Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Medicina (Kaunas) ; 59(6)2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37374342

RESUMO

Background and Objectives: Parkinson's disease (PD) is a clinically heterogeneous disorder with poorly understood pathological contributing factors. Depression presents one of the most frequent non-motor PD manifestations, and several genetic polymorphisms have been suggested that could affect the depression risk in PD. Therefore, in this review we have collected recent studies addressing the role of genetic factors in the development of depression in PD, aiming to gain insights into its molecular pathobiology and enable the future development of targeted and effective treatment strategies. Materials and Methods: we have searched PubMed and Scopus databases for peer-reviewed research articles published in English (pre-clinical and clinical studies as well as relevant reviews and meta-analyses) investigating the genetic architecture and pathophysiology of PD depression. Results: in particular, polymorphisms in genes related to the serotoninergic pathway (sodium-dependent serotonin transporter gene, SLC6A4, tryptophan hydrolase-2 gene, TPH2), dopamine metabolism and neurotransmission (dopamine receptor D3 gene, DRD3, aldehyde dehydrogenase 2 gene, ALDH2), neurotrophic factors (brain-derived neurotrophic factor gene, BDNF), endocannabinoid system (cannabinoid receptor gene, CNR1), circadian rhythm (thyrotroph embryonic factor gene, TEF), the sodium-dependent neutral amino acid transporter B(0)AT2 gene, SLC6A15), and PARK16 genetic locus were detected as altering susceptibility to depression among PD patients. However, polymorphisms in the dopamine transporter gene (SLC6A3), monoamine oxidase A (MAOA) and B (MAOB) genes, catechol-O-methyltransferase gene (COMT), CRY1, and CRY2 have not been related to PD depression. Conclusions: the specific mechanisms underlying the potential role of genetic diversity in PD depression are still under investigation, however, there is evidence that they may involve neurotransmitter imbalance, mitochondrial impairment, oxidative stress, and neuroinflammation, as well as the dysregulation of neurotrophic factors and their downstream signaling pathways.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros , Doença de Parkinson , Humanos , Doença de Parkinson/complicações , Doença de Parkinson/genética , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/uso terapêutico , Depressão/genética , Polimorfismo Genético , Fatores de Crescimento Neural , Predisposição Genética para Doença , Aldeído-Desidrogenase Mitocondrial/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas do Tecido Nervoso/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/uso terapêutico
2.
J Neurosci Res ; 100(2): 413-443, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34713909

RESUMO

Epilepsy is a devastating neurological disorder characterized by recurrent seizures attributed to the disruption of the dynamic excitatory and inhibitory balance in the brain. Epilepsy has emerged as a global health concern affecting about 70 million people worldwide. Despite recent advances in pre-clinical and clinical research, its etiopathogenesis remains obscure, and there are still no treatment strategies modifying disease progression. Although the precise molecular mechanisms underlying epileptogenesis have not been clarified yet, the role of ion channels as regulators of cellular excitability has increasingly gained attention. In this regard, emerging evidence highlights the potential implication of inwardly rectifying potassium (Kir) channels in epileptogenesis. Kir channels consist of seven different subfamilies (Kir1-Kir7), and they are highly expressed in both neuronal and glial cells in the central nervous system. These channels control the cell volume and excitability. In this review, we discuss preclinical and clinical evidence on the role of the several subfamilies of Kir channels in epileptogenesis, aiming to shed more light on the pathogenesis of this disorder and pave the way for future novel therapeutic approaches.


Assuntos
Epilepsia , Canais de Potássio Corretores do Fluxo de Internalização , Epilepsia/tratamento farmacológico , Humanos , Neurônios , Potássio , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Convulsões
3.
Cell Mol Life Sci ; 78(4): 1445-1453, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33052436

RESUMO

The exact etiology of Parkinson's disease (PD) remains obscure, lacking effective diagnostic and prognostic biomarkers. In search of novel molecular factors that may contribute to PD pathogenesis, emerging evidence highlights the multifunctional role of the calcium-binding protein S100B that is widely expressed in the brain and predominantly in astrocytes. Preclinical evidence points towards the possible time-specific contributing role of S100B in the pathogenesis of neurodegenerative disorders including PD, mainly by regulating neuroinflammation and dopamine metabolism. Although existing clinical evidence presents some contradictions, estimation of S100B in the serum and cerebrospinal fluid seems to hold a great promise as a potential PD biomarker, particularly regarding the severity of motor and non-motor PD symptoms. Furthermore, given the recent development of S100B inhibitors that are able to cross the blood brain barrier, novel opportunities are arising in the research field of PD therapeutics. In this review, we provide an update on recent advances in the implication of S100B protein in the pathogenesis of PD and discuss relevant studies investigating the biomarker potential of S100B in PD, aiming to shed more light on clinical targeting approaches related to this incurable disorder.


Assuntos
Encéfalo/metabolismo , Doenças Neurodegenerativas/genética , Doença de Parkinson/genética , Subunidade beta da Proteína Ligante de Cálcio S100/genética , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Encéfalo/patologia , Dopamina/genética , Dopamina/metabolismo , Humanos , Doenças Neurodegenerativas/patologia , Doença de Parkinson/patologia
4.
J Neurosci Res ; 99(9): 2117-2133, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34115895

RESUMO

The pathogenesis of Parkinson's disease (PD) remains elusive. There is still no available disease-modifying strategy against PD, whose management is mainly symptomatic. A growing amount of preclinical evidence shows that a complex interplay between autophagy dysregulation, mitochondrial impairment, endoplasmic reticulum stress, oxidative stress, and excessive neuroinflammation underlies PD pathogenesis. Identifying key molecules linking these pathological cellular processes may substantially aid in our deeper understanding of PD pathophysiology and the development of novel effective therapeutic approaches. Emerging preclinical evidence indicates that apelin, an endogenous neuropeptide acting as a ligand of the orphan G protein-coupled receptor APJ, may play a key neuroprotective role in PD pathogenesis, via inhibition of apoptosis and dopaminergic neuronal loss, autophagy enhancement, antioxidant effects, endoplasmic reticulum stress suppression, as well as prevention of synaptic dysregulation in the striatum, excessive neuroinflammation, and glutamate-induced excitotoxicity. Underlying signaling pathways involve phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin, extracellular signal-regulated kinase 1/2, and inositol requiring kinase 1α/XBP1/C/EBP homologous protein. Herein, we discuss the role of apelin/APJ axis and associated molecular mechanisms on the pathogenesis of PD in vitro and in vivo and provide evidence for its challenging therapeutic potential.


Assuntos
Antiparkinsonianos/metabolismo , Receptores de Apelina/metabolismo , Apelina/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Animais , Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , Apelina/farmacologia , Apelina/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Epilepsy Behav ; 115: 107701, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33412369

RESUMO

Epilepsy is a devastating neurological disorder characterized by the repeated occurrence of epileptic seizures. Epilepsy stands as a global health concern affecting around 70 million people worldwide. The mainstream antiepileptic drugs (AEDs) only exert symptomatic relief and drug-resistant epilepsy occurs in up to 33 percent of patients. Hence, the investigation of novel therapeutic strategies against epileptic seizures that could exert disease modifying effects is of paramount importance. In this context, compounds of natural origin with potential antiepileptic properties have recently gained increasing attention. Quercetin is a plant-derived flavonoid with several pharmacological activities. Emerging evidence has demonstrated the antiepileptic potential of quercetin as well. Herein, based on the available evidence, we discuss the neuroprotective effects of quercetin against epileptic seizures and further analyze the plausible underlying molecular mechanisms. Our review suggests that quercetin might be a potential therapeutic candidate against epilepsy that deserves further investigation, and paves the way for the development of plant-derived antiepileptic treatment approaches.


Assuntos
Epilepsia , Fármacos Neuroprotetores , Anticonvulsivantes/uso terapêutico , Epilepsia/tratamento farmacológico , Humanos , Fármacos Neuroprotetores/uso terapêutico , Quercetina/uso terapêutico , Convulsões/tratamento farmacológico
6.
Molecules ; 26(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34770864

RESUMO

Chrysin, a herbal bioactive molecule, exerts a plethora of pharmacological effects, including anti-oxidant, anti-inflammatory, neuroprotective, and anti-cancer. A growing body of evidence has highlighted the emerging role of chrysin in a variety of neurological disorders, including Alzheimer's and Parkinson's disease, epilepsy, multiple sclerosis, ischemic stroke, traumatic brain injury, and brain tumors. Based on the results of recent pre-clinical studies and evidence from studies in humans, this review is focused on the molecular mechanisms underlying the neuroprotective effects of chrysin in different neurological diseases. In addition, the potential challenges, and opportunities of chrysin's inclusion in the neurotherapeutics repertoire are critically discussed.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Flavonoides/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Antineoplásicos/química , Flavonoides/química , Humanos , Estrutura Molecular , Fármacos Neuroprotetores/química
7.
Pharmacol Res ; 152: 104593, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31843673

RESUMO

Parkinson's disease (PD) is a devastating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and Lewy pathology. PD is a major concern of today's aging population and has emerged as a global health burden. Despite the rapid advances in PD research over the past decades, the gold standard therapy provides only symptomatic relief and fails to halt disease progression. Therefore, exploring novel disease-modifying therapeutic strategies is highly demanded. Metformin, which is currently used as a first-line therapy for type 2 diabetes mellitus (T2DM), has recently demonstrated to exert a neuroprotective role in several neurodegenerative disorders including PD, both in vitro and in vivo. In this review, we explore the neuroprotective potential of metformin based on emerging evidence from pre-clinical and clinical studies. Regarding the underlying molecular mechanisms, metformin has been shown to inhibit α-synuclein (SNCA) phosphorylation and aggregation, prevent mitochondrial dysfunction, attenuate oxidative stress, modulate autophagy mainly via AMP-activated protein kinase (AMPK) activation, as well as prevent neurodegeneration and neuroinflammation. Overall, the neuroprotective effects of metformin in PD pathogenesis present a novel promising therapeutic strategy that might overcome the limitations of current PD treatment.


Assuntos
Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Animais , Autofagia/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Encefalite/tratamento farmacológico , Humanos , Mitocôndrias/efeitos dos fármacos
8.
Pharmacol Res ; 158: 104930, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32445958

RESUMO

Neuroinflammation plays a crucial role in the pathogenesis of Parkinson's disease (PD) with the dysregulation of microglial activity being tightly linked to dopaminergic degeneration. Fractalkine (CX3CL1), a chemokine mainly expressed by neurons, can modulate microglial activity through binding to its sole G-protein-coupled receptor (CX3CR1), expressed by microglia. Fractalkine/CX3CR1 signaling is one of the most important mediators of the communication between neurons and microglia, and its emerging role in neurodegenerative disorders including PD has been increasingly recognized. Pre-clinical evidence has revealed that fractalkine signaling axis exerts dual effects on PD-related inflammation and degeneration, which greatly depend on the isoform type (soluble or membrane-bound), animal model (mice or rats, toxin- or proteinopathy-induced), route of toxin administration, time course and specific brain region (striatum, substantia nigra). Furthermore, although existing clinical evidence is scant, it has been indicated that fractalkine may be possibly associated with PD progression, paving the way for future studies investigating its biomarker potential. In this review, we discuss recent evidence on the role of fractalkine/CX3CR1 signaling axis in PD pathogenesis, aiming to shed more light on the molecular mechanisms underlying the neuroinflammation commonly associated with the disease, as well as potential clinical and therapeutic implications.


Assuntos
Antiparkinsonianos/uso terapêutico , Quimiocina CX3CL1/metabolismo , Mediadores da Inflamação/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antiparkinsonianos/farmacologia , Quimiocina CX3CL1/genética , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Microglia/efeitos dos fármacos , Microglia/metabolismo , Doença de Parkinson/genética , Transdução de Sinais/fisiologia , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
9.
Pharmacol Res ; 156: 104792, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32278047

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating and rapidly progressing neurodegenerative disorder with no effective disease-modifying treatment up to date. The underlying molecular mechanisms of ALS are not yet completely understood. However, the critical role of the innate immune system and neuroinflammation in ALS pathogenesis has gained increased attention. High mobility group box 1 (HMGB1) is a typical damage-associated molecular pattern (DAMP) molecule, acting as a pro-inflammatory cytokine mainly through activation of its principal receptors, the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) which are crucial components of the innate immune system. HMGB1 is an endogenous ligand for both RAGE and TLR4 that mediate its biological effects. Herein, on the ground of pre-clinical findings we unravel the underlying mechanisms behind the plausible contribution of HMGB1 and its receptors (RAGE and TLR4) in the ALS pathogenesis. Furthermore, we provide an account of the therapeutic outcomes associated with inhibition/blocking of HMGB1 receptor signalling in preventing motor neuron's death and delaying disease progression in ALS experimental models. There is strong evidence that HMGB1, RAGE and TLR4 signaling axes might present potential targets against ALS, opening a novel headway in ALS research that could plausibly bridge the current treatment gap.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Neurônios Motores/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Medula Espinal/metabolismo , Receptor 4 Toll-Like/metabolismo , Esclerose Lateral Amiotrófica/imunologia , Esclerose Lateral Amiotrófica/patologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Humanos , Imunidade Inata , Ligantes , Neurônios Motores/imunologia , Neurônios Motores/patologia , Transdução de Sinais , Medula Espinal/imunologia , Medula Espinal/patologia
10.
Pharmacol Res ; 160: 105172, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32871246

RESUMO

Understanding the interplay between the innate immune system, neuroinflammation, and epilepsy might offer a novel perspective in the quest of exploring new treatment strategies. Due to the complex pathology underlying epileptogenesis, no disease-modifying treatment is currently available that might prevent epilepsy after a plausible epileptogenic insult despite the advances in pre-clinical and clinical research. Neuroinflammation underlies the etiopathogenesis of epilepsy and convulsive disorders with Toll-like receptor (TLR) signal transduction being highly involved. Among TLR family members, TLR4 is an innate immune system receptor and lipopolysaccharide (LPS) sensor that has been reported to contribute to epileptogenesis by regulating neuronal excitability. Herein, we discuss available evidence on the role of TLR4 and its endogenous ligands, the high mobility group box 1 (HMGB1) protein, the heat shock proteins (HSPs) and the myeloid related protein 8 (MRP8), in epileptogenesis and post-traumatic epilepsy (PTE). Moreover, we provide an account of the promising findings of TLR4 modulation/inhibition in experimental animal models with therapeutic impact on seizures.


Assuntos
Epilepsia/fisiopatologia , Imunidade Inata , Receptor 4 Toll-Like/efeitos dos fármacos , Animais , Epilepsia/patologia , Humanos , Inflamação/complicações , Inflamação/patologia , Ligantes
11.
Int J Mol Sci ; 21(13)2020 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-32610502

RESUMO

Brain injuries are devastating conditions, representing a global cause of mortality and morbidity, with no effective treatment to date. Increased evidence supports the role of neuroinflammation in driving several forms of brain injuries. High mobility group box 1 (HMGB1) protein is a pro-inflammatory-like cytokine with an initiator role in neuroinflammation that has been implicated in Traumatic brain injury (TBI) as well as in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Herein, we discuss the implication of HMGB1-induced neuroinflammatory responses in these brain injuries, mediated through binding to the receptor for advanced glycation end products (RAGE), toll-like receptor4 (TLR4) and other inflammatory mediators. Moreover, we provide evidence on the biomarker potential of HMGB1 and the significance of its nucleocytoplasmic translocation during brain injuries along with the promising neuroprotective effects observed upon HMGB1 inhibition/neutralization in TBI and EBI induced by SAH. Overall, this review addresses the current advances on neuroinflammation driven by HMGB1 in brain injuries indicating a future treatment opportunity that may overcome current therapeutic gaps.


Assuntos
Lesões Encefálicas/metabolismo , Proteína HMGB1/metabolismo , Animais , Lesões Encefálicas/patologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Citocinas/metabolismo , Humanos , Inflamação/metabolismo , Microglia/metabolismo , Neuroimunomodulação , Fármacos Neuroprotetores/farmacologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Receptor 4 Toll-Like/metabolismo
12.
Int J Mol Sci ; 21(7)2020 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-32260203

RESUMO

Epilepsy is a devastating neurological condition exhibited by repeated spontaneous and unpredictable seizures afflicting around 70 million people globally. The basic pathophysiology of epileptic seizures is still elusive, reflecting an extensive need for further research. Developing a novel animal model is crucial in understanding disease mechanisms as well as in assessing the therapeutic target. Most of the pre-clinical epilepsy research has been focused on rodents. Nevertheless, zebrafish disease models are relevant to human disease pathophysiology hence are gaining increased attention nowadays. The current study for the very first time developed a pilocarpine-induced chronic seizure-like condition in adult zebrafish and investigated the modulation in several neuroinflammatory genes and neurotransmitters after pilocarpine exposures. Seizure score analysis suggests that compared to a single dose, repeated dose pilocarpine produces chronic seizure-like effects maintaining an average seizure score of above 2 each day for a minimum of 10 days. Compared to the single dose pilocarpine treated group, there was increased mRNA expression of HMGB1, TLR4, TNF-α, IL-1, BDNF, CREB-1, and NPY; whereas decreased expression of NF-κB was upon the repeated dose of pilocarpine administration. In addition, the epileptic group demonstrates modulation in neurotransmitters levels such as GABA, Glutamate, and Acetylcholine. Moreover, proteomic profiling of the zebrafish brain from the normal and epileptic groups from LCMS/MS quantification detected 77 and 13 proteins in the normal and epileptic group respectively. Summing up, the current investigation depicted that chemically induced seizures in zebrafish demonstrated behavioral and molecular alterations similar to classical rodent seizure models suggesting the usability of adult zebrafish as a robust model to investigate epileptic seizures.


Assuntos
Redes Reguladoras de Genes/efeitos dos fármacos , Pilocarpina/efeitos adversos , Proteômica/métodos , Convulsões/genética , Animais , Cromatografia Líquida , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , NF-kappa B/genética , Neurotransmissores/metabolismo , Pilocarpina/administração & dosagem , Convulsões/induzido quimicamente , Convulsões/metabolismo , Espectrometria de Massas em Tandem , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
J Neurochem ; 151(5): 542-557, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30644560

RESUMO

Epilepsy is a serious neurological condition exhibiting complex pathology and deserving of more serious attention. More than 30% of people with epilepsy are not responsive to more than 20 anti-epileptic drugs currently available, reflecting an unmet clinical need for novel therapeutic strategies. Not much is known about the pathogenesis of epilepsy, but evidence indicates that neuroinflammation might contribute to the onset and progression of epilepsy following acquired brain insults. However, the molecular mechanisms underlying these pathophysiological processes are yet to be fully understood. The emerging research suggests that high-mobility group box protein 1 (HMGB1), a DNA-binding protein that is both actively secreted by inflammatory cells and released by necrotic cells, might contribute to the pathogenesis of epilepsy. HMGB1 as an initiator and amplifier of neuroinflammation, and its activation is implicated in the propagation of seizures in animal models. The current review will highlight the potential role of HMGB1 in the pathogenesis of epilepsy, and implications of HMGB1-targeted therapies against epilepsy. HMGB1 in this context is an emerging concept deserving further exploration. Increased understanding of HMGB1 in seizures and epilepsy will pave the way in designing novel and innovative therapeutic strategies that could modify the disease course or prevent its development.


Assuntos
Epilepsia/metabolismo , Proteína HMGB1/metabolismo , Animais , Humanos
14.
Pharmacol Res ; 150: 104515, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31707035

RESUMO

Parkinson's disease (PD) is a multifactorial disorder, attributed to a complex interplay between genetic and epigenetic factors. Although the exact etiology of the disease remains elusive, dysregulation of signaling pathways implicated in cell survival, apoptosis, protein aggregation, mitochondrial dysfunction, autophagy, oxidative damage and neuroinflammation, contributes to its pathogenesis. MicroRNAs (miRs) are endogenous short non-coding RNA molecules that negatively regulate gene expression at a post-transcriptional level. MiR-124 is one of the most abundantly expressed miRs in the brain that participates in neurogenesis, synapse morphology, neurotransmission, inflammation, autophagy and mitochondrial function. Accumulating pre-clinical evidence shows that miR-124 may act through calpain 1/p25/cyclin-dependent kinases 5 (CDK5), nuclear factor-kappa B (NF-κB), signal transducer and activator of transcription 3 (STAT3), Bcl-2-interacting mediator of cell death (Bim), 5' adenosine monophosphate-activated protein kinase (AMPK) and extracellular signal-regulated kinase (ERK)-mediated pathways to regulate cell survival, apoptosis, autophagy, mitochondrial dysfunction, oxidative damage and neuroinflammation in PD. Moreover, clinical evidence indicates that reduced plasma miR-124 levels may serve as a potential diagnostic biomarker in PD. This review provides an update of the pathogenic implication of miR-124 activity in PD and discusses its targeting potential for the development of future therapeutic strategies.


Assuntos
MicroRNAs , Doença de Parkinson/genética , Animais , Biomarcadores/sangue , Humanos , MicroRNAs/sangue , Neuroproteção , Doença de Parkinson/sangue , Doença de Parkinson/tratamento farmacológico
15.
J Bone Miner Metab ; 37(1): 36-42, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29380075

RESUMO

In view of the opposite effects of gut and brain serotonin in bone, the key role of Wnt ß/catenin pathway in osteoblastic proliferation and the controversial bony effects of selective serotonin reuptake inhibitors antidepressants, the present study investigated the effects of escitalopram alone and in combination with carbidopa (to block gut-derived serotonin) on markers of bone turnover and Wnt signaling and micro-CT in male Wistar rats. Escitalopram (2.0 mg/kg, p.o.) and carbidopa (10 mg/kg, p.o.) were administered daily for 40 days following which indicators of reduced (dickkopf-1, sclerostin), and increased (alkaline phosphatase) bone formation and bone resorption markers (receptor activator of nuclear factor κB ligand, tartrate-resistant acid phosphatase 5b) were determined. Our results indicated that escitalopram adversely affected bone as indicated by reduced bone formation and enhanced bone resorption. Further, the effects of escitalopram on bone formation were possibly mediated through gut serotonin while the mechanisms responsible for effects on resorption seem unrelated to gut serotonin. The promising effects of carbidopa on bone formation, as observed in our study, open up exciting possibilities for this drug requiring further investigations.


Assuntos
Biomarcadores/metabolismo , Osso e Ossos/metabolismo , Carbidopa/farmacologia , Citalopram/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Densidade Óssea/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Marcadores Genéticos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/fisiologia , Masculino , Ligante RANK/sangue , Ratos Wistar , Microtomografia por Raio-X
16.
Brain Sci ; 12(9)2022 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-36138926

RESUMO

Neurodegenerative disorders remain a major burden for our society, affecting millions of people worldwide [...].

17.
ACS Pharmacol Transl Sci ; 5(10): 838-848, 2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36268117

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease (AD), and its pathogenesis remains obscure. Current treatment approaches mainly including levodopa and dopamine agonists provide symptomatic relief but fail to halt disease progression, and they are often accompanied by severe side effects. In this context, natural phytochemicals have received increasing attention as promising preventive or therapeutic candidates for PD, given their multitarget pharmaceutical mechanisms of actions and good safety profile. Ginger (Zingiber officinale Roscoe, Zingiberaceae) is a very popular spice used as a medicinal herb throughout the world since the ancient years, for a wide range of conditions, including nausea, diabetes, dyslipidemia, and cancer. Emerging in vivo and in vitro evidence supports the neuroprotective effects of ginger and its main pharmaceutically active compounds (zingerone, 6-shogaol, and 6-gingerol) in PD, mainly via the regulation of neuroinflammation, oxidative stress, intestinal permeability, dopamine synaptic transmission, and possibly mitochondrial dysfunction. The regulation of several transcription factors and signaling pathways, including nuclear factor kappa B (NF-κB), p38 mitogen-activated protein kinase (MAPK), phosphatidylinositol-3-kinase (PI3K)/Ak strain transforming (Akt), extracellular signal-regulated kinase (ERK) 1/2, and AMP-activated protein kinase (AMPK)/proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) have been shown to contribute to the protective effects of ginger. Herein, we discuss recent findings on the beneficial role of ginger in PD as a preventive agent or potential supplement to current treatment strategies, focusing on potential underlying molecular mechanisms.

18.
Brain Sci ; 12(2)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35203939

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder with an unclear etiology and no disease-modifying treatment to date. PD is considered a multifactorial disease, since both genetic and environmental factors contribute to its pathogenesis, although the molecular mechanisms linking these two key disease modifiers remain obscure. In this context, epigenetic mechanisms that alter gene expression without affecting the DNA sequence through DNA methylation, histone post-transcriptional modifications, and non-coding RNAs may represent the key mediators of the genetic-environmental interactions underlying PD pathogenesis. Environmental exposures may cause chemical alterations in several cellular functions, including gene expression. Emerging evidence has highlighted that smoking, coffee consumption, pesticide exposure, and heavy metals (manganese, arsenic, lead, etc.) may potentially affect the risk of PD development at least partially via epigenetic modifications. Herein, we discuss recent accumulating pre-clinical and clinical evidence of the impact of lifestyle and environmental factors on the epigenetic mechanisms underlying PD development, aiming to shed more light on the pathogenesis and stimulate future research.

19.
Curr Res Neurobiol ; 3: 100032, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518345

RESUMO

Embelin is a neuroprotective compound with therapeutic benefit against experimental Alzheimer's disease (AD)-like condition. In the quest of untangling the underlying mechanism behind the neuroprotective effect of Embelin in AD, an in-vitro study of Embelin against neuronal damage induced by Streptozotocin (STZ) in rat hippocampal neuronal culture was performed. Current findings demonstrated that Embelin (2.5-10 µM) has efficiently protected hippocampal neurons against STZ (8 mM)-induced neurotoxicity. An increase in amyloid precursor protein (APP), microtubule-associated protein tau (MAPT), glycogen synthase kinase 3 alpha (GSK-3α) and glycogen synthase kinase 3 beta (GSK-3ß) expression levels was observed when STZ (8 mM) stimulation was done for 24 h in the hippocampal neurons. A significant downregulation in the mRNA expression levels of APP, MAPT, GSK-3α, and GSK-3ß upon pre-treatment with different doses of Embelin (2.5 µM, 5 µM and 10 µM) reflects that Embelin attenuated STZ-induced dysfunction of insulin signaling (IR). Embelin significantly modulated the mRNA expression of scavenger enzyme Superoxide dismutase (SOD1). Furthermore, STZ had significantly upregulates an expression of Aß. On the contrary, pre-treatment with three doses of Embelin reversed an Aß-induced neuronal death. Our findings suggest that, Embelin prevents Aß accumulation via SOD1 pathway to protect against AD-like condition.

20.
J Ethnopharmacol ; 292: 115234, 2022 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-35358621

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Parkinson's disease (PD) is the second most devastating age-related neurodegenerative diseases after Alzheimer diseases (AD) and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN) and aggregation of α-synuclein (α-syn). The precise etiology of PD is not yet fully understood and lacks the disease-modifying therapeutic strategies that could reverse the ongoing neurodegeneration. In the quest of exploring novel disease modifying therapeutic strategies, natural compounds from plant sources have gained much attention in recent days. Glycyrrhizin (GL) is the main active ingredient of the roots and rhizomes of licorice (Glycyrrhiza glabra L), which are generally used in the treatment of inflammatory diseases or as a tonifying herbal medicine. In Persia, GL is a conventional neuroprotective agent that are used to treat neurological disorders. The traditional use of GL in Japan is to treat chronic hepatitis B. In addition, GL is a natural inhibitor of high mobility group box 1 (HMGB1) which has exerted neuroprotective effect against several HMGB1 mediated pathological conditions. AIM OF THE STUDY: The study is aimed to evaluate therapeutic effect of GL against PD in zebrafish. MATERIAL AND METHODS: PD in zebrafish larvae is induced by administration of neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Apoptosis was assessed with TUNEL assay. Gene expression was performed to assess the modulation in genes related to neuroinflammatory and autophagy. RESULTS: We observed that GL co-treatment increased the length of DA neurons, decreased the number of apoptotic cells in zebrafish brain, and inhibited the loss of vasculature and disorganized vasculature induced by MPTP. GL co-treatment relieved the MPTP-induced locomotor impairment in zebrafish. GL co-treatment suppressed MPTP-induced upregulated mRNA expression of inflammatory markers such as hmgb1a, tlr4b, nfκb, il1ß, and il6. GL co-treatment suppressed the autophagy related genes α-syn and atg5 whereas increased the mRNA expression level of parkin and pink1. In addition, molecular docking study reveals that GL has binding interaction with HMGB1, TLR4, and RAGE. CONCLUSION: Hence, the effect of GL co-treatment on MPTP-induced PD-like condition in zebrafish is to alleviate apoptosis and autophagy, as well as suppress inflammatory responses.


Assuntos
Proteína HMGB1 , Fármacos Neuroprotetores , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/uso terapêutico , Animais , Modelos Animais de Doenças , Ácido Glicirrízico/farmacologia , Ácido Glicirrízico/uso terapêutico , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Neuroproteção , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , RNA Mensageiro , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA