Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nature ; 619(7970): 606-615, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37438521

RESUMEN

The specific loss of midbrain dopamine neurons (mDANs) causes major motor dysfunction in Parkinson's disease, which makes cell replacement a promising therapeutic approach1-4. However, poor survival of grafted mDANs remains an obstacle to successful clinical outcomes5-8. Here we show that the surgical procedure itself (referred to here as 'needle trauma') triggers a profound host response that is characterized by acute neuroinflammation, robust infiltration of peripheral immune cells and brain cell death. When midbrain dopamine (mDA) cells derived from human induced pluripotent stem (iPS) cells were transplanted into the rodent striatum, less than 10% of implanted tyrosine hydroxylase (TH)+ mDANs survived at two weeks after transplantation. By contrast, TH- grafted cells mostly survived. Notably, transplantation of autologous regulatory T (Treg) cells greatly modified the response to needle trauma, suppressing acute neuroinflammation and immune cell infiltration. Furthermore, intra-striatal co-transplantation of Treg cells and human-iPS-cell-derived mDA cells significantly protected grafted mDANs from needle-trauma-associated death and improved therapeutic outcomes in rodent models of Parkinson's disease with 6-hydroxydopamine lesions. Co-transplantation with Treg cells also suppressed the undesirable proliferation of TH- grafted cells, resulting in more compact grafts with a higher proportion and higher absolute numbers of TH+ neurons. Together, these data emphasize the importance of the initial inflammatory response to surgical injury in the differential survival of cellular components of the graft, and suggest that co-transplanting autologous Treg cells effectively reduces the needle-trauma-induced death of mDANs, providing a potential strategy to achieve better clinical outcomes for cell therapy in Parkinson's disease.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Neuronas Dopaminérgicas , Supervivencia de Injerto , Enfermedades Neuroinflamatorias , Enfermedad de Parkinson , Linfocitos T Reguladores , Tirosina 3-Monooxigenasa , Humanos , Dopamina/análogos & derivados , Dopamina/metabolismo , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/trasplante , Mesencéfalo/patología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/prevención & control , Enfermedades Neuroinflamatorias/terapia , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/cirugía , Enfermedad de Parkinson/terapia , Tirosina 3-Monooxigenasa/deficiencia , Tirosina 3-Monooxigenasa/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Animales , Ratones , Ratas , Oxidopamina/metabolismo , Supervivencia de Injerto/inmunología , Muerte Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Neostriado/metabolismo , Factores de Tiempo , Proliferación Celular , Resultado del Tratamiento
2.
J Neurosci ; 42(4): 702-716, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34876467

RESUMEN

The Parkinson's disease (PD) risk gene GTP cyclohydrolase 1 (GCH1) catalyzes the rate-limiting step in tetrahydrobiopterin (BH4) synthesis, an essential cofactor in the synthesis of monoaminergic neurotransmitters. To investigate the mechanisms by which GCH1 deficiency may contribute to PD, we generated a loss of function zebrafish gch1 mutant (gch1-/-), using CRISPR/Cas technology. gch1-/- zebrafish develop marked monoaminergic neurotransmitter deficiencies by 5 d postfertilization (dpf), movement deficits by 8 dpf and lethality by 12 dpf. Tyrosine hydroxylase (Th) protein levels were markedly reduced without loss of ascending dopaminergic (DAergic) neurons. L-DOPA treatment of gch1-/- larvae improved survival without ameliorating the motor phenotype. RNAseq of gch1-/- larval brain tissue identified highly upregulated transcripts involved in innate immune response. Subsequent experiments provided morphologic and functional evidence of microglial activation in gch1-/- The results of our study suggest that GCH1 deficiency may unmask early, subclinical parkinsonism and only indirectly contribute to neuronal cell death via immune-mediated mechanisms. Our work highlights the importance of functional validation for genome-wide association studies (GWAS) risk factors and further emphasizes the important role of inflammation in the pathogenesis of PD.SIGNIFICANCE STATEMENT Genome-wide association studies have now identified at least 90 genetic risk factors for sporadic Parkinson's disease (PD). Zebrafish are an ideal tool to determine the mechanistic role of genome-wide association studies (GWAS) risk genes in a vertebrate animal model. The discovery of GTP cyclohydrolase 1 (GCH1) as a genetic risk factor for PD was counterintuitive, GCH1 is the rate-limiting enzyme in the synthesis of dopamine (DA), mutations had previously been described in the non-neurodegenerative movement disorder dopa-responsive dystonia (DRD). Rather than causing DAergic cell death (as previously hypothesized by others), we now demonstrate that GCH1 impairs tyrosine hydroxylase (Th) homeostasis and activates innate immune mechanisms in the brain and provide evidence of microglial activation and phagocytic activity.


Asunto(s)
Encéfalo/enzimología , GTP Ciclohidrolasa/deficiencia , Homeostasis/fisiología , Inmunidad Innata/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/inmunología , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/inmunología , GTP Ciclohidrolasa/genética , Predisposición Genética a la Enfermedad/genética , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/inmunología , Análisis de Secuencia de ARN/métodos , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/genética , Pez Cebra
3.
Neurosci Lett ; 764: 136222, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34500002

RESUMEN

A growing body of evidence indicates an association between flavin-containing monooxygenase (FMO) and neurodegeneration, including Parkinson's disease (PD); however, the details of this association are unclear. We previously showed that the level of Fmo1 mRNA is decreased in an in vitro rotenone model of parkinsonism. To further explore the potential involvement of FMO1 deficiency in parkinsonism, we generated Fmo1 knockout (KO) mice and examined the survival of dopaminergic neurons and relative changes. Fmo1 KO mice exhibited loss of tyrosine hydroxylase-positive neurons, decreased levels of tyrosine hydroxylase and Parkin proteins, and increased levels of pro-inflammatory cytokines (IL1ß and IL6) in the nigrostriatal region. Moreover, the protein levels of PTEN induced kinase 1 (PINK1) and p62, and the Microtubule associated protein 1 light chain 3 (LC3)-II/I ratio were not significantly altered in Fmo1 KO mice (P > 0.05). FMO1 deficiency promotes neuroinflammation in dopaminergic neurons in mice, thus may plays a potential pathological role in dopaminergic neuronal loss. These findings may provide new insight into the pathogenesis of PD.


Asunto(s)
Neuronas Dopaminérgicas/patología , Enfermedades Neuroinflamatorias/inmunología , Oxigenasas/deficiencia , Enfermedad de Parkinson/inmunología , Sustancia Negra/patología , Animales , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Enfermedades Neuroinflamatorias/patología , Oxigenasas/genética , Enfermedad de Parkinson/patología , Proteínas Quinasas/análisis , Proteínas Quinasas/metabolismo , Proteína Sequestosoma-1/análisis , Proteína Sequestosoma-1/metabolismo , Sustancia Negra/citología , Sustancia Negra/inmunología , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/análisis , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina-Proteína Ligasas/análisis , Ubiquitina-Proteína Ligasas/metabolismo
4.
Nat Commun ; 12(1): 5382, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508096

RESUMEN

Pathways to control the spreading of α-synuclein (α-syn) and associated neuropathology in Parkinson's disease (PD), multiple system atrophy (MSA) and dementia with Lewy bodies (DLB) are unclear. Here, we show that preformed α-syn fibrils (PFF) increase the association between TLR2 and MyD88, resulting in microglial activation. The TLR2-interaction domain of MyD88 (wtTIDM) peptide-mediated selective inhibition of TLR2 reduces PFF-induced microglial inflammation in vitro. In PFF-seeded A53T mice, the nasal administration of the wtTIDM peptide, NEMO-binding domain (wtNBD) peptide, or genetic deletion of TLR2 reduces glial inflammation, decreases α-syn spreading, and protects dopaminergic neurons by inhibiting NF-κB. In summary, α-syn spreading depends on the TLR2/MyD88/NF-κB pathway and it can be reduced by nasal delivery of wtTIDM and wtNBD peptides.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Microglía/patología , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Humanos , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/patología , Ratones , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo , Atrofia de Múltiples Sistemas/genética , Atrofia de Múltiples Sistemas/patología , Mutagénesis Sitio-Dirigida , Mutación , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Regiones Promotoras Genéticas , Transducción de Señal/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , alfa-Sinucleína/genética
5.
Int Immunopharmacol ; 99: 108025, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34364303

RESUMEN

Microglia-mediated neuroinflammation is tightly correlated with the etiology and progression of neurodegenerative disorders, including Parkinson's disease (PD). Nilotinib, a c-Abl inhibitor used for chronic myeloid leukemia, has been proven effective in relieving PD progression. However, whether nilotinib could affect neuroinflammation is largely unknown. In this current study, we investigated the role of nilotinib in microglia-mediated neuroinflammatory response in Parkinson's disease. Lipopolysaccharide (LPS)-induced neuroinflammation in BV2 microglial cells and mouse brains were used as models for Parkinson's disease. Our results demonstrated that nilotinib significantly suppressed LPS-induced neuroinflammation by reducing the production of pro-inflammatory factors including iNOS, COX-2, IL-1ß, IL-6 and TNF-α in BV2 cells. Moreover, pretreatment of nilotinib attenuated the neurotoxicity of LPS-treated microglial conditioned medium to MES23.5 dopaminergic (DA) neurons. Mechanismly, nilotinib inhibited NF-κB signaling pathway and suppressed the nuclear translocation of p65 upon LPS stimulation. In LPS-injected mouse brains, nilotinib administration markedly suppressed the activation of microglia and down-regulated COX-2 as well as IL-1ß expression. Most importantly, nilotinib effectively protected against microglial activation-mediated mouse DA neuronal loss. Taken together, our study suggests that nilotinib exerts anti-neuroinflammatory effect and protects DA neurons from activated microglia-induced inflammatory damage through suppressing NF-κB signaling pathway, indicating its potential application in further clinical trials.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Pirimidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/patología , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Microglía/inmunología , Microglía/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos
6.
Exp Neurol ; 346: 113845, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34454938

RESUMEN

Viral infection of the central nervous system (CNS) can cause lasting neurological decline in surviving patients and can present with symptoms resembling Parkinson's disease (PD). The mechanisms underlying postencephalitic parkinsonism remain unclear but are thought to involve increased innate inflammatory signaling in glial cells, resulting in persistent neuroinflammation. We therefore studied the role of glial cells in regulating neuropathology in postencephalitic parkinsonism by studying the involvement of astrocytes in loss of dopaminergic neurons and aggregation of α-synuclein protein following infection with western equine encephalitis virus (WEEV). Infections were conducted in both wildtype mice and in transgenic mice lacking NFκB inflammatory signaling in astrocytes. For 2 months following WEEV infection, we analyzed glial activation, neuronal loss and protein aggregation across multiple brain regions, including the substantia nigra pars compacta (SNpc). These data revealed that WEEV induces loss of SNpc dopaminergic neurons, persistent activation of microglia and astrocytes that precipitates widespread aggregation of α-synuclein in the brain of C57BL/6 mice. Microgliosis and macrophage infiltration occurred prior to activation of astrocytes and was followed by opsonization of ⍺-synuclein protein aggregates in the cortex, hippocampus and midbrain by the complement protein, C3. Astrocyte-specific NFκB knockout mice had reduced gliosis, α-synuclein aggregate formation and neuronal loss. These data suggest that astrocytes play a critical role in initiating PD-like pathology following encephalitic infection with WEEV through innate immune inflammatory pathways that damage dopaminergic neurons, possibly by hindering clearance of ⍺-synuclein aggregates. Inhibiting glial inflammatory responses could therefore represent a potential therapy strategy for viral parkinsonism.


Asunto(s)
Astrocitos/metabolismo , Neuronas Dopaminérgicas/metabolismo , Encefalitis Viral/metabolismo , Mediadores de Inflamación/metabolismo , Agregado de Proteínas/fisiología , alfa-Sinucleína/metabolismo , Animales , Astrocitos/inmunología , Neuronas Dopaminérgicas/inmunología , Virus de la Encefalitis Equina del Oeste/inmunología , Virus de la Encefalitis Equina del Oeste/metabolismo , Encefalitis Viral/inmunología , Femenino , Humanos , Mediadores de Inflamación/inmunología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
7.
Aging (Albany NY) ; 13(8): 11455-11469, 2021 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-33839699

RESUMEN

BACKGROUND: Studies have revealed that lncRNA HOXA11-AS contributes to regulating inflammation, while the role of HOXA11-AS in Parkinson's disease (PD) remains unclear. METHODS: Both in vivo and in vitro PD models were induced. Gain- or loss-assays of HOXA11-AS and miR-124-3p were conducted. The neurological functions, dopaminergic neurons damage, microglia activation of PD mice were measured. Afterwards, the expressions of inflammatory factors were examined with RT-PCR. Western blot was employed to detect the level of FSTL1, NF-κB and NLRP3 inflammasome. Meanwhile, bioinformatics analysis and dual-luciferase reporter assay were utilized to confirm the targeting relationships among miR-124-3p, HOXA11-AS and FSTL1. RESULTS: HOXA11-AS promoted MPTP-mediated SH-SY5Y neuronal injury and LPS-induced microglia activation, while miR-124-3p had the opposite effects. Additionally, miR-124-3p was the target of HOXA11-AS and FSTL1. HOXA11-AS overexpression enhanced the expression of inflammatory factors and FSTL1, NF-κB and NLRP3 inflammasome, while inhibiting NF-κB weakened HOXA11-AS-mediated neuronal damage and microglia activation. Moreover, HOXA11-AS1 downregulation ameliorated MPTP-induced neurological damages and neuroinflammation in mice. CONCLUSION: Inhibition of HOXA11-AS protects mice against PD through repressing neuroinflammation and neuronal apoptosis through miR-124-3p-FSTL1-NF-κB axis.


Asunto(s)
Neuronas Dopaminérgicas/patología , MicroARNs/metabolismo , Trastornos Parkinsonianos/inmunología , ARN Largo no Codificante/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Animales , Apoptosis/genética , Apoptosis/inmunología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Inflamasomas/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Transducción de Señal/genética , Transducción de Señal/inmunología
8.
J Neuroinflammation ; 18(1): 20, 2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33422110

RESUMEN

BACKGROUND: Recent work has established that Parkinson's disease (PD) patients have an altered gut microbiome, along with signs of intestinal inflammation. This could help explain the high degree of gastric disturbances in PD patients, as well as potentially be linked to the migration of peripheral inflammatory factors into the brain. To our knowledge, this is the first study to examine microbiome alteration prior to the induction of a PD murine model. METHODS: We presently assessed whether pre-treatment with the probiotic, VSL #3, or the inflammatory inducer, dextran sodium sulphate (DSS), would influence the PD-like pathology provoked by a dual hit toxin model using lipopolysaccharide (LPS) and paraquat exposure. RESULTS: While VSL #3 has been reported to have anti-inflammatory effects, DSS is often used as a model of colitis because of the gut inflammation and the breach of the intestinal barrier that it induces. We found that VSL#3 did not have any significant effects (beyond a blunting of LPS paraquat-induced weight loss). However, the DSS treatment caused marked changes in the gut microbiome and was also associated with augmented behavioral and inflammatory outcomes. In fact, DSS markedly increased taxa belonging to the Bacteroidaceae and Porphyromonadaceae families but reduced those from Rikencellaceae and S24-7, as well as provoking colonic pro-inflammatory cytokine expression, consistent with an inflamed gut. The DSS also increased the impact of LPS plus paraquat upon microglial morphology, along with circulating lipocalin-2 (neutrophil marker) and IL-6. Yet, neither DSS nor VSL#3 influenced the loss of substantia nigra dopamine neurons or the astrocytic and cytoskeleton remodeling protein changes that were provoked by the LPS followed by paraquat treatment. CONCLUSIONS: These data suggest that disruption of the intestinal integrity and the associated microbiome can interact with systemic inflammatory events to promote widespread brain-gut changes that could be relevant for PD and at the very least, suggestive of novel neuro-immune communication.


Asunto(s)
Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/fisiología , Mediadores de Inflamación/metabolismo , Trastornos Parkinsonianos/metabolismo , Probióticos/administración & dosificación , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Mediadores de Inflamación/inmunología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/inmunología
9.
Neurobiol Dis ; 149: 105229, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33352233

RESUMEN

Alpha-synuclein (a-syn) can aggregate and form toxic oligomers and insoluble fibrils which are the main component of Lewy bodies. Intra-neuronal Lewy bodies are a major pathological characteristic of Parkinson's disease (PD). These fibrillar structures can act as seeds and accelerate the aggregation of monomeric a-syn. Indeed, recent studies show that injection of preformed a-syn fibrils (PFF) into the rodent brain can induce aggregation of the endogenous monomeric a-syn resulting in neuronal dysfunction and eventual cell death. We injected 8 µg of murine a-syn PFF, or soluble monomeric a-syn into the right striatum of rats. The animals were monitored behaviourally using the cylinder test, which measures paw asymmetry, and the corridor task that measures lateralized sensorimotor response to sugar treats. In vivo PET imaging was performed after 6, 13 and 22 weeks using [11C]DTBZ, a marker of the vesicular monoamine 2 transporter (VMAT2), and after 15 and 22 weeks using [11C]UCB-J, a marker of synaptic SV2A protein in nerve terminals. Histology was performed at the three time points using antibodies against dopaminergic markers, aggregated a-syn, and MHCII to evaluate the immune response. While the a-syn PFF injection caused only mild behavioural changes, [11C]DTBZ PET showed a significant and progressive decrease of VMAT2 binding in the ipsilateral striatum. This was accompanied by a small progressive decrease in [11C]UCB-J binding in the same area. In addition, our histological analysis revealed a gradual spread of misfolded a-syn pathology in areas anatomically connected to striatum that became bilateral with time. The striatal a-syn PFF injection resulted in a progressive unilateral degeneration of dopamine terminals, and an early and sustained presence of MHCII positive ramified microglia in the ipsilateral striatum and substantia nigra. Our study shows that striatal injections of a-syn fibrils induce progressive pathological synaptic dysfunction prior to cell death that can be detected in vivo with PET. We confirm that intrastriatal injection of a-syn PFFs provides a model of progressive a-syn pathology with loss of dopaminergic and synaptic function accompanied by neuroinflammation, as found in human PD.


Asunto(s)
Cuerpo Estriado/metabolismo , Progresión de la Enfermedad , Neuronas Dopaminérgicas/metabolismo , Tomografía de Emisión de Positrones/métodos , Agregado de Proteínas/fisiología , alfa-Sinucleína/toxicidad , Animales , Cuerpo Estriado/inmunología , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/patología , Femenino , Inyecciones Intraventriculares , Ratas , Ratas Sprague-Dawley , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/inmunología
10.
Int Immunopharmacol ; 91: 107269, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33340781

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease with complicated pathogenesis. A novel bibenzyl compound 2-[4-hydroxy-3-(4-hydroxyphenyl)benzyl]-4-(4-hydroxyphenyl)phenol (20C) has been shown to have some neuroprotective effects, and its mechanism still needs further research. In this study, we used a 6-hydroxydopamine (6-OHDA)-induced PD rat model to evaluate the protective effect of 20C. Our study found that 20C could improve behavioral defects in 6-OHDA-lesion rats, decrease neuroinflammation and protect their DA neurons. It could inhibit the activity of inducible nitric oxide synthase (iNOS) induced by 6-OHDA, and lead to a decrease in the expression of nitrated-α-synuclein. When exposed to AMT-an inhibitor of iNOS, the nitrated-α-synuclein in PC12 decreased, and 20C demonstrated the same function on nitrated-α-synuclein as AMT. Besides, we also found that nitrated-α-synuclein was displayed in microglia. And 20C could decrease the expression of antigen-presenting molecule major histocompatibility complex I (MHC I) in dopamine (DA) neurons and MHC II in microglia induced by 6-OHDA. So, these imply that nitrated-α-synuclein might act as an endogenous antigen activating adaptive immunity, and the neuroprotection of 20C might be associated with inhibiting the activity of iNOS, decreasing the expression of the antigen molecule nitrated-α-synuclein and the antigen presenting molecule MHC. Our results indicated that inhibiting iNOS might be an effective strategy to protect neurons from oxidative stress.


Asunto(s)
Bibencilos/farmacología , Encéfalo/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Animales , Antioxidantes/farmacología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Mediadores de Inflamación/metabolismo , Masculino , Microglía/inmunología , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxidopamina , Células PC12 , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , alfa-Sinucleína/metabolismo
11.
Mucosal Immunol ; 14(3): 652-666, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33184477

RESUMEN

Evidence from inflammatory bowel diseases (IBD) patients and animal models has indicated that gut inflammation is driven by effector CD4+ T-cell, including Th1 and Th17. Conversely, Treg seem to be dysfunctional in IBD. Importantly, dopamine, which is abundant in the gut mucosa under homoeostasis, undergoes a sharp reduction upon intestinal inflammation. Here we analysed the role of the high-affinity dopamine receptor D3 (DRD3) in gut inflammation. Our results show that Drd3 deficiency confers a stronger immunosuppressive potency to Treg, attenuating inflammatory colitis manifestation in mice. Mechanistic analyses indicated that DRD3-signalling attenuates IL-10 production and limits the acquisition of gut-tropism. Accordingly, the ex vivo transduction of wild-type Treg with a siRNA for Drd3 induced a potent therapeutic effect abolishing gut inflammation. Thus, our findings show DRD3-signalling as a major regulator of Treg upon gut inflammation.


Asunto(s)
Colitis/inmunología , Neuronas Dopaminérgicas/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Receptores de Dopamina D3/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , ARN Interferente Pequeño/genética , Receptores de Dopamina D3/genética , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal
12.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182554

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder, caused by, so far, unknown pathogenetic mechanisms. There is no doubt that pro-inflammatory immune-mediated mechanisms are pivotal to the pathogenicity and progression of the disease. In this review, we highlight the binary role of microglia activation in the pathophysiology of the disorder, both neuroprotective and neuromodulatory. We present how the expression of several cytokines implicated in dopaminergic neurons (DA) degeneration could be used as biomarkers for PD. Viral infections have been studied and correlated to the disease progression, usually operating as trigger factors for the inflammatory process. The gut-brain axis and the possible contribution of the peripheral bowel inflammation to neuronal death, mainly dopaminergic neurons, seems to be a main contributor of brain neuroinflammation. The role of the immune system has also been analyzed implicating a-synuclein in the activation of innate and adaptive immunity. We also discuss therapeutic approaches concerning PD and neuroinflammation, which have been studied in experimental and in vitro models and data stemming from epidemiological studies.


Asunto(s)
Enfermedad de Parkinson/etiología , Animales , Autoinmunidad , Biomarcadores/metabolismo , Citocinas/inmunología , Citocinas/fisiología , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/fisiología , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/fisiopatología , Microglía/inmunología , Microglía/fisiología , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Trastornos Parkinsonianos/etiología , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Transducción de Señal , Virosis/complicaciones , alfa-Sinucleína/inmunología , alfa-Sinucleína/fisiología
13.
J Neuroinflammation ; 17(1): 347, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213462

RESUMEN

BACKGROUND: The R1441G mutation in the leucine-rich repeat kinase 2 (LRRK2) gene results in late-onset Parkinson's disease (PD). Peripheral inflammation and gut microbiota are closely associated with the pathogenesis of PD. Chronic periodontitis is a common type of peripheral inflammation, which is associated with PD. Porphyromonas gingivalis (Pg), the most common bacterium causing chronic periodontitis, can cause alteration of gut microbiota. It is not known whether Pg-induced dysbiosis plays a role in the pathophysiology of PD. METHODS: In this study, live Pg were orally administrated to animals, three times a week for 1 month. Pg-derived lipopolysaccharide (LPS) was used to stimulate mononuclear cells in vitro. The effects of oral Pg administration on the gut and brain were evaluated through behaviors, morphology, and cytokine expression. RESULTS: Dopaminergic neurons in the substantia nigra were reduced, and activated microglial cells were increased in R1441G mice given oral Pg. In addition, an increase in mRNA expression of tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß) as well as protein level of α-synuclein together with a decrease in zonula occludens-1 (Zo-1) was detected in the colon in Pg-treated R1441G mice. Furthermore, serum interleukin-17A (IL-17A) and brain IL-17 receptor A (IL-17RA) were increased in Pg-treated R1441G mice. CONCLUSIONS: These findings suggest that oral Pg-induced inflammation may play an important role in the pathophysiology of LRRK2-associated PD.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Inmunidad/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/inmunología , Microglía/inmunología , Enfermedades Neurodegenerativas/inmunología , Porphyromonas gingivalis/inmunología , Administración Oral , Animales , Infecciones por Bacteroidaceae/genética , Infecciones por Bacteroidaceae/inmunología , Células Cultivadas , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/microbiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Ratones Transgénicos , Microglía/microbiología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/microbiología , Permeabilidad , Sustancia Negra/inmunología , Sustancia Negra/microbiología
14.
Nat Commun ; 11(1): 5163, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33057020

RESUMEN

Parkinson's disease-associated kinase LRRK2 has been linked to IFN type II (IFN-γ) response in infections and to dopaminergic neuronal loss. However, whether and how LRRK2 synergizes with IFN-γ remains unclear. In this study, we employed dopaminergic neurons and microglia differentiated from patient-derived induced pluripotent stem cells carrying LRRK2 G2019S, the most common Parkinson's disease-associated mutation. We show that IFN-γ enhances the LRRK2 G2019S-dependent negative regulation of AKT phosphorylation and NFAT activation, thereby increasing neuronal vulnerability to immune challenge. Mechanistically, LRRK2 G2019S suppresses NFAT translocation via calcium signaling and possibly through microtubule reorganization. In microglia, LRRK2 modulates cytokine production and the glycolytic switch in response to IFN-γ in an NFAT-independent manner. Activated LRRK2 G2019S microglia cause neurite shortening, indicating that LRRK2-driven immunological changes can be neurotoxic. We propose that synergistic LRRK2/IFN-γ activation serves as a potential link between inflammation and neurodegeneration in Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/inmunología , Interferón gamma/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Microglía/inmunología , Enfermedad de Parkinson/inmunología , Señalización del Calcio/genética , Diferenciación Celular , Citocinas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Técnicas de Inactivación de Genes , Glucólisis/genética , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Interferón gamma/inmunología , Microscopía Intravital , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Microglía/metabolismo , Microtúbulos/metabolismo , Mutación , Factores de Transcripción NFATC/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Transducción de Señal/genética , Transducción de Señal/inmunología , Células THP-1
15.
Cell Tissue Res ; 382(1): 135-146, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32870383

RESUMEN

The glial cell line-derived neurotrophic factor (GDNF) and its canonical receptor Ret can signal both in tandem and separately to exert many vital functions in the midbrain dopamine system. It is known that Ret has effects on maintenance, physiology, protection and regeneration in the midbrain dopamine system, with the physiological functions of GDNF still somewhat unclear. Notwithstanding, Ret ligands, such as GDNF, are considered as promising candidates for neuroprotection and/or regeneration in Parkinson's disease, although data from clinical trials are so far inconclusive. In this review, we discuss the current knowledge of GDNF/Ret signaling in the dopamine system in vivo as well as crosstalk with pathology-associated proteins and their signaling in mammals.


Asunto(s)
Neuronas Dopaminérgicas/inmunología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas Proto-Oncogénicas c-ret/genética , Animales , Humanos , Transducción de Señal
16.
Stem Cells Dev ; 29(13): 853-862, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32245345

RESUMEN

In recent years, great hope has arisen surrounding human stem cells, particularly human induced pluripotent stem (hiPS) cells, as nearly all human tissues can be derived from hiPS cells, using a specific protocol. Therefore, hiPS cells can be a source for replacing defective tissues and make up for the lack of organ donors. However, the alloreactivity of hiPS cells and their derivatives in the context of transplantation remain unclear. Although immunosuppressive drugs can inhibit the T cell compartment, these drugs inhibit partially or not at all natural killer (NK) cells activity. Therefore, the alloreactivity of NK cells against transplanted cells remains to be established. To partially answer this question, we choose, as a model, the potential of cellular therapy for Parkinson's disease (PD). First, we established the in vitro derivation of hiPS cells into mature dopaminergic (mDOPA) neurons, going through an intermediate step called neurosphere (NS) cells. These different cells population were cultured with or without interferon gamma (IFN-γ). They were characterized phenotypically regarding their morphology, and the expression of specific ligands for NK cell receptors expressed by these cells types was investigated. NK cells were isolated from the peripheral blood of healthy donors and cultured in the presence of interleukin 15, to be activated. To test NK cell alloreactivity, a cytotoxic assay was performed with hiPS cells, NS cells, and mDOPA neurons (IFN-γ treated or not) cocultured with allogenic NK cells. Our results show that allogenic NK cells kill hiPS cells (IFN-γ treated or not), but IFN-γ-treated NS cells were protected from killing by allogenic NK cells, compared with untreated NS cells. Finally, mDOPA neurons (IFN-γ treated or not) were partially protected against allogenic NK cell killing. These results indicate that derivatives of hiPS cells, especially NS cells, could be a good product for allogenic transplantation in cellular therapy for PD.


Asunto(s)
Autoinmunidad , Neuronas Dopaminérgicas/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Células Asesinas Naturales/inmunología , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Interferón gamma/farmacología , Neurogénesis
17.
FASEB J ; 34(5): 6570-6581, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32246801

RESUMEN

Dysfunction of the circadian rhythm is one of most common nonmotor symptoms in Parkinson's disease (PD), but the molecular role of the circadian rhythm in PD is unclear. We here showed that inactivation of brain and muscle ARNT-like 1 (BMAL1) in 1-methyl-4-phenyl-1,2,4,5-tetrahydropyridine (MPTP)-treated mice resulted in obvious motor functional deficit, loss of dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNpc), decrease of dopamine (DA) transmitter, and increased activation of microglia and astrocytes in the striatum. Time on the rotarod or calorie consumption, and food and water intake were reduced in the Bmal1-/- mice after MPTP treatment, suggesting that absence of Bmal1 may exacerbate circadian and PD motor function. We observed a significant reduction of DANs (~35%) in the SNpc, the tyrosine hydroxylase protein level in the striatum (~60%), the DA (~22%), and 3,4-dihydroxyphenylacetic acid content (~29%), respectively, in MPTP-treated Bmal1-/- mice. Loss of Bmal1 aggravated the inflammatory reaction both in vivo and in vitro. These findings suggest that BMAL1 may play an essential role in the survival of DANs and maintain normal function of the DA signaling pathway via regulating microglia-mediated neuroinflammation in the brain.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Factores de Transcripción ARNTL/fisiología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Inflamación/patología , Microglía/patología , Enfermedad de Parkinson/patología , Animales , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Inflamación/etiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Neurotoxinas/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo
18.
Toxicol Mech Methods ; 30(5): 350-357, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32189544

RESUMEN

Rotenone is a mitochondrial complex I inhibitor, which can cause the death of dopaminergic (DA) neurons and Parkinson's disease (PD). Currently, whether metformin has a protective effect on neurotoxicity induced by rotenone is unclear. The purpose of this study was to evaluate the potential protective effect of metformin against rotenone-induced neurotoxicity. PD animal model was established by unilateral rotenone injection into the right substantia nigra (SN) of C57BL/6 mice. The behavioral tests were performed by rotarod test and cylinder test. The numbers of TH-positive neurons and Iba-1 positive microglia in the SN were investigated by immunohistochemical staining. The mRNA levels of proinflammatory cytokines (TNF-α and IL-1ß) and molecules involved in endoplasmic reticulum (ER) stress (ATF4, ATF6, XBP1, Grp78, and CHOP) in the midbrain were detected by Quantitative real-time PCR. This study showed that 50 mg/kg metformin given orally daily, beginning 3 d before rotenone injection and continuing for 4 weeks following rotenone injection, significantly ameliorated dyskinesia, increased the number of TH-positive neurons, and mitigated the activation of microglia in the SN in rotenone-induced PD mice. Furthermore, 50 mg/kg metformin markedly downregulated the expression of proinflammatory cytokines (TNF-α and IL-1ß) and ER stress-related genes (ATF4, ATF6, XBP1, Grp78, and CHOP) in rotenone-induced PD mice. Metformin has a protective effect on DA neurons against rotenone-induced neurotoxicity through inhibiting neuroinflammation and ER stress in PD mouse model.


Asunto(s)
Conducta Animal/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Metformina/farmacología , Enfermedad de Parkinson Secundaria/prevención & control , Sustancias Protectoras/farmacología , Rotenona/toxicidad , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/inmunología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/inmunología , Inflamación , Interleucina-1beta/metabolismo , Masculino , Metformina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/inmunología , Sustancias Protectoras/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Neural Transm (Vienna) ; 127(2): 149-158, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32016606

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease associated with motor deficiency and rigidity. The genetic risks of the disease is reported to be between 5 and 10% depending on the background of the population. While PD is not considered an immune-mediated disease, amounting evidence in recent years suggests a major role of inflammation in the progression of PD. Markers of inflammation can be found around the regions of risk and adjacent to the appearance of Lewy bodies within the basal ganglia and the substantia nigra (SN) that are associated with PD pathology. Microglia, an important type of brain cell, has been reported to play a major role in mediating neuroinflammation and in PD disease pathology. This review aims to point out the potential role of microglia in disease progression and suggest that the interaction of microglia with the dopaminergic neurons may also facilitate the specificity of the disease in brain regions affected by PD.


Asunto(s)
Progresión de la Enfermedad , Neuronas Dopaminérgicas , Inflamación , Microglía , Enfermedad de Parkinson , Animales , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Microglía/inmunología , Microglía/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/metabolismo
20.
J Neuroimmune Pharmacol ; 15(1): 93-113, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31317376

RESUMEN

The overwhelming prevalence of obesity is a priority for public health compromising human lifespan and representing important economic burden worldwide. Obesity is characterized by a state of chronic low-grade inflammation associated to metabolic dysfunction. Although the efforts for unravelling the complex immunometabolic signaling pathways to explain the association of obesity with type 2 diabetes, cardiovascular diseases, cancer, neurodegenerative diseases and psychiatric disorders, we still do not have all the picture to design effective therapeutic to fight these immunometabolic disease clusters. Dopaminergic pathways apart from having a major role in the regulation of appetite and feeding behaviors are important immunoregulators in inflammation; thus, dopaminergic regulation is suggested to impact obesity- associated inflammation. Dopamine (DA) has been reported to modulate immune function and immune cells themselves produce endogenous DA. DA-induced immunomodulation is currently the focus of intense experimental research and dopaminergic pathways are increasingly considered a target for drug development in immune diseases. While the role of dopaminergic pathways in immune-mediated diseases has been intensively investigated in neurodegenerative diseases, dopaminergic immunomodulation in obesity-associated inflammation is largely unknown. This review will integrate the actual knowledge about dopaminergic pathways involved in obesity-associated inflammation with special focus on immune innate key cell players. We present an explanatory hypothesis with a model that integrate central and peripheral dopaminergic circuits in the relationship between neuroimmune and metabolic systems in obesity-associated inflammation. A perspective on the potential role of dopaminergic drugs in the context of obesity will be given. Graphical Abstract Graphical representation of central and peripheral dopaminergic pathways in obesity-associated inflammation.


Asunto(s)
Dopaminérgicos/uso terapéutico , Dopamina/inmunología , Neuronas Dopaminérgicas/inmunología , Obesidad/inmunología , Receptores Dopaminérgicos/inmunología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/metabolismo , Dopamina/metabolismo , Dopaminérgicos/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Receptores Dopaminérgicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...