Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Neurobiol Dis ; 197: 106528, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38740348

RESUMEN

BACKGROUND: Brain injury has been suggested as a risk factor for neurodegenerative diseases. Accordingly, defects in the brain's intrinsic capacity to repair injury may result in the accumulation of damage and a progressive loss of brain function. The G2019S (GS) mutation in LRRK2 (leucine rich repeat kinase 2) is the most prevalent genetic alteration in Parkinson's disease (PD). Here, we sought to investigate how this LRRK2-GS mutation affects repair of the injured brain. METHODS: Brain injury was induced by stereotaxic injection of ATP, a damage-associated molecular pattern (DAMP) component, into the striatum of wild-type (WT) and LRRK2-GS mice. Effects of the LRRK2-GS mutation on brain injury and the recovery from injury were examined by analyzing the molecular and cellular behavior of neurons, astrocytes, and monocytes. RESULTS: Damaged neurons express osteopontin (OPN), a factor associated with brain repair. Following ATP-induced damage, monocytes entered injured brains, phagocytosing damaged neurons and producing exosome-like vesicles (EVs) containing OPN through activation of the inflammasome and subsequent pyroptosis. Following EV production, neurons and astrocytes processes elongated towards injured cores. In LRRK2-GS mice, OPN expression and monocytic pyroptosis were decreased compared with that in WT mice, resulting in diminished release of OPN-containing EVs and attenuated elongation of neuron and astrocyte processes. In addition, exosomes prepared from injured LRRK2-GS brains induced neurite outgrowth less efficiently than those from injured WT brains. CONCLUSIONS: The LRRK2-GS mutation delays repair of injured brains through reduced expression of OPN and diminished release of OPN-containing EVs from monocytes. These findings suggest that the LRRK2-GS mutation may promote the development of PD by delaying the repair of brain injury.


Asunto(s)
Lesiones Encefálicas , Exosomas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Monocitos , Mutación , Osteopontina , Animales , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Exosomas/metabolismo , Exosomas/genética , Osteopontina/metabolismo , Osteopontina/genética , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/genética , Monocitos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Masculino , Astrocitos/metabolismo
2.
EMBO Rep ; 21(7): e48950, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32372484

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons located in the substantia nigra pars compacta and the presence of proteinaceous inclusions called Lewy bodies and Lewy neurites in numerous brain regions. Increasing evidence indicates that Lewy pathology progressively involves additional regions of the nervous system as the disease advances, and the prion-like propagation of α-synuclein (α-syn) pathology promotes PD progression. Accordingly, the modulation of α-syn transmission may be important for the development of disease-modifying therapies in patients with PD. Here, we demonstrate that α-syn fibrils induce c-src activation in neurons, which depends on the FcγRIIb-SHP-1/-2-c-src pathway and enhances signals for the uptake of α-syn into neurons. Blockade of c-src activation inhibits the uptake of α-syn and the formation of Lewy body-like inclusions. Furthermore, the blockade of c-src activation also inhibits the release of α-syn via activation of autophagy. The brain-permeable c-src inhibitor, saracatinib, efficiently reduces α-syn propagation into neighboring regions in an in vivo model system. These results suggest a new therapeutic target against progressive PD.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
Glia ; 69(4): 1037-1052, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33300228

RESUMEN

The brain has an intrinsic capacity to repair injury, but the specific mechanisms are largely unknown. In this study, we found that, despite their incipient death, damaged neurons play a key repair role with the help of monocytes infiltrated from blood. Monocytes phagocytosed damaged and/or dying neurons that expressed osteopontin (OPN), with possible subsequent activation of their inflammasome pathway, resulting in pyroptosis. During this process, monocytes released CD63-positive exosome-like vesicles containing OPN. Importantly, following the exosome-like vesicles, neuron and astrocyte processes elongated toward the injury core. In addition, exosomes prepared from the injured brain contained OPN, and enhanced neurite outgrowth of cultured neurons in an OPN-dependent manner. Thus, our results introduce the concept that neurons in the injured brain that are destined to die perceive the stressful condition and begin the regeneration processes through induction of OPN, ultimately executing the repair process with the help of monocytes recruited from the circulation.


Asunto(s)
Monocitos , Osteopontina , Encéfalo/metabolismo , Monocitos/metabolismo , Neuronas/metabolismo , Osteopontina/metabolismo , Fagocitosis
4.
J Biomed Sci ; 28(1): 51, 2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34229656

RESUMEN

BACKGROUND: Endoplasmic reticulum (ER) stress is a common feature of Parkinson's disease (PD), and several PD-related genes are responsible for ER dysfunction. Recent studies suggested LRRK2-G2019S, a pathogenic mutation in the PD-associated gene LRRK2, cause ER dysfunction, and could thereby contribute to the development of PD. It remains unclear, however, how mutant LRRK2 influence ER stress to control cellular outcome. In this study, we identified the mechanism by which LRRK2-G2019S accelerates ER stress and cell death in astrocytes. METHODS: To investigate changes in ER stress response genes, we treated LRRK2-wild type and LRRK2-G2019S astrocytes with tunicamycin, an ER stress-inducing agent, and performed gene expression profiling with microarrays. The XBP1 SUMOylation and PIAS1 ubiquitination were performed using immunoprecipitation assay. The effect of astrocyte to neuronal survival were assessed by astrocytes-neuron coculture and slice culture systems. To provide in vivo proof-of-concept of our approach, we measured ER stress response in mouse brain. RESULTS: Microarray gene expression profiling revealed that LRRK2-G2019S decreased signaling through XBP1, a key transcription factor of the ER stress response, while increasing the apoptotic ER stress response typified by PERK signaling. In LRRK2-G2019S astrocytes, the transcriptional activity of XBP1 was decreased by PIAS1-mediated SUMOylation. Intriguingly, LRRK2-GS stabilized PIAS1 by increasing the level of small heterodimer partner (SHP), a negative regulator of PIAS1 degradation, thereby promoting XBP1 SUMOylation. When SHP was depleted, XBP1 SUMOylation and cell death were reduced. In addition, we identified agents that can disrupt SHP-mediated XBP1 SUMOylation and may therefore have therapeutic activity in PD caused by the LRRK2-G2019S mutation. CONCLUSION: Our findings reveal a novel regulatory mechanism involving XBP1 in LRRK2-G2019S mutant astrocytes, and highlight the importance of the SHP/PIAS1/XBP1 axis in PD models. These findings provide important insight into the basis of the correlation between mutant LRRK2 and pathophysiological ER stress in PD, and suggest a plausible model that explains this connection.


Asunto(s)
Astrocitos/metabolismo , Estrés del Retículo Endoplásmico/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteína 1 de Unión a la X-Box/genética , Animales , Modelos Animales de Enfermedad , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Mutación , Enfermedad de Parkinson/fisiopatología , Receptores Citoplasmáticos y Nucleares/metabolismo , Sumoilación , Proteína 1 de Unión a la X-Box/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(7): 1629-1634, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29386384

RESUMEN

Mutations in DJ-1 (PARK7) are a known cause of early-onset autosomal recessive Parkinson's disease (PD). Accumulating evidence indicates that abnormalities of synaptic vesicle trafficking underlie the pathophysiological mechanism of PD. In the present study, we explored whether DJ-1 is involved in CNS synaptic function. DJ-1 deficiency impaired synaptic vesicle endocytosis and reavailability without inducing structural alterations in synapses. Familial mutants of DJ-1 (M26I, E64D, and L166P) were unable to rescue defective endocytosis of synaptic vesicles, whereas WT DJ-1 expression completely restored endocytic function in DJ-1 KO neurons. The defective synaptic endocytosis shown in DJ-1 KO neurons may be attributable to alterations in membrane cholesterol level. Thus, DJ-1 appears essential for synaptic vesicle endocytosis and reavailability, and impairment of this function by familial mutants of DJ-1 may be related to the pathogenesis of PD.


Asunto(s)
Endocitosis/fisiología , Terminaciones Nerviosas/patología , Proteína Desglicasa DJ-1/fisiología , Sinapsis/patología , Vesículas Sinápticas/patología , Animales , Células Cultivadas , Ratones , Ratones Noqueados , Mutación , Terminaciones Nerviosas/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo
6.
Korean J Physiol Pharmacol ; 25(6): 565-574, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34697267

RESUMEN

Astrocytes are activated in response to brain damage. Here, we found that expression of Kir4.1, a major potassium channel in astrocytes, is increased in activated astrocytes in the injured brain together with upregulation of the neural stem cell markers, Sox2 and Nestin. Expression of Kir4.1 was also increased together with that of Nestin and Sox2 in neurospheres formed from dissociated P7 mouse brains. Using the Kir4.1 blocker BaCl2 to determine whether Kir4.1 is involved in acquisition of stemness, we found that inhibition of Kir4.1 activity caused a concentration-dependent increase in sphere size and Sox2 levels, but had little effect on Nestin levels. Moreover, induction of differentiation of cultured neural stem cells by withdrawing epidermal growth factor and fibroblast growth factor from the culture medium caused a sharp initial increase in Kir4.1 expression followed by a decrease, whereas Sox2 and Nestin levels continuously decreased. Inhibition of Kir4.1 had no effect on expression levels of Sox2 or Nestin, or the astrocyte and neuron markers glial fibrillary acidic protein and ß-tubulin III, respectively. Taken together, these results indicate that Kir4.1 may control gain of stemness but not differentiation of stem cells.

7.
Glia ; 68(10): 2086-2101, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32176388

RESUMEN

Monocyte-derived macrophages play a role in the repair of the injured brain. We previously reported that a deficiency of the Parkinson's disease (PD)-associated gene DJ-1 delays repair of brain injury produced by stereotaxic injection of ATP, a component of damage-associated molecular patterns. Here, we show that a DJ-1 deficiency attenuates monocyte infiltration into the damaged brain owing to a decrease in C-C motif chemokine ligand 2 (CCL2) expression in astrocytes. Like DJ-1-knockout (KO) mice, CCL2 receptor (CCR2)-KO mice showed defects in monocyte infiltration and delayed recovery of brain injury, as determined by 9.4 T magnetic resonance imaging analysis and immunostaining for tyrosine hydroxylase and glial fibrillary acid protein. Notably, transcriptome analyses showed that genes related to regeneration and synapse formation were similarly downregulated in injured brains of DJ-1-KO and CCR2-KO mice compared with the injured wild-type brain. These results indicate that defective astrogliosis in DJ-1-KO mice is associated with decreased CCL2 expression and attenuated monocyte infiltration, resulting in delayed repair of brain injury. Thus, delayed repair of brain injury could contribute to the development of PD. MAIN POINTS: A DJ-1 deficiency attenuates infiltration of monocytes owing to a decrease in CCL2 expression in astrocytes, which in turn led to delay in repair of brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Quimiocina CCL2/biosíntesis , Monocitos/metabolismo , Proteína Desglicasa DJ-1/deficiencia , Animales , Astrocitos/patología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Proteína Desglicasa DJ-1/genética
8.
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
9.
Glia ; 66(2): 445-458, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29105838

RESUMEN

Defects in repair of damaged brain accumulate injury and contribute to slow-developing neurodegeneration. Here, we report that a deficiency of DJ-1, a Parkinson's disease (PD) gene, delays repair of brain injury due to destabilization of Sox9, a positive regulator of astrogliosis. Stereotaxic injection of ATP into the brain striatum produces similar size of acute injury in wild-type and DJ-1-knockout (KO) mice. However, recovery of the injury is delayed in KO mice, which is confirmed by 9.4T magnetic resonance imaging and tyrosine hydroxylase immunostaining. DJ-1 regulates neurite outgrowth from damaged neurons in a non-cell autonomous manner. In DJ-1 KO brains and astrocytes, Sox9 protein levels are decreased due to enhanced ubiquitination, resulting in defects in astrogliosis and glial cell-derived neurotrophic factor/ brain-derived neurotrophic factor expression in injured brain and astrocytes. These results indicate that DJ-1 deficiency causes defects in astrocyte-mediated repair of brain damage, which may contribute to the development of PD.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Gliosis/metabolismo , Proteína Desglicasa DJ-1/deficiencia , Factor de Transcripción SOX9/metabolismo , Animales , Astrocitos/patología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Gliosis/genética , Gliosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteína Desglicasa DJ-1/genética , Estabilidad Proteica , Factor de Transcripción SOX9/genética
10.
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
11.
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.
Mol Cell ; 35(6): 806-17, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19782030

RESUMEN

To unravel the roles of LXRs in inflammation and immunity, we examined the function of LXRs in development of IFN-gamma-mediated inflammation using cultured rat brain astrocytes. LXR ligands inhibit neither STAT1 phosphorylation nor STAT1 translocation to the nucleus but, rather, inhibit STAT1 binding to promoters and the expression of IRF1, TNFalpha, and IL-6, downstream effectors of STAT1 action. Immunoprecipitation data revealed that LXRbeta formed a trimer with PIAS1-pSTAT1, whereas LXRalpha formed a trimer with HDAC4-pSTAT1, mediated by direct ligand binding to the LXR proteins. In line with the fact that both PIAS1 and HDAC4 belong to the SUMO E3 ligase family, LXRbeta and LXRalpha were SUMO-conjugated by PIAS1 or HDAC4, respectively, and SUMOylation was blocked by transient transfection of appropriate individual siRNAs, reversing LXR-induced suppression of IRF1 and TNFalpha expression. Together, our data show that SUMOylation is required for the suppression of STAT1-dependent inflammatory responses by LXRs in IFN-gamma-stimulated brain astrocytes.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Interferón gamma/metabolismo , Procesamiento Proteico-Postraduccional , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Animales Recién Nacidos , Sitios de Unión , Células Cultivadas , Proteínas de Unión al ADN/genética , Histona Desacetilasas/metabolismo , Inflamación/genética , Inflamación/prevención & control , Factor 1 Regulador del Interferón/metabolismo , Interleucina-6/metabolismo , Ligandos , Receptores X del Hígado , Receptores Nucleares Huérfanos , Fosforilación , Regiones Promotoras Genéticas , Proteínas Inhibidoras de STAT Activados/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Factor de Transcripción STAT1/genética , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
14.
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
15.
Mol Pharmacol ; 88(1): 161-70, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25715797

RESUMEN

The timely resolution of inflammation prevents continued tissue damage after an initial insult. In the brain, the death of activated microglia by apoptosis has been proposed as one mechanism to resolve brain inflammation. How microglial death is regulated after activation is still unclear. We reported that exposure to lipopolysaccharide (LPS) and interleukin (IL)-13 together initially activates and then kills rat microglia in culture by a mechanism dependent on cyclooxygenase-2 (COX-2). We show here that activation of the E prostanoid receptor 2 (EP2, PTGER2) for prostaglandin E2 mediates microglial death induced by LPS/IL-13, and that EP2 activation by agonist alone kills microglia. Both EP2 antagonists and reactive oxygen scavengers block microglial death induced by either LPS/IL-13 or EP2 activation. By contrast, the homeostatic induction of heme oxygenase 1 (Hmox1) by LPS/IL-13 or EP2 activation protects microglia. Both the Hmox1 inducer cobalt protoporphyrin and a compound that releases the Hmox1 product carbon monoxide (CO) attenuated microglial death produced by LPS/IL-13. Whereas CO reduced COX-2 protein expression, EP2 activation increased Hmox1 and COX-2 expression at both the mRNA and protein level. Interestingly, caspase-1 inhibition prevented microglial death induced by either LPS/IL-13 or low (but not high) concentrations of butaprost, suggestive of a predominantly pyroptotic mode of death. Butaprost also caused the expression of activated caspase-3 in microglia, pointing to apoptosis. These results indicate that EP2 activation, which initially promotes microglial activation, later causes delayed death of activated microglia, potentially contributing to the resolution phase of neuroinflammation.


Asunto(s)
Apoptosis , Microglía/patología , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Estado Epiléptico/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Interleucina-13/inmunología , Lipopolisacáridos/inmunología , Ratones , Microglía/metabolismo , Pilocarpina , Embarazo , Ratas , Ratas Sprague-Dawley , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/inducido químicamente
16.
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
17.
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
18.
J Immunol ; 188(10): 5132-41, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22504638

RESUMEN

Emerging evidence has established that astrocytes, once considered passive supporting cells that maintained extracellular ion levels and served as a component of the blood-brain barrier, play active regulatory roles during neurogenesis and in brain pathology. In the current study, we demonstrated that astrocytes sense H(2)O(2) by rapidly phosphorylating the transcription factor STAT6, a response not observed in microglia. STAT6 phosphorylation was induced by generators of other reactive oxygen species (ROS) and reactive nitrogen species, as well as in the reoxygenation phase of hypoxia/reoxygenation, during which ROS are generated. Src-JAK pathways mediated STAT6 phosphorylation upstream. Experiments using lipid raft disruptors and analyses of detergent-fractionated cells demonstrated that H(2)O(2)-induced STAT6 phosphorylation occurred in lipid rafts. Under experimental conditions in which H(2)O(2) did not affect astrocyte viability, H(2)O(2)-induced STAT6 phosphorylation resulted in STAT6-dependent cyclooxygenase-2 expression and subsequent release of PGE(2) and prostacyclin, an effect also observed in hypoxia/reoxygenation. Finally, PGs released from H(2)O(2)-stimulated astrocytes inhibited microglial TNF-α expression. Accordingly, our results indicate that ROS-induced STAT6 phosphorylation in astrocytes can modulate the functions of neighboring cells, including microglia, through cyclooxygenase-2 induction and subsequent release of PGs. Differences in the sensitivity of STAT6 in astrocytes (highly sensitive) and microglia (insensitive) to phosphorylation following brief exposure to H(2)O(2) suggest that astrocytes can act as sentinels for certain stimuli, including H(2)O(2) and ROS, refining the canonical notion that microglia are the first line of defense against external stimuli.


Asunto(s)
Astrocitos/metabolismo , Ciclooxigenasa 2/biosíntesis , Peróxido de Hidrógeno/metabolismo , Microglía , Prostaglandinas/metabolismo , Factor de Transcripción STAT6/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/fisiología , Masculino , Microglía/enzimología , Microglía/inmunología , Microglía/metabolismo , Fosforilación/inmunología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA