Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Signal ; 118: 111125, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38432574

RESUMO

BACKGROUND: Parkinson's disease patients on chronic levodopa often suffer from motor complications, which tend to reduce their quality of life. Levodopa-induced dyskinesia (LID) is one of the most prevalent motor complications, often characterized by abnormal involuntary movements, and the pathogenesis of LID is still unclear but recent studies have suggested the involvement of autophagy. METHODS: The onset of LID was mimicked by chronic levodopa treatment in a unilateral 6-hydroxydopamine (6-OHDA) -lesion rat model. Overexpression of ΔFosB in HEK293 cells to mimic the state of ΔFosB accumulation. The modulation of the AMP-activated protein kinase (AMPK)-mediated autophagy pathway using by metformin, AICAR (an AMPK activator), Compound C (an AMPK inhibitor) and chloroquine (an autophagy pathway inhibitor). The severity of LID was assessed by axial, limb, and orofacial (ALO) abnormal involuntary movements (AIMs) score and in vivo electrophysiology. The activity of AMPK pathway as well as autophagy markers and FosB-ΔFosB levels were detected by western blotting. RT-qPCR was performed to detect the transcription level of FosB-ΔFosB. The mechanism of autophagy dysfunction was further explored by immunofluorescence and transmission electron microscopy. RESULTS: In vivo experiments demonstrated that chronic levodopa treatment reduced AMPK phosphorylation, impaired autophagosome-lysosomal fusion and caused FosB-ΔFosB accumulation in the striatum of PD rats. Long-term metformin intervention improved ALO AIMs scores as well as reduced the mean power of high gamma (hγ) oscillations and the proportion of striatal projection neurons unstable in response to dopamine for LID rats. Moreover, the intervention of metformin promoted AMPK phosphorylation, ameliorated the impairment of autophagosome-lysosomal fusion, thus, promoting FosB-ΔFosB degradation to attenuate its accumulation in the striatum of LID rats. However, the aforementioned roles of metformin were reversed by Compound C and chloroquine. The results of in vitro studies demonstrated the ability of metformin and AICAR to attenuate ΔFosB levels by promoting its degradation, while Compound C and chloroquine could block this effect. CONCLUSIONS: In conclusion, our results suggest that long-term metformin treatment could promote ΔFosB degradation and thus attenuate the development of LID through activating the AMPK-mediated autophagy pathway. Overall, our results support the AMPK-mediated autophagy pathway as a novel therapeutic target for LID and also indicate that metformin is a promising therapeutic candidate for LID.


Assuntos
Discinesia Induzida por Medicamentos , Metformina , Humanos , Ratos , Animais , Levodopa/farmacologia , Levodopa/uso terapêutico , Antiparkinsonianos/farmacologia , Proteínas Quinases Ativadas por AMP , Células HEK293 , Qualidade de Vida , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Oxidopamina/uso terapêutico , Autofagia , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Metformina/farmacologia , Modelos Animais de Doenças
2.
Biol Trace Elem Res ; 202(3): 1115-1125, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37386228

RESUMO

Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder characterized by the accumulation of accumulated alpha-synuclein (α-Syn) in substantia nigra. Research has shown that selenium (Se) can protect neural cells through the actions of selenoproteins, including selenoprotein P (SelP) and selenoprotein S (SelS), which participate in endoplasmic reticulum-associated protein degradation (ERAD). In this study, we investigated the potential protective role of Se in a pre-clinical PD rat model.We aimed to evaluate the therapeutic effects of Se administration in the 6-hydroxydopamine (6-OHDA) induced unilateral rat PD model. Male Wistar rats were utilised for unilateral PD animal model which were subjected to stereotaxic surgery and injected with 20 µg 6-OHDA/5 µl 0.2% ascorbate saline. After confirming the model, the rats were intraperitoneally injected with 0.1, 0.2, and 0.3 mg/kg of sodium selenite for 7 days. We then performed behavioral tests, including apomorphine-induced rotation, hanging, and rotarod tests. Following sacrifice, we analysed the substantia nigra area of the brain and serum for protein quantification, element analysis, and gene expression analysis.Our results indicate that the administration of 0.3 mg/kg of Se improved the motor deficiency in hanging, rotarod, and apomorphine-induced rotational tests. While there was no significant improvement in the expression of α-Syn, Se increased the expression of selenoproteins. Additionally, levels of selenoproteins, Se, and α-Syn both brain and serum were re-established by the treatment, suggesting the role of Se on the α-Syn accumulation. Furthermore, Se improved PD-induced biochemical deficits by increasing the levels of SelS and SelP (p<0.005).In conclusion, our findings suggest that Se may have a protective role in PD. 0.3 mg/kg dosage of Se increased the expression of selenoproteins, reduced the accumulation of α-Syn in the brain, and improved PD-induced motor deficits. These results suggest that Se may be a potential therapeutic option for PD treatment.


Assuntos
Doença de Parkinson , Selênio , Ratos , Masculino , Animais , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/uso terapêutico , Parte Compacta da Substância Negra/metabolismo , Selênio/metabolismo , Apomorfina/metabolismo , Apomorfina/uso terapêutico , Oxidopamina/farmacologia , Oxidopamina/metabolismo , Oxidopamina/uso terapêutico , Ratos Wistar , Selenoproteínas/metabolismo , Modelos Animais de Doenças
3.
Chem Biol Drug Des ; 103(1): e14358, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37749299

RESUMO

Parkinson's disease (PD) is the commonest neurodegenerative disorder. It reduces motor and cognitive function in patients. Vinpocetine (Vinp) has the effects of anti-inflammatory and antioxidant, and could improve cognitive function in patients. This study was aimed to investigating the therapeutic effects of Vinp on dyskinesia in a 6-Hydroxydopamine hydrobromide (6-OHDA)-induced PD rat model. We constructed a PD rat model by injecting 6-OHDA, and intervened with Vinp for 7 days. The motor function of the rats was evaluated by an open-field test and rotation test. Besides, H&E staining was applied to observe the changes of dopaminergic neurons in the striatum. The levels of superoxide dismutase (SOD) and malondialdehyde (MDA) in the rat striatum were detected. We assessed the impact of Vinp on α-synuclein (α-Syn) and Wnt/ß-catenin signaling pathway-related molecules by western blot and qRT-PCR. Rats in the PD group showed reduced horizontal movement frequency and number of squares crossed, increased contact time and rotation frequency, and reduced number of dopaminergic neurons accompanied by severe morphological damage. Vinp treatment increased the horizontal movement frequency and number of squares crossed, reduced the contact time, and rotation frequency in PD rats. Also, Vinp downregulated α-Syn protein expression and MDA level, while upregulated SOD activity in the striatum of PD rats. Furthermore, Vinp treatment activated the Wnt/ß-catenin signaling pathway in the striatum of PD rats. In conclusion, Vinp improved the dyskinesia in 6-OHDA-induced PD rats by alleviating oxidative stress, and these effects may be associated with activating the Wnt/ß-catenin signaling pathway.


Assuntos
Discinesias , Doença de Parkinson , Alcaloides de Vinca , Humanos , Ratos , Animais , Doença de Parkinson/tratamento farmacológico , Via de Sinalização Wnt/fisiologia , Oxidopamina/farmacologia , Oxidopamina/uso terapêutico , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Modelos Animais de Doenças
4.
J Neurochem ; 167(5): 633-647, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37916541

RESUMO

L-Dopa, while treating motor symptoms of Parkinson's disease, can lead to debilitating L-Dopa-induced dyskinesias, limiting its use. To investigate the causative relationship between neuro-inflammation and dyskinesias, we assessed if striatal M1 and M2 microglia numbers correlated with dyskinesia severity and whether the anti-inflammatories, minocycline and indomethacin, reverse these numbers and mitigate against dyskinesia. In 6-OHDA lesioned mice, we used stereology to assess numbers of striatal M1 and M2 microglia populations in non-lesioned (naïve) and lesioned mice that either received no L-Dopa (PD), remained non-dyskinetic even after L-Dopa (non-LID) or became dyskinetic after L-Dopa treatment (LID). We also assessed the effect of minocycline/indomethacin treatment on striatal M1 and M2 microglia and its anti-dyskinetic potential via AIMs scoring. We report that L-Dopa treatment leading to LIDs exacerbates activated microglia numbers beyond that associated with the PD state; the severity of LIDs is strongly correlated to the ratio of the striatal M1 to M2 microglial numbers; in non-dyskinetic mice, there is no M1/M2 microglia ratio increase above that seen in PD mice; and reducing M1/M2 microglia ratio using anti-inflammatories is anti-dyskinetic. Parkinson's disease is associated with increased inflammation, but this is insufficient to underpin dyskinesia. Given that L-Dopa-treated non-LID mice show the same ratio of M1/M2 microglia as PD mice that received no L-Dopa, and, given minocycline/indomethacin reduces both the ratio of M1/M2 microglia and dyskinesia severity, our data suggest the increased microglial M1/M2 ratio that occurs following L-Dopa treatment is a contributing cause of dyskinesias.


Assuntos
Discinesias , Doença de Parkinson , Ratos , Camundongos , Animais , Levodopa/efeitos adversos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/etiologia , Microglia , Minociclina/farmacologia , Minociclina/uso terapêutico , Ratos Sprague-Dawley , Corpo Estriado , Discinesias/complicações , Oxidopamina/toxicidade , Oxidopamina/uso terapêutico , Inflamação/complicações , Anti-Inflamatórios/farmacologia , Indometacina/farmacologia , Indometacina/uso terapêutico , Antiparkinsonianos/farmacologia
5.
Pharmacology ; 108(6): 550-564, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37820589

RESUMO

INTRODUCTION: Oxidative stress and inflammation are major factors contributing to the progressive death of dopaminergic neurons in Parkinson's disease (PD). Recent studies have demonstrated that morphine's biosynthetic pathway, coupled with nitric oxide (NO) release, is evolutionarily conserved throughout animals and humans. Moreover, dopamine is a key precursor for morphine biosynthesis. METHOD: The present study evaluated a series of preclinical experiments to evaluate the effects of low-level morphine treatment upon neuro-immune tissues exposed to rotenone and 6-OHDA as models of PD, followed by an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide cell proliferation assay and cell/tissue computer-assisted imaging analyses to assess cell/neuronal viability. RESULTS: Morphine at normal physiological concentrations (i.e., 10-6 M and 10-7 M) provided neuroprotection, as it significantly inhibited rotenone and 6-OHDA dopaminergic insults; thereby, reducing and/or forestalling cell death in invertebrate ganglia and human nerve cells. To ensure that morphine caused this neuroprotective effect, naloxone, a potent opiate receptor antagonist, was employed and the results showed that it blocked morphine's neuroprotective effects. Additionally, co-incubation of NO synthase inhibitor L-NAME also blocked morphine's neuroprotective effects against rotenone and 6-OHDA insults. CONCLUSIONS: Taken together, the present preclinical study showed that while morphine can attenuate lipopolysaccharide-induced inflammation and cell death, both naloxone and L-NAME can abolish this effect. Preincubation of morphine precursors (i.e., L-3,4-dihydroxyphenylalanine, reticuline, and trihexyphenidyl [THP] at physiological concentrations) mimics the observed morphine effect. However, high concentrations of THP, a precursor of the morphine biosynthetic pathway, induced cell death, indicating the physiological importance of morphine biosynthesis in neural tissues. Thus, understanding the morphine biosynthetic pathway coupled with a NO signaling mechanism as a molecular target for neuroprotection against oxidative stress and inflammation in other preclinical models of PD is warranted.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Humanos , Doença de Parkinson/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Oxidopamina/metabolismo , Oxidopamina/farmacologia , Oxidopamina/uso terapêutico , NG-Nitroarginina Metil Éster/farmacologia , Rotenona/farmacologia , Rotenona/metabolismo , Rotenona/uso terapêutico , Estresse Oxidativo , Morfina/farmacologia , Naloxona/farmacologia , Neurônios Dopaminérgicos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Transdução de Sinais
6.
Clinics (Sao Paulo) ; 78: 100243, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37459671

RESUMO

AIMS: Although reduced life expectancy in Parkinson's Disease (PD) patients has been related to severe cardiac arrhythmias due to autonomic dysfunctions, its molecular mechanisms remain unclear. To investigate the role of cardiac ß1-Adrenergic (ß1AR) and A1-Adenosine (A1R) receptors in these dysfunctions, the pharmacological effects of stimulation of cardiac ß1AR (isoproterenol, ISO), in the absence and presence of cardiac ß1AR (atenolol, AT) or A1R (1,3-dipropyl-8-cyclopentyl xanthine, DPCPX) blockade, on the arrhythmias induced by Ischemia/Reperfusion (CIR) in an animal PD model were studied. METHODS: PD was produced by dopaminergic lesions (confirmed by immunohistochemistry analysis) caused by the injection of 6-hydroxydopamine (6-OHDA, 6 µg) in rat striatum. CIR was produced by a surgical interruption for 10 min followed by reestablishment of blood circulation in the descendent left coronary artery. On the incidence of CIR-Induced Ventricular Arrhythmias (VA), Atrioventricular Block (AVB), and Lethality (LET), evaluated by Electrocardiogram (ECG) analysis, the effects of intravenous treatment with ISO, AT and DPCPX (before CIR) were studied. RESULTS: VA, AVB and LET incidences were significantly higher in 6-OHDA (83%, 92%, 100%, respectively) than in control rats (58%, 67% and 67%, respectively). ISO treatment significantly reduced these incidences in 6-OHDA (33%, 33% and 42%, respectively) and control rats (25%, 25%, 33%, respectively), indicating that stimulation of cardiac ß1AR induced cardioprotection. This response was prevented by pretreatment with AT and DPCPX, confirming the involvement of cardiac ß1AR and A1R. CONCLUSION: Pharmacological modulation of cardiac ß1AR and A1R could be a potential therapeutic strategy to reduce severe arrhythmias and increase life expectancy in PD patients.


Assuntos
Adrenérgicos , Doença de Parkinson , Ratos , Animais , Adrenérgicos/uso terapêutico , Oxidopamina/uso terapêutico , Arritmias Cardíacas/etiologia , Receptores Purinérgicos P1/uso terapêutico
7.
Mol Omics ; 19(3): 218-228, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36723117

RESUMO

The most common treatment strategies for Parkinson's disease (PD) aim to slow down the neurodegeneration process or control the symptoms. In this study, using an in vitro PD model we carried out a transcriptome-based drug target prediction strategy. We identified novel drug target candidates by mapping genes upregulated in 6-OHDA-treated cells on a human protein-protein interaction network. Among the predicted targets, we show that AKR1C3 and CEBPB are promising in validating our bioinformatics approach since their known ligands, rutin and quercetin, respectively, act as neuroprotective drugs that effectively decrease cell death, and restore the expression profiles of key genes upregulated in 6-OHDA-treated cells. We also show that these two genes upregulated in our in vitro PD model are downregulated to basal levels upon drug administration. As a further validation of our methodology, we further confirm that the potential target genes identified with our bioinformatics approach are also upregulated in post-mortem transcriptome samples of PD patients from the literature. Therefore, we propose that this methodology predicts novel drug targets AKR1C3 and CEBPB, which are relevant to future clinical applications as potential drug repurposing targets for PD. Our systems-based computational approach to predict candidate drug targets can be employed in identifying novel drug targets in other diseases without a priori assumption.


Assuntos
Doença de Parkinson , Humanos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Transcriptoma/genética , Oxidopamina/farmacologia , Oxidopamina/uso terapêutico , Preparações Farmacêuticas , Mapas de Interação de Proteínas/genética
8.
Turk Neurosurg ; 33(4): 568-575, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36300579

RESUMO

AIM: To investigate the effect of stellate ganglion block (SGB) on nociception in Parkinson's disease (PD) rat models, and to clarify the associated mechanism. MATERIAL AND METHODS: To generate PD nociception rat model 6-hydroxydopamine (6-OHDA) injection method was used. Paw withdrawal threshold (PWT) and paw retraction latency (PWL) was used to reflect mechanical stimulation and thermal stimulation, respectively, at pre-modeling and 1, 2, 3, 4 weeks post modeling. The preventive and therapeutic effects of SGB treatment on nociception were observed in Naive, Vehicle, and 6-OHDA group (model). Levels of IL-1ß, IL-6, and TNF-α in striatum and periaqueductal gray (PAG) were detected with ELISA. RESULTS: 6-OHDA injection induced obvious reduction of bilateral PWT from 2 to 4 weeks post modeling, suggesting that PD nociception rat model was successfully established. Continuous SGB prevention inhibited mechanical hyperalgesia at 2, 3 and 4 weeks post modeling, and significantly reversed mechanical hyperalgesia at 3 and 4 weeks post modeling, compared with those of Saline group (p < 0.05). These results suggest that continuous SGB could effectively prevent and alleviate pain of PD rats. SGB treatment remarkably suppressed levels of inflammatory factors (IL-1 ß, IL-6, and TNF-α) in striatum and PAG of PD rats compared with those of rats in Vehicle group (p < 0.05). CONCLUSION: Continuous SGB effectively inhibited and reversed mechanical hyperalgesia of PD nociception rats through inhibiting inflammatory response in striatum and PAG.


Assuntos
Hiperalgesia , Doença de Parkinson , Ratos , Animais , Hiperalgesia/tratamento farmacológico , Substância Cinzenta Periaquedutal , Limiar da Dor/fisiologia , Citocinas , Fator de Necrose Tumoral alfa , Interleucina-6 , Oxidopamina/toxicidade , Oxidopamina/uso terapêutico , Nociceptividade
9.
Anat Rec (Hoboken) ; 306(4): 820-830, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35476228

RESUMO

PURPOSE: Parkinson's disease (PD) is the most common age-related neurodegenerative disease worldwide. S-adenosyl methionine (SAMe), a methyl donor that plays an important role in DNA methylation, could replenish the cellular antioxidant glutathione (GSH). Herein, we investigated the neuroprotective effects of SAMe in 6-hydroxydopamine (6-OHDA) rat models of PD and elucidated the underlying mechanism. METHODS: PD model rats were developed by injecting 6-OHDA stereotaxically into the striatum. In Phase 1 of the study, we performed the neurobehavioral tests, GSH assay, and histopathology to evaluate the neuroprotective effects of SAMe. The animals were treated with SAMe (150 or 300 mg/kg body weight) orally for 28 days. The positive control group received selegiline (5 mg/kg), whereas the disease control group received normal saline. In Phase 2, we evaluated the striatal dopamine levels and performed DNA methylation assay to uncover the mechanism of action of SAMe. In this phase, a higher dose of SAMe (300 mg/kg) was used. RESULTS: SAMe (300 mg/kg) treatment for 4 weeks significantly attenuated the abnormal circling behavior in PD rats (p < 0.05). Moreover, SAMe at both doses (150 and 300 mg/kg) enhanced the performance of PD rats in the open field test and stepping test (p < 0.05). SAMe treatment significantly increased the GSH levels, and at high dose, SAMe restricted neuronal loss in the striatum of PD-model rats (p < 0.05). Moreover, SAMe treatment led to a significant recovery in the dopamine levels and improved the DNA methylation status in the dopaminergic neurons (p < 0.05) of PD model rats. CONCLUSION: SAMe exhibits antioxidant activity and DNA methylation modulating effects in 6-OHDA model PD rats. Moreover, SAMe prevents neuronal loss in PD rats suggesting that SAMe has therapeutic potential in preventing PD development. The neuroprotective potential of SAMe is greater at high doses.


Assuntos
Doenças Neurodegenerativas , Fármacos Neuroprotetores , Doença de Parkinson , Ratos , Animais , Dopamina , Oxidopamina/toxicidade , Oxidopamina/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Metilação de DNA , Substância Negra/patologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Encéfalo/metabolismo , Estresse Oxidativo , Antioxidantes/farmacologia , Glutationa/metabolismo , Metionina/farmacologia , Metionina/uso terapêutico , Modelos Animais de Doenças
10.
Expert Opin Ther Targets ; 26(10): 837-851, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36469635

RESUMO

INTRODUCTION: Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. PD patients exhibit a classic spectrum of motor symptoms, arising when dopamine neurons in the substantia nigra pars compacta are reduced by 60%. The dopamine precursor L-DOPA represents the most effective therapy for improving PD motor dysfunctions, thus far available. Unfortunately, long-term treatment with L-DOPA is associated with the development of severe side effects, resulting in abnormal involuntary movements termed levodopa-induced dyskinesia (LID). Amantadine is the only drug currently approved for the treatment of LID indicating that LID management is still an unmet need in PD and encouraging the search for novel anti-dyskinetic drugs or the assessment of combined therapies with different molecular targets. AREAS COVERED: This review provides an overview of the main preclinical models used to study LID and of the latest preclinical evidence on experimental and clinically available pharmacological approaches targeting non-dopaminergic systems. EXPERT OPINION: LIDs are supported by complex molecular and neurobiological mechanisms that are still being studied today. This complexity suggests the need of developing personalized pharmacological approach to obtain an effective amelioration of LID condition and improve the quality of life of PD patients.


Assuntos
Discinesia Induzida por Medicamentos , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Doença de Parkinson/tratamento farmacológico , Levodopa/efeitos adversos , Qualidade de Vida , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/etiologia , Oxidopamina/uso terapêutico , Modelos Animais de Doenças , Antiparkinsonianos/efeitos adversos
11.
Cytotherapy ; 24(11): 1105-1120, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35973920

RESUMO

BACKGROUND AIMS: Gingival mesenchymal stem cells (GMSCs) demonstrate high proliferation, trilineage differentiation and immunomodulatory properties. Parkinson disease (PD) is the second most common type of neurodegenerative disease. This study aimed to explore the effect and mechanism of GMSC-based therapy in 6-hydroxydopamine-induced PD rats. METHODS: RNA sequencing and quantitative proteomics technology was used to validate the neuroprotective role of GMSCs therapeutic in 6-Hydroxydopamine -induced PD model in vitro and in vivo. Western blotting, immunofluorescence and real-time quantitative PCR verified the molecular mechanism of GMSCs treatment. RESULTS: Intravenous injection of GMSCs improved rotation and forelimb misalignment behavior, enhanced the anti-apoptotic B-cell lymphoma 2/B-cell lymphoma 2-associated X axis, protected tyrosine hydroxylase neurons, decreased the activation of astrocytes and reduced the astrocyte marker glial fibrillary acidic protein and microglia marker ionized calcium-binding adaptor molecule 1 in the substantia nigra and striatum of PD rats. The authors found that GMSCs upregulated nerve regeneration-related molecules and inhibited metabolic disorders and the activation of signal transducer and activator of transcription 3. GMSCs showed a strong ability to protect neurons and reduce mitochondrial membrane potential damage and reactive oxygen species accumulation. The safety of GMSC transplantation was confirmed by the lack of tumor formation following subcutaneous transplantation into nude mice for up to 8 weeks. CONCLUSIONS: The authors' research helps to explain the mechanism of GMSC-based therapeutic strategies and promote potential clinical application in Parkinson disease.


Assuntos
Células-Tronco Mesenquimais , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Cálcio/metabolismo , Gengiva , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Camundongos Nus , Neurônios/metabolismo , Oxidopamina/metabolismo , Oxidopamina/farmacologia , Oxidopamina/uso terapêutico , Doença de Parkinson/terapia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/farmacologia , Fator de Transcrição STAT3/uso terapêutico , Tirosina 3-Mono-Oxigenase/metabolismo , Tirosina 3-Mono-Oxigenase/farmacologia , Tirosina 3-Mono-Oxigenase/uso terapêutico
12.
Mol Ther ; 30(10): 3176-3192, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-35689381

RESUMO

Parkinson's disease is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra with no effective cure available. MicroRNA-124 has been regarded as a promising therapeutic entity for Parkinson's disease due to its pro-neurogenic and neuroprotective roles. However, its efficient delivery to the brain remains challenging. Here, we used umbilical cord blood mononuclear cell-derived extracellular vesicles as a biological vehicle to deliver microRNA (miR)-124-3p and evaluate its therapeutic effects in a mouse model of Parkinson's disease. In vitro, miR-124-3p-loaded small extracellular vesicles induced neuronal differentiation in subventricular zone neural stem cell cultures and protected N27 dopaminergic cells against 6-hydroxydopamine-induced toxicity. In vivo, intracerebroventricularly administered small extracellular vesicles were detected in the subventricular zone lining the lateral ventricles and in the striatum and substantia nigra, the brain regions most affected by the disease. Most importantly, although miR-124-3p-loaded small extracellular vesicles did not increase the number of new neurons in the 6-hydroxydopamine-lesioned striatum, the formulation protected dopaminergic neurons in the substantia nigra and striatal fibers, which fully counteracted motor behavior symptoms. Our findings reveal a novel promising therapeutic application of small extracellular vesicles as delivery agents for miR-124-3p in the context of Parkinson's disease.


Assuntos
Vesículas Extracelulares , MicroRNAs , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos , Camundongos , MicroRNAs/farmacologia , Oxidopamina/farmacologia , Oxidopamina/uso terapêutico , Doença de Parkinson/genética , Doença de Parkinson/terapia , Substância Negra
13.
J Parkinsons Dis ; 12(6): 1881-1896, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35466951

RESUMO

BACKGROUND: First-in-human studies to test the efficacy and safety of human embryonic stem cells (hESC)-derived dopaminergic cells in the treatment of Parkinson's disease (PD) are imminent. Pre-clinical studies using hESC-derived dopamine neuron transplants in rat models have indicated that the benefits parallel those shown with fetal tissue but have thus far failed to consider how ongoing L-DOPA administration might impact on the graft. OBJECTIVE: To determine whether L-DOPA impacts on survival and functional recovery following grafting of hESC-derived dopaminergic neurons. METHODS: Unilateral 6-OHDA lesioned rats were administered with either saline or L-DOPA prior to, and for 18 weeks following surgical implantation of dopaminergic neural progenitors derived from RC17 hESCs according to two distinct protocols in independent laboratories. RESULTS: Grafts from both protocols elicited reduction in amphetamine-induced rotations. Reduced L-DOPA-induced dyskinesia preceded the improvement in amphetamine-induced rotations. Furthermore, L-DOPA had no effect on overall survival (HuNu) or dopaminergic neuron content of the graft (TH positive cells) but did lead to an increase in the number of GIRK2 positive neurons. CONCLUSION: Critically, we found that L-DOPA was not detrimental to graft function, potentially enhancing graft maturation and promoting an A9 phenotype. Early improvement of L-DOPA-induced dyskinesia suggests that grafts may support the handling of exogenously supplied dopamine earlier than improvements in amphetamine-induced behaviours indicate. Given that one of the protocols will be employed in the production of cells for the European STEM-PD clinical trial, this is vital information for the management of patients and achieving optimal outcomes following transplantation of hESC-derived grafts for PD.


Assuntos
Discinesia Induzida por Medicamentos , Células-Tronco Embrionárias Humanas , Doença de Parkinson , Anfetaminas/uso terapêutico , Animais , Antiparkinsonianos/uso terapêutico , Modelos Animais de Doenças , Dopamina , Discinesia Induzida por Medicamentos/tratamento farmacológico , Humanos , Levodopa/uso terapêutico , Oxidopamina/uso terapêutico , Oxidopamina/toxicidade , Doença de Parkinson/tratamento farmacológico , Ratos , Ratos Sprague-Dawley
14.
Ultrasound Med Biol ; 48(2): 265-274, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34740497

RESUMO

Low-intensity pulsed ultrasound (LIPUS) is a promising non-invasive neuromodulation tool for deep brain stimulation. Here, we investigated the impact of LIPUS, including neuroprotective effects, on the pathology of Parkinson's disease (PD) in an animal model. Sprague-Dawley rats were injected with 6-hydroxydopamine (6-OHDA) at two sites in the right striatum. LIPUS (1 MHz, 5% duty cycle, 1-Hz pulse repetition frequency, 15 min/d) stimulation was then applied to some of the rats (the 6-OHDA + LIPUS group) beginning 2 wk after the 6-OHDA administration, while the remaining rats (the 6-OHDA group) received no LIPUS stimulation. The 6-OHDA-induced inflammatory responses and expressions of neurotrophic factors were quantified with immunofluorescence activity. The safety of LIPUS was assessed using hematoxylin and eosin and Nissl staining. LIPUS treatment significantly inhibited 6-OHDA-induced glial activation and the phosphorylation of nuclear factor-κB p65 in the substantia nigra pars compacta. Further study revealed that LIPUS effectively preserved the levels of neurotrophic factors, dopamine transporter and tight junction proteins of the blood-brain barrier in the 6-OHDA + LIPUS group compared with the 6-OHDA group. These results indicate that LIPUS acts via multiple neuroprotective mechanisms in the PD rat model and suggest that LIPUS can be viewed as a potential treatment for PD.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Fármacos Neuroprotetores/uso terapêutico , Oxidopamina/uso terapêutico , Doença de Parkinson/terapia , Ratos , Ratos Sprague-Dawley , Substância Negra
15.
J Pain ; 23(3): 472-486, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34699985

RESUMO

Chronic pain frequently develops after limb injuries, and its pathogenesis is poorly understood. We explored the hypothesis that the autonomic nervous system regulates adaptive immune system activation and nociceptive sensitization in a mouse model of chronic post-traumatic pain with features of complex regional pain syndrome (CRPS). In studies sympathetic signaling was reduced using 6-hydroxydopamine (6-OHDA) or lofexidine, while parasympathetic signaling was augmented by nicotine administration. Hindpaw allodynia, unweighting, skin temperature, and edema were measured at 3 and 7 weeks after fracture. Hypertrophy of regional lymph nodes and IgM deposition in the skin of injured limbs were followed as indices of adaptive immune system activation. Passive transfer of serum from fracture mice to recipient B cell deficient (muMT) mice was used to assess the formation of pain-related autoantibodies. We observed that 6-OHDA or lofexidine reduced fracture-induced hindpaw nociceptive sensitization and unweighting. Nicotine had similar effects. These treatments also prevented IgM deposition, hypertrophy of popliteal lymph nodes, and the development of pronociceptive serum transfer effects. We conclude that inhibiting sympathetic or augmenting parasympathetic signaling inhibits pro-nociceptive immunological changes accompanying limb fracture. These translational results support the use of similar approaches in trials potentially alleviating persistent post-traumatic pain and, possibly, CRPS. PERSPECTIVE: Selective treatments aimed at autonomic nervous system modulation reduce fracture-related nociceptive and functional sequelae. The same treatment strategies limit pain-supporting immune system activation and the production of pro-nociceptive antibodies. Thus, the therapeutic regulation of autonomic activity after limb injury may reduce the incidence of chronic pain.


Assuntos
Dor Crônica , Síndromes da Dor Regional Complexa , Fraturas Ósseas , Animais , Sistema Nervoso Autônomo , Dor Crônica/complicações , Modelos Animais de Doenças , Fraturas Ósseas/complicações , Hipertrofia/complicações , Imunoglobulina M/uso terapêutico , Camundongos , Nicotina , Nociceptividade/fisiologia , Oxidopamina/uso terapêutico , Oxidopamina/toxicidade
16.
Oxid Med Cell Longev ; 2021: 2492286, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34880964

RESUMO

In this study, we investigated whether chemical 6-hydroxydopamine (6-OHDA) stimuli caused cardiac sympathetic denervation (SD), and we analyzed gene expression profiles to determine the changes in the lncRNA/circRNAs-miRNA-mRNA network in the affected spinal cord segments to identify putative target genes and molecular pathways in rats with myocardial ischemia-reperfusion injury (MIRI). Our results showed that cardiac sympathetic denervation induced by 6-OHDA alleviated MIRI. Compared with the ischemia reperfusion (IR, MIRI model) group, there were 148 upregulated and 51 downregulated mRNAs, 165 upregulated and 168 downregulated lncRNAs, 70 upregulated and 52 downregulated circRNAs, and 12 upregulated and 11 downregulated miRNAs in the upper thoracic spinal cord of the SD-IR group. Furthermore, we found that the differential genes related to cellular components were mainly enriched in extracellular and cortical cytoskeleton, and molecular functions were mainly enriched in chemokine activity. Pathway analysis showed that the differentially expressed genes were mainly related to the interaction of cytokines and cytokine receptors, sodium ion reabsorption, cysteine and methionine metabolism, mucoglycan biosynthesis, cGMP-PKG signaling pathway, and MAPK signaling pathway. In conclusion, the lncRNA/circRNAs-miRNA-mRNA networks in the upper thoracic spinal cord play an important role in the preventive effect of cardiac sympathetic denervation induced by 6-OHDA on MIRI, which offers new insights into the pathogenesis of MIRI and provides new targets for MIRI.


Assuntos
Redes Reguladoras de Genes/genética , Traumatismo por Reperfusão Miocárdica/patologia , Oxidopamina/farmacologia , Medula Espinal/metabolismo , Animais , Quimiocinas/metabolismo , Vasos Coronários/cirurgia , Regulação para Baixo/efeitos dos fármacos , Ontologia Genética , Masculino , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oxidopamina/uso terapêutico , Mapas de Interação de Proteínas/genética , RNA Circular/metabolismo , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Simpatectomia , Regulação para Cima/efeitos dos fármacos
17.
Cell Transplant ; 30: 9636897211052300, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34743572

RESUMO

Amphetamine-type stimulants have become important and popular abused drugs worldwide. Methamphetamine (Meth) sensitization, characterized by a progressive increase in behavioral responses after repeated administration, has been reported in rodents and patients. This behavioral effect has been used as a laboratory model to study drug addiction and schizophrenia. The mesolimbic dopaminergic pathway plays a significant role in the development of Meth behavioral sensitization. Previous studies have reported that the ablation of nucleus accumbens (NAc) by electrolytic or thermal lesioning attenuates addictive behavior to opioids in animals. However, these studies were only conducted in opioid addictive rodents. Furthermore, these ablation procedures also damaged the non-dopaminergic neurons and fibers passing through the NAc. The purpose of this study was to examine the therapeutic effect of NAc lesioning by a selective dopaminergic toxin in Meth-sensitized animals. Adult mice received repeated administration of Meth for 7 days. Open-field locomotor activity and stereotype behavior were significantly increased after Meth treatment, suggesting behavior sensitization. A partial lesion of dopaminergic terminals was made through stereotaxic administration of dopaminergic toxin 6-hydroxydopamine (6-OHDA) to the NAc in the Meth -sensitized mice. Meth behavioral sensitization was significantly antagonized after the lesioning. Brain tissue was collected for qRT-PCR analysis. Repeated administration of Meth increased the expression of tyrosine hydroxylase (TH), BDNF, and Shati, a marker for Meth sensitization, in the NAc. Treatment with 6-OHDA significantly antagonized the upregulation of TH and Shati. Taken together, these data suggest that local administration of 6-OHDA mitigated Meth sensitization in chronic Meth-treated animals. Our data support a new surgical treatment strategy for Meth abuse.


Assuntos
Estimulantes do Sistema Nervoso Central/administração & dosagem , Dopamina/metabolismo , Metanfetamina/administração & dosagem , Núcleo Accumbens/fisiopatologia , Oxidopamina/uso terapêutico , Animais , Humanos , Masculino , Camundongos , Oxidopamina/farmacologia
18.
Oxid Med Cell Longev ; 2020: 9260748, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32377313

RESUMO

Parkinson's disease (PD) patients can benefit from antioxidant supplementation, and new efficient antioxidants are needed. The aim of this study was to evaluate the protective effect of selected nitroxide-containing redox nanoparticles (NRNPs) in a cellular model of PD. Antioxidant properties of NRNPs were studied in cell-free systems by protection of dihydrorhodamine 123 against oxidation by 3-morpholino-sydnonimine and protection of fluorescein against bleaching by 2,2-azobis(2-amidinopropane) hydrochloride and sodium hypochlorite. Model blood-brain barrier penetration was studied using hCMEC/D3 cells. Human neuroblastoma SH-SY5Y cells, exposed to 6-hydroxydopamine (6-OHDA), were used as an in vitro model of PD. Cells were preexposed to NRNPs or free nitroxides (TEMPO or 4-amino-TEMPO) for 2 h and treated with 6-OHDA for 1 h and 24 h. The reactive oxygen species (ROS) level was estimated with dihydroethidine 123 and Fluorimetric Mitochondrial Superoxide Activity Assay Kit. Glutathione level (GSH) was measured with ortho-phtalaldehyde, ATP by luminometry, changes in mitochondrial membrane potential with JC-1, and mitochondrial mass with 10-Nonyl-Acridine Orange. NRNP1, TEMPO, and 4-amino-TEMPO (25-150 µM) protected SH-SY5Y cells from 6-OHDA-induced viability loss; the protection was much higher for NRNP1 than for free nitroxides. NRNP1 were better antioxidants in vitro and permeated better the model BBB than free nitroxides. Exposure to 6-OHDA decreased the GSH level after 1 h and increased it considerably after 24 h (apparently a compensatory overresponse); NRNPs and free nitroxides prevented this increase. NRNP1 and free nitroxides prevented the decrease in ATP level after 1 h and increased it after 24 h. 6-OHDA increased the intracellular ROS level and mitochondrial superoxide level. Studied antioxidants mostly decreased ROS and superoxide levels. 6-OHDA decreased the mitochondrial potential and mitochondrial mass; both effects were prevented by NRNP1 and nitroxides. These results suggest that the mitochondria are the main site of 6-OHDA-induced cellular damage and demonstrate a protective effect of NRNP1 in a cellular model of PD.


Assuntos
Nanopartículas/metabolismo , Neuroblastoma/tratamento farmacológico , Oxidopamina/uso terapêutico , Linhagem Celular Tumoral , Humanos , Oxirredução , Oxidopamina/farmacologia , Transdução de Sinais
19.
Stem Cells Transl Med ; 8(5): 490-499, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30706999

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder affecting millions of people worldwide. At present, there is no effective cure for PD; treatments are symptomatic and do not halt progression of neurodegeneration. Extracellular vesicles (EVs) can cross the blood-brain barrier and represent promising alternative to the classical treatment strategies. In the present study, we examined therapeutic effects of intranasal administration of EVs derived from human exfoliated deciduous teeth stem cells (SHEDs) on unilateral 6-hydroxydopamine (6-OHDA) medial forebrain bundle (MFB) rat model of PD. CatWalk gait tests revealed that EVs effectively suppressed 6-OHDA-induced gait impairments. All tested gait parameters (stand, stride length, step cycle, and duty cycle) were significantly improved in EV-treated animals when compared with 6-OHDA-lesion group rats. Furthermore, EVs slowed down numbers of 6-OHDA-induced contralateral rotations in apomorphine test. Improvements in motor function correlated with normalization of tyrosine hydroxylase expression in the striatum and substantia nigra. In conclusion, we demonstrated, for the first time, the therapeutic efficacy of intranasal administration of EVs derived from SHEDs in a rat model of PD induced by 6-OHDA intra-MFB lesion. Our findings could be potentially exploited for the development of new treatment strategies against PD.


Assuntos
Administração Intranasal/métodos , Vesículas Extracelulares/metabolismo , Microscopia Eletrônica de Transmissão/métodos , Oxidopamina/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Células-Tronco/metabolismo , Dente/fisiopatologia , Tirosina 3-Mono-Oxigenase/metabolismo , Idoso , Animais , Corpo Estriado/patologia , Modelos Animais de Doenças , Humanos , Masculino , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Ratos , Ratos Wistar , Substância Negra/patologia
20.
Bratisl Lek Listy ; 119(1): 22-27, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29405726

RESUMO

OBJECTIVE: The present study aimed to explore the role and the underlying mechanism of miR-221 in Parkinson's Disease. MATERIALS AND METHODS: To perform our investigation, a PD cell model was created by using 6-OHDA. Cell viability and proliferation assays, and flow cytometry analysis were performed to detect cell viability and apoptosis. The qRT-PCR and western blotting were used for gene and protein level detection. RESULTS: We found that the expression of miRNA-221 is significantly lower in 6-OHDA treated PC12 pheochromocytoma cells compared to the normal cells. The results of further analysis indicated that miR-221 mimic significantly promoted the cell viability and proliferation of PC12 cells treated with 6-OHDA. MiR-221 mimic significantly inhibited 6-OHDA-treated PC12 cells from apoptosis. These effects were eliminated by PTEN over-expression. We also revealed that PTEN was a direct target gene of miR-221. Moreover, we found miR-221 mimic significantly promoted the phosphorylation of AKT in PC12 cells treated with 6-OHDA, and over-expression of PTEN could eliminate this effect. CONCLUSIONS: MiR-221 plays a protective role in Parkinson's Disease via regulating PC12 cell viability and apoptosis by targeting PTEN. Therefore, miR-221 may serve as a potential therapeutic target for Parkinson's disease treatment (Fig. 3, Ref. 27).


Assuntos
MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Doença de Parkinson/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , MicroRNAs/metabolismo , Oxidopamina/uso terapêutico , Células PC12/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...