Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biol Pharm Bull ; 47(10): 1637-1643, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39370268

RESUMO

Depressive disorder is the most common mental disorder with significant economic burden and limited treatments. Traditional Chinese medicine monomer has emerged as a promising non-pharmacological treatment for reducing depressive symptoms. The aim of this study was to investigate the antidepressant-like effects of asperuloside (ASP) and its mechanism. The depression-like behaviors of chronic unpredictable mild stress (CUMS)-exposed rats were evaluated by behavioral tests. At the same time, the behaviors of rats treated with different concentrations of ASP (10, 20, 40 mg/kg) were also evaluated. RNA sequencing was performed to screen for dysregulated genes following ASP treatment. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was performed to state the enriched pathways. Protein expression was detected by Western blotting. With the increase of ASP concentration (over 20 mg/kg), the depression-like behaviors of the rats were alleviated, which was manifested as the increase of the number of entries in the central zone, decrease of immobility time, and the increase of swimming time, sucrose preference, and body weight. ASP activated the Wnt3α/glycogen synthase kinase 3ß (GSK-3ß)/ß-catenin signaling pathway in vivo. Knockdown of ß-catenin reversed the effects of ASP on regulating depression-like behaviors. ASP alleviates depression-like behaviors by activating the Wnt3α/GSK-3ß/ß-catenin signaling pathway, indicating that ASP may be a potential therapeutic drug for treatment of depression.


Assuntos
Antidepressivos , Depressão , Glicogênio Sintase Quinase 3 beta , Ratos Sprague-Dawley , Animais , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Proteína Wnt3/metabolismo , Proteína Wnt3/genética , beta Catenina/metabolismo , Comportamento Animal/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/tratamento farmacológico , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos
2.
Int J Biol Markers ; : 3936155241281076, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39233606

RESUMO

BACKGROUND: Exploring effect biomarkers that monitor tumor progression and predict the prognosis could benefit the clinical management of bladder cancer and improve the postoperative life of patients. This study aimed to estimate the function of long non-coding (lnc)RNA RHPN1-AS1 (RHPN1-AS1) in bladder cancer and the potential molecular mechanism. METHODS: The expression of RHPN1-AS1 was evaluated in bladder cancer tissues from 115 patients and cells by polymerase chain reaction. The clinical significance of RHPN1-AS1 was assessed and its effect was also estimated in cell proliferation, migration, and invasion. The underlying molecular mechanism was explored by the dual-luciferase reporter assay. RESULTS: The expression of RHPN1-AS1 was 2.91-fold elevated in bladder cancer, which showed a close correlation with advanced tumor node metastasis stage (P = 0.013) and the presence of lymph node metastasis (P = 0.018). RHPN1-AS1 also served as a poor prognostic indicator (hazard ratio = 2.563) for bladder cancer. The knockdown of RHPN1-AS1 significantly suppressed the proliferation and metastasis ability of bladder cancer cells. Moreover, miR-485-5p was found to mediate the function of RHPN1-AS1 in bladder cancer, which was considered the underlying regulatory mechanism. CONCLUSIONS: RHPN1-AS1 serves as a prognostic biomarker and tumor promoter in bladder cancer via modulating miR-485-5p, which might be a reliable target of bladder cancer therapy.

3.
Stem Cell Res Ther ; 15(1): 299, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39267160

RESUMO

BACKGROUND: The established association between Alzheimer's disease (AD) and compromised neural regeneration is well-documented. In addition to the mitigation of apoptosis in neural stem cells (NSCs), the induction of neurogenesis has been proposed as a promising therapeutic strategy for AD. Our previous research has demonstrated the effective inhibition of NSC injury induced by microglial activation through the repression of oxidative stress and mitochondrial dysfunction by Sirtuin 3 (SIRT3). Nonetheless, the precise role of SIRT3 in neurogenesis remains incompletely understood. METHODS: In vivo, SIRT3 overexpression adenovirus was firstly injected by brain stereotaxic localization to affect the hippocampal SIRT3 expression in APP/PS1 mice, and then behavioral experiments were performed to investigate the cognitive improvement of SIRT3 in APP/PS1 mice, as well as neurogenic changes in hippocampal region by immunohistochemistry and immunofluorescence. In vitro, under the transwell co-culture condition of microglia and neural stem cells, the mechanism of SIRT3 improving neurogenesis of neural stem cells through DVL/GSK3/ISL1 axis was investigated by immunoblotting, immunofluorescence and other experimental methods. RESULTS: Our findings indicate that the overexpression of SIRT3 in APP/PS1 mice led to enhanced cognitive function and increased neurogenesis. Additionally, SIRT3 was observed to promote the differentiation of NSCs into neurons during retinoic acid (RA)-induced NSC differentiation in vitro, suggesting a potential role in neurogenesis. Furthermore, we observed the activation of the Wnt/ß-catenin signaling pathway during this process, with Glycogen Synthase Kinase-3a (GSK3a) primarily governing NSC proliferation and GSK3ß predominantly regulating NSC differentiation. Moreover, the outcomes of our study demonstrate that SIRT3 exerts a protective effect against microglia-induced apoptosis in neural stem cells through its interaction with DVLs. CONCLUSIONS: Our results show that SIRT3 overexpressing APP/PS1 mice have improved cognition and neurogenesis, as well as improved neurogenesis of NSC in microglia and NSC transwell co-culture conditions through the DVL/GSK3/ISL1 axis.


Assuntos
Doença de Alzheimer , Células-Tronco Neurais , Neurogênese , Transdução de Sinais , Sirtuína 3 , Animais , Sirtuína 3/metabolismo , Sirtuína 3/genética , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Doença de Alzheimer/genética , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Desgrenhadas/metabolismo , Proteínas Desgrenhadas/genética , Camundongos Transgênicos , Microglia/metabolismo , Diferenciação Celular , Hipocampo/metabolismo
4.
BMC Urol ; 24(1): 144, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38997703

RESUMO

BACKGROUND: Prostate cancer, characterized by its insidious onset and short overall survival, and has seen a rise in incidence over recent decades. This study aims to investigate the expression and molecular mechanism of lncRNA PTCSC3 (PTCSC3) in prostate cancer in order to develop new prognostic and therapeutic biomarkers. METHODS: The level of PTCSC3 in serum and cell samples of prostate cancer was quantitatively measured using RT-qPCR assays. The correlation between the variation in PTCSC3 levels and clinical indicators of patients was evaluated. The survival status of the prostate cancer patients included in the study was evaluated using Kaplan-Meier curve and multivariable Cox analysis. The impact of PTCSC3 overexpression on cell growth and activity was revealed by CCK-8 and Transwell assays. The targeting relationship between PTCSC3 and miR-182-5p was determined by bioinformatics prediction and luciferase activity. RESULTS: PTCSC3 was found to be downregulated in prostate cancer, and its low levels were associated with short overall survival in patients. It influenced the progression of prostate cancer by targeting miR-182-5p. Increasing PTCSC3 levels suppressed the proliferation, migration and invasion levels of cells, and miR-182-5p mimic counteracted PTCSC3's effects on cells. CONCLUSIONS: As a potential prognostic biological factor for prostate cancer, PTCSC3 may regulate the progression of prostate cancer by sponging miR-182-5p and affect the prognosis of patients.


Assuntos
MicroRNAs , Neoplasias da Próstata , RNA Longo não Codificante , Masculino , Humanos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , MicroRNAs/genética , MicroRNAs/sangue , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/sangue , Pessoa de Meia-Idade , Idoso , Taxa de Sobrevida , Regulação para Baixo
5.
Discov Oncol ; 15(1): 113, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38605206

RESUMO

PURPOSE: The purpose of this study was to investigate the role of lncRNA DSCAM-AS1 in prostate cancer to find new therapeutic targets and promote the research progress of prostate cancer. METHODS: RT-qPCR was used to detect DSCAM-AS1 expression in prostate cancer tissues, normal tissues, human normal prostate epithelial cells (RWPE), and four prostate cancer cell lines. The clinical and prognostic role of DSCAM-AS1 was evaluated by the Kaplan-Meier curve and chi-square test. Secondly, a dual luciferase reporter gene assay was used to study the regulatory mechanism between miR-338-3p and DSCAM-AS1. Finally, the roles of DSCAM-AS1 and miR-338-3p in prostate cancer cell proliferation and metastasis were explored by CCK-8 and Transwell assays. RESULTS: It was found that DSCAM-AS1 upregulation could serve as a warning of deterioration and poor prognosis in prostate cancer patients, and that knockdown of DSCAM-AS1 expression inhibited the progression of prostate cancer cells. In addition, miR-338-3p, a target of DSCAM-AS1, was found to be down-regulated in prostate cancer cells and miR-338-3p knockdown could reverse the inhibitory effect of DSCAM-AS1 silencing on prostate cancer. CONCLUSION: DSCAM-AS1 is up-regulated in prostate cancer and regulates the progression of prostate cancer cells by targeting miR-338-3p.

6.
Medicine (Baltimore) ; 102(34): e34687, 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37653795

RESUMO

BACKGROUND: Paroxetine therapy has been used for treatment of patients with depression and Parkinson's disease (dPD) in many clinical studies, but, the effects of paroxetine in dPD patients are not completely understood. The aim of this study was to systematically evaluate the effects of paroxetine therapy on depressive symptom and motor function in the treatment of dPD, in order to confer a reference for clinical practice. METHODS: Randomized controlled trials (RCTs) of paroxetine for dPD published up to October, 2022 were retrieved. Standardised mean difference (SMD), odds ratio (OR), and 95% confidence interval (CI) were calculated and heterogeneity was measured with the I2 test. The outcomes of interest were as follows: the efficacy, Hamilton depression rating scale score, unified Parkinson's disease rating scale score, Hamilton anxiety rating scale score or adverse events. RESULTS: Thirty-four RCTs with 2819 participants were included. Compared with control group, the pooled effects of paroxetine therapy on depression were (22 trials; OR 3.62, 95% CI 2.63 to 4.98, P < .00001) for antidepressant response (25 trials; SMD -2.14, 95% CI -2.73 to -1.56, P < .00001) for Hamilton depression rating scale score, the pooled effects of paroxetine therapy on motor function were (10 trials; OR 4.63, 95% CI 3.15 to 6.79, P < .00001) for anti-PD efficacy (18 trials; SMD -2.02, 95% CI -2.48 to -1.55, P < .00001) for total unified Parkinson's disease rating scale score. The Hamilton anxiety rating scale score showed significant decrease in the paroxetine treatment group compared to control group (10 trials; SMD -1.93, 95% CI -2.65 to -1.22, P < .00001). In addition, paroxetine therapy reduced the number of any adverse events obviously in dPD patients (twenty trials; OR 0.42, 95% CI 0.31 to 0.57, P < .00001). CONCLUSIONS: Paroxetine therapy has clinical benefits for improvement of depressive symptom and motor function in dPD patients, moreover, it is of high drug safety. Further well-designed, multi-center RCTs needed to identify these findings.


Assuntos
Doença de Parkinson , Paroxetina , Humanos , Paroxetina/uso terapêutico , Doença de Parkinson/complicações , Doença de Parkinson/tratamento farmacológico , Depressão/tratamento farmacológico , Depressão/etiologia , Grupos Controle , Testes de Estado Mental e Demência
7.
Exp Ther Med ; 24(5): 662, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36168411

RESUMO

The mitochondrial protein sirtuin 3 (SIRT3) can counteract cell damage caused by oxidative stress and inflammation, and contribute to cell survival primarily by improving mitochondrial function. However, the effects of SIRT3 in dopaminergic neuronal cells (DACs) remain unclear. In our previous studies, microglia activation-associated cytotoxicity was observed to promote the apoptosis of DACs, along with the decrease of SIRT3 expression. The aim of the present study was to explore the potential neuroprotective effect of SIRT3 expression against dopaminergic neuron injury caused by microglia activation, and clarify its possible mechanisms. SIRT3 overexpression in DACs reduced the production of intracellular reactive oxygen species (ROS), cell apoptosis rate, mitochondrial membrane potential (ΔΨm) depolarization, opening of mitochondrial permeability transition pore (mPTP) and cyclophilin D (CypD) protein level, and promoted cell cycle progression. However, SIRT3 siRNA-mediated knockdown further aggravated microglia activation-mediated cytotoxicity, including ROS accumulation, increased cell apoptosis and mPTP opening, elevated the CypD level, enhanced mitochondrial ΔΨm depolarization, concomitant to cell cycle arrest at G0/G1 phase. The mechanisms of SIRT3 mitigated microglia activation-induced DAC dysfunction, which included decreased mPTP opening and Bax/Bcl-2 ratio, inhibition of mitochondrial cytochrome c release to the cytoplasm, reduced caspase-3/9 activity, increased LC3II/LC3I and beclin-1 protein expression levels, and decreased nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing protein 3 (NLRP3), caspase-1, IL-1ß and IL-18 protein expression. In conclusion, these results indicated that SIRT3 expression attenuated cell damage caused by microglia activation through the mitochondrial apoptosis pathway in DACs. The mitophagy-NLRP3 inflammasome pathway may also be associated with this neuroprotection. These findings may provide new intervention targets for the survival of dopaminergic neurons and the prevention and treatment of Parkinson's disease.

8.
Medicine (Baltimore) ; 100(44): e27511, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34871213

RESUMO

BACKGROUND: Pramipexole (P) or levodopa (L) treatment has been suggested as a therapeutic method for Parkinson disease (PD) in many clinical studies. Nonetheless, the combined effects of 2 drugs for PD patients are not completely understood.The aim of this research was to evaluate the clinical efficacy and safety of P plus L (P+L) combination therapy in the treatment of PD compared to that of L monotherapy, in order to confer a reference for clinical practice. METHODS: Randomized controlled trials (RCTs) of P+L for PD published up to April, 2020 were retrieved. Standardized mean difference (SMD), odds ratio (OR), and 95% confidence interval (CI) were calculated and heterogeneity was measured with the I2 test. Sensitivity analysis was also carried out. The outcomes of interest were as follows: the efficacy, unified Parkinson disease rating scale (UPDRS) scores, Hamilton depression rating scale score or adverse events. RESULTS: Twenty-four RCTs with 2171 participants were included. Clinical efficacy of P+L combination therapy was significantly better than L monotherapy (9 trials; OR 4.29, 95% CI 2.78 to 6.64, P < .00001). Compared with L monotherapy, the pooled effects of P+L combination therapy on UPDRS score were (22 trials; SMD -1.31, 95% CI -1.57 to -1.04, P < .00001) for motor UPDRS score, (16 trials; SMD -1.26, 95% CI -1.49 to -1.03, P < .00001) for activities of daily living UPDRS score, (12 trials; SMD -1.02, 95% CI -1.27 to -0.77, P < .00001) for mental UPDRS score, (10 trials; SMD -1.54, 95% CI -1.93 to -1.15, P < .00001) for complication UPDRS score. The Hamilton depression rating scale score showed significant decrease in the P+L combination therapy compared to L monotherapy (12 trials; SMD -1.56, 95% CI -1.90 to -1.22, P < .00001). In contrast to L monotherapy, P+L combination therapy reduced the number of any adverse events obviously in PD patients (16 trials; OR 0.36, 95% CI 0.27 to 0.50, P < .00001). CONCLUSIONS: P+L combination therapy is superior to L monotherapy for improvement of clinical symptoms in PD patients. Moreover, the safety profile of P+L combination therapy is better than that of L monotherapy. Further well-designed, multicenter RCTs needed to identify these findings.


Assuntos
Antiparkinsonianos/uso terapêutico , Levodopa/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Pramipexol/uso terapêutico , Antiparkinsonianos/efeitos adversos , Terapia Combinada , Humanos , Levodopa/efeitos adversos , Estudos Multicêntricos como Assunto , Pramipexol/efeitos adversos , Resultado do Tratamento
9.
BMC Endocr Disord ; 21(1): 178, 2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34465338

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is one of the most serious microvascular complications of diabetes, valsartan and α-lipoic acid alone or in combination has been used for the treatment of patients with DN. However, some results in these clinical reports were still controversial. The purpose of this study was to evaluate the efficacy of valsartan combined with α-lipoic acid on renal function in patients with DN. METHODS: We searched the electronic databases including PubMed, Sciencedirect, EMBASE, Cochrane library, Chinese national knowledge infrastructure (CNKI) and Wanfang databases, and the publication deadline was limited to January 2020. Randomized controlled trials (RCTs) evaluating the effects of valsartan combined with α-lipoic acid in DN patients were included. Pooled estimates were conducted using a fixed or random effect model. The outcomes included urinary albumin excretion rate (UAER), and the level of urinary albumin, ß2-microglobulin (ß2-MG), hypersensitive C-reactive protein (hs-CRP) and oxidative stress. RESULTS: 11 studies with 1294 participants were included in this study. The pooled analysis indicated that α-lipoic acid combined with valsartan could remarkably reduce UAER (P < 0.00001, SMD = -1.95, 95%CI = -2.55 to - 1.20; P = 0.03, SMD = -0.85, 95%CI = -1.59 to - 0.1) and the level of urinary albumin (P = 0.001, SMD = -1.48, 95%CI = - 2.38 to - 0.58; P = 0.01, SMD = -1.67, 95%CI = -3.00 to - 0.33), ß2-MG (P < 0.001,SMD = - 2.59, 95%CI = -3.78 to - 1.40; P = 0.03, SMD = -0.48, 95%CI = -0.93 to - 0.04) when compared with valsartan or lipoic acid monotherapy in patients with DN. However, there was no significant difference in the level of hs-CRP among the three therapies (P = 0.06, SMD = -2.80, 95%CI = -5.67 to 0.07; P = 0.10, SMD = -0.42, 95%CI = - 0.92 to 0.08). In addition, α-lipoic acid combined with valsartan markedly increased the level of SOD (P = 0.03, SMD = 1.24, 95%CI = 0.32 to 1.03; P = 0.0002, SMD = 0.68, 95%CI = 0.32 to 1.03) and T-AOC (P < 0.00001, SMD = 0.89, 95%CI = 0.62 to 1.16; P = 0.02, SMD = 0.58, 95%CI = 0.10 to1.07), and reduced the level of MDA(P = 0.0002, SMD = -1.99, 95%CI = -3.02 to - 0.96; P = 0.0001, SMD = -0.69, 95%CI = -1.04 to - 0.34). CONCLUSIONS: α-lipoic acid combined with valsartan could significantly reduce the level of urinary albumin and oxidative stress, increase antioxidant capacity and alleviate renal function damage in patients with DN, and this will provide a reference for the selection of treatment drugs for DN.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Rim/fisiologia , Ácido Tióctico/uso terapêutico , Valsartana/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Antioxidantes/uso terapêutico , Nefropatias Diabéticas/patologia , Quimioterapia Combinada , Humanos , Rim/efeitos dos fármacos
10.
Medicine (Baltimore) ; 100(25): e26512, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34160472

RESUMO

ABSTRACT: To explore the expressions of calculus-related functional proteins in the ureteral calculus-adhered polyp tissues and investigate the role of these proteins in the formation of adhesions between the calculus and polyp.Patients with ureteral calculi and polyps who underwent ureteroscopic lithotripsy for the excision of polyps between January 2019 and June 2019 were enrolled. Polyps obtained from each patient were divided into 2 groups using a matched pairs design: observation group (polyps adhered to calculus) and control group (polyps not adhered to calculus). Histopathological examination of polyps was performed using hematoxylin and eosin staining. Polyp tissues were immunohistochemically stained to assess the expressions of calculus-related functional proteins, that is, annexin A1, calcium-binding protein S100A9 (S100A9), uromodulin, and osteopontin. Furthermore, quantitative analysis was performed using the H-score of tissue staining; Pearson correlation analysis was performed for proteins with high expression.Overall, 40 polyp specimens were collected from 20 patients with ureteral calculi combined with polyps (observation group, 20 specimens; control group, 20 specimens). Hematoxylin and eosin staining revealed obvious epithelial cell proliferation in polyps of both groups; crystals were observed in the epithelial cells of the polyp tissue in the observation group. The expression levels of annexin A1 and S100A9 in the observation group were significantly greater than those in the control group (P < .05). However, no obvious expression of osteopontin or uromodulin was observed in the polyp tissues of both groups. There was a strong correlation between the increased expressions of annexin A1 and S100A9 in the observation group (R = 0.741, P = .022).We documented increased expressions of annexin A1 and S100A9 in the ureteral calculus-adhered polyp tissues. Annexin A1 and S100A9 may play an essential role in the adhesion of calculus and polyp and the growth of calculi.


Assuntos
Anexina A1/metabolismo , Calgranulina B/metabolismo , Pólipos/patologia , Cálculos Ureterais/complicações , Obstrução Ureteral/patologia , Adulto , Anexina A1/análise , Calgranulina B/análise , Feminino , Humanos , Litotripsia/métodos , Masculino , Pessoa de Meia-Idade , Pólipos/cirurgia , Ureter/patologia , Ureter/cirurgia , Cálculos Ureterais/imunologia , Cálculos Ureterais/patologia , Cálculos Ureterais/cirurgia , Obstrução Ureteral/etiologia , Obstrução Ureteral/cirurgia , Ureteroscopia/métodos
11.
Asian J Psychiatr ; 61: 102691, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33992852

RESUMO

OBJECTIVE: The aim of this report was to evaluate the clinical efficacy and safety of pramipexole therapy for patients with depression and Parkinson's disease (dPD), in order to confer a reference for clinical practice. METHODS: Randomized controlled trials (RCTs) of pramipexole for dPD published up to June 2020 were retrieved. Standardised mean difference (SMD), risk ratio (RR), and 95 % confidence interval (CI) were calculated. The outcomes included efficacy, Hamilton depression rating scale (HAMD) score, unified Parkinson's disease rating scale (UPDRS) scores, self-rating depression scale (SDS) score, self-rating anxiety scale (SAS) score or adverse events. RESULTS: Eighteen RCTs with 1789 participants were included. Clinical efficacy in pramipexole treatment group was significantly better than control group (RR 1.26, 95 % CI 1.20-1.33, P < 0.00001). Compared with control group, the pooled effects of pramipexole therapy on depression were (SMD -1.90, 95 % CI -2.58 to -1.23, P < 0.00001) for HAMD score, (SMD -3.94, 95 % CI -4.73 to -3.15, P < 0.00001) for SDS score, pramipexole therapy also decreased SAS score markedly (P < 0.0001). Compared with control group, the pooled effects of pramipexole on motor UPDRS score and activities of daily living UPDRS score were statistically significant (P < 0.01). Furthermore, pramipexole therapy didn't increase the number of any adverse events in dPD patients (RR 0.72, 95 % CI 0.37-1.41, P = 0.34). CONCLUSIONS: Pramipexole therapy can alleviate depressive symptoms and motor dysfunction in dPD patients, and there were no more side effects associated with drug intervention. These findings should be further validated by high-quality and well-designed RCTs.


Assuntos
Doença de Parkinson , Depressão/tratamento farmacológico , Humanos , Doença de Parkinson/tratamento farmacológico , Pramipexol , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
12.
Naunyn Schmiedebergs Arch Pharmacol ; 394(9): 1893-1905, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33959780

RESUMO

The purpose of this research was to evaluate the clinical efficacy and safety of pramipexole plus levodopa/benserazide (P+LB) combination therapy in the treatment of Parkinson's disease (PD) compared to that of LB monotherapy, in order to confer a reference for clinical practice. Randomized controlled trials (RCTs) of P+LB for PD published up to April 2020 were retrieved. Heterogeneity and sensitivity analysis were executed. Twenty-nine RCTs with 3017 participants were included. Clinical efficacy of P+LB combination therapy was significantly better than LB monotherapy (RR 1.27, 95% CI 1.22 to 1.32, P<0.00001). Compared with LB monotherapy, the pooled effects of P+LB combination therapy on UPDRS score were (SMD -1.41, 95% CI -1.71 to -1.11, P<0.00001) for motor UPDRS score, (SMD -1.65, 95% CI -2.25 to -1.04, P<0.00001) for activities of daily living UPDRS score, (SMD -2.20, 95% CI -3.32 to -1.09, P=0.0001) for mental UPDRS score, and (SMD -1.60, 95% CI -2.06 to -1.15, P<0.00001) for complication UPDRS score. The HAMD score showed significant decrease in the P+LB combination therapy compared to LB monotherapy (SMD -1.32, 95% CI -1.80 to -0.84, P<0.00001). In contrast to LB monotherapy, P+LB combination therapy decreased the number of any adverse events obviously in PD patients (RR 0.53, 95% CI 0.45 to 0.63, P<0.00001). In conclusion, P+LB combination therapy is superior to LB monotherapy for improvement of clinical symptoms in PD patients. Moreover, the safety profile of P+LB combination therapy is better than that of LB monotherapy. Further well-designed, multi-center RCTs needed to identify these findings.


Assuntos
Benserazida/administração & dosagem , Levodopa/administração & dosagem , Doença de Parkinson/tratamento farmacológico , Pramipexol/administração & dosagem , Antiparkinsonianos/administração & dosagem , Antiparkinsonianos/farmacologia , Benserazida/farmacologia , Combinação de Medicamentos , Quimioterapia Combinada , Humanos , Levodopa/farmacologia , Pramipexol/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
13.
Medicine (Baltimore) ; 99(48): e23322, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33235094

RESUMO

BACKGROUND: Many studies have evaluated the association between aldosterone synthase (CYP11B2) C-344T polymorphism and preeclampsia (PE) susceptibility, however, the results from different studies are inconsistent. OBJECTIVE: The study aimed to derive a more precise estimation of this association. METHODS: We searched PubMed, Embase, Chinese National Knowledge Infrastructure, China Biological Medicine, and Wanfang Database. The association was evaluated by calculating the odds ratios (ORs) with the corresponding 95% confidence intervals (CIs). RESULTS: Seven case-control studies with a total of 720 cases and 766 controls were eligible to be included in this meta-analysis. Overall, there was no significant association between CYP11B2 C-344T polymorphism and PE (for the allele model T vs.C: OR=0.78, 95%CI 0.60-1.01, p=0.06; for the codominant model CT vs. CC: OR=1.08, 95%CI 0.80-1.46, p=0.63; for the dominant model TT + CT vs. CC: OR=0.91, 95%CI 0.68-1.20, p=0.49). Similar results were obtained in sensitivity analysis. CONCLUSION: In summary, the present meta-analysis suggests that CYP11B2 C-344T polymorphism may not be associated with genetic susceptibility of PE, but the association remains indeterminate due to the insufficient evidence.


Assuntos
Citocromo P-450 CYP11B2/genética , Pré-Eclâmpsia/genética , Alelos , Estudos de Casos e Controles , China , Feminino , Humanos , Polimorfismo de Nucleotídeo Único , Gravidez , Fatores de Risco
14.
Medicine (Baltimore) ; 99(48): e23432, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33235126

RESUMO

BACKGROUND: It has been reported that polymorphisms of transferrin (TF) G258A and transferrin receptor (TFR) A82G might be associated with susceptibility to Parkinson disease (PD). OBJECTIVE: Owing to limitation of sample size and inconclusive results, we conducted a meta-analysis to clarify the association. METHODS: By searching PubMed, Embase, Chinese National Knowledge Infrastructure, China Biological Medicine Database, and Wanfang Databases, the published articles about studies of the association of the TF G258A, TFR A82G gene polymorphisms with the risk of PD were collected. Q-statistics and I statistics were calculated to examine heterogeneity and summary odds ratios (ORs) and 95% confidence intervals (95%CI) were evaluated the association. RESULTS: Five studies assessed the relationship between TF G258A and risk of PD. A significant increased protective of A allele and AA genotype was observed in allele model and recessive model (the allele model A vs G: OR = 0.54, 95%CI 0.40-0.72, P < .001; the recessive model AA vs GA + GG: OR = 0.32, 95%CI 0.20-0.52, P < .001). The remaining models of the TF G258A genotype showed no significant association with PD risk, while the protective tendency were increased (the heterozygote model GA vs GG: OR = 0.93, 95%CI 0.61-1.43, P = .75; the homozygous model AA vs GG: OR = 0.47, 95%CI 0.21-1.04, P = .06; the dominant model GA + AA vs GG: OR = 0.75, 95%CI 0.50-1.11, P = .15). There was also a lack of association between TFR A82G polymorphism and PD (the allele model G vs A: OR = 0.92, 95%CI 0.75-1.13, P = .43; the heterozygote model AG vs AA: OR = 1.17, 95%CI 0.79-1.71, P = .43; the homozygous model GG vs AA: OR = 0.91, 95%CI 0.60-139, P = .66; the dominant model AG + GG vs AA: OR = 1.05, 95%CI 0.73-1.49, P = .81; the recessive model GG vs AG +AA: OR = 0.80, 95%CI 0.59-1.09, P = .16). CONCLUSION: Our study suggests that TF G258A polymorphism may be associated with PD, while TFR A82G polymorphism may not contribute to PD based on the current evidence.


Assuntos
Predisposição Genética para Doença , Doença de Parkinson/genética , Polimorfismo de Nucleotídeo Único , Receptores da Transferrina/genética , Transferrina/genética , Alelos , Genótipo , Humanos , Modelos Genéticos
15.
Aging Clin Exp Res ; 32(5): 769-779, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31175606

RESUMO

BACKGROUND: Selegiline or levodopa treatment has been suggested as a therapeutic method for Parkinson's disease (PD) in many clinical trial reports. However, the combined effects of two drugs still remain controversial. The aim of this report was to evaluate the clinical efficacy and safety of selegiline plus levodopa (S + L) combination therapy in the treatment of PD compared to that of L monotherapy, to provide a reference resource for rational drug use. METHODS: Randomized controlled trials (RCTs) of S + L for PD published up to September, 2018 were searched. Mean difference (MD), odds ratio (OR), and 95% confidence interval (CI) were calculated and heterogeneity was assessed with the I2 test. Sensitivity analysis was also performed. The outcomes measured were as follows: the unified Parkinson's disease rating scale (UPDRS) scores, modified Webster score, adverse events and mortality. RESULTS: Fourteen RCTs with 2008 participants were included. Compared with L monotherapy, the pooled effects of S + L combination therapy on UPDRS score were (eleven trials; MD - 7.00, 95% CI - 8.35 to - 5.65, P < 0.00001) for total UPDRS score (nine trials; MD - 5.74, 95% CI - 7.71 to - 3.77, P < 0.00001) for motor UPDRS score (seven trials; MD - 1.61, 95% CI - 2.18 to - 1.04, P < 0.00001) for activities of daily living UPDRS score (three trials; MD - 0.38, 95% CI - 0.61 to - 0.14, P = 0.002) for mental UPDRS score. The Webster score showed significant decrease in the S + L combination therapy compared to L monotherapy (four trials; MD - 5.71, 95% CI - 7.11 to - 4.32, P < 0.00001). Compared with L monotherapy, S + L combination therapy did not increase the number of any adverse events significantly in PD patients (ten trials; OR 1.58, 95% CI 0.83-3.00, P = 0.16). CONCLUSIONS: S + L combination therapy is superior to L monotherapy for the improvement of clinical symptoms in PD patients. Moreover, the safety profile of S + L combination therapy is comparable with that of L monotherapy.


Assuntos
Levodopa/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Selegilina/uso terapêutico , Atividades Cotidianas , Terapia Combinada , Quimioterapia Combinada , Humanos , Testes de Estado Mental e Demência , Resultado do Tratamento
16.
Neurol Sci ; 41(1): 101-109, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31446579

RESUMO

OBJECTIVE: The aim of this report was to systematically evaluate the efficacy and safety of rasagiline (R) plus levodopa (L) (R + L) for the treatment of Parkinson's disease (PD) compared with that of L monotherapy, in order to provide a reference resource for rational drug use. METHODS: Randomized controlled trials (RCTs) of R + L for PD published up to September 2018 were searched. Sensitivity analyses were also performed. RESULTS: Fourteen RCTs with 2531 participants were included. Compared with L monotherapy, the pooled effects of R + L combination therapy on unified Parkinson's disease rating scale (UPDRS) score were (SMD - 0.50, 95% CI - 0.70 to - 0.30, P < 0.00001) for UPDRS motor score, (SMD - 0.59, 95% CI - 0.79 to - 0.39, P < 0.00001) for UPDRS activities of daily living (ADL) score, (SMD - 0.65, 95% CI - 0.81 to - 0.49, P < 0.00001) for UPDRS total score. R + L combination therapy was better than L monotherapy in reducing daily off-time (SMD - 1.15, 95% CI - 2.13 to - 0.17, P = 0.02), but there was a statistically nonsignificant result in daily on-time increase (SMD 1.39, 95% CI - 0.69 to 3.48, P = 0.19). There were no statistical differences in number of adverse events (OR 1.33, 95% CI 0.97 to 1.82, P = 0.07) and number of dropout (OR 0.88, 95% CI 0.65 to 1.19, P = 0.39) between R + L combination therapy and L monotherapy. CONCLUSIONS: R + L combination therapy was superior to L monotherapy for improvement of UPDRS scores and off-time in PD patients. Moreover, R + L combination therapy and L monotherapy were similar in terms of safety and tolerability.


Assuntos
Antiparkinsonianos/administração & dosagem , Indanos/administração & dosagem , Levodopa/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Doença de Parkinson/tratamento farmacológico , Quimioterapia Combinada , Humanos , Doença de Parkinson/diagnóstico , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos
17.
J Neural Transm (Vienna) ; 126(11): 1465-1470, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31471711

RESUMO

Studies investigating the impact of polymorphisms on monocyte chemotactic protein-1 (MCP-1) and CC chemokine receptor (CCR2) on the susceptibility of Parkinson's disease (PD) have reported inconsistent results. Owing to mixed and inconclusive results, we conducted a meta-analysis to systematically summarize and clarify the association between the two gene polymorphisms and PD risk. We performed a meta-analysis of five eligible studies to summarize the data describing the association between PD risk and polymorphisms in MCP-1 A2518G and CCR2 V64I. The association was evaluated by calculating the odds ratios (ORs) with the corresponding 95% confidence intervals (CIs). A significant increased risk of PD was observed in the MCP-1 A2518G polymorphism in allele model (G vs. A: OR 1.12, 95% CI 1.01-1.25, p = 0.03). The dominant model of MCP-1 A2518G genotype showed no significant association with PD risk, while the risk tendency was increased (AG + GG vs. AA: OR 1.20, 95% CI 1.00-1.42, p = 0.05). In addition, CCR2 V64I polymorphism showed no significant association with PD risk (I vs. V: OR 0.33, 95% CI 0.06-1.92, p = 0.22; VI + II vs. VV: OR 1.00, 95% CI 0.83-1.21, p = 0.99). In subgroup analysis by ethnicity, no significant difference was found in both Caucasians and Asians between CCR2 V64I polymorphism and PD risk, while a significant statistical association was identified in Asians between MCP-1 A2518G polymorphism and PD risk. When the data were stratified by study area, the increased risk of PD was observed only in studies conducted in China. In summary, the present meta-analysis suggests that genetic polymorphisms of MCP-1 A2518G may influence the susceptibility of PD in Asian countries, especially in China. However, CCR2 V64I polymorphism is not correlated with PD risk. The results should be interpreted with caution due to limited sample and heterogeneity. Large scale and well-designed studies are needed to validate our findings.


Assuntos
Quimiocina CCL2/genética , Predisposição Genética para Doença/etnologia , Doença de Parkinson/etnologia , Doença de Parkinson/genética , Receptores CCR2/genética , Humanos , Polimorfismo de Nucleotídeo Único , Risco
18.
J Neural Transm (Vienna) ; 126(5): 559-568, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31004314

RESUMO

Microglia activation-mediated neuroinflammation plays an important role in the progression of Parkinson's disease (PD). However, effects of microglia activation on dopaminergic neuronal cell (DAC) fate are still poorly understood. The objective of this study was to explore the neurotoxic effects of microglia activation-mediated oxidative injury in DACs and its possible mechanisms. In the present study, microglia-DACs co-culture systems (murine BV-2 and MN9D cells, or primary microglia and mesencephalic neurons) were used to display the crosstalk between both cell types. The cytotoxicity of lipopolysaccharide-induced microglia activation led to the accumulation of intracellular reactive oxygen species, increased cell apoptosis rate, reduced number of DACs, concomitant to cell cycle arrest at G1 phase. Molecular mechanisms of apoptosis caused by microglia activation-induced oxidative injury included the increased opening of mitochondrial permeability transition pore and enhanced membrane potential depolarization in MN9D cells, down-regulation of Bcl-2 and up-regulation of Bax, caspase-3 expression in DACs. In addition, microglia activation made a significant reduction of SIRT3 and superoxide dismutase 2 gene expression in DACs. Taken together, these data imply that microglia activation promotes cell apoptosis through mitochondrial pathway and decreases SIRT3 expression in DACs, which may provide some support for PD progression promoted by neuroinflammation.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Microglia/metabolismo , Estresse Oxidativo/fisiologia , Sirtuína 3/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Neurônios Dopaminérgicos/patologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ratos , Ratos Sprague-Dawley
19.
Medicine (Baltimore) ; 97(44): e13020, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30383660

RESUMO

BACKGROUND: Prostaglandin E1 (P) or methylcobalamin (M) treatment has been suggested as a therapeutic approach for diabetic peripheral neuropathy (DPN) in many clinical trial reports. However, the combined effects of 2 drugs still remain dubious. OBJECTIVE: The aim of this report was to evaluate the efficacy of M plus P (M + P) for the treatment of DPN compared with that of P monotherapy, in order to provide a reference resource for rational drug use. METHODS: Randomized controlled trials (RCTs) of M + P for DPN published up to September 2017 were searched. Risk ratio (RR), mean difference (MD), and 95% confidence interval (CI) were calculated and heterogeneity was assessed with the I test. Subgroup and sensitivity analyses were also performed. The outcomes measured were as follows: the clinical efficacy, median motor nerve conduction velocities (MNCV), median sensory nerve conduction velocity (SNCV), peroneal MNCV, peroneal SNCV, and adverse effects. RESULTS: Sixteen RCTs with 1136 participants were included. Clinical efficacy of M + P combination therapy was significantly better than P monotherapy (fifteen trials; RR 1.25, 95% CI 1.18-1.32, P < .00001, I = 27%). Compared with P monotherapy, the pooled effects of M + P combination therapy on nerve conduction velocity were (MD 6.29, 95% CI 4.63-7.94, P < .00001, I = 90%) for median MNCV, (MD 5.68, 95% CI 3.53-7.83, P < .00001, I = 94%) for median SNCV, (MD 5.36, 95% CI 3.86-6.87, P < .00001, I = 92%) for peroneal MNCV, (MD 4.62, 95% CI 3.48-5.75, P < .00001, I = 86%) for peroneal SNCV. There were no serious adverse events associated with drug intervention. CONCLUSIONS: M + P combination therapy was superior to P monotherapy for improvement of neuropathic symptoms and NCVs in DPN patients. Moreover, no serious adverse events occur in combination therapy.


Assuntos
Alprostadil/administração & dosagem , Neuropatias Diabéticas/tratamento farmacológico , Vitamina B 12/análogos & derivados , Alprostadil/efeitos adversos , Quimioterapia Combinada , Feminino , Humanos , Masculino , Condução Nervosa/efeitos dos fármacos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento , Vitamina B 12/administração & dosagem , Vitamina B 12/efeitos adversos
20.
J Neural Transm (Vienna) ; 125(12): 1781-1786, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30284076

RESUMO

The -2518A/G polymorphism in monocyte chemotactic protein-1 (MCP-1) has been extensively investigated for association with Alzheimer's disease (AD); however, the results from different studies are inconsistent. The aim of this study was to draw an accurate conclusion of the association. All eligible case-control studies were searched in PubMed, Embase, Chinese National Knowledge Infrastructure, China Biological Medicine Databases, and Wanfang Databases. Eight case-control studies with a total of 2370 cases and 2413 controls were eligible to be included in this meta-analysis. The association was evaluated by calculating the odds ratios (ORs) with the corresponding 95% confidence intervals (CIs). Overall, there was no significant association between MCP-1-2518A/G polymorphism and AD risk in all genetic models (the allele model G vs. A: OR = 1.15, 95% CI 0.92-1.45, p = 0.22; the co-dominant model GG vs. AA: OR = 1.38, 95% CI 0.80-2.36, p = 0.25; the dominant model AG + GG vs. AA: OR = 1.14, 95% CI 0.89-1.46, p = 0.31; the recessive model GG vs. AG + AA: OR = 1.35, 95% CI 0.87-2.09, p = 0.18). In subgroup analysis by ethnicity, a significant difference was not detected in both Caucasians and Asians. In allele model (G vs. A), the required sample size of 31858 was calculated by applying trial sequential analysis. Cumulative z curve is always below the trial sequential monitoring boundary and is nominally statistically significant (Z = 1.96). A consistent result was obtained in other genetic models. In summary, the present meta-analysis suggests that MCP-1-2518A/G polymorphism may not be associated with genetic susceptibility of AD in general population, but the association remains indeterminate due to the insufficient evidence.


Assuntos
Doença de Alzheimer/genética , Quimiocina CCL2/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Alelos , Estudos de Associação Genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA