Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38464024

RESUMO

Mitochondrial dysfunction is a central aspect of Parkinson's disease (PD) pathology, yet the underlying mechanisms are not fully understood. This study investigates the link between α-Synuclein (α-Syn) pathology and the loss of translocase of the outer mitochondrial membrane 40 (TOM40), unraveling its implications for mitochondrial dysfunctions in neurons. We discovered that TOM40 protein depletion occurs in the brains of patients with Guam Parkinsonism Dementia (Guam PD) and cultured neurons expressing α-Syn proteinopathy, notably, without corresponding changes in TOM40 mRNA levels. Cultured neurons expressing α-Syn mutants, with or without a mitochondria-targeting signal (MTS) underscore the role of α-Syn's mitochondrial localization in inducing TOM40 degradation. Parkinson's Disease related etiological factors, such as 6-hydroxy dopamine or ROS/metal ions stress, which promote α-Syn oligomerization, exacerbate TOM40 depletion in PD patient-derived cells with SNCA gene triplication. Although α-Syn interacts with both TOM40 and TOM20 in the outer mitochondrial membrane, degradation is selective for TOM40, which occurs via the ubiquitin-proteasome system (UPS) pathway. Our comprehensive analyses using Seahorse technology, mitochondrial DNA sequencing, and damage assessments, demonstrate that mutant α-Syn-induced TOM40 loss results in mitochondrial dysfunction, characterized by reduced membrane potential, accumulation of mtDNA damage, deletion/insertion mutations, and altered oxygen consumption rates. Notably, ectopic supplementation of TOM40 or reducing pathological forms of α-Syn using ADP-ribosylation inhibitors ameliorate these mitochondrial defects, suggesting potential therapeutic avenues. In conclusion, our findings provide crucial mechanistic insights into how α-Syn accumulation leads to TOM40 degradation and mitochondrial dysfunction, offering insights for targeted interventions to alleviate mitochondrial defects in PD.

2.
Comb Chem High Throughput Screen ; 25(7): 1134-1147, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33645478

RESUMO

BACKGROUND: Red mamey is the fruit of P. sapota, a tree found in Mesoamerica and Asia. This fruit is considered a nutraceutical due to its multiple beneficial health including antiamyloidogenic activity and potential anti-tumorigenic property. Red mamey contain a variety of carotenoids including novel ketocarotenoids such as sapotexanthin and cryptocapsin. A ketocarotenoid is a chemical compound with a carbonyl group present in the ß-ring or in the double bond chain of a carotenoid. In red mamey, the 3'-deoxy-k-end group in sapotexanthin has proven to be an important pro-vitamin A source, which is essential for maintaining a healthy vision and cognitive processes. OBJECTIVE: This work reviews the current knowledge about the chemistry and biological activities of carotenoids in red mamey. METHOD: An exhaustive extraction is the most usual methodology to isolate and thoroughly characterize the carotenoids present in this fruit. High performance liquid chromatography is used to determine the profile of total carotenoids and its purity, while atmospheric pressure chemical ionization was used to determine their molecular weight and nuclear magnetic resonance determined their structure. RESULT: For each 100 g of fresh weight, 0.12 mg of total carotenoid from this fruit can be obtained. Out of the more than 47 reported carotenoids in red mamey, only 34 have a detailed characterization. CONCLUSION: It is important to continue studying the chemical composition and biological activity of this unique tropical fruit with commercial and nutritional value.


Assuntos
Pouteria , Carotenoides/química , Carotenoides/farmacologia , Cromatografia Líquida de Alta Pressão , Frutas , Espectroscopia de Ressonância Magnética , Pouteria/química
4.
Front Psychiatry ; 11: 585893, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33250794

RESUMO

In today's ever-growing concerns about the coronavirus disease (COVID-19) pandemic, many experience sleep insufficiencies, such as difficulty falling or staying asleep, sleep-related behavioral symptoms, and out-of-phase circadian rhythmicity despite the lack of history of earlier such symptoms. Meanwhile, the disruption in sleep bioparameters is experienced more in people with a history of sleep disorders. The behavioral sleep disorders in the current situations are prevalent given the today's amount of anxiety everyone is feeling about COVID-19. On the other hand, evidences indicated that the cross-link between impaired sleep efficiency and disrupted innate immunity makes people susceptible to viral infections. The present brief review highlights the links between psychosocial stress, sleep insufficiency, and susceptibility to viral infections in relevance to COVID-19 situation. The stress management measures, including addressing sleep-related disorders and sleep hygiene, will have a notable impact by harnessing immune response and thus reducing the susceptibility to viral infections.

5.
Proc Natl Acad Sci U S A ; 117(14): 8154-8165, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32205441

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is a dominantly inherited neurodegenerative disease caused by CAG (encoding glutamine) repeat expansion in the Ataxin-3 (ATXN3) gene. We have shown previously that ATXN3-depleted or pathogenic ATXN3-expressing cells abrogate polynucleotide kinase 3'-phosphatase (PNKP) activity. Here, we report that ATXN3 associates with RNA polymerase II (RNAP II) and the classical nonhomologous end-joining (C-NHEJ) proteins, including PNKP, along with nascent RNAs under physiological conditions. Notably, ATXN3 depletion significantly decreased global transcription, repair of transcribed genes, and error-free double-strand break repair of a 3'-phosphate-containing terminally gapped, linearized reporter plasmid. The missing sequence at the terminal break site was restored in the recircularized plasmid in control cells by using the endogenous homologous transcript as a template, indicating ATXN3's role in PNKP-mediated error-free C-NHEJ. Furthermore, brain extracts from SCA3 patients and mice show significantly lower PNKP activity, elevated p53BP1 level, more abundant strand-breaks in the transcribed genes, and degradation of RNAP II relative to controls. A similar RNAP II degradation is also evident in mutant ATXN3-expressing Drosophila larval brains and eyes. Importantly, SCA3 phenotype in Drosophila was completely amenable to PNKP complementation. Hence, salvaging PNKP's activity can be a promising therapeutic strategy for SCA3.


Assuntos
Ataxina-3/genética , Reparo do DNA por Junção de Extremidades , Enzimas Reparadoras do DNA/metabolismo , Doença de Machado-Joseph/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , RNA Polimerase II/metabolismo , Proteínas Repressoras/genética , Idoso de 80 Anos ou mais , Animais , Animais Geneticamente Modificados , Ataxina-3/metabolismo , Encéfalo/patologia , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Modelos Animais de Doenças , Drosophila , Feminino , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Pluripotentes Induzidas , Doença de Machado-Joseph/metabolismo , Doença de Machado-Joseph/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Peptídeos/genética , RNA Interferente Pequeno/metabolismo
6.
ACS Nano ; 14(3): 2827-2846, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32049495

RESUMO

Therapy for intracerebral hemorrhage (ICH) remains elusive, in part dependent on the severity of the hemorrhage itself as well as multiple deleterious effects of blood and its breakdown products such as hemin and free iron. While oxidative injury and genomic damage have been seen following ICH, the details of this injury and implications remain unclear. Here, we discovered that, while free iron produced mostly reactive oxygen species (ROS)-related single-strand DNA breaks, hemin unexpectedly induced rapid and persistent nuclear and mitochondrial double-strand breaks (DSBs) in neuronal and endothelial cell genomes and in mouse brains following experimental ICH comparable to that seen with γ radiation and DNA-complexing chemotherapies. Potentially as a result of persistent DSBs and the DNA damage response, hemin also resulted in senescence phenotype in cultured neurons and endothelial cells. Subsequent resistance to ferroptosis reported in other senescent cell types was also observed here in neurons. While antioxidant therapy prevented senescence, cells became sensitized to ferroptosis. To address both senescence and resistance to ferroptosis, we synthesized a modified, catalytic, and rapidly internalized carbon nanomaterial, poly(ethylene glycol)-conjugated hydrophilic carbon clusters (PEG-HCC) by covalently bonding the iron chelator, deferoxamine (DEF). This multifunctional nanoparticle, DEF-HCC-PEG, protected cells from both senescence and ferroptosis and restored nuclear and mitochondrial genome integrity in vitro and in vivo. We thus describe a potential molecular mechanism of hemin/iron-induced toxicity in ICH that involves a rapid induction of DSBs, senescence, and the consequent resistance to ferroptosis and provide a mechanistic-based combinatorial therapeutic strategy.


Assuntos
Carbono/farmacologia , Hemorragia Cerebral/tratamento farmacológico , Nanopartículas/química , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Dano ao DNA , Desferroxamina/farmacologia , Hemina/antagonistas & inibidores , Hemina/farmacologia , Humanos , Ferro/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo
7.
Prog Neurobiol ; 185: 101729, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31863801

RESUMO

α-Synuclein is a hallmark amyloidogenic protein component of the Lewy bodies (LBs) present in dopaminergic neurons affected by Parkinson's disease (PD). Despite an enormous increase in emerging knowledge, the mechanism(s) of α-synuclein neurobiology and crosstalk among pathological events that are critical for PD progression remains enigmatic, creating a roadblock for effective intervention strategies. One confounding question is about the potential link between α-synuclein toxicity and genome instability in PD. We previously reported that pro-oxidant metal ions, together with reactive oxygen species (ROS), act as a "double whammy" in dopaminergic neurons by not only inducing genome damage but also inhibiting their repair. Our recent studies identified a direct role for chromatin-bound, oxidized α-synuclein in the induction of DNA strand breaks, which raised the question of a paradoxical role for α-synuclein's DNA binding in neuroprotection versus neurotoxicity. Furthermore, recent advances in our understanding of α-synuclein mediated mitochondrial dysfunction warrants revisiting the topics of α-synuclein pathophysiology in order to devise and assess the efficacy of α-synuclein-targeted interventions. In this review article, we discuss the multi-faceted neurotoxic role of α-synuclein in the nucleus and mitochondria with a particular emphasis on the role of α-synuclein in DNA damage/repair defects. We utilized a protein-DNA binding simulation to identify potential residues in α-synuclein that could mediate its binding to DNA and may be critical for its genotoxic functions. These emerging insights and paradigms may guide new drug targets and therapeutic modalities.


Assuntos
Cromatina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Mitocôndrias/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animais , Dano ao DNA/genética , Humanos , alfa-Sinucleína/genética
8.
ACS Chem Neurosci ; 10(10): 4250-4263, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31545596

RESUMO

Alzheimer's disease (AD) is the most common form of dementia among the elderly and has become a leading public health concern worldwide. It represents a huge economic and psychological burden to caregivers and families. The presence of extracellular amyloid beta (Aß) plaques is one of the hallmarks of this neurodegenerative disorder. Amyloid plaques are comprised of aggregates of Aß peptides, mainly Aß42, originated by the cleavage of the amyloid precursor protein (APP). Aß is a crucial target for the treatment of AD, but to date, no effective treatment for the clearance of Aß has been found. We have identified four new hexahydropyrroloindoles (HPI) synthetic compounds that are able to inhibit the aggregation of Aß42 and/or disaggregate the fibril. Docking experiments suggest that the nonpolar component of the interaction of compounds with Aß42 contributes favorably to the binding free energy of each complex. Molecular dynamics simulations suggested fibril disaggregating activity of compounds 1 via interaction with hydrophobic moieties of the fibril. Consistently, compounds 1 and 2 were able to mitigate Aß42 fibrils induced death in rat pheochromocytoma cells (PC 12). One of the compounds reduces the formation of Aß aggregates in vivo and the paralysis associated with Aß toxicity in Caenorhabditis elegans. Our study thus augments efforts for the identification and characterization of new agents that may help stop or delay the progression of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Indóis/uso terapêutico , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/tratamento farmacológico , Pirróis/uso terapêutico , Doença de Alzheimer/metabolismo , Animais , Indóis/farmacologia , Células PC12 , Agregação Patológica de Proteínas/metabolismo , Pirróis/farmacologia , Ratos
9.
Proc Natl Acad Sci U S A ; 116(10): 4696-4705, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30770445

RESUMO

Genome damage and their defective repair have been etiologically linked to degenerating neurons in many subtypes of amyotrophic lateral sclerosis (ALS) patients; however, the specific mechanisms remain enigmatic. The majority of sporadic ALS patients feature abnormalities in the transactivation response DNA-binding protein of 43 kDa (TDP-43), whose nucleo-cytoplasmic mislocalization is characteristically observed in spinal motor neurons. While emerging evidence suggests involvement of other RNA/DNA binding proteins, like FUS in DNA damage response (DDR), the role of TDP-43 in DDR has not been investigated. Here, we report that TDP-43 is a critical component of the nonhomologous end joining (NHEJ)-mediated DNA double-strand break (DSB) repair pathway. TDP-43 is rapidly recruited at DSB sites to stably interact with DDR and NHEJ factors, specifically acting as a scaffold for the recruitment of break-sealing XRCC4-DNA ligase 4 complex at DSB sites in induced pluripotent stem cell-derived motor neurons. shRNA or CRISPR/Cas9-mediated conditional depletion of TDP-43 markedly increases accumulation of genomic DSBs by impairing NHEJ repair, and thereby, sensitizing neurons to DSB stress. Finally, TDP-43 pathology strongly correlates with DSB repair defects, and damage accumulation in the neuronal genomes of sporadic ALS patients and in Caenorhabditis elegans mutant with TDP-1 loss-of-function. Our findings thus link TDP-43 pathology to impaired DSB repair and persistent DDR signaling in motor neuron disease, and suggest that DSB repair-targeted therapies may ameliorate TDP-43 toxicity-induced genome instability in motor neuron disease.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , Humanos , Neurônios Motores/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
10.
J Alzheimers Dis ; 66(2): 453-460, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30320583

RESUMO

Altered expression of α-synuclein is linked to Parkinson's disease (PD). A major challenge to explore how the increased α-synuclein affect neurotoxicity is the lack of a suitable human neuronal cell model that mimics this scenario. Its expression in neural precursors affects their differentiation process, in addition to the neuronal adaptability and variability in maintaining a constant level of expression across passages. Here, we describe an SH-SY5Y line harboring Tet-ON SNCA cDNA cassette that allows for induction of controlled α-synuclein expression after neuronal differentiation, which can be an important tool for PD research.


Assuntos
Regulação da Expressão Gênica/genética , alfa-Sinucleína/metabolismo , Antibacterianos/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Colina O-Acetiltransferase/metabolismo , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , alfa-Sinucleína/química , alfa-Sinucleína/genética
11.
J Alzheimers Dis ; 60(s1): S133-S150, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28731447

RESUMO

Alpha-synuclein (α-Syn) overexpression and misfolding/aggregation in degenerating dopaminergic neurons have long been implicated in Parkinson's disease (PD). The neurotoxicity of α-Syn is enhanced by iron (Fe) and other pro-oxidant metals, leading to generation of reactive oxygen species in PD brain. Although α-Syn is predominantly localized in presynaptic nerve terminals, a small fraction exists in neuronal nuclei. However, the functional and/or pathological role of nuclear α-Syn is unclear. Following up on our earlier report that α-Syn directly binds DNA in vitro, here we confirm the nuclear localization and chromatin association of α-Syn in neurons using proximity ligation and chromatin immunoprecipitation analysis. Moderate (∼2-fold) increase in α-Syn expression in neural lineage progenitor cells (NPC) derived from induced pluripotent human stem cells (iPSCs) or differentiated SHSY-5Y cells caused DNA strand breaks in the nuclear genome, which was further enhanced synergistically by Fe salts. Furthermore, α-Syn required nuclear localization for inducing genome damage as revealed by the effect of nucleus versus cytosol-specific mutants. Enhanced DNA damage by oxidized and misfolded/oligomeric α-Syn suggests that DNA nicking activity is mediated by the chemical nuclease activity of an oxidized peptide segment in the misfolded α-Syn. Consistent with this finding, a marked increase in Fe-dependent DNA breaks was observed in NPCs from a PD patient-derived iPSC line harboring triplication of the SNCA gene. Finally, α-Syn combined with Fe significantly promoted neuronal cell death. Together, these findings provide a novel molecular insight into the direct role of α-Syn in inducing neuronal genome damage, which could possibly contribute to neurodegeneration in PD.


Assuntos
Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , alfa-Sinucleína/metabolismo , Anexina A5/metabolismo , Morte Celular/fisiologia , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Ensaio Cometa , Sulfato de Cobre/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Ferro/farmacologia , Nestina/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sulfetos/farmacologia , alfa-Sinucleína/genética
12.
Mech Ageing Dev ; 161(Pt A): 163-176, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27663141

RESUMO

A foremost challenge for the neurons, which are among the most oxygenated cells, is the genome damage caused by chronic exposure to endogenous reactive oxygen species (ROS), formed as cellular respiratory byproducts. Strong metabolic activity associated with high transcriptional levels in these long lived post-mitotic cells render them vulnerable to oxidative genome damage, including DNA strand breaks and mutagenic base lesions. There is growing evidence for the accumulation of unrepaired DNA lesions in the central nervous system (CNS) during accelerated aging and progressive neurodegeneration. Several germ line mutations in DNA repair or DNA damage response (DDR) signaling genes are uniquely manifested in the phenotype of neuronal dysfunction and are etiologically linked to many neurodegenerative disorders. Studies in our lab and elsewhere revealed that pro-oxidant metals, ROS and misfolded amyloidogenic proteins not only contribute to genome damage in CNS, but also impede their repair/DDR signaling leading to persistent damage accumulation, a common feature in sporadic neurodegeneration. Here, we have reviewed recent advances in our understanding of the etiological implications of DNA damage vs. repair imbalance, abnormal DDR signaling in triggering neurodegeneration and potential of DDR as a target for the amelioration of neurodegenerative diseases.


Assuntos
Quebras de DNA de Cadeia Dupla , Genoma Humano , Doenças Neurodegenerativas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Humanos , Doenças Neurodegenerativas/genética , Oxirredução
13.
Mol Neurobiol ; 51(3): 1417-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25139280

RESUMO

α-Synuclein, an abundant and conserved presynaptic brain protein, is implicated as a critical factor in Parkinson's disease (PD). The aggregation of α-synuclein is believed to be a critical event in the disease process. α-Synuclein is characterized by a remarkable conformational plasticity, adopting different conformations depending on the environment. Therefore, it is classified as an "intrinsically disordered protein." Recently, a debate has challenged the view on the intrinsically disordered behavior of α-synuclein in the cell. It has been proposed that α-synuclein is a stable tetramer with a low propensity for aggregation; however, its destabilization leads to protein misfolding and its aggregation kinetics. In our critical analysis, we discussed about major issues: (i) why α-synuclein conformational behavior does not fit into the normal secondary structural characteristics of proteins, (ii) potential amino acids involved in the complexity of misfolding in α-synuclein that leads to aggregation, and (iii) the role of metals in misfolding and aggregation. To evaluate the above critical issues, we developed bioinformatics models related to secondary and tertiary conformations, Ramachandran plot, free energy change, intrinsic disordered prediction, solvent accessibility, and FoldIndex pattern. To the best of our knowledge, this is a novel critical assessment to understand the misfolding biology of synuclein and its relevance to Parkinson's disease.


Assuntos
Doença de Parkinson/metabolismo , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína/fisiologia , alfa-Sinucleína/metabolismo , Animais , Humanos , Cinética
14.
Neurol Res Ther ; 1(2)2014.
Artigo em Inglês | MEDLINE | ID: mdl-25717476

RESUMO

Excessive accumulation of pro-oxidant metals, observed in affected brain regions, has consistently been implicated as a contributor to the brain pathology including neurodegenerative diseases and acute injuries such as stroke. Furthermore, the potential interactions between metal toxicity and other commonly associated etiological factors, such as misfolding/aggregation of amyloidogenic proteins or genomic damage, are poorly understood. Decades of research provide compelling evidence implicating metal overload in neurological diseases and stroke. However, the utility of metal toxicity as a therapeutic target is controversial, possibly due to a lack of comprehensive understanding of metal dyshomeostasis-mediated neuronal pathology. In this article, we discuss the current understanding of metal toxicity and the challenges associated with metal-targeted therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...