Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Mol Brain ; 17(1): 47, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075534

RESUMEN

In this study, we examined how systemic inflammation affects repair of brain injury. To this end, we created a brain-injury model by stereotaxic injection of ATP, a damage-associated molecular pattern component, into the striatum of mice. Systemic inflammation was induced by intraperitoneal injection of lipopolysaccharide (LPS-ip). An analysis of magnetic resonance images showed that LPS-ip reduced the initial brain injury but slowed injury repair. An immunostaining analysis using the neuronal marker, NeuN, showed that LPS-ip delayed removal of dead/dying neurons, despite the fact that LPS-ip enhanced infiltration of monocytes, which serve to phagocytize dead cells/debris. Notably, infiltrating monocytes showed a widely scattered distribution. Bulk RNAseq analyses showed that LPS-ip decreased expression of genes associated with phagocytosis, with PCR and immunostaining of injured brains confirming reduced levels of Cd68 and Clec7a, markers of phagocytic activity, in monocytes. Collectively, these results suggest that systemic inflammation affects properties of blood monocytes as well as brain cells, resulting in delay in clearing damaged cells and activating repair processes.


Asunto(s)
Encéfalo , Inflamación , Lipopolisacáridos , Ratones Endogámicos C57BL , Monocitos , Fagocitosis , Animales , Fagocitosis/efectos de los fármacos , Monocitos/metabolismo , Inflamación/patología , Encéfalo/patología , Masculino , Lipopolisacáridos/farmacología , Lesiones Encefálicas/patología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Neuronas/metabolismo , Neuronas/patología , Neuronas/efectos de los fármacos , Lectinas Tipo C/metabolismo , Cicatrización de Heridas , Ratones , Adenosina Trifosfato/metabolismo , Molécula CD68
2.
Nanoscale ; 16(2): 833-847, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38093712

RESUMEN

Astrocytes are highly activated following brain injuries, and their activation influences neuronal survival. Additionally, SOX9 expression is known to increase in reactive astrocytes. However, the role of SOX9 in activated astrocytes following ischemic brain damage has not been clearly elucidated yet. Therefore, in the present study, we investigated the role of SOX9 in reactive astrocytes using a poly-lactic-co-glycolic acid (PLGA) nanoparticle plasmid delivery system in a photothrombotic stroke animal model. We designed PLGA nanoparticles to exclusively enhance SOX9 gene expression in glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Our observations indicate that PLGA nanoparticles encapsulated with GFAP:SOX9:tdTOM reduce ischemia-induced neurological deficits and infarct volume through the prostaglandin D2 pathway. Thus, the astrocyte-targeting PLGA nanoparticle plasmid delivery system provides a potential opportunity for stroke treatment. Since the only effective treatment currently available is reinstating the blood supply, cell-specific gene therapy using PLGA nanoparticles will open a new therapeutic paradigm for brain injury patients in the future.


Asunto(s)
Lesiones Encefálicas , Nanopartículas , Accidente Cerebrovascular , Humanos , Animales , Astrocitos/metabolismo , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Lesiones Encefálicas/metabolismo , Péptidos/farmacología , Encéfalo/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/farmacología
3.
Theranostics ; 12(13): 5856-5876, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35966584

RESUMEN

Preconditioning nerve injury enhances axonal regeneration of dorsal root ganglia (DRG) neurons in part by driving pro-regenerative perineuronal macrophage activation. How these macrophages influence the neuronal capacity of axon regeneration remains elusive. We report that oncomodulin (ONCM) is produced from the regeneration-associated macrophages and strongly influences regeneration of DRG sensory axons. We also attempted to promote sensory axon regeneration by nanogel-mediated delivery of ONCM to DRGs. Methods:In vitro neuron-macrophage interaction model and preconditioning sciatic nerve injury were used to verify the necessity of ONCM in preconditioning injury-induced neurite outgrowth. We developed a nanogel-mediated delivery system in which electrostatic encapsulation of ONCM by a reducible epsilon-poly(L-lysine)-nanogel (REPL-NG) enabled a controlled release of ONCM. Results: Sciatic nerve injury upregulated ONCM in DRG macrophages. ONCM in macrophages was necessary to produce pro-regenerative macrophages in the in vitro model of neuron-macrophage interaction and played an essential role in preconditioning-induced neurite outgrowth. ONCM increased neurite outgrowth in cultured DRG neurons by activating a distinct gene set, particularly neuropeptide-related genes. Increasing extracellularly secreted ONCM in DRGs sufficiently enhanced the capacity of neurite outgrowth. Intraganglionic injection of REPL-NG/ONCM complex allowed sustained ONCM activity in DRG tissue and achieved a remarkable long-range regeneration of dorsal column sensory axons beyond spinal cord lesion. Conclusion: NG-mediated ONCM delivery could be exploited as a therapeutic strategy for promoting sensory axon regeneration following spinal cord injury.


Asunto(s)
Axones , Traumatismos de los Nervios Periféricos , Axones/fisiología , Proteínas de Unión al Calcio , Humanos , Macrófagos/fisiología , Nanogeles , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/patología , Médula Espinal
4.
FEBS J ; 289(19): 5914-5932, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35429212

RESUMEN

Interleukin-6 (IL-6) is involved in many inflammatory diseases. IL-6 binds to membrane-bound IL-6 receptor α (IL-6Rα) (classic signaling) or soluble IL-6Rα (trans-signaling); this complex then associates with the signal-transducing membrane protein gp130. IL-6Rα and gp130 float on membrane (i.e., lipid) rafts; however, how membrane rafts regulate IL-6 signaling remains unclear. Here, we demonstrate that both IL-6 classic signaling and trans-signaling depend on membrane cholesterol, an essential raft component. Super-resolution fluorescence imaging using perfringolysin O D4 fragments that selectively bind to high cholesterol concentrations revealed that IL-6 and hyper-IL-6, a fusion protein of IL-6 and soluble IL-6Rα, induce the alteration of membrane rafts. IL-6 and hyper-IL-6 induced D4-positive raft (D4 raft) formation without affecting cholera toxin subunit B (CTB)-positive rafts (CTB rafts). Receptor clustering of IL-6Rα and gp130 and STAT3 phosphorylation occurred in D4 rafts. These results indicate that D4 rafts serve as platforms for the assembly of functional IL-6 receptor complexes. We found that Eps15 homology domain-containing protein 1 (EHD1) mediates the formation of functional IL-6 receptor complexes through D4 rafts. Overall, we uncover a novel regulatory mechanism of the EHD1-mediated alteration of membrane raft in IL-6 signaling.


Asunto(s)
Toxina del Cólera , Interleucina-6 , Toxina del Cólera/metabolismo , Colesterol/metabolismo , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo
5.
Acta Neuropathol Commun ; 7(1): 149, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530290

RESUMEN

Multifunctional signal transducer and activator of transcription (STAT) proteins play important roles in cancer. Here, we have shown that STAT6 is epigenetically silenced in some cases of malignant glioblastoma, which facilitates cancer cell survival in a hypoxic microenvironment. This downregulation results from hypermethylation of CpG islands within the STAT6 promoter by DNA methyltransferases. STAT6 interacts with Rheb under hypoxia and inhibits mTOR/S6K/S6 signaling, in turn, inducing increased HIF-1α translation. STAT6 silencing and consequent tumor-promoting effects are additionally observed in glioma stem-like cells (GSC). Despite recent advances in cancer treatment, survival rates have shown little improvement. This is particularly true in the case of glioma, where multimodal treatment and precision medicine is needed. Our study supports the application of epigenetic restoration of STAT6 with the aid of DNA methyltransferase inhibitors, such as 5-aza-2-deoxycytidine, for treatment of STAT6-silenced gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor de Transcripción STAT6/metabolismo , Línea Celular Tumoral , Supervivencia Celular , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , Regulación hacia Abajo , Humanos , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Hipoxia Tumoral
6.
Neurobiol Dis ; 127: 482-491, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30954702

RESUMEN

Dysfunctional regulation of inflammation may contribute to the progression of neurodegenerative diseases. The results of this study revealed that DJ-1, a Parkinson's disease (PD) gene, regulated expression of prostaglandin D2 synthase (PTGDS) and production of prostaglandin D2 (PGD2), by which DJ-1 enhanced anti-inflammatory function of astrocytes. In injured DJ-1 knockout (KO) brain, expression of tumor necrosis factor-alpha (TNF-α) was more increased, but that of anti-inflammatory heme oxygenase-1 (HO-1) was less increased compared with that in injured wild-type (WT) brain. Similarly, astrocyte-conditioned media (ACM) prepared from DJ-1-KO astrocytes less induced HO-1 expression and less inhibited expression of inflammatory mediators in microglia. With respect to the underlying mechanism, we found that PTGDS that induced expression of HO-1 was lower in DJ-1 KO astrocytes and brains compared with their WT counterparts. In addition, PTGDS levels increased in the injured brain of WT mice, but barely in that of KO mice. We also found that DJ-1 regulated PTGDS expression through Sox9. Thus, Sox9 siRNAs reduced PTGDS expression in WT astrocytes, and Sox9 overexpression rescued PTGDS expression in DJ-1 KO astrocytes. In agreement with these results, ACM from Sox9 siRNA-treated astrocytes and that from Sox9-overexpression astrocytes exerted opposite effects on HO-1 expression and anti-inflammation. These findings suggest that DJ-1 positively regulates anti-inflammatory functions of astrocytes, and that DJ-1 dysfunction contributes to the excessive inflammatory response in PD development.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Proteína Desglicasa DJ-1/genética , Animales , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inflamación/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
7.
Biochim Biophys Acta ; 1859(8): 1056-70, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27206966

RESUMEN

MAP kinase phosphatase (MKP)-1 plays a pivotal role in controlling MAP kinase (MAPK)-dependent (patho) physiological processes. Although MKP-1 gene expression is tightly regulated at multiple levels, the underlying mechanistic details remain largely unknown. In this study, we demonstrate that MKP-1 expression is regulated at the post-transcriptional level by 22(R)-hydroxycholesterol [22(R)-HC] through a novel mechanism. 22(R)-HC induces Hu antigen R (HuR) phosphorylation, cytoplasmic translocation and binding to MKP-1 mRNA, resulting in stabilization of MKP-1 mRNA. The resulting increase in MKP-1 leads to suppression of JNK-mediated inflammatory responses in brain astrocytes. We further demonstrate that 22(R)-HC-induced phosphorylation of nuclear HuR is mediated by PKCα, which is activated in the cytosol by increases in intracellular Ca(2+) levels mediated by the phospholipase C/inositol 1,4,5-triphosphate receptor (PLC/IP3R) pathway and translocates from cytoplasm to nucleus. In addition, pharmacological interventions reveal that metabotropic glutamate receptor5 (mGluR5) is responsible for the increases in intracellular Ca(2+) that underlie these actions of 22(R)-HC. Collectively, our findings identify a novel anti-inflammatory mechanism of 22(R)-HC, which acts through PKCα-mediated cytoplasmic shuttling of HuR to post-transcriptionally regulate MKP-1 expression. These findings provide an experimental basis for the development of a RNA-targeted therapeutic agent to control MAPK-dependent inflammatory responses.


Asunto(s)
Astrocitos/metabolismo , Fosfatasa 1 de Especificidad Dual/genética , Proteína 1 Similar a ELAV/genética , Hidroxicolesteroles/farmacología , Proteína Quinasa C-alfa/genética , ARN Mensajero/genética , Receptor del Glutamato Metabotropico 5/genética , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Proteína 1 Similar a ELAV/agonistas , Proteína 1 Similar a ELAV/metabolismo , Regulación de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Unión Proteica , Proteína Quinasa C-alfa/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Ratas , Receptor del Glutamato Metabotropico 5/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo
8.
Exp Neurobiol ; 25(1): 14-23, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26924929

RESUMEN

PTEN-induced putative kinase 1 (PINK1) is a Parkinson's disease (PD) gene. We examined miRNAs regulated by PINK1 during brain development and neural stem cell (NSC) differentiation, and found that lvels of miRNAs related to tumors and inflammation were different between 1-day-old-wild type (WT) and PINK1-knockout (KO) mouse brains. Notably, levels of miR-326, miR-330 and miR-3099, which are related to astroglioma, increased during brain development and NSC differentiation, and were significantly reduced in the absence of PINK1. Interestingly, in the presence of ciliary neurotrophic factor (CNTF), which pushes differentiation of NSCs into astrocytes, miR-326, miR-330, and miR-3099 levels in KO NSCs were also lower than those in WT NSCs. Furthermore, mimics of all three miRNAs increased expression of the astrocytic marker glial fibrillary acidic protein (GFAP) during differentiation of KO NSCs, but inhibitors of these miRNAs decreased GFAP expression in WT NSCs. Moreover, these miRNAs increased the translational efficacy of GFAP through the 3'-UTR of GFAP mRNA. Taken together, these results suggest that PINK1 deficiency reduce expression levels of miR-326, miR-330 and miR-3099, which may regulate GFAP expression during NSC differentiation and brain development.

9.
Neurobiol Dis ; 83: 90-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342897

RESUMEN

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. Although the etiology of PD has not yet been fully understood, accumulating evidence indicates that neuroinflammation plays a critical role in the progression of PD. α-Synuclein (α-Syn) has been considered to be a key player of the pathogenesis of PD, and recent reports that prion-like propagation of misfolded α-syn released from neurons may play an important role in the progression of PD have led to increased attention to the studies elucidating the roles of extracellular α-syn in the CNS. Extracellular α-syn has also been reported to regulate microglial inflammatory response. In this study, we demonstrated that aggregated α-syn inhibited microglial phagocytosis by activating SHP-1. SHP-1 activation was also observed in A53T α-syn transgenic mice. In addition, aggregated α-syn bound to FcγRIIB on microglia, inducing SHP-1 activation, further inhibiting microglial phagocytosis. Aggregated α-syn upregulated FcγRIIB expression in microglia and upregulated FcγRIIB was also observed in A53T α-syn transgenic mice. These data suggest that aggregated α-syn released from neurons dysregulates microglial immune response through inhibiting microglial phagocytosis, further causing neurodegeneration observed in PD. The interaction of aggregated α-syn and FcγRIIB and further SHP-1 activation can be a new therapeutic target against PD.


Asunto(s)
Encéfalo/metabolismo , Microglía/metabolismo , Microglía/fisiología , Fagocitosis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de IgG/metabolismo , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregado de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
10.
Biochim Biophys Acta ; 1849(6): 612-25, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25805336

RESUMEN

In the present study, we demonstrate a mechanism through which 15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2) induces MKP-1 expression in rat primary astrocytes, leading to the regulation of inflammatory responses. We show that 15d-PGJ2 enhances the efficiency of MKP-1 pre-mRNA processing (constitutive splicing and 3'-end processing) and increases the stability of the mature mRNA. We further report that this occurs via the RNA-binding protein, Hu antigen R (HuR). Our experiments show that HuR knockdown abrogates the 15d-PGJ2-induced increases in the pre-mRNA processing and mature mRNA stability of MKP-1, whereas HuR overexpression further enhances the 15d-PGJ2-induced increases in these parameters. Using cysteine (Cys)-mutated HuR proteins, we show that the Cys-245 residue of HuR (but not Cys-13 or Cys-284) is critical for the direct binding of HuR with 15d-PGJ2 and the effects downstream of this interaction. Collectively, our data show that HuR is a novel target of 15d-PGJ2 and reveal HuR-mediated pre-mRNA processing and mature mRNA stabilization as important regulatory steps in the 15d-PGJ2-induced expression of MKP-1. The potential to use a small molecule such as 15d-PGJ2 to regulate the induction of MKP-1 at multiple levels of gene expression could be exploited as a novel therapeutic strategy aimed at combating a diverse range of MKP-1-associated pathologies.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/genética , Proteínas ELAV/genética , Inflamación/genética , Prostaglandina D2/análogos & derivados , Animales , Astrocitos/metabolismo , Astrocitos/patología , Fosfatasa 1 de Especificidad Dual/biosíntesis , Proteínas ELAV/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Cultivo Primario de Células , Prostaglandina D2/administración & dosificación , Prostaglandina D2/metabolismo , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN/genética , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , Ratas
11.
EMBO Mol Med ; 7(5): 510-25, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25759364

RESUMEN

Use of the physiological mechanisms promoting midbrain DA (mDA) neuron survival seems an appropriate option for developing treatments for Parkinson's disease (PD). mDA neurons are specifically marked by expression of the transcription factors Nurr1 and Foxa2. We show herein that Nurr1 and Foxa2 interact to protect mDA neurons against various toxic insults, but their expression is lost during aging and degenerative processes. In addition to their proposed cell-autonomous actions in mDA neurons, forced expression of these factors in neighboring glia synergistically protects degenerating mDA neurons in a paracrine mode. As a consequence of these bimodal actions, adeno-associated virus (AAV)-mediated gene delivery of Nurr1 and Foxa2 in a PD mouse model markedly protected mDA neurons and motor behaviors associated with nigrostriatal DA neurotransmission. The effects of the combined gene delivery were dramatic, highly reproducible, and sustained for at least 1 year, suggesting that expression of these factors is a promising approach in PD therapy.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Terapia Genética/métodos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Mesencéfalo/patología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Factor Nuclear 3-beta del Hepatocito/genética , Masculino , Ratones Endogámicos ICR , Neuroglía/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Resultado del Tratamiento
12.
Hum Mol Genet ; 24(4): 1127-41, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305081

RESUMEN

Deciphering the molecular basis of neuronal cell death is a central issue in the etiology of neurodegenerative diseases, such as Parkinson's and Alzheimer's. Dysregulation of p53 levels has been implicated in neuronal apoptosis. The role of histone deacetylase 3 (HDAC3) in suppressing p53-dependent apoptosis has been recently emphasized; however, the molecular basis of modulation of p53 function by HDAC3 remains unclear. Here we show that PTEN-induced putative kinase 1 (PINK1), which is linked to autosomal recessive early-onset familial Parkinson's disease, phosphorylates HDAC3 at Ser-424 to enhance its HDAC activity in a neural cell-specific manner. PINK1 prevents H2O2-induced C-terminal cleavage of HDAC3 via phosphorylation of HDAC3 at Ser-424, which is reversed by protein phosphatase 4c. PINK1-mediated phosphorylation of HDAC3 enhances its direct association with p53 and causes subsequent hypoacetylation of p53. Genetic deletion of PINK1 partly impaired the suppressive role of HDAC3 in regulating p53 acetylation and transcriptional activity. However, depletion of HDAC3 fully abolished the PINK1-mediated p53 inhibitory loop. Finally, ectopic expression of phosphomometic-HDAC3(S424E) substantially overcomes the defective action of PINK1 against oxidative stress in dopaminergic neuronal cells. Together, our results uncovered a mechanism by which PINK1-HDAC3 network mediates p53 inhibitory loop in response to oxidative stress-induced damage.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Quinasas/metabolismo , Acetilación/efectos de los fármacos , Animales , Caspasa 7/metabolismo , Muerte Celular/genética , Línea Celular , Citoplasma/metabolismo , Neuronas Dopaminérgicas/patología , Activación Enzimática , Histona Desacetilasas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Especificidad de Órganos , Fosforilación , Proteínas Quinasas/genética , Proteolisis , Proteína p53 Supresora de Tumor/metabolismo
13.
Neurobiol Dis ; 60: 1-10, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23969237

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative movement disorder caused by the death of dopaminergic neurons in the substantia nigra. Importantly, altered astrocyte and microglial functions could contribute to neuronal death in PD. In this study, we demonstrate a novel mechanism by which DJ-1 (PARK7), an early onset autosomal-recessive PD gene, negatively regulates inflammatory responses of astrocytes and microglia by facilitating the interaction between STAT1 and its phosphatase, SHP-1 (Src-homology 2-domain containing protein tyrosine phosphatase-1). Astrocytes and microglia cultured from DJ-1-knockout (KO) mice exhibited increased expression of inflammatory mediators and phosphorylation levels of STAT1 (p-STAT1) in response to interferon-gamma (IFN-γ) compared to cells from wild-type (WT) mice. DJ-1 deficiency also attenuated IFN-γ-induced interactions of SHP-1 with p-STAT1 and STAT1, measured 1 and 12h after IFN-γ treatment, respectively. Subsequent experiments showed that DJ-1 directly interacts with SHP-1, p-STAT1, and STAT1. Notably, DJ-1 bound to SHP-1 independently of IFN-γ, whereas the interactions of DJ-1 with p-STAT1 and STAT1 were dependent on IFN-γ. Similar results were obtained in brain slice cultures, where IFN-γ induced much stronger STAT1 phosphorylation and inflammatory responses in KO slices than in WT slices. Moreover, IFN-γ treatment induced neuronal damage in KO slices. Collectively, these findings suggest that DJ-1 may function as a scaffold protein that facilitates SHP-1 interactions with p-STAT1 and STAT1, thereby preventing extensive and prolonged STAT1 activation. Thus, the loss of DJ-1 function may increase the risk of PD by enhancing brain inflammation.


Asunto(s)
Astrocitos/metabolismo , Microglía/metabolismo , Proteínas Oncogénicas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Antiinflamatorios/metabolismo , Encéfalo/metabolismo , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Peroxirredoxinas , Fosforilación , Proteína Desglicasa DJ-1
14.
Hum Mol Genet ; 22(23): 4805-17, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23847046

RESUMEN

Parkinson's disease (PD) is the second most common progressive neurodegenerative disease. Several genes have been associated with familial type PD, providing tremendous insights into the pathogenesis of PD. Gathering evidence supports the view that these gene products may operate through common molecular pathways. Recent reports suggest that many PD-associated gene products, such as α-synuclein, LRRK2, parkin and PINK1, associate with lipid rafts and lipid rafts may be associated with neurodegeneration. Here, we observed that DJ-1 protein also associated with lipid rafts. Palmitoylation of three cysteine residues (C46/53/106) and C-terminal region of DJ-1 were required for this association. Lipopolysaccharide (LPS) induced the localization of DJ-1 into lipid rafts in astrocytes. The LPS-TLR4 signaling was more augmented in DJ-1 knock-out astrocytes by the impairment of TLR4 endocytosis. Furthermore, lipid rafts-dependent endocytosis including the endocytosis of CD14, which play a major role in regulating TLR4 endocytosis was also impaired, but clathrin-dependent endocytosis was not. This study provides a novel function of DJ-1 in lipid rafts, which may contribute the pathogenesis of PD. Moreover, it also provides the possibility that many PD-related proteins may operate through common molecular pathways in lipid rafts.


Asunto(s)
Astrocitos/fisiología , Endocitosis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microdominios de Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Oncogénicas/metabolismo , Enfermedad de Parkinson/fisiopatología , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Cisteína/metabolismo , Humanos , Lipopolisacáridos/fisiología , Lipoilación , Ratones Noqueados , Enfermedad de Parkinson/metabolismo , Peroxirredoxinas , Proteína Desglicasa DJ-1 , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Receptores Toll-Like/fisiología
15.
J Am Chem Soc ; 135(26): 9915-23, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23745510

RESUMEN

Hydrogen sulfide (H2S) is a multifunctional signaling molecule that exerts neuroprotective effects in oxidative stress. In this article, we report a mitochondria-localized two-photon probe, SHS-M2, that can be excited by 750 nm femtosecond pulses and employed for ratiometric detection of H2S in live astrocytes and living brain slices using two-photon microscopy (TPM). SHS-M2 shows bright two-photon-excited fluorescence and a marked change in emission color from blue to yellow in response to H2S, low cytotoxicity, easy loading, and minimum interference from other biologically relevant species including reactive sulfur, oxygen, and nitrogen species, thereby allowing quantitative analysis of H2S levels. Molecular TPM imaging with SHS-M2 in astrocytes revealed that there is a correlation between the ratiometric analysis and expression levels of cystathionine ß-synthase (CBS), the major enzyme that catalyzes H2S production. In studies involving DJ-1, a Parkinson's disease (PD) gene, attenuated H2S production in comparison with wild-type controls was observed in DJ-1-knockout astrocytes and brain slices, where CBS expression was decreased. These findings demonstrate that reduced H2S levels in astrocytes may contribute to the development of PD and that SHS-M2 may be useful as a marker to detect a risk of neurodegenerative diseases, including PD.


Asunto(s)
Astrocitos/efectos de los fármacos , Colorantes Fluorescentes/farmacología , Sulfuro de Hidrógeno/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Oncogénicas/metabolismo , Enfermedad de Parkinson/metabolismo , Protones , Astrocitos/metabolismo , Colorantes Fluorescentes/química , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Mitocondrias/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Oxidación-Reducción , Enfermedad de Parkinson/genética , Proteína Desglicasa DJ-1
16.
Neurobiol Dis ; 58: 249-57, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23759293

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) has been shown to enhance the recovery of brain functions following ischemic injury. Although immune modulation has been suggested to be one of the mechanisms, the molecular mechanisms underlying improved recovery has not been clearly identified. Here, we report that MSCs secrete transforming growth factor-beta (TGF-ß) to suppress immune propagation in the ischemic rat brain. Ischemic stroke caused global death of resident cells in the infarcted area, elevated the monocyte chemoattractant protein-1 (MCP-1) level, and evoked massive infiltration of circulating CD68+ immune cells through the impaired blood-brain barrier. Transplantation of MSCs at day 3 post-ischemia blocked the subsequent upregulation of MCP-1 in the ischemic area and the infiltration of additional CD68+ immune cells. MSC-conditioned media decreased the migration and MCP-1 production of freshly isolated immune cells in vitro, and this effect was blocked by an inhibitor of TGF-ß signaling or an anti-TGF-ß neutralizing antibody. Finally, transplantation of TGF-ß1-silenced MSCs failed to attenuate the infiltration of CD68+ cells into the ischemic brain, and was associated with only minor improvements in motor function. These results indicate that TGF-ß is key to the ability of MSCs to beneficially attenuate immune reactions in the ischemic brain. Our findings offer insight into the interactions between allogeneic MSCs and the host immune system, reinforcing the prospective clinical value of using MSCs in the treatment of neurological disorders involving inflammation-mediated secondary damage.


Asunto(s)
Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/cirugía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antígenos CD/metabolismo , Barrera Hematoencefálica/fisiopatología , Infarto Encefálico/etiología , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Encefalitis/etiología , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Proteínas de Microfilamentos/metabolismo , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología
17.
Cell Signal ; 25(9): 1861-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23707391

RESUMEN

Fibrillar amyloid-beta (fAß) peptide causes neuronal cell death, which is known as Alzheimer's disease. One of the mechanisms for neuronal cell death is the activation of microglia which releases toxic compounds like reactive oxygen species (ROS) in response to fAß. We observed that fAß rather than soluble form blocked BV2 cell proliferation of microglial cell line BV2, while N-acetyl-l-cysteine (NAC), a scavenger of superoxide, prevented the cells from death, suggesting that cell death is induced by ROS. Indeed, both fAß1-42 and fAß25-35 induced superoxide production in BV2 cells. fAß25-35 produced superoxide, although fAß25-35 is not phagocytosed into BV2 cells. Thus, superoxide production by fAß does not seem to be dependent on phagocytosis of fAß. Herein we studied how fAß produces superoxide in BV2. Transfection of dominant negative (DN) RhoA (N19) cDNA plasmid, small hairpin (sh)-RhoA forming plasmid, and Y27632, an inhibitor of Rho-kinase, abrogated the superoxide formation in BV2 cells stimulated by fAß. Furthermore, fAß elevated GTP-RhoA level as well as Rac1 and Cdc42. Tat-C3 toxin, sh-RhoA, and Y27632 inhibited the phosphorylation of p47(PHOX). Moreover, peritoneal macrophages from p47(PHOX) (-/-) knockout mouse could not produce superoxide in response to fAß. These results suggest that RhoA closely engages in the regulation of superoxide production induced by fAß through phosphorylation of p47(PHOX) in microglial BV2 cells.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/citología , Superóxidos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Animales , Línea Celular , Ratones , Microglía/metabolismo , Datos de Secuencia Molecular , NADPH Oxidasas/metabolismo , Fosforilación
18.
Glia ; 61(5): 800-12, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23440919

RESUMEN

PINK1 (PTEN induced putative kinase 1), a familial Parkinson's disease (PD)-related gene, is expressed in astrocytes, but little is known about its role in this cell type. Here, we found that astrocytes cultured from PINK1-knockout (KO) mice exhibit defective proliferative responses to epidermal growth factor (EGF) and fetal bovine serum. In PINK1-KO astrocytes, basal and EGF-induced p38 activation (phosphorylation) were increased whereas EGF receptor (EGFR) expression and AKT activation were decreased. p38 inhibition (SB203580) or knockdown with small interfering RNA (siRNA) rescued EGFR expression and AKT activation in PINK1-KO astrocytes. Proliferation defects in PINK1-KO astrocytes appeared to be linked to mitochondrial defects, manifesting as decreased mitochondrial mass and membrane potential, increased intracellular reactive oxygen species level, decreased glucose-uptake capacity, and decreased ATP production. Mitochondrial toxin (oligomycin) and a glucose-uptake inhibitor (phloretin) mimicked the PINK1-deficiency phenotype, decreasing astrocyte proliferation, EGFR expression and AKT activation, and increasing p38 activation. In addition, the proliferation defect in PINK1-KO astrocytes resulted in a delay in the wound healing process. Taken together, these results suggest that PINK1 deficiency causes astrocytes dysfunction, which may contribute to the development of PD due to delayed astrocytes-mediated repair of microenvironment in the brain.


Asunto(s)
Astrocitos/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Mitocondrias/metabolismo , Proteínas Quinasas/deficiencia , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Animales , Astrocitos/patología , Bovinos , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Regulación hacia Arriba/genética
19.
PLoS One ; 7(4): e34693, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496842

RESUMEN

LRRK2, a Parkinson's disease associated gene, is highly expressed in microglia in addition to neurons; however, its function in microglia has not been evaluated. Using Lrrk2 knockdown (Lrrk2-KD) murine microglia prepared by lentiviral-mediated transfer of Lrrk2-specific small inhibitory hairpin RNA (shRNA), we found that Lrrk2 deficiency attenuated lipopolysaccharide (LPS)-induced mRNA and/or protein expression of inducible nitric oxide synthase, TNF-α, IL-1ß and IL-6. LPS-induced phosphorylation of p38 mitogen-activated protein kinase and stimulation of NF-κB-responsive luciferase reporter activity was also decreased in Lrrk2-KD cells. Interestingly, the decrease in NF-κB transcriptional activity measured by luciferase assays appeared to reflect increased binding of the inhibitory NF-κB homodimer, p50/p50, to DNA. In LPS-responsive HEK293T cells, overexpression of the human LRRK2 pathologic, kinase-active mutant G2019S increased basal and LPS-induced levels of phosphorylated p38 and JNK, whereas wild-type and other pathologic (R1441C and G2385R) or artificial kinase-dead (D1994A) LRRK2 mutants either enhanced or did not change basal and LPS-induced p38 and JNK phosphorylation levels. However, wild-type LRRK2 and all LRRK2 mutant variants equally enhanced NF-κB transcriptional activity. Taken together, these results suggest that LRRK2 is a positive regulator of inflammation in murine microglia, and LRRK2 mutations may alter the microenvironment of the brain to favor neuroinflammation.


Asunto(s)
Encéfalo/metabolismo , Encefalitis/metabolismo , Microglía/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Interleucina-1beta/biosíntesis , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 4/metabolismo , Ratones , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Neuroinflammation ; 9: 34, 2012 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-22339770

RESUMEN

BACKGROUND: The peroxisome proliferator-activated receptor (PPAR)-α activator, 5,8,11,14-eicosatetraynoic acid (ETYA), is an arachidonic acid analog. It is reported to inhibit up-regulation of pro-inflammatory genes; however, its underlying mechanism of action is largely unknown. In the present study, we focused on the inhibitory action of ETYA on the expression of the chemokine, CCL2/MCP-1, which plays a key role in the initiation and progression of inflammation. METHODS: To determine the effect of ETYA, primary cultured rat astrocytes and microglia were stimulated with IFN-γ in the presence of ETYA and then, expression of CCL2/MCP-1 and MAPK phosphatase (MKP-1) were determined using RT-PCR and ELISA. MKP-1 mRNA stability was evaluated by treating actinomycin D. The effect of MKP-1 and human antigen R (HuR) was analyzed by using specific siRNA transfection system. The localization of HuR was analyzed by immunocytochemistry and subcellular fractionation experiment. RESULTS: We found that ETYA suppressed CCL2/MCP-1 transcription and secretion of CCL2/MCP-1 protein through up-regulation of MKP-1mRNA levels, resulting in suppression of c-Jun N-terminal kinase (JNK) phosphorylation and activator protein 1 (AP1) activity in IFN-γ-stimulated brain glial cells. Moreover, these effects of ETYA were independent of PPAR-α. Experiments using actinomycin D revealed that the ETYA-induced increase in MKP-1 mRNA levels reflected an increase in transcript stability. Knockdown experiments using small interfering RNA demonstrated that this increase in MKP-1 mRNA stability depended on HuR, an RNA-binding protein known to promote enhanced mRNA stability. Furthermore, ETYA-induced, HuR-mediated mRNA stabilization resulted from HuR-MKP-1 nucleocytoplasmic translocation, which served to protect MKP-1 mRNA from the mRNA degradation machinery. CONCLUSION: ETYA induces MKP-1 through HuR at the post-transcriptional level in a receptor-independent manner. The mechanism revealed here suggests eicosanoids as potential therapeutic modulators of inflammation that act through a novel target.


Asunto(s)
Ácido 5,8,11,14-Eicosatetrainoico/farmacología , Astrocitos/efectos de los fármacos , Quimiocina CCL2/metabolismo , Fosfatasa 1 de Especificidad Dual/genética , Interferón gamma/farmacología , ARN Mensajero/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Inmunoprecipitación de Cromatina , Proteínas ELAV , Ensayo de Cambio de Movilidad Electroforética , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Microglía/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA