Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 483
Filtrar
1.
Pharmacol Res ; 182: 106338, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35781057

RESUMO

The lysosomal cysteine hydrolase N-acylethanolamine acid amidase (NAAA) deactivates palmitoylethanolamide (PEA), a lipid-derived PPAR-α agonist that is critically involved in the control of pain and inflammation. In this study, we asked whether NAAA-regulated PEA signaling might contribute to dopamine neuron degeneration and parkinsonism induced by the mitochondrial neurotoxins, 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In vitro experiments showed that 6-OHDA and MPTP enhanced NAAA expression and lowered PEA content in human SH-SY5Y cells. A similar effect was observed in mouse midbrain dopamine neurons following intra-striatal 6-OHDA injection. Importantly, deletion of the Naaa gene or pharmacological inhibition of NAAA activity substantially attenuated both dopamine neuron death and parkinsonian symptoms in mice treated with 6-OHDA or MPTP. Moreover, NAAA expression was elevated in postmortem brain cortex and premortem blood-derived exosomes from persons with Parkinson's disease compared to age-matched controls. The results identify NAAA-regulated PEA signaling as a molecular control point for dopaminergic neuron survival and a potential target for neuroprotective intervention.


Assuntos
Neuroblastoma , Transtornos Parkinsonianos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Amidoidrolases , Animais , Modelos Animais de Doenças , Dopamina , Neurônios Dopaminérgicos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Degeneração Neural/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Oxidopamina , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico
2.
Molecules ; 26(16)2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34443417

RESUMO

Sambucus nigra flowers (elderflower) have been widely used in traditional medicine for the relief of early symptoms of common cold. Its chemical composition mainly consists of polyphenolic compounds such as flavonoids, hydroxycinnamic acids, and triterpenes. Although the antioxidant properties of polyphenols are well known, the aim of this study is to assess the antioxidant and protective potentials of Sambucus nigra flowers in the human neuroblastoma (SH-SY5Y) cell line using different in vitro approaches. The antioxidant capacity is first evaluated by the oxygen radical absorbance capacity (ORAC) and the free radical scavenging activity (DPPH) methods. Cell viability is assessed by the crystal violet method; furthermore, the intracellular ROS formation (DCFH-DA method) is determined, together with the effect on the cell antioxidant defenses: reduced glutathione (GSH) and antioxidant enzyme activities (GPx, GR). On the other hand, mTORC1 hyperactivation and autophagy blockage have been associated with an increase in the formation of protein aggregates, this promoting the transference and expansion of neurodegenerative diseases. Then, the ability of Sambucus nigra flowers in the regulation of mTORC1 signaling activity and the reduction in oxidative stress through the activation of autophagy/mitophagy flux is also examined. In this regard, search for different molecules with a potential inhibitory effect on mTORC1 activation could have multiple positive effects either in the molecular pathogenic events and/or in the progression of several diseases including neurodegenerative ones.


Assuntos
Técnicas de Cultura de Células , Degeneração Neural/tratamento farmacológico , Sambucus nigra/química , Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Compostos de Bifenilo/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Flores/química , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Degeneração Neural/patologia , Picratos/química , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
J Pharm Pharmacol ; 73(4): 522-534, 2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33793839

RESUMO

OBJECTIVES: This study aimed to evaluate the effect of duloxetine (10 and 20 mg/kg) against chronic immobilisation stress (CIS)-induced anxiety, depression, cognitive impairment and neurodegeneration in mice. METHODS: CIS, 2 h/10 days (11:00 AM-1:00 PM) was applied after 30 min of pretreatment with saline, duloxetine 10 mg/kg and 20 mg/kg to the respective groups of animals, except the control group. Animals were examined for physiological (body weight, locomotion and grip strength), psychological (memory impairment, anxiety and depression), neurochemical (GABA and glutamate), biochemical (MDA, catalase, glutathione, superoxide dismutase) and histopathological changes. KEY FINDINGS: CIS exposure revealed anxiety-like behaviour, depression-like behaviour, motor in-coordination and learning and memory impairment in mice. Besides, CIS induction decreased the antioxidant enzymes (GSH, SOD and catalase), GABA and the viable neuronal cell count, whereas CIS exposure significantly elevated the MDA, AChE activity and glutamate content in the cortex and hippocampus. Pretreatment with duloxetine10 and 20 mg/kg showed dose-dependent ameliorated effect against the CIS-induced alterations in mice. CONCLUSION: In conclusion, the results of this study demonstrated the protective effect of duloxetine against neuropsychiatric symptoms, memory impairment caused by CIS-induction through inhibition of oxidative stress, AChE activity and glutamate release.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Cloridrato de Duloxetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Catalase/metabolismo , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutationa/metabolismo , Camundongos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo , Psicotrópicos/farmacologia , Superóxido Dismutase/metabolismo , Resultado do Tratamento
4.
Int J Mol Sci ; 22(2)2021 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-33445535

RESUMO

This study aimed to investigate whether the Protaetia brevitarsis seulensis (PB)' water extract (PBWE) ameliorates trimethyltin (TMT)-induced seizures and hippocampal neurodegeneration. To investigate the potential neuroprotective effect of the PBWE in vitro, a lactate dehydrogenase (LDH) assay was conducted in TMT-treated primary cultures of mouse hippocampal neurons. In TMT-treated adult C57BL/6 mice, behavioral and histopathological changes were evaluated by seizure scoring and Fluoro-Jade C staining, respectively. In our in vitro assay, we observed that pretreating mice hippocampal neuron cultures with the PBWE reduced TMT-induced cytotoxicity, as indicated by the decreased LDH release. Furthermore, pretreatment with the PBWE alleviated seizures and hippocampal neurodegeneration in TMT-treated mice. The antioxidant activity of the PBWE increased in a dose-dependent manner; moreover, pretreatment with the PBWE mitigated the TMT-induced Nrf2 stimulation. In addition, six major compounds, including adenine, hypoxanthine, uridine, adenosine, inosine, and benzoic acid, were isolated from the PBWE, and among them, inosine and benzoic acid have been confirmed to have an essential antioxidative activity. In conclusion, the PBWE ameliorated TMT-induced toxicity in hippocampal neurons in both in vitro and in vivo assays, through a potential antioxidative effect. Our findings suggest that the PBWE may have pharmacotherapeutic potential in neurodegenerative diseases such as seizures or epilepsy.


Assuntos
Produtos Biológicos/química , Produtos Biológicos/farmacologia , Besouros/química , Hipocampo/efeitos dos fármacos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Animais , Produtos Biológicos/isolamento & purificação , Biomarcadores , Cromatografia Líquida de Alta Pressão , Besouros/anatomia & histologia , Besouros/classificação , Besouros/genética , Modelos Animais de Doenças , Imunofluorescência , Genes de Insetos , Testes Genéticos , Hipocampo/metabolismo , Hipocampo/patologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/isolamento & purificação , Estresse Oxidativo , Fenótipo , Convulsões/tratamento farmacológico , Convulsões/etiologia , Compostos de Trimetilestanho/efeitos adversos
5.
Muscle Nerve ; 63(3): E18-E21, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33290581

Assuntos
Doenças Cerebelares/induzido quimicamente , Neoplasias Cerebelares/terapia , Inibidores de Checkpoint Imunológico/efeitos adversos , Síndrome Miastênica de Lambert-Eaton/induzido quimicamente , Degeneração Neural/induzido quimicamente , Tumores Neuroendócrinos/terapia , Nivolumabe/efeitos adversos , Amifampridina/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Canais de Cálcio Tipo P , Canais de Cálcio Tipo Q , Doenças Cerebelares/tratamento farmacológico , Doenças Cerebelares/imunologia , Doenças Cerebelares/fisiopatologia , Neoplasias Cerebelares/secundário , Feminino , Glucocorticoides/uso terapêutico , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Síndrome Miastênica de Lambert-Eaton/tratamento farmacológico , Síndrome Miastênica de Lambert-Eaton/imunologia , Síndrome Miastênica de Lambert-Eaton/fisiopatologia , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Linfonodos/diagnóstico por imagem , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Tumores Neuroendócrinos/secundário , Fármacos Neuromusculares/uso terapêutico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Prednisona/uso terapêutico , Radiocirurgia , Radioterapia , Rituximab/uso terapêutico , Carcinoma de Pequenas Células do Pulmão/diagnóstico por imagem , Carcinoma de Pequenas Células do Pulmão/secundário , Carcinoma de Pequenas Células do Pulmão/terapia , Tomografia Computadorizada por Raios X
6.
Molecules ; 25(22)2020 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-33207644

RESUMO

Functional nutrition is a valuable supplementation to dietary therapy. Functional foods are enriched with biologically active substances. Plant polyphenols attract particular attention due to multiple beneficial properties attributed to their high antioxidant and other biological activities. We assessed the effect of grape polyphenols on the life span of C57BL/6 mice and on behavioral and neuroinflammatory alterations in a transgenic mouse model of Parkinson disease (PD) with overexpression of the A53T-mutant human α-synuclein. C57BL/6 mice were given a dietary supplement containing grape polyphenol concentrate (GPC-1.5 mL/kg/day) with drinking water from the age of 6-8 weeks for life. Transgenic PD mice received GPC beginning at the age of 10 weeks for four months. GPC significantly influenced the cumulative proportion of surviving and substantially augmented the average life span in mice. In the transgenic PD model, the grape polyphenol (GP) diet enhanced memory reconsolidation and diminished memory extinction in a passive avoidance test. Behavioral effects of GP treatment were accompanied by a decrease in α-synuclein accumulation in the frontal cortex and a reduction in the expression of neuroinflammatory markers (IBA1 and CD54) in the frontal cortex and hippocampus. Thus, a GP-rich diet is recommended as promising functional nutrition for aging people and patients with neurodegenerative disorders.


Assuntos
Encéfalo/patologia , Inflamação/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Polifenóis/uso terapêutico , Vitis/química , Animais , Comportamento Animal/efeitos dos fármacos , Suplementos Nutricionais , Inflamação/complicações , Inflamação/patologia , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Degeneração Neural/complicações , Doença de Parkinson/complicações , Polifenóis/farmacologia , Aumento de Peso/efeitos dos fármacos , alfa-Sinucleína/metabolismo
7.
Mol Biol Rep ; 47(12): 9733-9738, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33249542

RESUMO

Akebia Saponin D (ASD), a triterpenoid saponin, was shown to have protective effects in certain neuronal cells. The purpose of the present study was to investigate the possibility of ASD to prevent tumor necrosis factor (TNF)-induced axonal loss and the ASD modulation of the biologic process of autophagy in optic nerves. Rats were given intravitreal administration of TNF, simultaneous administration of 2, 20, or 200 pmol ASD and TNF, or ASD alone. LC3-II and p62 expression, which is a marker of autophagic flux, and phosphorylated p38 (p-p38) expression in optic nerves were examined by immunoblot analysis. Morphometric analysis revealed a significant ameliorated effect of ASD against TNF-induced optic nerve damage. p62 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was totally prevented by ASD. The ASD alone injection showed significant reduction of p62 levels compared with the PBS-treated control eyes. LC3-II was significantly increased by ASD treatment in the TNF-injected eyes. p-p38 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was completely prevented by ASD. The protective effects of ASD may be associated with enhanced autophagy activation and inhibition of p-p38.


Assuntos
Glaucoma/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores , Nervo Óptico/efeitos dos fármacos , Saponinas , Animais , Autofagia/efeitos dos fármacos , Axônios/efeitos dos fármacos , Axônios/patologia , Glaucoma/patologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Nervo Óptico/patologia , Ratos , Ratos Wistar , Saponinas/administração & dosagem , Saponinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
J Neuroinflammation ; 17(1): 323, 2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33115477

RESUMO

BACKGROUND: The neuronal ceroid lipofuscinoses (CLN diseases) are fatal lysosomal storage diseases causing neurodegeneration in the CNS. We have previously shown that neuroinflammation comprising innate and adaptive immune reactions drives axonal damage and neuron loss in the CNS of palmitoyl protein thioesterase 1-deficient (Ppt1-/-) mice, a model of the infantile form of the diseases (CLN1). Therefore, we here explore whether pharmacological targeting of innate immune cells modifies disease outcome in CLN1 mice. METHODS: We applied treatment with PLX3397 (150 ppm in the chow), a potent inhibitor of the colony stimulating factor-1 receptor (CSF-1R) to target innate immune cells in CLN1 mice. Experimental long-term treatment was non-invasively monitored by longitudinal optical coherence tomography and rotarod analysis, as well as analysis of visual acuity, myoclonic jerks, and survival. Treatment effects regarding neuroinflammation, neural damage, and neurodegeneration were subsequently analyzed by histology and immunohistochemistry. RESULTS: We show that PLX3397 treatment attenuates neuroinflammation in CLN1 mice by depleting pro-inflammatory microglia/macrophages. This leads to a reduction of T lymphocyte recruitment, an amelioration of axon damage and neuron loss in the retinotectal system, as well as reduced thinning of the inner retina and total brain atrophy. Accordingly, long-term treatment with the inhibitor also ameliorates clinical outcomes in CLN1 mice, such as impaired motor coordination, visual acuity, and myoclonic jerks. However, we detected a sex- and region-biased efficacy of CSF-1R inhibition, with male microglia/macrophages showing higher responsiveness toward depletion, especially in the gray matter of the CNS. This results in a better treatment outcome in male Ppt1-/- mice regarding some histopathological and clinical readouts and reflects heterogeneity of innate immune reactions in the diseased CNS. CONCLUSIONS: Our results demonstrate a detrimental impact of innate immune reactions in the CNS of CLN1 mice. These findings provide insights into CLN pathogenesis and may guide in the design of immunomodulatory treatment strategies.


Assuntos
Aminopiridinas/uso terapêutico , Encéfalo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Pirróis/uso terapêutico , Aminopiridinas/farmacologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Macrófagos/patologia , Masculino , Camundongos , Microglia/patologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/patologia , Lipofuscinoses Ceroides Neuronais/imunologia , Lipofuscinoses Ceroides Neuronais/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Pirróis/farmacologia , Retina/efeitos dos fármacos , Retina/patologia , Fatores Sexuais , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Tomografia de Coerência Óptica
9.
Eur J Pharmacol ; 886: 173412, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32771668

RESUMO

Neurodegenerative diseases (NDs) are characterized by disorders with progressive deterioration of the structure and/or function of neurons. Genetic mutations can lead to many NDs. Nevertheless, neurodegeneration can also take place due to several biological processes. The pathogenesis of several NDs including Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases are associated with oxidative stress (OS). In order to maintain the normal functions of neurons, lower levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important, since their increased levels can cause neuronal cell death. It has been found that OS-mediated neurodegeneration involves a number of events including mitochondrial dysfunction, Ca2+ overload, and excitotoxicity. A growing number of studies are suggesting the benefit of using polyphenols for the treatment of neurodegenerative disorders. Indeed, in order to treat most of the NDs, synthetic drugs are extensively used which are found to exert side effects in the course of the treatment. There is mounting evidence that researchers have identified several naturally-occurring chemical compounds in plants, which are used for the management of NDs. Overall, polyphenolic phytochemicals are safer in nature and have negligible side effects. In this article, we have focused on the potential efficacy of polyphenols such as epigallocatechin-3-gallate, curcumin, resveratrol, quercetin and methylated polyphenols berberine against the most common neurodegenerative disorders.


Assuntos
Degeneração Neural/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Polifenóis/farmacologia , Animais , Humanos , Fármacos Neuroprotetores/uso terapêutico , Polifenóis/uso terapêutico
10.
Metab Brain Dis ; 35(7): 1145-1156, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32653975

RESUMO

Morin hydrate (MH) is the major flavonoid constituent of Morus alba acclaimed to have antioxidant, anti-inflammatory, anti-stress and neuroprotective properties. However, report on the effect of MH on memory performance and the underlying mechanism following chronic stress exposure is lacking. The current study aimed at investigating the neuroprotective effect of MH on chronic unpredictable stress (CUS)-induced memory impairment in mice using the Y maze test. Mice were subjected to unpredicted stress for 14 days, during which MH (5, 10 and 20 mg/kg i.p) or 25 mg/kg Ginseng was administered to them. On the 14th day, 1 h after treatment, learning and memory deficit was evaluated using the Y maze test and thereafter brains were harvested for the estimation of glutathione (GSH), lipid peroxidation product; malondialdehyde (MDA) and nitrite. Levels of inflammatory mediators tumor necrosis factor-alpha (TNF-α) and interleukin1-beta (IL-1ß), inducible nitric oxide synthase (iNOS) and nuclear factor-kappa B (NF-кB) expressions were also determined. The hippocampus was stained with hematoxylin-eosin (H&E) to examine any morphological changes in the neurons. Mice exposed to CUS showed evidence of impaired memory and increase levels of MDA, nitrite, TNF-α and IL-1ß. Furthermore, CUS reduced GSH level, increased the expressions of iNOS and NFкB immune-positive cells and produced loss of neuronal cells in the hippocampus. The MH treatment however improved memory, reduced MDA and nitrite levels, and enhanced brain GSH levels in CUS-mice. Besides, MH reduced brain levels of TNF-α and IL-1ß levels, down regulated the expressions of iNOS and NF-кB and rescue neurons in the hippocampal CA3 region of mice exposed to CUS. The results of the study indicate that MH improved CUS-induced memory impairment, which may be related to its ability to boost antioxidant defense system and suppress neuroinflammatory pathways.


Assuntos
Flavonoides/farmacologia , Hipocampo/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Memória/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Citocinas/metabolismo , Flavonoides/uso terapêutico , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
11.
Neuroreport ; 31(6): 456-465, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32168102

RESUMO

Onjisaponin B (OB) is the main active ingredient of the traditional Chinese medicinal herb polygala, which is effective against neurodegenerative disorders. However, the target of OB is currently unknown. Neuroinflammation and oxidative stress are both risk factors for the pathogenesis and progression of Parkinson's disease (PD). Here, we used a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced subacute mouse model of PD to explore the efficacy and neuroprotective mechanism of OB in PD. Immunohistochemistry was used to mark dopaminergic (DA) neurons and microglia in the substantia nigra pars compact. Administration of OB (20 and 40 mg/kg) prevented the degeneration of DA neurons and improved motor impairment in the rotarod test. Furthermore, OB attenuated microglia over-activation and reduced the secretion of inflammatory factors including tumor necrosis factor-alpha, interleukin-1 beta (IL-1ß) and interleukin-6 (IL-6), as determined by ELISA. Meanwhile, the activities of superoxide dismutase and malondialdehyde were used to measure the level of oxidative stress in brain homogenates and suppression of excessive lipid epoxidation and increased antioxidant enzyme activity were found in OB-treated PD mice. Finally, OB inhibits the expression of the p65 subunit of NF-κB in the nucleus and attenuated expression of the RhoA and ROCK2 proteins in PD mice. Consequently, our results show that OB ameliorates DA neurodegeneration in a MPTP-induced mouse model of PD through anti-oxidant and anti-inflammatory activities mediated via the RhoA/ROCK2 signaling pathway. This finding demonstrates that OB may be a promising drug for DA neuron degeneration, which may provide a new therapeutic agent for future discovery of drugs for PD.See video abstract: http://links.lww.com/WNR/A580.


Assuntos
Diterpenos do Tipo Caurano/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Transtornos Parkinsonianos/tratamento farmacológico , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Diterpenos do Tipo Caurano/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Destreza Motora/efeitos dos fármacos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Teste de Desempenho do Rota-Rod
12.
Sci Rep ; 10(1): 2767, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-32066745

RESUMO

Current therapeutic strategies for Parkinson's disease (PD) aim to delay progression or replace damaged neurons by restoring the original neuronal structures. The poor regenerative capacity of neural tissue highlights the need for the development of cellular environments to model the pathogenesis of PD. In the current work, we have characterised the growth, survival and response to PD mimetics of human SH-SY5Y neuroblastoma and U-87MG glioblastoma cell lines cultured on polyacrylonitrile (PAN) and Jeffamine® doped polyacrylonitrile (PJ) nano-scaffolds. Differentiation induced by a range of agents was evaluated by immunoassays of neural protein biomarkers. PAN and PJ nanofibre scaffolds provided suitable three-dimensional (3D) environment to support the growth, differentiation and network formation of dopaminergic neuron- and astrocyte-like cell populations, respectively. The scaffolds selectively supported the survival and differentiation of both cell populations with prolonged neuronal survival when exposed to PD mimetics in the presence of astrocytes in a co-culture model. Such 3D nanoscaffold-based assays could aid our understanding of the molecular basis of PD mimetic-induced Parkinsonism and the discovery of neuroprotective agents.


Assuntos
Resinas Acrílicas/farmacologia , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Astrócitos/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Humanos , Nanofibras/química , Degeneração Neural/patologia , Fármacos Neuroprotetores/química , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Alicerces Teciduais
13.
Nutr Neurosci ; 23(3): 183-189, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29973113

RESUMO

Objectives: Green tea infusion contains a complex mixture of polyphenolic compounds that were shown to provide health benefits. It was previously demonstrated that (-)-epigallocatechin-3-gallate, one of the major polyphenols present in green tea, has a suppressing effect on various aspects of pathogenesis in models of Huntington's disease (HD), an inherited neurodegenerative disorder. In this study, we aimed to investigate, whether green tea infusion prepared as for human consumption has similar positive effects.Methods: We used a transgenic Drosophila model of HD to study the effects of green tea on mutant Huntingtin induced phenotypes. We tested the effects of green tea infusion on mutant Huntingtin induced neurodegeneration, impaired motor performance, reduced viability and lifespan by pseudopupil assay, climbing assay, eclosion and survival tests, respectively. We used immunoblots to measure Huntingtin protein levels and tested generic health benefits of green tea by longevity analysis.Results: We found that green tea supplementation reduced mutant Huntingtin induced neurodegeneration in Drosophila and positively impacted the longevity of mutant Huntingtin expressing flies. However, green tea did not rescue reduced viability of Drosophila expressing mutant Huntingtin or increased longevity of wild-type fruit flies.Discussion: Our results indicate that green tea consumption might have a modest positive effect on symptoms of HD.


Assuntos
Animais Geneticamente Modificados , Drosophila/genética , Proteína Huntingtina/genética , Degeneração Neural/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Chá , Animais , Feminino , Expressão Gênica , Proteína Huntingtina/análise , Doença de Huntington/tratamento farmacológico , Doença de Huntington/fisiopatologia , Longevidade/efeitos dos fármacos , Masculino
14.
Anat Rec (Hoboken) ; 303(3): 626-633, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30632702

RESUMO

The olfactory epithelium (OE) is the peripheral organ for the sense of smell, housing primary sensory neurons that project axons from the nose to the brain. Due to the presence of a basal stem cell niche, the adult mammalian OE is a dynamic tissue capable of replacing neurons following their loss. Nonetheless, certain conditions, such as blunt head trauma, can result in persistent olfactory loss, thought to be due to shearing of olfactory nerve filaments at the skull base, degeneration, and failures in proper regeneration/reinnervation. The identification of new treatment strategies aimed at preventing degeneration of olfactory neurons is, therefore, needed. In considering potential therapies, we have focused on N-acetylcysteine (NAC), a glutathione substrate shown to be neuroprotective, with a record of safe clinical use. Here, we have tested the use of NAC in an animal model of olfactory degeneration. Administered acutely, we found that NAC (100 mg/kg, twice daily) resulted in a reduction of olfactory neuronal loss from the OE of the nose following surgical ablation of the olfactory bulb. At 1 week postlesion, we identified 54 ± 8.1 mature neurons per 0.5 mm epithelium in NAC-treated animals vs. 28 ± 4.2 in vehicle-treated controls (P = 0.02). Furthermore, in an olfactory cell culture model, we have identified significant alterations in the expression of several genes involved in oxidative stress pathways following NAC exposure. Our results provide evidence supporting the potential therapeutic utility for NAC acutely following head trauma-induced olfactory loss. Anat Rec, 303:626-633, 2020. © 2019 American Association for Anatomy.


Assuntos
Acetilcisteína/uso terapêutico , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Mucosa Olfatória/efeitos dos fármacos , Neurônios Receptores Olfatórios/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Camundongos , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Bulbo Olfatório/lesões , Mucosa Olfatória/patologia , Neurônios Receptores Olfatórios/patologia
15.
J Clin Invest ; 129(10): 4539-4549, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31524631

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disease that lacks therapies to prevent progressive neurodegeneration. Impaired energy metabolism and reduced ATP levels are common features of PD. Previous studies revealed that terazosin (TZ) enhances the activity of phosphoglycerate kinase 1 (PGK1), thereby stimulating glycolysis and increasing cellular ATP levels. Therefore, we asked whether enhancement of PGK1 activity would change the course of PD. In toxin-induced and genetic PD models in mice, rats, flies, and induced pluripotent stem cells, TZ increased brain ATP levels and slowed or prevented neuron loss. The drug increased dopamine levels and partially restored motor function. Because TZ is prescribed clinically, we also interrogated 2 distinct human databases. We found slower disease progression, decreased PD-related complications, and a reduced frequency of PD diagnoses in individuals taking TZ and related drugs. These findings suggest that enhancing PGK1 activity and increasing glycolysis may slow neurodegeneration in PD.


Assuntos
Glicólise/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Prazosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Progressão da Doença , Dopamina/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/tratamento farmacológico , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/metabolismo , Fosfoglicerato Quinase/metabolismo , Prazosina/farmacologia , Ratos
16.
JCI Insight ; 4(17)2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31484833

RESUMO

Chemotherapy-induced peripheral neuropathy is one of the most prevalent dose-limiting toxicities of anticancer therapy. Development of effective therapies to prevent chemotherapy-induced neuropathies could be enabled by a mechanistic understanding of axonal breakdown following exposure to neuropathy-causing agents. Here, we reveal the molecular mechanisms underlying axon degeneration induced by 2 widely used chemotherapeutic agents with distinct mechanisms of action: vincristine and bortezomib. We showed previously that genetic deletion of SARM1 blocks vincristine-induced neuropathy and demonstrate here that it also prevents axon destruction following administration of bortezomib in vitro and in vivo. Using cultured neurons, we found that vincristine and bortezomib converge on a core axon degeneration program consisting of nicotinamide mononucleotide NMNAT2, SARM1, and loss of NAD+ but engage different upstream mechanisms that closely resemble Wallerian degeneration after vincristine and apoptosis after bortezomib. We could inhibit the final common axon destruction pathway by preserving axonal NAD+ levels or expressing a candidate gene therapeutic that inhibits SARM1 in vitro. We suggest that these approaches may lead to therapies for vincristine- and bortezomib-induced neuropathies and possibly other forms of peripheral neuropathy.


Assuntos
Proteínas do Domínio Armadillo/efeitos dos fármacos , Proteínas do Domínio Armadillo/metabolismo , Axônios/metabolismo , Bortezomib/farmacologia , Proteínas do Citoesqueleto/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Degeneração Neural/metabolismo , Vincristina/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose , Proteínas do Domínio Armadillo/genética , Axônios/patologia , Proteínas do Citoesqueleto/genética , Tratamento Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/tratamento farmacológico , Degeneração Neural/genética , Degeneração Neural/patologia , Neurônios/metabolismo , Mononucleotídeo de Nicotinamida , Nicotinamida-Nucleotídeo Adenililtransferase
17.
Cells ; 8(8)2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31426448

RESUMO

: Accumulative evidence indicated that the pathologically accumulated metal ions (iron species and Mn3+) and abnormally up-regulated monoamine oxidase B (MAOB) activity induced oxidation of endogenous dopamine (DA) can lead to mitochondria impairment, lysosome dysfunction, proteasome inhibition, and selective DA neuron vulnerability, which is implicated in the pathogenesis of Parkinson's disease (PD). The DA oxidation can generate deleterious reactive oxygen species (ROS) and highly reactive DA quinones (DAQ) to induce DA-related toxicity, which can be alleviated by DA oxidation suppressors, ROS scavengers, DAQ quenchers, and MAOB inhibitors. On the other hand, the nuclear factor erythroid 2-related factor 2 (Nrf2)-Keap1 and Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) anti-oxidative and proliferative signaling pathways play roles in anti-oxidative cell defense and mitochondria biogenesis, which is implicated in DA neuron protections. Therefore, agents with capabilities to suppress DA-related toxicity including inhibition of DA oxidation, scavenge of ROS, detoxification of DAQ, inhibition of MAOB, and modulations of anti-oxidative signaling pathways can be protective to DA neurons. Accumulative evidence shows that tea or coffee consumptions and smoking are related to deceased PD prevalence with unknown mechanisms. In this study, we investigate the protective capabilities of tea polyphenols and other PD relevant agents to inhibit DA-related toxicity and protect against environmental or genetic factors induced DA neuron degeneration in vitro and in vivo. We find that tea polyphenols can significantly suppress DA-related toxicity to protect DA neurons. The tea polyphenols can protect DA neurons via inhibition of DA oxidation, conjugation with DAQ, scavenge of ROS, inhibition of MAOB, and modulations of Nrf2-Keap1 and PGC-1α anti-oxidative signaling pathways. The tea polyphenols with more phenolic hydroxyl groups and ring structures have stronger protective functions. The protective capabilities of tea polyphenols is further strengthened by evidence that phenolic hydroxyl groups can directly conjugate with DAQ. However, GSH and other sulfhydyl groups containing agents have weaker capabilities to abrogate DA oxidation, detoxify ROS and DAQ and inhibit MAOB; whereas nicotine (NICO) and caffeine (CAF) can only modulate Nrf2-Keap1 and PGC-1α pathways to protect DA neurons weakly. The tea polyphenols are identified to protect against overexpression of mutant A30P α-synuclein (α-syn) induced DA neuron degeneration and PD-like symptoms in transgenic Drosophila. Based on achievements from current studies, the excellent and versatile protective capabilities of tea polyphenols are highlighted, which will contribute and benefit to future anti-PD therapy.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Doença de Parkinson , Polifenóis/farmacologia , Animais , Dopamina/análogos & derivados , Dopamina/toxicidade , Drosophila , Células HEK293 , Humanos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Extratos Vegetais , Chá
18.
J Agric Food Chem ; 67(30): 8348-8360, 2019 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-31304751

RESUMO

We have recently demonstrated that tau hyperphosphorylation causes diabetic synaptic neurodegeneration of retinal ganglion cells (RGCs), which might be the earliest affair during the pathogenesis of diabetic retinopathy (DR). Thus, there is a pressing need to seek therapeutic agents possessing neuroprotective effects against tau hyperphosphorylation in RGCs for arresting the progression of DR. Here, using a well-characterized diabetes model of db/db mouse, we discovered that topical ocular application of 10 mg/kg/day of ginsenoside Rg1 (GRg1), one of the major active ingredients extracted from Panax ginseng and Panax notoginseng, ameliorated hyperphosphorylated tau-triggered RGCs synaptic neurodegeneration in diabetic mice. The neuroprotective effects of GRg1 on diabetic retinae were abrogated when retinal IRS-1 or Akt was suppressed by intravitreal injection with si-IRS-1 or topically coadministered with a specific inhibitor of Akt, respectively. However, selective repression of retinal GSK3ß by intravitreal administration of si-GSK3ß rescued the neuroprotective properties of GRg1 when Akt was inactivated. Therefore, the present study showed for the first time that GRg1 can prevent hyperphosphorylated tau-induced synaptic neurodegeneration of RGCs via activation of IRS-1/Akt/GSK3ß signaling in the early phase of DR. Moreover, our data clarify the potential therapeutic significance of GRg1 for neuroprotective intervention strategies of DR.


Assuntos
Retinopatia Diabética/tratamento farmacológico , Ginsenosídeos/administração & dosagem , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Proteínas tau/metabolismo , Animais , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Proteínas Substratos do Receptor de Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/tratamento farmacológico , Degeneração Neural/genética , Degeneração Neural/metabolismo , Panax notoginseng/química , Fosforilação , Extratos Vegetais/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/genética , Retina/patologia , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/genética
19.
Int Rev Cell Mol Biol ; 343: 129-218, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30712672

RESUMO

Post-synaptic density-95, disks-large and zonula occludens-1 (PDZ) domains are small globular protein-protein interaction domains widely conserved from yeast to humans. They are composed of ∼90 amino acids and form a classical two α-helical/six ß-strand structure. The prototypical ligand is the C-terminus of partner proteins; however, they also bind internal peptide sequences. Recent findings indicate that PDZ domains also bind phosphatidylinositides and cholesterol. Through their ligand interactions, PDZ domain proteins are critical for cellular trafficking and the surface retention of various ion channels. In addition, PDZ proteins are essential for neuronal signaling, memory, and learning. PDZ proteins also contribute to cytoskeletal dynamics by mediating interactions critical for maintaining cell-cell junctions, cell polarity, and cell migration. Given their important biological roles, it is not surprising that their dysfunction can lead to multiple disease states. As such, PDZ domain-containing proteins have emerged as potential targets for the development of small molecular inhibitors as therapeutic agents. Recent data suggest that the critical binding function of PDZ domains in cell signaling is more than just glue, and their binding function can be regulated by phosphorylation or allosterically by other binding partners. These studies also provide a wealth of structural and biophysical data that are beginning to reveal the physical features that endow this small modular domain with a central role in cell signaling.


Assuntos
Neoplasias/tratamento farmacológico , Domínios PDZ/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Humanos , Ligantes , Neoplasias/metabolismo , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo
20.
J Neuroinflammation ; 16(1): 32, 2019 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755225

RESUMO

BACKGROUND: The inflammatory process has been described as a crucial mechanism in the pathophysiology of temporal lobe epilepsy. The anti-inflammatory protein annexin A1 (ANXA1) represents an interesting target in the regulation of neuroinflammation through the inhibition of leukocyte transmigration and the release of proinflammatory mediators. In this study, the role of the ANXA1-derived peptide Ac2-26 in an experimental model of status epilepticus (SE) was evaluated. METHODS: Male Wistar rats were divided into Naive, Sham, SE and SE+Ac2-26 groups, and SE was induced by intrahippocampal injection of pilocarpine. In Sham animals, saline was applied into the hippocampus, and Naive rats were only handled. Three doses of Ac2-26 (1 mg/kg) were administered intraperitoneally (i.p.) after 2, 8 and 14 h of SE induction. Finally, 24 h after the experiment-onset, rats were euthanized for analyses of neuronal lesion and inflammation. RESULTS: Pilocarpine induced generalised SE in all animals, causing neuronal damage, and systemic treatment with Ac2-26 decreased neuronal degeneration and albumin levels in the hippocampus. Also, both SE groups showed an intense influx of microglia, which was corroborated by high levels of ionised calcium binding adaptor molecule 1(Iba-1) and monocyte chemoattractant protein-1 (MCP-1) in the hippocampus. Ac2-26 reduced the astrocyte marker (glial fibrillary acidic protein; GFAP) levels, as well as interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and growth-regulated alpha protein (GRO/KC). These effects of the peptide were associated with the modulation of the levels of formyl peptide receptor 2, a G-protein-coupled receptor that binds to Ac2-26, and the phosphorylated extracellular signal-regulated kinase (ERK) in the hippocampal neurons. CONCLUSIONS: The data suggest a neuroprotective effect of Ac2-26 in the epileptogenic processes through downregulation of inflammatory mediators and neuronal loss.


Assuntos
Anexina A1/uso terapêutico , Citocinas/metabolismo , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Estado Epiléptico/complicações , Estado Epiléptico/tratamento farmacológico , Animais , Anexina A1/metabolismo , Anticonvulsivantes/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Diazepam/uso terapêutico , Modelos Animais de Doenças , Gliose/etiologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Agonistas Muscarínicos/toxicidade , Degeneração Neural/etiologia , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/patologia , Neurônios/efeitos dos fármacos , Pilocarpina/toxicidade , Ratos , Ratos Wistar , Receptores de Lipoxinas/metabolismo , Estado Epiléptico/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA