons, reduced p-tau and ß-amyloid (Aß) peptide-stained neurons, and improved memory. The BBB-bypassing Nrf2-activating polysaccharide reported here may be effective in reducing oxidative stress and neurodegeneration in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antioxidantes/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Polisacáridos Bacterianos/administración & dosificación , Administración Intranasal , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
4.
Mol Cell Biochem ; 442(1-2): 29-38, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28913673

RESUMEN

The ADAM (a disintegrin and metalloprotease) protein family uniquely exhibits both catalytic and adhesive properties. In the well-defined process of ectodomain shedding, ADAMs transform latent, cell-bound substrates into soluble, biologically active derivatives to regulate a spectrum of normal and pathological processes. In contrast, the integrin ligand properties of ADAMs are not fully understood. Emerging models posit that ADAM-integrin interactions regulate shedding activity by localizing or sequestering the ADAM sheddase. Interestingly, 8 of the 21 human ADAMs are predicted to be catalytically inactive. Unlike their catalytically active counterparts, integrin recognition of these "dead" enzymes has not been largely reported. The present study delineates the integrin ligand properties of a group of non-catalytic ADAMs. Here we report that human ADAM11, ADAM23, and ADAM29 selectively support integrin α4-dependent cell adhesion. This is the first demonstration that the disintegrin-like domains of multiple catalytically inactive ADAMs are ligands for a select subset of integrin receptors that also recognize catalytically active ADAMs.


Asunto(s)
Proteínas ADAM/metabolismo , Integrina alfa4/metabolismo , Proteínas ADAM/genética , Animales , Células CHO , Adhesión Celular/fisiología , Cricetulus , Humanos , Integrina alfa4/genética , Células Jurkat , Células K562 , Ligandos
5.
Rapid Commun Mass Spectrom ; 32(22): 1941-1948, 2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30117207

RESUMEN

RATIONALE: Preclinical studies in the search for treatments for several neurodegenerative diseases have identified lanthionine ketimine (LK) and its monoethyl ester derivative (LKE) as potential candidates. An ultrahigh-pressure liquid chromatography/tandem mass spectrometry (UHPLC/MS/MS) assay was developed to evaluate bioavailability by measuring these compounds in mouse serum, whole blood and brain tissue. METHODS: Following administration of LKE to mice for 3 days in chow at 300 ppm, the animals were sacrificed, and LKE was extracted from serum, whole blood and brain tissues through protein precipitation using cold methanol. To enhance chromatographic separation and electrospray ionization, LK was methylated using diazomethane. Separations were carried out using C18 reversed-phase UHPLC, and quantitative measurements were obtained using on-line triple-quadruple mass spectrometry with positive ion electrospray ionization, collision-induced dissociation and selected reaction monitoring. Tolbutamide was used as internal standard. RESULTS: LKE showed good recovery ranging from 77-90% in serum and 82-88% in brain tissue. An eight-point standard curve ranging from 0.005 to 4.6 µM was linear (R2 0.998). The average LKE detected in mouse serum was 277.42 nM, while the concentration in whole blood was 38 nM. Neither LK nor LKE was detected in brain tissues. CONCLUSIONS: A rapid quantitative method to measure LKE in mouse serum, whole blood and brain tissues using UHPLC/MS/MS was developed and validated following FDA guidelines. This method is suitable for bioavailability and pharmacokinetic studies.


Asunto(s)
Aminoácidos Sulfúricos/sangre , Aminoácidos Sulfúricos/farmacocinética , Encéfalo/metabolismo , Espectrometría de Masas en Tándem/métodos , Animales , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión/métodos , Ésteres/sangre , Ésteres/farmacocinética , Límite de Detección , Ratones
6.
Bioorg Med Chem Lett ; 28(4): 562-565, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29398540

RESUMEN

The multiple-step, one-pot procedure for a series of 2-substituted-3-phosphono-1-thia-4-aza-2-cyclohexene-5-carboxylates, analogues of the natural, sulfur amino acid metabolite lanthionine ketimine (LK), its 5-ethyl ester (LKE) and 2-substituted LKEs is described. Initiating the synthesis with the Michaelis-Arbuzov preparation of α-ketophosphonates allows for a wide range of functional variation at the 2-position of the products. Nine new compounds were synthesized with overall yields range from 40 to 62%. In addition, the newly prepared 2-isopropyl-LK-P, 2-n-hexyl-LKE-P and 2-ethyl-LKE were shown to stimulate autophagy in cultured cells better than that of the parent compound, LKE.


Asunto(s)
Aminoácidos Sulfúricos/farmacología , Ciclohexenos/farmacología , Ésteres/farmacología , Ácidos Fosforosos/farmacología , Tiazinas/farmacología , Aminoácidos Sulfúricos/síntesis química , Animales , Autofagia/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ciclohexenos/síntesis química , Ésteres/síntesis química , Humanos , Macrólidos/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Ácidos Fosforosos/síntesis química , Ratas , Tiazinas/síntesis química
7.
Biochem Biophys Res Commun ; 483(1): 759-764, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-27965088

RESUMEN

The mammalian central nervous system (CNS) has limited regenerative ability after injury, largely due to scar formation and axonal growth inhibitors. Experimental suppression of neuroinflammation encourages recovery from spinal cord injury (SCI), yet practical means for pharmacologically treating SCI have remained elusive. Lanthionine ketimine (LK) is a natural brain sulfur amino acid metabolite with demonstrated anti-neuroinflammatory and neurotrophic activities. LK and its synthetic brain-penetrating ethyl ester (LKE) promote growth factor-dependent neurite extension in cultured cell and suppress microglial activation in animal models of neurodegeneration. Thus far however, LKE has not been explored as a potential therapy for SCI. The present study investigated the hypothesis that systemic LKE could improve motor functional recovery after SCI in a mouse model. Intraperitoneal administration of LKE (100 mg/kg/d) after near-complete transect of spinal cord at the T7 level significantly improved motor function over a 4-week time course. Vehicle-treated mice, in contrast, demonstrated negligible functional recovery. In terms of histology, LKE treatment reduced pro-neuroinflammatory microglia/macrophage activation evidenced by quantitative Iba1 labeling and shifted the microglial phenotype toward a more neurotrophic M2 character evidenced by changes in the M2 marker arginase-1. This was correlated with less dense scar formation and more extensive axonal regrowth across the transection site demonstrated by 5-hydroxytryptamine (5HT) immunolabeling of raphespinal tract axons. These data provide evidence that LKE or similar compounds have potential therapeutic value for recovery after certain forms of SCI.


Asunto(s)
Aminoácidos Sulfúricos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Médula Espinal/fisiopatología , Aminoácidos Sulfúricos/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Axones/efectos de los fármacos , Axones/fisiología , Biomarcadores/análisis , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Inflamación/tratamiento farmacológico , Inflamación/patología , Locomoción/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/patología , Microglía/fisiología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Serotonina/metabolismo , Médula Espinal/efectos de los fármacos , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología
8.
Amino Acids ; 49(4): 747-759, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28044206

RESUMEN

Collapsin response mediator protein 2 (CRMP-2) is a neuronal protein involved in axonal pathfinding. Intense research is focusing on its role in various neurological diseases. Despite a wealth of studies, not much is known about the molecular mechanisms of CRMP-2 function in vivo. The detailed structure-function relationships of CRMP-2 have also largely remained unknown, in part due to the fact that the available crystal structures lack the C-terminal tail, which is known to be a target for many post-translational modifications and protein interactions. Although CRMP-2, and other CRMPs, belong to the dihydropyrimidinase family, they have lost the enzymatic active site. Drug candidates for CRMP-2-related processes have come up during the recent years, but no reports of CRMP-2 complexes with small molecules have emerged. Here, CRMP-2 was studied at 1.25-Å resolution using X-ray crystallography. In addition, ligands were docked into the homotetrameric structure, and the C-terminal tail of CRMP-2 was produced recombinantly and analyzed. We have obtained the human CRMP-2 crystal structure at atomic resolution and could identify small-molecule binding pockets in the protein. Structures obtained in different crystal forms highlight flexible regions near possible ligand-binding pockets. We also used the CRMP-2 structure to analyze known or suggested post-translational modifications at the 3D structural level. The high-resolution CRMP-2 structure was also used for docking experiments with the sulfur amino acid metabolite lanthionine ketimine and its ester. We show that the C-terminal tail is intrinsically disordered, but it has conserved segments that may act as interaction sites. Our data provide the most accurate structural data on CRMPs to date and will be useful in further computational and experimental studies on CRMP-2, its function, and its binding to small-molecule ligands.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/química , Proteínas del Tejido Nervioso/química , Procesamiento Proteico-Postraduccional , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química
9.
Biomarkers ; 22(3-4): 372-382, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28055279

RESUMEN

CONTEXT: Quantitative changes of salivary proteins due to acute stress were detected. OBJECTIVE: To explore protein markers of stress in saliva of eight medical residents who performed emergency medicine simulations. MATERIALS AND METHODS: Saliva was collected before the simulations, after the simulations, and following morning upon waking. Proteins were separated by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE), identified by mass spectrometry (MS), and relatively quantified by densitometry. RESULTS: Salivary alpha-amylase and S-type cystatins significantly increased, while the ∼26 kDa and low-molecular weight (MW) (<10 kDa) SDS-PAGE bands exhibited changes after stress. DISCUSSION AND CONCLUSION: Alpha-amylase and cystatins are potential salivary markers of acute stress, but further validation should be performed using larger sample populations.


Asunto(s)
Proteómica/métodos , Proteínas y Péptidos Salivales/metabolismo , Estrés Psicológico/metabolismo , Adulto , Electroforesis en Gel de Poliacrilamida , Servicios Médicos de Urgencia/métodos , Femenino , Humanos , Internado y Residencia , Masculino , Espectrometría de Masas , Proyectos Piloto , Cistatinas Salivales/análisis , Proteínas y Péptidos Salivales/análisis , Adulto Joven , alfa-Amilasas/análisis
10.
Genes Dev ; 23(12): 1387-92, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19528316

RESUMEN

Eukaryotic lanthionine synthetase C-like protein 1 (LanCL1) is homologous to prokaryotic lanthionine cyclases, yet its biochemical functions remain elusive. We report the crystal structures of human LanCL1, both free of and complexed with glutathione, revealing glutathione binding to a zinc ion at the putative active site formed by conserved GxxG motifs. We also demonstrate by in vitro affinity analysis that LanCL1 binds specifically to the SH3 domain of a signaling protein, Eps8. Importantly, expression of LanCL1 mutants defective in Eps8 interaction inhibits nerve growth factor (NGF)-induced neurite outgrowth, providing evidence for the biological significance of this novel interaction in cellular signaling and differentiation.


Asunto(s)
Glutatión/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Escherichia coli/genética , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Mutación , Factor de Crecimiento Nervioso/farmacología , Neuritas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Células PC12 , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal , Zinc/metabolismo
11.
Biochim Biophys Acta ; 1840(2): 722-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23644035

RESUMEN

BACKGROUND: Immuno-spin trapping (IST) is based on the reaction of a spin trap with a free radical to form a stable nitrone adduct, followed by the use of antibodies, rather than traditional electron paramagnetic resonance spectroscopy, to detect the nitrone adduct. IST has been successfully applied to mechanistic in vitro studies, and recently, macromolecule-centered radicals have been detected in models of drug-induced agranulocytosis, hepatotoxicity, cardiotoxicity, and ischemia/reperfusion, as well as in models of neurological, metabolic and immunological diseases. SCOPE OF THE REVIEW: To critically evaluate advances, challenges, and pitfalls as well as the scientific opportunities of IST as applied to the study of protein-centered free radicals generated in stressed organelles, cells, tissues and animal models of disease and exposure. MAJOR CONCLUSIONS: Because the spin trap has to be present at high enough concentrations in the microenvironment where the radical is formed, the possible effects of the spin trap on gene expression, metabolism and cell physiology have to be considered in the use of IST and in the interpretation of results. These factors have not yet been thoroughly dealt with in the literature. GENERAL SIGNIFICANCE: The identification of radicalized proteins during cell/tissue response to stressors will help define their role in the complex cellular response to stressors and pathogenesis; however, the fidelity of spin trapping/immuno-detection and the effects of the spin trap on the biological system should be considered. This article is part of a Special Issue entitled Current methods to study reactive oxygen species - pros and cons and biophysics of membrane proteins. Guest Editor: Christine Winterbourn.


Asunto(s)
Radicales Libres/análisis , Inmunoglobulina G/inmunología , Óxidos de Nitrógeno/química , Proteínas/inmunología , Detección de Spin/métodos , Animales , Bioquímica , Radicales Libres/aislamiento & purificación , Humanos , Óxidos de Nitrógeno/inmunología
12.
J Neurochem ; 134(2): 302-14, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25846048

RESUMEN

Lanthionine ketimine (LK) is a natural sulfur amino acid metabolite which binds to collapsin response mediator protein-2 (CRMP2), an abundant brain protein that interacts with multiple partners to regulate microtubule dynamics, neurite growth and retraction, axonal transport, and neurotransmitter release. LK ethyl-ester (LKE) is a cell-permeable synthetic derivative that promotes neurogenesis, suppresses nitric oxide production from microglia, and reduces neurotoxicity of microglia-conditioned medium. These properties led us to test the effects of LKE in experimental autoimmune encephalomyelitis (EAE), a commonly used mouse model of multiple sclerosis. Female C57Bl/6 mice were immunized with myelin oligodendrocyte glycoprotein peptide 35-55 to develop a chronic disease. LKE was provided in the chow at 100 ppm, ad libitum beginning when the mice reached moderate clinical signs. Over the following 4 weeks the LKE-treated mice showed a significant reduction in clinical signs compared to vehicle-treated mice. LKE dose dependently reduced IFNγ production from splenic T cells, but had no effect on IL-17 production suggesting protective effects were mediated within the CNS. Electron microscopy revealed that, compared to sham mice, EAE mice had significant neurodegeneration in both the optic nerve and spinal cord, which was reduced in the LKE-treated mice. In contrast only minimal disruption of myelin was observed at this time point. In the optic nerve, measurements of axon caliber and myelin thickness showed little changes between sham and EAE mice, however, treatment with LKE increased the percentage of axons with thicker myelin and with larger axon calibers. In the spinal cord, only smaller effects of LKE on myelin thickness were observed. The effects of LKE were associated with a reduced relative level of phosphorylated CRMP2 to CRMP2. Together, these results demonstrate that LKE reduces neurodegeneration in a chronic EAE model of MS, which could have translation potential for treatment of progressive forms of MS.


Asunto(s)
Aminoácidos Sulfúricos/farmacología , Axones/patología , Encefalomielitis Autoinmune Experimental/patología , Degeneración Nerviosa/patología , Fármacos Neuroprotectores/farmacología , Animales , Axones/efectos de los fármacos , Western Blotting , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
13.
Neurobiol Dis ; 84: 50-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25771170

RESUMEN

Autophagy and redox biochemistry are two major sub disciplines of cell biology which are both coming to be appreciated for their paramount importance in the etiology of neurodegenerative diseases including Alzheimer's disease (AD). Thus far, however, there has been relatively little exploration of the interface between autophagy and redox biology. Autophagy normally recycles macro-molecular aggregates produced through oxidative-stress mediated pathways, and also may reduce the mitochondrial production of reactive oxygen species through recycling of old and damaged mitochondria. Conversely, dysfunction in autophagy initiation, progression or clearance is evidenced to increase aggregation-prone proteins in neural and extraneural tissues. Redox mechanisms of autophagy regulation have been documented at the level of cross-talk between the Nrf2/Keap1 oxidant and electrophilic defense pathway and p62/sequestosome-1 (SQSTM1)-associated autophagy, at least in extraneural tissue; but other mechanisms of redox autophagy regulation doubtless remain to be discovered and the relevance of such processes to maintenance of neural homeostasis remains to be determined. This review summarizes current knowledge regarding the relationship of redox signaling, autophagy control, and oxidative stress as these phenomena relate to neurodegenerative disease. AD is specifically addressed as an example of the theme and as a promising indication for new therapies that act through engagement of autophagy pathways. To exemplify one such novel therapeutic entity, data is presented that the antioxidant and neurotrophic agent lanthionine ketimine-ethyl ester (LKE) affects autophagy pathway proteins including beclin-1 in the 3xTg-AD model of Alzheimer's disease where the compound has been shown to reduce pathological features and cognitive dysfunction.


Asunto(s)
Autofagia , Encéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Humanos , Oxidación-Reducción
14.
Neurobiol Dis ; 84: 60-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25779968

RESUMEN

Autophagy is a fundamental cellular recycling process vulnerable to compromise in neurodegeneration. We now report that a cell-penetrating neurotrophic and neuroprotective derivative of the central nervous system (CNS) metabolite, lanthionine ketimine (LK), stimulates autophagy in RG2 glioma and SH-SY5Y neuroblastoma cells at concentrations within or below pharmacological levels reported in previous mouse studies. Autophagy stimulation was evidenced by increased lipidation of microtubule-associated protein 1 light chain 3 (LC3) both in the absence and presence of bafilomycin-A1 which discriminates between effects on autophagic flux versus blockage of autophagy clearance. LKE treatment caused changes in protein level or phosphorylation state of multiple autophagy pathway proteins including mTOR; p70S6 kinase; unc-51-like-kinase-1 (ULK1); beclin-1 and LC3 in a manner essentially identical to effects observed after rapamycin treatment. The LKE site of action was near mTOR because neither LKE nor the mTOR inhibitor rapamycin affected tuberous sclerosis complex (TSC) phosphorylation status upstream from mTOR. Confocal immunofluorescence imaging revealed that LKE specifically decreased mTOR (but not TSC2) colocalization with LAMP2(+) lysosomes in RG2 cells, a necessary event for mTORC1-mediated autophagy suppression, whereas rapamycin had no effect. Suppression of the LK-binding adaptor protein CRMP2 (collapsin response mediator protein-2) by means of shRNA resulted in diminished autophagy flux, suggesting that the LKE action on mTOR localization may occur through a novel mechanism involving CRMP2-mediated intracellular trafficking. These findings clarify the mechanism-of-action for LKE in preclinical models of CNS disease, while suggesting possible roles for natural lanthionine metabolites in regulating CNS autophagy.


Asunto(s)
Aminoácidos Sulfúricos/farmacología , Autofagia/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Fármacos Neuroprotectores/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos Sulfúricos/química , Animales , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Inmunosupresores/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Ratas , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
16.
J Neurosci Res ; 91(9): 1183-90, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825043

RESUMEN

Lanthionine ketimine (LK) is a natural sulfur amino acid metabolite with potent neurotrophic activity. Proteomics indicate that LK interacts with collapsin response mediator protein-2 (CRMP2/DPYSL2/UNC-33), a brain-enriched protein that was shown to regulate cytoskeletal remodeling, neuronal morphology, and synaptic function. To elucidate further the molecular interplay and biological action of LK and UNC-33, we began examining the nervous system of Caenorhabditis elegans nematodes in which both LK concentrations and UNC-33 protein were manipulated. To this end, a cell-permeable LK-ester (LKE) was administered to developing C. elegans engineered to express yellow fluorescent protein (YFP) in cholinergic neurons (strain RM3128) or green fluorescent protein (GFP) in GABAergic neurons (strain CZ1200), and neural morphology was assessed. Fluorescent imaging analyses show that LKE exposure to wild-type animals induced neural commissure outgrowth, crossing over, and bundling in both neurites from GABAergic and cholinergic motor neurons. Additionally, when unc-33(e204) hypomorph mutant nematodes (D389N substitution mutants) were exposed to LKE, both the neuroanatomical defects of incomplete dorsoventral neural commissures and the ventral nerve cord gaps were partially rescued. In contrast, LKE did not rescue ventral nerve cord gaps found in unc-33(mn407) null mutant. Together these data suggest possible functions for LK as a regulator of neuritic elongation, corroborate roles for UNC-33/CRMP2 in the mechanism of LKE activity, and suggest the potential of LKE as a therapeutic molecule for neurological diseases involving CRMP2 dysfunction.


Asunto(s)
Aminoácidos Sulfúricos/uso terapéutico , Encefalopatías/tratamiento farmacológico , Proteínas de Caenorhabditis elegans/genética , Discapacidades del Desarrollo/tratamiento farmacológico , Mutación/genética , Factores de Crecimiento Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Factores de Edad , Aminoácidos Sulfúricos/química , Aminoácidos Sulfúricos/farmacología , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Proteínas Bacterianas/genética , Encefalopatías/complicaciones , Encefalopatías/genética , Caenorhabditis elegans , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Modelos Animales de Enfermedad , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Locomoción/efectos de los fármacos , Locomoción/genética , Longevidad/efectos de los fármacos , Longevidad/genética , Proteínas Luminiscentes/genética , Sistema Nervioso/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
17.
J Neuroinflammation ; 9: 115, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22651808

RESUMEN

Over the past two decades, it has become increasingly apparent that Alzheimer's disease neuropathology is characterized by activated microglia (brain resident macrophages) as well as the classic features of amyloid plaques and neurofibrillary tangles. The intricacy of microglial biology has also become apparent, leading to a heightened research interest in this particular cell type. Over the years a number of different microglial cell culturing techniques have been developed to study either primary mammalian microglia, or immortalized cell lines. Each microglial system has advantages and disadvantages and should be selected for its appropriateness in a particular research context. This review summarizes several of the most common microglial cell culture systems currently being employed in Alzheimer's research including primary microglia; BV2 and N9 retroviral immortalized microglia; human immortalized microglia (HMO6); and spontaneously immortalized rodent microglial lines (EOC lines and HAPI cells). Particularities of cell culture requirements and characteristics of microglial behavior, especially in response to applied inflammogen stimuli, are compared and discussed across these cell types.


Asunto(s)
Enfermedad de Alzheimer/patología , Técnicas de Cultivo de Célula/métodos , Microglía/patología , Microglía/fisiología , Enfermedad de Alzheimer/genética , Animales , Línea Celular Transformada , Células Cultivadas , Humanos
18.
Free Radic Biol Med ; 178: 161-173, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34863876

RESUMEN

Although we can thoroughly describe individual neurodegenerative diseases from the molecular level through cell biology to histology and clinical presentation, our understanding of them and hence treatment gains have been depressingly limited, partly due to difficulty conceptualizing different diseases as variations within the same overarching pathological rubric. This review endeavors to create such rubric by knitting together the seemingly disparate phenomena of oxidative stress, dysregulated proteostasis, and neuroinflammation into a cohesive triad that highlights mechanistic connectivities. We begin by considering that brain metabolic demands necessitate careful control of oxidative homeostasis, largely through sulfur redox chemistry and glutathione (GSH). GSH is essential for brain antioxidant defense, but also for redox signaling and thus neuroinflammation. Delicate regulation of neuroinflammatory pathways (NFκB, MAPK-p38, and NLRP3 particularly) occurs through S-glutathionylation of protein phosphatases but also through redox-sensing elements like ASK1; the 26S proteasome and cysteine deubiquitinases (DUBs). The relationship amongst triad elements is underscored by our discovery that LanCL1 (lanthionine synthetase-like protein-1) protects against oxidant toxicity; mediates GSH-dependent reactivation of oxidized DUBs; and antagonizes the pro-inflammatory cytokine, tumor necrosis factor-α (TNFα). We highlight currently promising pharmacological efforts to modulate key triad elements and suggest nexus points that might be exploited to further clinical advantage.


Asunto(s)
Enfermedades Neuroinflamatorias , Proteostasis , Glutatión/metabolismo , Humanos , Oxidación-Reducción , Estrés Oxidativo , Transducción de Señal , Azufre
19.
ASN Neuro ; 14: 17590914221112352, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35791633

RESUMEN

Although over 20 disease modifying therapies are approved to treat Multiple Sclerosis (MS), these do not increase remyelination of demyelinated axons or mitigate axon damage. Previous studies showed that lanthionine ketenamine ethyl ester (LKE) reduces clinical signs in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS and increased maturation of oligodendrocyte (OL) progenitor cells (OPCs) in vitro. In the current study, we used the cuprizone (CPZ) demyelination model of MS to test if LKE could increase remyelination. The corpus callosum (CC) and somatosensory cortex was examined by immunohistochemistry (IHC), electron microscopy and for mRNA expression changes in mice provided 5 weeks of CPZ diet followed by 2 weeks of normal diet in the presence of LKE or vehicle. A significant increase in the number of myelinated axons, and increased myelin thickness was observed in the CC of LKE-treated groups compared to vehicle-treated groups. LKE also increased myelin basic protein and proteolipid protein expression in the CC and cortex, and increased the number of mature OLs in the cortex. In contrast, LKE did not increase the percentage of proliferating OPCs suggesting effects on OPC survival and differentiation but not proliferation. The effects of LKE on OL maturation and remyelination were supported by similar changes in their relative mRNA levels. Interestingly, LKE did not have significant effects on GFAP or Iba1 immunostaining or mRNA levels. These findings suggest that remyelinating actions of LKE can potentially be formulated to induce remyelination in neurological diseases associated with demyelination including MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Remielinización , Aminoácidos Sulfúricos , Animales , Cuprizona/toxicidad , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ésteres/farmacología , Ratones , Esclerosis Múltiple/tratamiento farmacológico , ARN Mensajero
20.
J Neurosci ; 30(8): 2979-88, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20181595

RESUMEN

Lanthionine ketimine (LK) represents a poorly understood class of thioethers present in mammalian CNS. Previous work has indicated high-affinity interaction of LK with synaptosomal membrane protein(s), but neither LK binding partners nor specific bioactivities have been reported. In this study, LK was chemically synthesized and used as an affinity agent to capture binding partners from mammalian brain lysate. Liquid chromatography with electrospray ionization-mass spectrometry of electrophoretically separated, LK-bound proteins identified polypeptides implicated in axon remodeling or vesicle trafficking and diseases including Alzheimer's disease and schizophrenia: collapsin response mediator protein-2/dihydropyrimidinase-like protein-2 (CRMP2/DRP2/DPYSL2), myelin basic protein, and syntaxin-binding protein-1 (STXBP1/Munc-18). Also identified was the recently discovered glutathione-binding protein lanthionine synthetase-like protein-1. Functional consequences of LK:CRMP2 interactions were probed through immunoprecipitation studies using brain lysate wherein LK was found to increase CRMP2 coprecipitation with its partner neurofibromin-1 but decreased CRMP2 coprecipitation with beta-tubulin. Functional studies of NSC-34 motor neuron-like cells indicated that a cell-permeable LK-ester, LKE, was nontoxic and protective against oxidative challenge with H(2)O(2). LKE-treated NSC-34 cells significantly increased neurite number and length in a serum concentration-dependent manner, consistent with a CRMP2 interaction. Finally, LKE antagonized the activation of EOC-20 microglia by inflammogens. The results are discussed with reference to possible biochemical origins, paracrine functions, neurological significance, and pharmacological potential of lanthionyl compounds.


Asunto(s)
Aminoácidos Sulfúricos/metabolismo , Aminoácidos Sulfúricos/farmacología , Encéfalo/metabolismo , Microglía/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Aminoácidos Sulfúricos/síntesis química , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Bovinos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Transformada , Células Cultivadas , Ésteres/farmacología , Hidroliasas/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Microglía/metabolismo , Estructura Molecular , Neuronas Motoras/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas Munc18/metabolismo , Proteína Básica de Mielina/metabolismo , Neuroquímica/métodos , Neurofibromina 1/metabolismo , Proteómica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA