Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.492
Filtrar
1.
Sci Transl Med ; 16(767): eadl3438, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39356746

RESUMO

Oxidative stress has long been implicated in Parkinson's disease (PD) pathogenesis, although the sources and regulation of reactive oxygen species (ROS) production are poorly defined. Pathogenic mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are associated with increased kinase activity and a greater risk of PD. The substrates and downstream consequences of elevated LRRK2 kinase activity are still being elucidated, but overexpression of mutant LRRK2 has been associated with oxidative stress, and antioxidants reportedly mitigate LRRK2 toxicity. Here, using CRISPR-Cas9 gene-edited HEK293 cells, RAW264.7 macrophages, rat primary ventral midbrain cultures, and PD patient-derived lymphoblastoid cells, we found that elevated LRRK2 kinase activity was associated with increased ROS production and lipid peroxidation and that this was blocked by inhibitors of either LRRK2 kinase or NADPH oxidase 2 (NOX2). Oxidative stress induced by the pesticide rotenone was ameliorated by LRRK2 kinase inhibition and was absent in cells devoid of LRRK2. In a rat model of PD induced by rotenone, a LRRK2 kinase inhibitor prevented the lipid peroxidation and NOX2 activation normally seen in nigral dopaminergic neurons in this model. Mechanistically, LRRK2 kinase activity was shown to regulate phosphorylation of serine-345 in the p47phox subunit of NOX2. This, in turn, led to translocation of p47phox from the cytosol to the membrane-associated gp91phox (NOX2) subunit, activation of the NOX2 enzyme complex, and production of ROS. Thus, LRRK2 kinase activity may drive cellular ROS production in PD through the regulation of NOX2 activity.


Assuntos
Modelos Animais de Doenças , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , NADPH Oxidase 2 , Estresse Oxidativo , Doença de Parkinson , Espécies Reativas de Oxigênio , Rotenona , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/genética , Células HEK293 , Estresse Oxidativo/efeitos dos fármacos , Camundongos , NADPH Oxidase 2/metabolismo , NADPH Oxidase 2/genética , Rotenona/farmacologia , Ratos , Peroxidação de Lipídeos , Fosforilação/efeitos dos fármacos , Células RAW 264.7 , Ratos Sprague-Dawley , NADPH Oxidases
2.
PLoS One ; 19(10): e0311824, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39383141

RESUMO

The exploration of novel natural products for Parkinson's disease (PD) is a focus of current research, as there are no definitive drugs to cure or stop the disease. Campsis grandiflora (Thunb.) K. Schum (Lingxiaohua) is a traditional Chinese medicine (TCM), and the exact active constituents and putative mechanisms for treating PD are unknown. Through data mining and network pharmacology, apigenin (APi) was identified as the main active ingredient of Lingxiaohua, and key targets (TNF, AKT1, INS, TP53, CASP3, JUN, BCL2, MMP9, FOS, and HIF1A) of Lingxiaohua for the treatment of PD were discovered. The primary routes implicated were identified as PI3K/AKT, Apoptosis, TNF, and NF-κB pathways. Subsequently, therapeutic potential of APi in PD and its underlying mechanism were experimentally evaluated. APi suppressed the release of mediators of inflammation and initiation of NF-κB pathways in MES23.5 cells induced by MPP+. APi suppressed caspase-3 activity and apoptosis and elevated p-AKT levels in MES23.5 cells. Pretreatment with LY294002, a PI3K inhibitor, resulted in APi treatment blocking the activation of NF-κB pathway and expression of inflammatory factors in MES23.5 cells by activating the PI3K/AKT pathway. In conclusion, APi protects dopaminergic neurons by controlling the PI3K/AKT/NF-κB pathway, giving novel insights into the pharmacological mechanism of Lingxiaohua in treating PD.


Assuntos
Apigenina , NF-kappa B , Farmacologia em Rede , Doença de Parkinson , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Apigenina/farmacologia , NF-kappa B/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Flores/química , Camundongos , Humanos , Apoptose/efeitos dos fármacos , Linhagem Celular
3.
Sci Adv ; 10(41): eado4893, 2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39383232

RESUMO

α-Synuclein (α-syn), a crucial molecule in Parkinson's disease (PD), is known for its interaction with lipid membranes, which facilitates vesicle trafficking and modulates its pathological aggregation. Deciphering the complexity of the membrane-binding behavior of α-syn is crucial to understand its functions and the pathology of PD. Here, we used single-molecule imaging to show that α-syn forms multimers on lipid membranes with huge intermultimer distances. The multimers are characterized by self-limiting growth, manifesting in concentration-dependent exchanges of monomers, which are fast at micromolar concentrations and almost stop at nanomolar concentrations. We further uncovered movement patterns of α-syn's occasional trapping on membranes, which may be attributed to sparse lipid packing defects. Mutations such as E46K and E35K may disrupt the limit on the growth, resulting in larger multimers and accelerated amyloid fibril formation. This work emphasizes sophisticated regulation of α-syn multimerization on membranes as a critical underlying factor in the PD pathology.


Assuntos
Membrana Celular , Doença de Parkinson , Multimerização Proteica , alfa-Sinucleína , alfa-Sinucleína/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/patologia , Humanos , Membrana Celular/metabolismo , Mutação , Imagem Individual de Molécula , Amiloide/metabolismo , Amiloide/química
4.
Drug Res (Stuttg) ; 74(8): 365-378, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39353579

RESUMO

Sphingosine-1-phosphate (S1P) is a cellular signalling molecule derived from sphingosine, which is a pro-apoptotic sphingolipid. Sphingolipids control various cellular actions like growth, homeostasis, and stress-related responses. The main sources of S1P in our body are erythrocytes. S1P controls both cellular mediators and other second messengers intracellularly. The S1P receptor also helps in inflammatory and neuroprotective effects (required to manage of Parkinson's). A large number of anti-Parkinson drugs are available, but still, there is a need for more effective and safer drugs. S1P and its receptors could be targeted as novel drugs due to their involvement in neuro-inflammation and Parkinson's. The present review effort to explore the biological role of S1P and related receptors, for their possible involvement in PD; furthermore. Overall, S1P and other related metabolizing enzymes have significant therapeutic opportunities for Parkinson's disease along with other neurological disorders.


Assuntos
Lisofosfolipídeos , Doença de Parkinson , Transdução de Sinais , Esfingosina , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores
5.
Life Sci ; 357: 123088, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39357796

RESUMO

Parkinson's disease (PD), a neurodegenerative disorder characterized by impaired motor function, is typically treated with medications and surgery. However, recent studies have validated physical exercise as an effective adjunct therapy, significantly improving both motor and non-motor symptoms in PD patients. Irisin, a myokine, has garnered increasing attention for its beneficial effects on the nervous system. Research has shown that irisin plays a crucial role in regulating metabolic balance, optimizing autophagy, maintaining mitochondrial quality, alleviating oxidative stress and neuroinflammation, and regulating cell death-all processes intricately linked to the pathogenesis of PD. This review examines the mechanisms through which irisin may counteract PD, provides insights into its biological effects, and considers its potential as a target for therapeutic strategies.


Assuntos
Fibronectinas , Estresse Oxidativo , Doença de Parkinson , Humanos , Doença de Parkinson/terapia , Doença de Parkinson/metabolismo , Fibronectinas/metabolismo , Animais , Autofagia/fisiologia , Mitocôndrias/metabolismo
6.
Nat Cell Biol ; 26(10): 1639-1651, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39358449

RESUMO

Mitophagy mediated by the recessive Parkinson's disease genes PINK1 and Parkin responds to mitochondrial damage to preserve mitochondrial function. In the pathway, PINK1 is the damage sensor, probing the integrity of the mitochondrial import pathway, and activating Parkin when import is blocked. Parkin is the effector, selectively marking damaged mitochondria with ubiquitin for mitophagy and other quality-control processes. This selective mitochondrial quality-control pathway may be especially critical for dopamine neurons affected in Parkinson's disease, in which the mitochondrial network is widely distributed throughout a highly branched axonal arbor. Here we review the current understanding of the role of PINK1-Parkin in the quality control of mitophagy, including sensing of mitochondrial distress by PINK1, activation of Parkin by PINK1 to induce mitophagy, and the physiological relevance of the PINK1-Parkin pathway.


Assuntos
Mitocôndrias , Mitofagia , Doença de Parkinson , Proteínas Quinases , Ubiquitina-Proteína Ligases , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Humanos , Proteínas Quinases/metabolismo , Proteínas Quinases/genética , Mitocôndrias/metabolismo , Mitocôndrias/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Animais , Transdução de Sinais
7.
J Clin Invest ; 134(20)2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39403921

RESUMO

Parkinson's disease (PD) is characterized by age-dependent neurodegeneration and the accumulation of toxic phosphorylated α-synuclein (pS129-α-syn). The mechanisms underlying these crucial pathological changes remain unclear. Mutations in parkin RBR E3 ubiquitin protein ligase (PARK2), the gene encoding parkin that is phosphorylated by PTEN-induced putative kinase 1 (PINK1) to participate in mitophagy, cause early onset PD. However, current parkin-KO mouse and pig models do not exhibit neurodegeneration. In the current study, we utilized CRISPR/Cas9 technology to establish parkin-deficient monkey models at different ages. We found that parkin deficiency leads to substantia nigra neurodegeneration in adult monkey brains and that parkin phosphorylation decreases with aging, primarily due to increased insolubility of parkin. Phosphorylated parkin is important for neuroprotection and the reduction of pS129-α-syn. Consistently, overexpression of WT parkin, but not a mutant form that cannot be phosphorylated by PINK1, reduced the accumulation of pS129-α-syn. These findings identify parkin phosphorylation as a key factor in PD pathogenesis and suggest it as a promising target for therapeutic interventions.


Assuntos
Modelos Animais de Doenças , Doença de Parkinson , Ubiquitina-Proteína Ligases , alfa-Sinucleína , Animais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/deficiência , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Fosforilação , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Humanos , Substância Negra/metabolismo , Substância Negra/patologia , Macaca fascicularis , Camundongos , Masculino
8.
J Chem Phys ; 161(15)2024 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-39412059

RESUMO

The behavior of hydration water around familial Parkinson's disease linked mutants of α-synuclein may be linked to the early-onset of Parkinson's disease. For the first time, this study compares the local structure and dynamics of hydration water around different segments of some of the natural mutants of α-synuclein, i.e., E46K, G51D, A30P, and A53E, with that of the wild-type protein through explicit water MD simulations. The results show that the C-terminal segments of the fast aggregating mutants such as E46K and A30P are less exposed to water, while those of the slow aggregating ones such as A53E and G51D are more exposed to water relative to that of the wild-type protein. In addition, the water molecules are found to be more ordered around the C-terminal segment of the A53E and G51D mutants as compared to the wild-type protein. This is due to an increase in the overall charge of α-syn upon A53E and G51D mutations. The translational and rotational motions of water molecules in the hydration shell of the C-terminal segment of A53E and G51D mutants are found to be faster relative to that of the wild-type protein. This study validates the differential hydration environment around the C-terminal segment for the causative and protective mutants of α-synuclein.


Assuntos
Simulação de Dinâmica Molecular , Mutação , Água , alfa-Sinucleína , alfa-Sinucleína/química , alfa-Sinucleína/genética , Água/química , Humanos , Doença de Parkinson/genética , Doença de Parkinson/metabolismo
9.
CNS Neurosci Ther ; 30(10): e70076, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39412224

RESUMO

AIMS: Insulin-like growth factor binding protein 2 (IGFBP2) is implicated in various neurodegenerative diseases. However, its role in Parkinson's disease (PD) is unclear. METHODS: PD rat model was established by 6-OHDA injection. After 3 weeks, mRNA-seq was conducted. Rats received rIGFBP2 via intra-MFB injection 6 h prior to 6-OHDA infusion, and the effect of IGFBP2 in PD rats was investigated by western blotting, IHC, specific kits, JC-1 staining, and TUNEL analysis. In vitro, PC12 cells were treated with 6-OHDA, and CCK-8, specific kits, Hoechst 33258 staining, Western blotting, and JC-1 staining were performed to assess the IGFBP2's role. RESULTS: mRNA-seq revealed DEGs in PD, with attention to downregulated IGFBP2. rIGFBP2 treatment aggravated neurobehavioral deficits, decreased TH expression, Ψm, ATP level and SOD, GSH-Px activities but increased α-synuclein, ROS, MDA, mitochondrial cytochrome c contents, cell apoptosis in 6-OHDA-lesioned rats, which might be mediated through inactivating IGF-1R/AKT pathway. In 6-OHDA-treated PC12 cells, rIGFBP2 aggravated cell injury, demonstrated by decreased cell viability and increased apoptosis, oxidative stress, and mitochondrial dysfunction. Co-treatment with rIGFBP2 and rIGF-1 partially reversed the effect of rIGFBP2 on cell damage. CONCLUSION: IGFBP2 exacerbates neurodegeneration in PD through increasing oxidative stress, mitochondrial dysfunction, and apoptosis via inhibiting IGF-1R/AKT pathway.


Assuntos
Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina , Oxidopamina , Animais , Ratos , Células PC12 , Oxidopamina/toxicidade , Masculino , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Ratos Sprague-Dawley , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Receptor IGF Tipo 1/metabolismo , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
10.
Proc Natl Acad Sci U S A ; 121(42): e2406009121, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39388267

RESUMO

Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease, PD); hence, it is an urgent therapeutic target. To identify correctors of severe protein misfolding and trafficking obstruction manifested by the pathogenic L444P-variant of GCase, we developed a suite of quantitative, high-throughput, cell-based assays. First, we labeled GCase with a small proluminescent HiBiT peptide reporter tag, enabling quantitation of protein stabilization in cells while faithfully maintaining target biology. TALEN-based gene editing allowed for stable integration of a single HiBiT-GBA1 transgene into an intragenic safe-harbor locus in GBA1-knockout H4 (neuroglioma) cells. This GD cell model was amenable to lead discovery via titration-based quantitative high-throughput screening and lead optimization via structure-activity relationships. A primary screen of 10,779 compounds from the NCATS bioactive collections identified 140 stabilizers of HiBiT-GCase-L444P, including both pharmacological chaperones (ambroxol and noninhibitory chaperone NCGC326) and proteostasis regulators (panobinostat, trans-ISRIB, and pladienolide B). Two complementary high-content imaging-based assays were deployed to triage hits: The fluorescence-quenched substrate LysoFix-GBA captured functional lysosomal GCase activity, while an immunofluorescence assay featuring antibody hGCase-1/23 directly visualized GCase lysosomal translocation. NCGC326 was active in both secondary assays and completely reversed pathological glucosylsphingosine accumulation. Finally, we tested the concept of combination therapy by demonstrating synergistic actions of NCGC326 with proteostasis regulators in enhancing GCase-L444P levels. Looking forward, these physiologically relevant assays can facilitate the identification, pharmacological validation, and medicinal chemistry optimization of small molecules targeting GCase, ultimately leading to a viable therapeutic for GD and PD.


Assuntos
Doença de Gaucher , Glucosilceramidase , Ensaios de Triagem em Larga Escala , Doença de Parkinson , Dobramento de Proteína , Glucosilceramidase/metabolismo , Glucosilceramidase/genética , Humanos , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/genética , Doença de Gaucher/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Ensaios de Triagem em Larga Escala/métodos , Dobramento de Proteína/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Linhagem Celular Tumoral
11.
Int J Mol Sci ; 25(19)2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39408728

RESUMO

Parkinson's disease (PD) affects movement; however, most patients with PD also develop nonmotor symptoms, such as hyposmia, sleep disorder, and depression. Dopamine levels in the brain have a critical influence on movement control, but other neurotransmitters are also involved in the progression of PD. This study analyzed the fluctuation of neurotransmitters in PC12 cells during neurogenesis and neurodegeneration by performing mass spectrometry. We found that the dopaminergic metabolism pathway of PC12 cells developed vigorously during the neuron differentiation process and that the neurotransmitters were metabolized into 3-methoxytyramine, which was released from the cells. The regulation of the intracellular and extracellular concentrations of adenosine indicated that adenine nucleotides were actively utilized in neural differentiation. Moreover, we exposed the differentiated PC12 cells to rotenone, which is a suitable material for modeling PD. The cells exposed to rotenone in the early stage of differentiation exhibited stimulated serotoninergic metabolism, and the contents of the serotoninergic neurotransmitters returned to their normal levels in the late stage of differentiation. Interestingly, the nondifferentiated cells can resist the toxicant rotenone and produce normal dopaminergic metabolites. However, when differentiated neuron cells were exposed to rotenone, they were seriously damaged, leading to a failure to produce dopaminergic neurotransmitters. In the low-dosage damage process, the amino acids that functioned as dopaminergic pathway precursors could not be absorbed by the cells, and dopamine and L-dopa were secreted and unable to be reuptaken to trigger the cell damage.


Assuntos
Diferenciação Celular , Neurogênese , Neurotransmissores , Rotenona , Células PC12 , Animais , Ratos , Neurogênese/efeitos dos fármacos , Neurotransmissores/metabolismo , Rotenona/farmacologia , Espectrometria de Massas , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Dopamina/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo
12.
Int J Mol Sci ; 25(19)2024 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-39408813

RESUMO

The human brain is highly dependent on oxygen, utilizing approximately 20% of the body's oxygen at rest. Oxygen deprivation to the brain can lead to loss of consciousness within seconds and death within minutes. Recent studies have identified regions of the brain with spontaneous episodic hypoxia, referred to as "hypoxic pockets". Hypoxia can also result from impaired blood flow due to conditions such as heart disease, blood clots, stroke, or hemorrhage, as well as from reduced oxygen intake or excessive oxygen consumption caused by factors like low ambient oxygen, pulmonary diseases, infections, inflammation, and cancer. Severe hypoxia in the brain can manifest symptoms similar to Parkinson's disease (PD), including cerebral edema, mood disturbances, and cognitive impairments. Additionally, the development of PD appears to be closely associated with hypoxia and hypoxic pathways. This review seeks to investigate the molecular interactions between hypoxia and PD, emphasizing the pathological role of hypoxic pathways in PD and exploring their potential as therapeutic targets.


Assuntos
Hipóxia , Doença de Parkinson , Humanos , Doença de Parkinson/metabolismo , Hipóxia/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Transdução de Sinais , Oxigênio/metabolismo
13.
Int J Mol Sci ; 25(19)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39409112

RESUMO

Multiple system atrophy and Lewy body diseases (LBDs) such as Parkinson's disease, dementia with Lewy bodies, and Parkinson's disease with dementia, known as synucleinopathies, are defined neuropathologically by the accumulation and deposition of aberrant protein aggregates, primarily in neuronal cells. Seeding aggregation assays (SAA) have significant potential as biomarkers for early diagnosis, monitoring disease progression, and evaluating treatment efficacy for these diseases. Real-time quaking-induced conversion (RT-QuIC) and Protein Misfolding Cyclic Amplification (PMCA) assays represent two ultrasensitive protein amplification techniques that were initially tested for the field of prion disorders. Although the fundamental idea behind the creation of these two methods is very similar, their technical differences resulted in different levels of diagnostic accuracy for the identification of prion proteins, making the RT-QuIC assay the most trustworthy and effective instrument for the detection of suspected cases of LBDs and prion-like diseases.


Assuntos
Doença por Corpos de Lewy , Humanos , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/diagnóstico , Doença por Corpos de Lewy/patologia , Biomarcadores , Animais , Proteínas Priônicas/metabolismo , Agregação Patológica de Proteínas/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/diagnóstico , Doença de Parkinson/patologia , Dobramento de Proteína , alfa-Sinucleína/metabolismo , alfa-Sinucleína/análise , Agregados Proteicos
14.
Mol Neurodegener ; 19(1): 69, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39379975

RESUMO

BACKGROUND: Pathological accumulation of aggregated α-synuclein (aSYN) is a common feature of Parkinson's disease (PD). However, the mechanisms by which intracellular aSYN pathology contributes to dysfunction and degeneration of neurons in the brain are still unclear. A potentially relevant target of aSYN is the mitochondrion. To test this hypothesis, genetic and physiological methods were used to monitor mitochondrial function in substantia nigra pars compacta (SNc) dopaminergic and pedunculopontine nucleus (PPN) cholinergic neurons after stereotaxic injection of aSYN pre-formed fibrils (PFFs) into the mouse brain. METHODS: aSYN PFFs were stereotaxically injected into the SNc or PPN of mice. Twelve weeks later, mice were studied using a combination of approaches, including immunocytochemical analysis, cell-type specific transcriptomic profiling, electron microscopy, electrophysiology and two-photon-laser-scanning microscopy of genetically encoded sensors for bioenergetic and redox status. RESULTS: In addition to inducing a significant neuronal loss, SNc injection of PFFs induced the formation of intracellular, phosphorylated aSYN aggregates selectively in dopaminergic neurons. In these neurons, PFF-exposure decreased mitochondrial gene expression, reduced the number of mitochondria, increased oxidant stress, and profoundly disrupted mitochondrial adenosine triphosphate production. Consistent with an aSYN-induced bioenergetic deficit, the autonomous spiking of dopaminergic neurons slowed or stopped. PFFs also up-regulated lysosomal gene expression and increased lysosomal abundance, leading to the formation of Lewy-like inclusions. Similar changes were observed in PPN cholinergic neurons following aSYN PFF exposure. CONCLUSIONS: Taken together, our findings suggest that disruption of mitochondrial function, and the subsequent bioenergetic deficit, is a proximal step in the cascade of events induced by aSYN pathology leading to dysfunction and degeneration of neurons at-risk in PD.


Assuntos
Neurônios Colinérgicos , Neurônios Dopaminérgicos , Mitocôndrias , Doença de Parkinson , alfa-Sinucleína , Animais , alfa-Sinucleína/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Camundongos , Camundongos Endogâmicos C57BL
15.
Sci Adv ; 10(41): eadp9150, 2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39383230

RESUMO

Here, we report a magnetogenetic system, based on a single anti-ferritin nanobody-TRPV1 receptor fusion protein, which regulated neuronal activity when exposed to magnetic fields. Adeno-associated virus (AAV)-mediated delivery of a floxed nanobody-TRPV1 into the striatum of adenosine-2a receptor-Cre drivers resulted in motor freezing when placed in a magnetic resonance imaging machine or adjacent to a transcranial magnetic stimulation device. Functional imaging and fiber photometry confirmed activation in response to magnetic fields. Expression of the same construct in the striatum of wild-type mice along with a second injection of an AAVretro expressing Cre into the globus pallidus led to similar circuit specificity and motor responses. Last, a mutation was generated to gate chloride and inhibit neuronal activity. Expression of this variant in the subthalamic nucleus in PitX2-Cre parkinsonian mice resulted in reduced c-fos expression and motor rotational behavior. These data demonstrate that magnetogenetic constructs can bidirectionally regulate activity of specific neuronal circuits noninvasively in vivo using clinically available devices.


Assuntos
Dependovirus , Terapia Genética , Animais , Camundongos , Terapia Genética/métodos , Dependovirus/genética , Doença de Parkinson/terapia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Vetores Genéticos/genética , Humanos , Núcleo Subtalâmico/metabolismo , Campos Magnéticos , Globo Pálido/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Neurônios/metabolismo , Corpo Estriado/metabolismo , Canais de Cátion TRPV
16.
Proc Natl Acad Sci U S A ; 121(42): e2403450121, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39388265

RESUMO

Aging is the biggest risk factor for Parkinson's disease (PD), suggesting that age-related changes in the brain promote dopamine neuron vulnerability. It is unclear, however, whether aging alone is sufficient to cause significant dopamine neuron loss, and if so, how this intersects with PD-related neurodegeneration. Here, through examining a large collection of naturally varying Drosophila strains, we find a strong relationship between life span and age-related dopamine neuron loss. Strains with naturally short-lived animals exhibit a loss of dopamine neurons without generalized neurodegeneration, while animals from long-lived strains retain dopamine neurons across age. Metabolomic profiling reveals lower glutathione levels in short-lived strains which is associated with elevated levels of reactive oxygen species (ROS), sensitivity to oxidative stress, and vulnerability to silencing the familial PD gene parkin. Strikingly, boosting neuronal glutathione levels via glutamate-cysteine ligase (Gcl) overexpression is sufficient to normalize ROS levels, extend life span, and block dopamine neurons loss in short-lived backgrounds, demonstrating that glutathione deficiencies are central to neurodegenerative phenotypes associated with short longevity. These findings may be relevant to human PD pathogenesis, where glutathione depletion is reported to occur in the idiopathic PD patient brain through unknown mechanisms. Building on this, we find reduced expression of the Gcl catalytic subunit in both Drosophila strains vulnerable to age-related dopamine neuron loss and in the human brain from familial PD patients harboring the common LRRK2 G2019S mutation. Our study across Drosophila and human PD systems suggests that glutathione synthesis and levels play a conserved role in regulating age-related dopamine neuron health.


Assuntos
Envelhecimento , Neurônios Dopaminérgicos , Proteínas de Drosophila , Glutationa , Longevidade , Doença de Parkinson , Espécies Reativas de Oxigênio , Animais , Glutationa/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Espécies Reativas de Oxigênio/metabolismo , Drosophila melanogaster/metabolismo , Estresse Oxidativo , Humanos , Glutamato-Cisteína Ligase/metabolismo , Glutamato-Cisteína Ligase/genética , Degeneração Neural/patologia , Degeneração Neural/metabolismo , Degeneração Neural/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Drosophila/metabolismo , Masculino
17.
Sci Rep ; 14(1): 23861, 2024 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-39394439

RESUMO

Parkinson's disease (PD) is the most common progressive neurodegenerative movement disorder and results from the selective loss of dopaminergic neurons in the substantia nigra pars compacta. Pink1 and Parkin are proteins that function together in mitochondrial quality control, and when they carry loss-of-function mutations lead to familial forms of PD. While much research has focused on central nervous system alterations in PD, peripheral contributions to PD pathogenesis are increasingly appreciated. We report Pink1/Parkin regulate glycolytic and mitochondrial oxidative metabolism in peripheral blood mononuclear cells (PBMCs) from rats. Pink1/Parkin deficiency induces changes in the circulating lymphocyte populations, namely increased CD4 + T cells and decreased CD8 + T cells and B cells. Loss of Pink1/Parkin leads to elevated platelet counts in the blood and increased platelet-T cell aggregation. Platelet-lymphocyte aggregates are associated with increased thrombosis risk suggesting targeting the Pink1/Parkin pathway in the periphery might have therapeutic potential.


Assuntos
Plaquetas , Mitocôndrias , Proteínas Quinases , Ubiquitina-Proteína Ligases , Animais , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/deficiência , Proteínas Quinases/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/deficiência , Ratos , Plaquetas/metabolismo , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Masculino , Glicólise , Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Leucócitos Mononucleares/metabolismo , Agregação Plaquetária
18.
Sci Rep ; 14(1): 23414, 2024 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-39379677

RESUMO

IMPHY000797 derivatives have been well known for their efficacy in various diseases. Moreover, IMPHY000797 derivatives have been found to modulate such genes involved in multiple neurological disorders. Hence, this study seeks to identify such genes and the probable molecular mechanism that could be involved in the pathogenesis of Parkinson's disease. The study utilized various biological tools such as DisGeNET, STRING, Swiss target predictor, Cytoscape, AutoDock 4.2, Schrodinger suite, ClueGo, and GUSAR. All the reported genes were obtained using DisGeNET, and further, the common genes were incorporated into the STRING to get the KEGG pathway, and all the data was converted to a protein/pathway network via Cytoscape. The clustering of the genes was performed for the gene-enriched data using two-sided hypergeometrics (p-value). The binding affinity of the IMPHY000797 was verified with the highest regulated 25 proteins via utilizing the "Monte Carlo iterated search technique" and the "Emodel and Glide score" function. Three thousand five hundred eighty-three genes were identified for Parkinson's disease and 31 genes for IMPHY000797 compound, among which 25 common genes were identified. Further, the "FOXO-signaling pathway" was identified to be a modulated pathway. Among the 25 proteins, the highest modulated genes and highest binding affinity were exhibited by SIRT3, FOXO1, and PPARGC1A with the compound IMPHY000797. Further, rat toxicity analysis provided the efficacy and safety of the compound. The study was required to identify the probable molecular mechanism, which needs more confirmation from other studies, which is still a significant hit-back.


Assuntos
Doença de Parkinson , Doença de Parkinson/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Humanos , Animais , Ratos , Biologia de Sistemas/métodos , Farmacologia em Rede , Simulação por Computador , Redes Reguladoras de Genes/efeitos dos fármacos , Mapas de Interação de Proteínas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
19.
J Transl Med ; 22(1): 919, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390613

RESUMO

BACKGROUND: Propagation of neuronal α-synuclein aggregate pathology to the cortex and hippocampus correlates with cognitive impairment in Parkinson's disease (PD) dementia and dementia with Lewy body disease. Previously, we showed accumulation of the parkin substrate aminoacyl-tRNA synthetase interacting multifunctional protein-2 (AIMP2) in the temporal lobe of postmortem brains of patients with advanced PD. However, the potential pathological role of AIMP2 accumulation in the cognitive dysfunction of patients with PD remains unknown. METHODS: We performed immunofluorescence imaging to examine cellular distribution and accumulation of AIMP2 in brains of conditional AIMP2 transgenic mice and postmortem PD patients. The pathological role of AIMP2 was investigated in the AIMP2 transgenic mice by assessing Nissl-stained neuron counting in the hippocampal area and Barnes maze to determine cognitive functions. Potential secretion and cellular uptake of AIMP2 was monitored by dot blot analysis and immunofluorescence. The utility of AIMP2 as a new PD biomarker was evaluated by dot blot and ELISA measurement of plasma AIMP2 collected from PD patients and healthy control followed by ROC curve analysis. RESULTS: We demonstrated that AIMP2 is toxic to the dentate gyrus neurons of the hippocampus and that conditional AIMP2 transgenic mice develop progressive cognitive impairment. Moreover, we found that neuronal AIMP2 expression levels correlated with the brain endothelial expression of AIMP2 in both AIMP2 transgenic mice and in the postmortem brains of patients with PD. AIMP2, when accumulated, was released from the neuronal cell line SH-SY5Y cells. Secreted AIMP2 was taken up by human umbilical vein endothelial cells. Consistent with the fact that AIMP2 can be released into the extracellular space, we showed that AIMP2 transgenic mice have higher levels of plasma AIMP2. Finally, ELISA-based assessment of AIMP2 in plasma samples from patients with PD and controls, and subsequent ROC curve analysis proved that high plasma AIMP2 expression could serve as a reliable molecular biomarker for PD diagnosis. CONCLUSIONS: The pathological role in the hippocampus and the cell-to-cell transmissibility of AIMP2 provide new therapeutic avenues for PD treatment, and plasma AIMP2 combined with α-synuclein may improve the accuracy of PD diagnosis in the early stages.


Assuntos
Encéfalo , Camundongos Transgênicos , Doença de Parkinson , Animais , Humanos , Doença de Parkinson/patologia , Doença de Parkinson/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Idoso , Feminino , Disfunção Cognitiva/metabolismo , Biomarcadores/metabolismo , Biomarcadores/sangue , alfa-Sinucleína/metabolismo , Camundongos , Hipocampo/metabolismo , Hipocampo/patologia
20.
Commun Biol ; 7(1): 1294, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390051

RESUMO

IP3 receptor (IP3R)-mediated Ca2+ transfer at the mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) drives mitochondrial Ca2+ uptake and oxidative metabolism and is linked to different pathologies, including Parkinson's disease (PD). The dependence of Ca2+ transfer efficiency on the ER-mitochondria distance remains unexplored. Employing molecular rulers that stabilize ER-mitochondrial distances at 5 nm resolution, and using genetically encoded Ca2+ indicators targeting the ER lumen and the sub-mitochondrial compartments, we now show that a distance of ~20 nm is optimal for Ca2+ transfer and mitochondrial oxidative metabolism due to enrichment of IP3R at MERCS. In human iPSC-derived astrocytes from PD patients, 20 nm MERCS were specifically reduced, which correlated with a reduction of mitochondrial Ca2+ uptake. Stabilization of the ER-mitochondrial interaction at 20 nm, but not at 10 nm, fully rescued mitochondrial Ca2+ uptake in PD astrocytes. Our work determines with precision the optimal distance for Ca2+ flux between ER and mitochondria and suggests a new paradigm for fine control over mitochondrial function.


Assuntos
Astrócitos , Cálcio , Retículo Endoplasmático , Mitocôndrias , Doença de Parkinson , Mitocôndrias/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Cálcio/metabolismo , Astrócitos/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Sinalização do Cálcio , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Oxirredução , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...