Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Nat Med ; 30(4): 1096-1103, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38622249

RESUMO

Prasinezumab, a monoclonal antibody that binds aggregated α-synuclein, is being investigated as a potential disease-modifying therapy in early-stage Parkinson's disease. Although in the PASADENA phase 2 study, the primary endpoint (Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) sum of Parts I + II + III) was not met, prasinezumab-treated individuals exhibited slower progression of motor signs than placebo-treated participants (MDS-UPDRS Part III). We report here an exploratory analysis assessing whether prasinezumab showed greater benefits on motor signs progression in prespecified subgroups with faster motor progression. Prasinezumab's potential effects on disease progression were assessed in four prespecified and six exploratory subpopulations of PASADENA: use of monoamine oxidase B inhibitors at baseline (yes versus no); Hoehn and Yahr stage (2 versus 1); rapid eye movement sleep behavior disorder (yes versus no); data-driven subphenotypes (diffuse malignant versus nondiffuse malignant); age at baseline (≥60 years versus <60 years); sex (male versus female); disease duration (>12 months versus <12 months); age at diagnosis (≥60 years versus <60 years); motor subphenotypes (akinetic-rigid versus tremor-dominant); and motor subphenotypes (postural instability gait dysfunction versus tremor-dominant). In these subpopulations, the effect of prasinezumab on slowing motor signs progression (MDS-UPDRS Part III) was greater in the rapidly progressing subpopulations (for example, participants who were diffuse malignant or taking monoamine oxidase B inhibitors at baseline). This exploratory analysis suggests that, in a trial of 1-year duration, prasinezumab might reduce motor progression to a greater extent in individuals with more rapidly progressing Parkinson's disease. However, because this was a post hoc analysis, additional randomized clinical trials are needed to validate these findings.


Assuntos
Doença de Parkinson , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Tremor/tratamento farmacológico , Antiparkinsonianos/uso terapêutico , Monoaminoxidase/uso terapêutico , Progressão da Doença
2.
CNS Drugs ; 37(11): 941-956, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37973769

RESUMO

BACKGROUND AND OBJECTIVE: In Parkinson's disease, safinamide and zonisamide are novel monoamine oxidase-B inhibitors with a dual mechanism of action involving the inhibition of sodium and calcium channels and the subsequent release of glutamate. The aim of this systematic review and meta-analysis was to examine the efficacy and safety of both drugs compared with placebo on motor symptoms, cognitive function, and quality of life in patients with Parkinson's disease. METHODS: We searched MEDLINE, EMBASE, Cochrane Central, Scopus, PsycINFO, and trials registries up to March 2023 for randomized controlled trials of adults with Parkinson's disease administered either safinamide or zonisamide and published in English. We excluded single-arm trials or if neither the efficacy nor safety outcomes of interest were reported. Primary outcomes were the change from baseline in Unified Parkinson's Disease Rating Scale section III (UPDRS-III) and serious adverse events. Secondary outcomes included a change from baseline in OFF-time, Parkinson's Disease Questionnaire 39 to evaluate quality of life, and Mini-Mental State Examination for cognitive function assessment. The meta-analysis was conducted using Review Manager 5.4.1. Random-effect models were used to calculate the pooled mean differences (MDs) and risk ratios with 95% confidence intervals (CIs). Subgroup analyses by medication, doses, Parkinson's disease stage, and risk of bias were conducted. We assessed the risk of bias using the Cochrane's risk of bias tool. Sensitivity analysis was conducted, and publication bias were evaluated. This meta-analysis was not externally funded, and the protocol is available on the Open Science Framework Registration ( https://doi.org/10.17605/OSF.IO/AMNP5 ). RESULTS: Of 3570 screened citations, 16 trials met inclusion criteria (4314 patients with Parkinson's disease). Ten safinamide trials were conducted in several countries. Six zonisamide trials were included, five of which were conducted in Japan and one in India. UPDRS Part III scores were significantly lower with both monoamine oxidase-B inhibitors than with placebo (MD = -  2.18; 95% CI -  2.88 to -  1.49; I 2 =63%; n = 14 studies). A subgroup analysis showed a significant improvement in UPDRS-III in safinamide (MD = -  2.10; 95% CI -  3.09 to -  1.11; I2 = 71%; n = 8 studies) and zonisamide (MD = -  2.31; 95% CI -  3.35 to -  1.27; I2 = 52%; n = 6 studies) compared with placebo. Monoamine oxidase-B inhibitors significantly decreased OFF-time compared with placebo. No significant differences in cognitive function (Mini-Mental State Examination), whereas an improvement in quality of life (Parkinson's Disease Questionnaire 39 scores) was observed. There was no significant difference in incidence rates of serious adverse events among all examined doses of zonisamide and safinamide compared with placebo. Two trials were reported as a high risk of bias and sensitivity analyses confirmed the primary analysis results. CONCLUSIONS: Evidence suggests that novel monoamine oxidase-B inhibitors not only improve motor symptoms but also enhance patients' quality of life. The meta-analysis showed that both medications have a similar safety profile to placebo with regard to serious adverse events. The overall findings emphasize the effectiveness of safinamide and zonisamide in the treatment of Parkinson's disease as adjunct therapy. Further long-term studies examining the impact of these medications on motor and non-motor symptoms are necessary.


Assuntos
Doença de Parkinson , Adulto , Humanos , Doença de Parkinson/tratamento farmacológico , Zonisamida/efeitos adversos , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Dopaminérgicos/uso terapêutico , Monoaminoxidase/uso terapêutico
3.
Inflammopharmacology ; 31(6): 3367-3370, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37558921

RESUMO

There is a group of enzymes called monoamine oxidase(s) (MAOs) that help with the oxidation of amines found in both our diet and our bodies. Currently, monoamine oxidase inhibitors (MAO-Is) are utilized to manage conditions like depression, Parkinson's disease, and other psychiatric disorders. Rheumatoid arthritis (RA) is an auto-immune disease that has been linked to negative changes in mental health, such as depression. When depression co-occurs with RA, it can further worsen the outcome of the disease. Inhibiting monoamine oxidases could potentially treat RA by improving its pathological markers. Using existing pre-clinical and clinical data on safety and toxicity makes drug re-purposing advantageous. Hence, the pre-clinical validation of MAO-I's effectiveness in managing RA requires urgent regulatory intervention to commence clinical trials. Back in 1983, a clinical case report put forward the idea of repurposing MAO-I for RA treatment. Although MAO-I had been used for depression, it was observed to have a significant reduction in joint pain and stiffness. However, no significant clinical research has been conducted on this matter since then. In this commentary article, we provide a summary of the pre-clinical data that is currently available. The main focus of our discussion is on the significance of clinical trials for MAO-I, repurposing it for RA, and using it for the simultaneous management of depression and RA.


Assuntos
Artrite Reumatoide , Doenças Autoimunes , Doença de Parkinson , Humanos , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/uso terapêutico , Monoaminoxidase/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico
4.
J Mol Graph Model ; 122: 108471, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37087882

RESUMO

An in silico consensus molecular docking approach and in vitro evaluations were adopted in the present study to explore a dataset of FDA-approved drugs as novel multitarget MAO-B/AChE agents in the treatment of Alzheimer's disease (AD). GOLD 5.3 and Glide were employed in the virtual assessments and consensus superimpositions of the obtained poses were applied to increase the reliability of the docking protocols. Furthermore, the top ranked molecules were subjected to binding free energy calculations using MM/GBSA, Induced fit docking (IFD) simulations, and a literature review. Consequently, the top four multitarget drugs were examined for their in vitro MAO-B and AChE inhibition effects. The consensus molecular docking identified Dolutegravir, Rebamipide, Loracarbef and Diflunisal as potential multitarget drugs. The biological data demonstrated that most of the docking scores were in good correlation with the in vitro experiments, however the theoretical simulations in the active site of MAO-B identified two false-positives - Rebamipide and Diflunisal. Dolutegravir and Loracarbef were accessed as active MAO-B inhibitors, while Dolutegravir, Rebamapide and Diflunisal as potential AChE inhibitors. The antiretroviral agent Dolutegravir exhibited the most potent multitarget activity - 41% inhibition of MAO-B (1 µM) and 68% inhibition of AChE (10 µM). Visualizations of the intermolecular interactions of Dolutegravir in the active sites of MAO-B and AChE revealed the formation of several stable hydrogen bonds. Overall, Dolutegravir was identified as a potential anti-AD drug, however further in vivo evaluations should be considered.


Assuntos
Doença de Alzheimer , Diflunisal , Humanos , Monoaminoxidase/química , Monoaminoxidase/metabolismo , Monoaminoxidase/uso terapêutico , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/química , Inibidores da Monoaminoxidase/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Simulação de Acoplamento Molecular , Diflunisal/uso terapêutico , Reposicionamento de Medicamentos , Reprodutibilidade dos Testes , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/química , Acetilcolinesterase/química
5.
Cancer Lett ; 563: 216188, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37076041

RESUMO

Monoamine oxidase A (MAOA) is a mitochondrial enzyme that catalyzes the oxidative deamination of monoamine neurotransmitters and dietary amines. Previous studies have shown that MAOA is clinically associated with prostate cancer (PCa) progression and plays a key role in almost each stage of PCa, including castrate-resistant prostate cancer, neuroendocrine prostate cancer, metastasis, drug resistance, stemness, and perineural invasion. Moreover, MAOA expression is upregulated not only in cancer cells but also in stromal cells, intratumoral T cells, and tumor-associated macrophages; thus, targeting MAOA can be a multi-pronged approach to disrupt tumor promoting interactions between PCa cells and tumor microenvironment. Furthermore, targeting MAOA can disrupt the crosstalk between MAOA and the androgen receptor (AR) to restore enzalutamide sensitivity, blocks glucocorticoid receptor (GR)- and AR-dependent PCa cell growth, and is a potential strategy for immune checkpoint inhibition, thereby alleviating immune suppression and enhancing T cell immunity-based cancer immunotherapy. MAOA is a promising target for PCa therapy, which deserves further exploration in preclinical and clinical settings.


Assuntos
Monoaminoxidase , Neoplasias da Próstata , Masculino , Humanos , Monoaminoxidase/metabolismo , Monoaminoxidase/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Próstata/patologia , Proliferação de Células , Receptores Androgênicos , Linhagem Celular Tumoral , Microambiente Tumoral
6.
Inflammopharmacology ; 31(3): 1305-1317, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36826724

RESUMO

OBJECTIVE: This study aims to investigate the anti-inflammatory mechanism of monoamine oxidase inhibitor (MAOI) in carrageenan (CARR) induced inflammation models to reprofile their use. We also aimed to explore the role of monoamine oxidase (MAO)-mediated H2O2-NF-κB-COX-2 pathway in acute inflammation. METHODS: In vitro anti-inflammatory activity and hydrogen peroxide (H2O2) scavenging activity were performed according to the established procedure. Inflammation was induced using CARR in BALB/c mice at the foot paw and peritoneal cavity. Hourly measurement of paw swelling was performed. The level of nitric oxide (NO), myeloperoxidase (MPO), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) and nuclear factor κB (NF-κB) was determined using enzyme-linked immunosorbent assay (ELISA). Peritoneal fluid was collected to investigate total count, differential count of leukocytes, and capillary permeability. RESULTS: In vitro anti-inflammatory evaluations revealed the potential role of MAOI to inhibit heat-induced protein denaturation and human red cell membrane destabilization. H2O2 inhibition activity of MAOI also proved their powerful role as an H2O2 scavenger. Treatment with MAOI in CARR-induced mice significantly reduced paw edema, leukocyte extravasation, and total and differential leukocyte count. The result of ELISA showed MAOI effectively reduce the level of COX-2, PGE2 and NF-κB in inflamed tissue. CONCLUSIONS: In short, this study demonstrates that inhibition of H2O2 by MAOI alleviates CARR-induced paw edema possibly by inhibiting the H2O2-mediated NF-κB-COX-2 pathway. The present investigation identifies MAOI might reprofile for the treatment of acute inflammation also, the MAO enzyme may use as a novel therapeutic target to design and develop new class of anti-inflammatory agents.


Assuntos
Peróxido de Hidrogênio , NF-kappa B , Camundongos , Humanos , Animais , NF-kappa B/metabolismo , Ciclo-Oxigenase 2/metabolismo , Peróxido de Hidrogênio/metabolismo , Inibidores da Monoaminoxidase/efeitos adversos , Transdução de Sinais , Carragenina/farmacologia , Inflamação/metabolismo , Anti-Inflamatórios/uso terapêutico , Edema/metabolismo , Monoaminoxidase/metabolismo , Monoaminoxidase/farmacologia , Monoaminoxidase/uso terapêutico , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo
7.
J Clin Psychiatry ; 83(6)2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36300995

RESUMO

Objective: Ketamine is increasingly prescribed for treatment-resistant depression (TRD), often as add-on to regular antidepressants. Augmentation of ketamine to monoamine oxidase inhibitors (MAOIs) is advised against, as this practice might increase blood pressure or cause serotonin syndrome. Despite the potential relevance for patients, little is known about actual side effects of combined use. We summarize literature on the safety and add results of our case series.Evidence Review: PubMed and Embase were searched from inception to July 2021 for English-language articles describing concomitant use of ketamine and MAOIs. The search strategy included terms for "ketamine" AND "monoamine oxidase inhibitor" including generic and brand names. Additionally, we describe the safety of twice weekly oral esketamine administration over the course of 5 weeks to 9 months in 8 TRD patients using MAOIs.Findings: After deduplication, we screened 138 articles and assessed 43 full texts. Twelve studies were included with a total of 39 patients receiving ketamine and MAOIs. Blood pressure and heart rate increased in multiple cases, though this was deemed clinically insignificant in all but 1 patient. No signs of hypertensive crisis or serotonin syndrome were observed. In our case series, we observed minor elevations in blood pressure and heart rate and no serious adverse events.Conclusions and Relevance: The results suggest that combined use of MAOIs and esketamine is less prone to severe side effects than presumed. The investigated sample size was small, and prescribed doses of MAOIs were relatively low. Further research is required before definite conclusions about the safety of this combination can be drawn.


Assuntos
Transtorno Depressivo Resistente a Tratamento , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Síndrome da Serotonina , Humanos , Inibidores da Monoaminoxidase/efeitos adversos , Síndrome da Serotonina/induzido quimicamente , Síndrome da Serotonina/tratamento farmacológico , Depressão/tratamento farmacológico , Transtorno Depressivo Resistente a Tratamento/tratamento farmacológico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/tratamento farmacológico , Monoaminoxidase/uso terapêutico
8.
J Integr Bioinform ; 19(4)2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36112816

RESUMO

Parkinson's disease is considered the second most frequent neurodegenerative disease. It is described by the loss of dopaminergic neurons in the mid-brain. For many decades, L-DOPA has been considered as the gold standard for treating Parkinson's disease motor symptoms, however, due to the decrease of efficacy, in the long run, there is an urgent need for novel antiparkinsonian drugs. Caffeine derivatives have been reported several times for their neuroprotective properties and dual blockade of monoamine oxidase (MAO) and adenosine A2A receptors (AA2AR). Natural products are currently attracting more focus due to structural diversity and safety in contrast to synthetic drugs. In the present work, computational studies were conducted on natural product-like caffeine derivatives to search for novel potent candidates acting as dual MAO-B inhibitors/AA2AR antagonists for Parkinson's disease. Our findings revealed two natural products among the top hits: CNP0202316 and CNP0365210 fulfill the requirements of drugs acting on the brain. The selected lead compounds were further studied using molecular dynamics simulation to assess their stability with MAO-B. Current findings might shift the interest towards natural-based compounds and could be exploited to further optimize caffeine derivatives into a successful dual-target-directed drug for managing and halting the neuronal damage in Parkinson's disease patients.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Cafeína/farmacologia , Cafeína/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/uso terapêutico , Inibidores da Monoaminoxidase/química , Doenças Neurodegenerativas/tratamento farmacológico , Antagonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Antagonistas do Receptor A2 de Adenosina/química , Monoaminoxidase/uso terapêutico
9.
Biomed Pharmacother ; 155: 113718, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36152409

RESUMO

Parkinson's disease is the second most prevalent neurodegenerative disease after Alzheimer's disease, mostly happened in the elder population and the prevalence gradually increased with age. Parkinson's disease is a movement disorder that severely affects patients' daily life. The mechanism of Parkinson's disease still remains unknown, however, studies already proved that the damage or absence of dopaminergic neurons located in the substantia nigra and the decreased dopamine in the striatum are significantly related to Parkinson's disease. To date, the mainstream treatment of Parkinson's disease has been achieved by alleviating its associated morbid symptoms, such as the use of levodopa, carbidopa, dopamine receptor agonists, monoamine oxidase type B inhibitors, anticholinergic drugs, etc. However, strong side effects, even toxicity, have been reported after using these drugs, with reduced effectiveness over time. Plant compounds have shown good therapeutic effects in neurodegenerative diseases as a less toxic treatment. In this review, we have compiled several natural plant compounds and classified the currently reported compounds for therapeutic use based on their structural parent nuclei and constituent elements. We wish to inspire new ideas for the treatment of Parkinson's disease by summarizing their mechanisms.


Assuntos
Produtos Biológicos , Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Idoso , Doença de Parkinson/tratamento farmacológico , Levodopa , Carbidopa , Dopamina , Agonistas de Dopamina/uso terapêutico , Produtos Biológicos/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Monoaminoxidase/uso terapêutico , Antagonistas Colinérgicos
10.
J Pain ; 23(12): 2092-2109, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35940543

RESUMO

MAO-B inhibitors have been implicated to reverse neuropathic pain behaviors. Our previous study has demonstrated that KDS2010 (KDS), a newly developed reversible MAO-B inhibitor, could attenuate Paclitaxel (PTX)-induced tactile hypersensitivity in mice through suppressing reactive oxidant species (ROS)-decreased inhibitory GABA synaptic transmission in the spinal cord. In this study, we evaluated the analgesic effect of KDS under a new approach, in which KDS acts on dorsal horn sensory neurons to reduce excitatory transmission. Oral administration of KDS effectively enhanced mechanical thresholds in the spinal nerve ligation (SNL) induced neuropathic pain in rats. Moreover, we discovered that although treatment with KDS increased brain-derived neurotrophic factor (BDNF) levels, KDS inhibited Tropomyosin receptor kinase B (TrkB) receptor activation, suppressing increased p-NR2B-induced hyperexcitability in spinal dorsal horn sensory neurons after nerve injury. In addition, KDS showed its anti-inflammatory effects by reducing microgliosis and astrogliosis and the activation of MAPK and NF-ᴋB inflammatory pathways in these glial cells. The levels of ROS production in the spinal cords after the SNL procedure were also decreased with KDS treatment. Taken together, our results suggest that KDS may represent a promising therapeutic option for treating neuropathic pain. PERSPECTIVE: Our study provides evidence suggesting the mechanisms by which KDS, a novel MAO-B inhibitor, can be effective in pain relief. KDS, by targeting multiple mechanisms involved in BDNF/TrkB/NR2B-related excitatory transmission and neuroinflammation, may represent the next future of pain medicine.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Neuralgia , Ratos , Camundongos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Espécies Reativas de Oxigênio/uso terapêutico , Ratos Sprague-Dawley , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Nervos Espinhais , Medula Espinal , Monoaminoxidase/metabolismo , Monoaminoxidase/farmacologia , Monoaminoxidase/uso terapêutico
11.
Eur J Cancer ; 165: 81-96, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35219026

RESUMO

BACKGROUND: Although programmed cell death protein 1 (PD-1) inhibitors have revolutionised treatment for advanced melanoma, not all patients respond. We previously showed that inhibition of the flavoprotein renalase (RNLS) in preclinical melanoma models decreases tumour growth. We hypothesised that RNLS inhibition promotes tumour rejection by effects on the tumour microenvironment (TME). METHODS: We used two distinct murine melanoma models, studied in RNLS knockout (KO) or wild-type (WT) mice. WT mice were treated with the anti-RNLS antibody, m28, with or without anti-PD-1. 10X single-cell RNA-sequencing was used to identify transcriptional differences between treatment groups, and tumour cell content was interrogated by flow cytometry. Samples from patients treated with immunotherapy were examined for RNLS expression by quantitative immunofluorescence. RESULTS: RNLS KO mice injected with wild-type melanoma cells reject their tumours, supporting the importance of RNLS in cells in the TME. This effect was blunted by anti-cluster of differentiation 3. However, MØ-specific RNLS ablation was insufficient to abrogate tumour formation. Anti-RNLS antibody treatment of melanoma-bearing mice resulted in enhanced T cell infiltration and activation and resulted in immune memory on rechallenging mice with injection of melanoma cells. At the single-cell level, treatment with anti-RNLS antibodies resulted in increased tumour density of MØ, neutrophils and lymphocytes and increased expression of IFNγ and granzyme B in natural killer cells and T cells. Intratumoural Forkhead Box P3 + CD4 cells were decreased. In two distinct murine melanoma models, we showed that melanoma-bearing mice treated with anti-RNLS antibodies plus anti-PD-1 had superior tumour shrinkage and survival than with either treatment alone. Importantly, in pretreatment samples from patients treated with PD-1 inhibitors, high RNLS expression was associated with decreased survival (log-rank P = 0.006), independent of other prognostic variables. CONCLUSIONS: RNLS KO results in melanoma tumour regression in a T-cell-dependent fashion. Anti-RNLS antibodies enhance anti-PD-1 activity in two distinct aggressive murine melanoma models resistant to PD-1 inhibitors, supporting the development of anti-RNLS antibodies with PD-1 inhibitors as a novel approach for melanomas poorly responsive to anti-PD-1.


Assuntos
Inibidores de Checkpoint Imunológico , Melanoma , Animais , Humanos , Imunoterapia , Melanoma/tratamento farmacológico , Camundongos , Monoaminoxidase/uso terapêutico , Microambiente Tumoral
12.
J Parkinsons Dis ; 12(2): 477-493, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34957948

RESUMO

Monoamine oxidase-B (MAO-B) inhibitors are commonly used for the symptomatic treatment of Parkinson's disease (PD). MAO-B inhibitor monotherapy has been shown to be effective and safe for the treatment of early-stage PD, while MAO-B inhibitors as adjuvant drugs have been widely applied for the treatment of the advanced stages of the illness. MAO-B inhibitors can effectively improve patients' motor and non-motor symptoms, reduce "OFF" time, and may potentially prevent/delay disease progression. In this review, we discuss the effects of MAO-B inhibitors on motor and non-motor symptoms in PD patients, their mechanism of action, and the future development of MAO-B inhibitor therapy.


Assuntos
Doença de Parkinson , Dopaminérgicos/uso terapêutico , Humanos , Monoaminoxidase/uso terapêutico , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Selegilina/uso terapêutico
13.
Nutr Neurosci ; 25(9): 1898-1908, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33856270

RESUMO

BACKGROUND: Naringenin is a powerful antioxidant and anti-inflammatory flavonoid which has been widely used as a therapeutic agent in various toxic models. However, few studies have clearly discussed the neuromodulatory effects of naringenin against different neurodegenerative disorders. AIM: We investigated the neuroprotective efficacy of naringenin against 3-nitropropionic acid (3-NP)-induced neurobehavioral, biochemical and histopathological alterations in rats. METHODS: Albino Wistar rats were randomly divided into three experimental groups. Group 1, the vehicle administered group, received saline. Group 2 received 3-NP (20 mg/kg body weight, i.p.) for 4 consecutive days. Group 3 received naringenin (50 mg/kg body weight, p.o.) twice daily for a period of 4 days, 30 min before and 6 h after the 3-NP administration. On the 5th day, neurobehavioral experiments were performed to access the behavioral outcomes and the striatum tissue was used for analysis of the monoamine oxidase (MAO) activity and serotonin (5-HT) levels. In addition, astrocytes activation was observed by glial fibrillary acidic protein (GFAP) immunostaining. RESULTS: Our results showed that naringenin co-treatment provides neuroprotection against 3-NP-induced neurological disorders. Naringenin also increased the MAO activity and 5-HT levels in the striatum. Moreover, co-treatment with naringenin reduced the expression of GFAP protein in the striatal part and significantly attenuated the neuronal cell death. The findings of the present study suggest that naringenin provides neuroprotection and mitigates neurobehavioral alterations in experimental rats. CONCLUSION: The results show that co-treatment with naringenin ameliorates 3-NP-induced HD-like symptoms in rats.


Assuntos
Flavanonas , Doença de Huntington , Fármacos Neuroprotetores , Animais , Antioxidantes/uso terapêutico , Peso Corporal , Corpo Estriado , Modelos Animais de Doenças , Flavanonas/uso terapêutico , Proteína Glial Fibrilar Ácida/metabolismo , Doença de Huntington/induzido quimicamente , Doença de Huntington/tratamento farmacológico , Doença de Huntington/prevenção & controle , Monoaminoxidase/metabolismo , Monoaminoxidase/farmacologia , Monoaminoxidase/uso terapêutico , Atividade Motora , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Nitrocompostos/toxicidade , Propionatos/toxicidade , Ratos , Ratos Wistar , Serotonina/metabolismo
14.
J Geriatr Psychiatry Neurol ; 35(5): 639-654, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34964392

RESUMO

BACKGROUND: Orthostatic hypotension (OH) is multifactorial in Parkinson's disease (PD). Antiparkinsonian medication can contribute to OH, leading to increased risk of falls, weakness and fatigue. METHODS: We conducted a systematic review and meta-analysis of randomised controlled trials (RCTs) of antiparkinsonian drugs associated with OH as an adverse effect, compared to placebo. We searched EMBASE, MEDLINE and Web of Science databases until November 2020. Analysis used fixed-effects models and the GRADE tool to rate quality of evidence. Meta-analysis was performed if 3 or more studies of a drug group were available. RESULTS: Twenty-one RCTs including 3783 patients were included comparing 6 PD drug groups to placebo (MAO-B inhibitors, dopamine agonists, levodopa, COMT inhibitors, levodopa and adenosine receptor antagonists). OH was recorded as an adverse event or measurement of vital signs, without further specification on how this was defined or operationalised. Meta-analysis was performed for MAO-B inhibitors and dopamine agonists, as there were 3 or more studies for these drug groups. In this analysis, compared with placebo, neither MAO-B inhibitors or dopamine agonists were associated with increased risk of OH, (OR 2.28 [95% CI:0.81-6.46]), (OR 1.39 [95% CI:0.97-1.98]). CONCLUSIONS: Most studies did not specifically report OH, or reporting of OH was limited, including how and when it was measured. Furthermore, studies specifically reporting OH included participants that were younger than typical PD populations without multimorbidity. Future trials should address this, for example,, by including individuals over the age of 75, to improve estimations of how antiparkinsonian medications affect risk of OH.


Assuntos
Hipotensão Ortostática , Doença de Parkinson , Antiparkinsonianos/efeitos adversos , Agonistas de Dopamina/efeitos adversos , Humanos , Hipotensão Ortostática/induzido quimicamente , Hipotensão Ortostática/complicações , Hipotensão Ortostática/tratamento farmacológico , Levodopa/efeitos adversos , Monoaminoxidase/uso terapêutico , Doença de Parkinson/complicações , Doença de Parkinson/tratamento farmacológico
15.
Brain Res Bull ; 168: 165-177, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33387637

RESUMO

The discovery and development of safinamide, an alpha-aminoamide, has been a valuable addition to the existing clinical management of Parkinson's disease (PD). The journey of safinamide dates back to the year 1983, when an alpha-aminoamide called milacemide showed a weak anticonvulsant activity. Milacemide was then structurally modified to give rise to safinamide, which in turn produced robust anticonvulsant activity. The underlying mechanism behind this action of safinamide is attributed to the inhibition of voltage gated calcium and sodium channels. Moreover, owing to the importance of ion channels in maintaining neuronal circuitry and neurotransmitter release, numerous studies explored the potential of safinamide in neurological diseases including PD, stroke, multiple sclerosis and neuromuscular disorders such as Duchenne muscular dystrophy and non-dystrophic myotonias. Nevertheless, evidence from multiple preclinical studies suggested a potent, selective and reversible inhibitory activity of safinamide against monoamine oxidase (MAO)-B enzyme which is responsible for degrading dopamine, a neurotransmitter primarily implicated in the pathophysiology of PD. Therefore, clinical studies were conducted to assess safety and efficacy of safinamide in PD. Indeed, results from various Phase 3 clinical trials suggested strong evidence of safinamide as an add-on therapy in controlling the exacerbation of PD. This review presents a thorough developmental history of safinamide in PD and provides comprehensive insight into plausible mechanisms via which safinamide can be explored in other neurological and muscular diseases.


Assuntos
Alanina/análogos & derivados , Antiparkinsonianos/farmacologia , Benzilaminas/farmacologia , Doenças do Sistema Nervoso/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Alanina/farmacologia , Animais , Anticonvulsivantes/uso terapêutico , Humanos , Monoaminoxidase/efeitos dos fármacos , Monoaminoxidase/uso terapêutico
16.
Biomed Pharmacother ; 131: 110712, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32916539

RESUMO

Cardiovascular disease (CVD) is prevalent worldwide and remains a leading cause of death. Although substantial progress has been made in the diagnosis and treatment of CVD, the prognosis remains unsatisfactory. Renalase is a newly discovered cytokine that is synthesized by the kidney and then secreted into blood. Numerous studies have suggested the efficacy of renalase in treating CVD by metabolizing catecholamines in the circulatory system. As a new biomarker of heart disease, renalase is normally recognized as a signalling molecule that activates cytoprotective intracellular signals to lower blood pressure, protect ischaemic heart muscle and promote atherosclerotic plaque stability in CVD, which subsequently improves cardiac function. Due to its important regulatory role in the circulatory system, renalase has gradually become a potential target in the treatment of CVD. This review summarizes the structure, mechanism and function of renalase in CVD, thereby providing preclinical evidence for alternative approaches and new prospects in the development of renalase-related drugs against CVD.


Assuntos
Doenças Cardiovasculares/etiologia , Monoaminoxidase/fisiologia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Doença da Artéria Coronariana/etiologia , Insuficiência Cardíaca/etiologia , Humanos , Hipertensão/etiologia , Monoaminoxidase/química , Monoaminoxidase/uso terapêutico
17.
Bioorg Chem ; 101: 104032, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32599370

RESUMO

The aim of this study was to effectively obtain monoamine oxidase A (MAO-A) inhibitory peptides from in vitro simulated gastrointestinal digestion and to assess the correspondences between in silico prediction and in vitro confirmation. Fractions (<3 kDa) from ultrafiltration of pepsin and simulated gastrointestinal enzymes hydrolysates exhibited the highest MAO-A inhibitory activity with IC50 values of 0.61 and 2.54 mg/mL, respectively. After sequencing and then screening by HPEPDOCK, 11 high-score peptides and 2 low-score peptides were selected for further synthesis. Remarkable correlation was found between (-)docking scores and MAO-A inhibitory activity of the synthesized peptides, and among which VVFEVFW showed the highest MAO-A inhibitory activity (IC50 = 0.405 mM). Current research suggested that in silico is an effective method to screen MAO-A inhibitory peptides from hairtail protein hydrolysates, and these peptides can be used as functional ingredients for MAO-A inhibition or potential alternatives for antidepressant.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Monoaminoxidase/uso terapêutico , Animais , Simulação por Computador , Peixes , Humanos , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade
18.
Curr Pharm Biotechnol ; 21(6): 528-541, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31820688

RESUMO

BACKGROUND/AIMS: This case series presents the novel Genetic Addiction Risk Score (GARS®) coupled with a customized pro-dopamine regulator matched to polymorphic reward genes having a hypodopaminergic risk. METHODS: The proband is a female with a history of drug abuse and alcoholism. She experienced a car accident under the influence and voluntarily entered treatment. Following an assessment, she was genotyped using the GARS, and started a neuronutrient with a KB220 base indicated by the identified polymorphisms. She began taking it in April 2018 and continues. RESULTS: She had success in recovery from Substance Use Disorder (SUD) and improvement in socialization, family, economic status, well-being, and attenuation of Major Depression. She tested negative over the first two months in treatment and a recent screening. After approximately two months, her parents also decided to take the GARS and started taking the recommended variants. The proband's father (a binge drinker) and mother (no SUD) both showed improvement in various behavioral issues. Finally, the proband's biological children were also GARS tested, showing a high risk for SUD. CONCLUSION: This three-generation case series represents an example of the impact of genetic information coupled with an appropriate DNA guided "Pro-Dopamine Regulator" in recovery and enhancement of life.


Assuntos
Comportamento Aditivo/genética , Dopamina/deficiência , Dopamina/genética , Transtornos Relacionados ao Uso de Substâncias/genética , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/psicologia , Catecolaminas/uso terapêutico , Criança , Feminino , Predisposição Genética para Doença , Humanos , Monoaminoxidase/uso terapêutico , Neprilisina/uso terapêutico , Núcleo Familiar , Polimorfismo Genético , Recompensa , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/psicologia
19.
Life Sci ; 222: 78-87, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30797821

RESUMO

AIMS: Acute kidney injury (AKI) can limit the clinical use of cisplatin in cancer treatment. The drivers of cisplatin-induced AKI include oxidative stress, mitochondrial dysfunction and apoptosis. Previous studies showed renalase protected cultured human renal proximal tubular cell (HK-2) against cisplatin induced necrosis, and renalase-knockout mice subjected to cisplatin showed exacerbated kidney injury. Therefore, it is necessary to determine the exact mechanisms of renalase in cisplatin-induced nephrotoxicity. MAIN METHODS: To study the protective effect of renalase on cell viability, renal function, apoptosis, reactive oxygen species (ROS) production and mitochondrial dynamics, cultured HK-2 cells and male mice were subjected to cisplatin. Signaling proteins related to apoptosis, survival, and mitochondrial fission were analyzed by Western blot. KEY FINDINGS: In this study, we showed that the protective effect of recombinant renalase in cisplatin-induced AKI was associated with the regulation of ROS production, mitochondrial dynamics and sirtuin-3 (Sirt3) levels in vivo and in vitro. After cisplatin treatment, recombinant renalase restored Sirt3 expression, reduced mitochondrial fission and ROS generation. In HK-2 cells, downregulation of endogenous Sirt3 expression by siRNA transfection abrogated the renalase cytoprotection. SIGNIFICANCE: Our study suggests that renalase protects against cisplatin-induced AKI by improving mitochondrial function and inhibiting oxidative stress, and in vitro, it functions in a Sirt3-dependent manner.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Cisplatino/toxicidade , Dinâmica Mitocondrial/efeitos dos fármacos , Monoaminoxidase/uso terapêutico , Sirtuína 3/biossíntese , Injúria Renal Aguda/induzido quimicamente , Animais , Antineoplásicos/toxicidade , Linhagem Celular , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dinâmica Mitocondrial/fisiologia , Monoaminoxidase/farmacologia , Distribuição Aleatória , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/antagonistas & inibidores
20.
J Behav Addict ; 7(1): 192-203, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29316800

RESUMO

Background Addictive-like behaviors (e.g., hoarding and shopping) may be the result of the cumulative effects of dopaminergic and other neurotransmitter genetic variants as well as elevated stress levels. We, therefore, propose that dopamine homeostasis may be the preferred goal in combating such challenging and unwanted behaviors, when simple dopaminergic activation through potent agonists may not provide any resolution. Case presentation C.J. is a 38-year-old, single, female, living with her mother. She has a history of substance use disorder as well as attention deficit hyperactivity disorder, inattentive type. She had been stable on buprenorphine/naloxone combination and amphetamine, dextroamphetamine mixed salts for many years when unexpectedly she lost her job for oversleeping and not calling into work. KB200z (a pro-dopamine compound) was added to her regimen for complaints of low drive and motivation. After taking this nutraceutical for 4 weeks, she noticed a marked improvement in her mental status and many behaviors. She noted that her shopping and hoarding addictions had appreciably decreased. Furthermore, her lifelong history of terrifying lucid dreams was eliminated. Finally, she felt more in control; her locus of control shifted from external to more internal. Discussion The hypothesis is that C.J.'s reported, behavioral, and psychological benefits resulted from the pro-dopamine-regulating effect of KB220Z across the brain reward system. Conclusions This effect, we surmise, could be the result of a new dopamine balance, across C.J.'s brain reward system. Dopamine homeostasis is an effect of KB220Z seen in both animal and human placebo-controlled fMRI experiments.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Catecolaminas/uso terapêutico , Dopaminérgicos/uso terapêutico , Colecionismo/tratamento farmacológico , Monoaminoxidase/uso terapêutico , Neprilisina/uso terapêutico , Psicotrópicos/uso terapêutico , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Adulto , Transtorno do Deficit de Atenção com Hiperatividade/complicações , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Feminino , Colecionismo/complicações , Humanos , Transtornos Relacionados ao Uso de Substâncias/complicações , Transtornos Relacionados ao Uso de Substâncias/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA