Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Glia ; 69(9): 2133-2145, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33956370

RESUMEN

Our recent finding has demonstrated that astrocytes confer neuroprotection by endogenously producing ciliary neurotrophic factor (CNTF) via transient receptor potential vanilloid 1 (TRPV1) in Parkinson's disease (PD). In this study, the possible molecular target for TRPV1-mediated CNTF production and its neuroprotective effects on dopamine neurons were further investigated. For comparison, glial cell-line derived neurotrophic factor (GDNF) was also examined. The results show that TRPV1-ribosomal protein 70 S6 kinase (p70S6K) signaling on astrocytes produces endogenous CNTF in the SN of MPP+ -lesioned rat. By marked contrast, the expression of GDNF on astrocytes is independent of TRPV1-p70S6K signaling. Administration of a TRPV1 agonist, capsaicin, increases levels of phosphorylated p70S6K (p-p70S6K; activation of p70S6K) on astrocytes, resulting in the survival of dopamine neurons and behavioral recovery through endogenous production of CNTF in the MPP+ -lesioned rat model of PD. Immunohistochemical analysis reveals expression of p-p70S6K on astrocytes in the SN of PD patients, indicating relevance to human PD. The present in vivo data is the first to demonstrate that astrocytic TRPV1-p70S6K signaling plays a pivotal role as endogenous neuroprotective, and it may constitute a novel therapeutic target for treating PD.


Asunto(s)
Neuronas Dopaminérgicas , Fármacos Neuroprotectores , 1-Metil-4-fenilpiridinio/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Astrocitos/metabolismo , Neuronas Dopaminérgicas/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Fármacos Neuroprotectores/farmacología , Ratas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/farmacología , Sustancia Negra/metabolismo
2.
J Cell Biochem ; 119(1): 300-313, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569433

RESUMEN

Human glioblastomas express higher levels of matrix metalloprotease-2 (MMP-2) than low-grade brain tumors and normal brain tissues. Ascochlorin (ASC) has anti-metastatic, anti-angiogenic, and synergistic effect in various types of cancer cells. However, it remains unknown whether ASC can affect cell migration and invasion in malignant human glioma cells. In this study, we found that ASC indeed inhibits cell migration and invasion in U373MG and A172. ASC significantly suppresses the MMP-2 gelatinolytic activity and expression in U373MG and A172. To determine the molecular mechanism by which ASC suppressed cell migration and invasion, we investigated whether ASC could modulate metastasis via focal adhesion kinase (FAK) and janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, a potential drug target. ASC strongly inhibits the phosphorylation of FAK, and treatment with a FAK inhibitor significantly suppresses cancer cell migration in the presence of ASC. In addition, ASC significantly decreased phosphorylation of JAK2/STAT3, cancer cell migration and nuclear translocation of STAT3. Taken together, these results suggest that ASC inhibits cell migration and invasion by blocking FAK and JAK/STAT signaling, resulting in reduced MMP-2 activity. J. Cell. Biochem. 119: 300-313, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Alquenos/farmacología , Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Glioblastoma/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Fenoles/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Invasividad Neoplásica
3.
J Neuroinflammation ; 15(1): 271, 2018 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-30231870

RESUMEN

BACKGROUND: The FDA-approved small-molecule drug ibrutinib is an effective targeted therapy for patients with chronic lymphocytic leukemia (CLL). Ibrutinib inhibits Bruton's tyrosine kinase (BTK), a kinase involved in B cell receptor signaling. However, the potential regulation of neuroinflammatory responses in the brain by ibrutinib has not been comprehensively examined. METHODS: BV2 microglial cells were treated with ibrutinib (1 µM) or vehicle (1% DMSO), followed by lipopolysaccharide (LPS; 1 µg/ml) or PBS. RT-PCR, immunocytochemistry, and subcellular fractionation were performed to examine the effects of ibrutinib on neuroinflammatory responses. In addition, wild-type mice were sequentially injected with ibrutinib (10 mg/kg, i.p.) or vehicle (10% DMSO, i.p.), followed by LPS (10 mg/kg, i.p.) or PBS, and microglial and astrocyte activations were assessed using immunohistochemistry. RESULTS: Ibrutinib significantly reduced LPS-induced increases in proinflammatory cytokine levels in BV2 microglial and primary microglial cells but not in primary astrocytes. Ibrutinib regulated TLR4 signaling to alter LPS-induced proinflammatory cytokine levels. In addition, ibrutinib significantly decreased LPS-induced increases in p-AKT and p-STAT3 levels, suggesting that ibrutinib attenuates LPS-induced neuroinflammatory responses by inhibiting AKT/STAT3 signaling pathways. Interestingly, ibrutinib also reduced LPS-induced BV2 microglial cell migration by inhibiting AKT signaling. Moreover, ibrutinib-injected wild-type mice exhibited significantly reduced microglial/astrocyte activation and COX-2 and IL-1ß proinflammatory cytokine levels. CONCLUSIONS: Our data provide insights on the mechanisms of a potential therapeutic strategy for neuroinflammation-related diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Adenina/análogos & derivados , Animales , Animales Recién Nacidos , Línea Celular Transformada , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Compuestos Heterocíclicos con 3 Anillos/farmacología , Inflamación/inducido químicamente , Lipopolisacáridos/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Piperidinas , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Pirimidinas/química , Ratas , Transducción de Señal/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
4.
J Neuroinflammation ; 15(1): 286, 2018 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-30309372

RESUMEN

BACKGROUND: Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer's disease (AD). Thus, modulating the neuroinflammatory response represents a potential therapeutic strategy for treating neurodegenerative diseases. Several recent studies have shown that dopamine (DA) and its receptors are expressed in immune cells and are involved in the neuroinflammatory response. Thus, we recently developed and synthesized a non-self-polymerizing analog of DA (CA140) and examined the effect of CA140 on neuroinflammation. METHODS: To determine the effects of CA140 on the neuroinflammatory response, BV2 microglial cells were pretreated with lipopolysaccharide (LPS, 1 µg/mL), followed by treatment with CA140 (10 µM) and analysis by reverse transcription-polymerase chain reaction (RT-PCR). To examine whether CA140 alters the neuroinflammatory response in vivo, wild-type mice were injected with both LPS (10 mg/kg, intraperitoneally (i.p.)) and CA140 (30 mg/kg, i.p.), and immunohistochemistry was performed. In addition, familial AD (5xFAD) mice were injected with CA140 or vehicle daily for 2 weeks and examined for microglial and astrocyte activation. RESULTS: Pre- or post-treatment with CA140 differentially regulated proinflammatory responses in LPS-stimulated microglia and astrocytes. Interestingly, CA140 regulated D1R levels to alter LPS-induced proinflammatory responses. CA140 significantly downregulated LPS-induced phosphorylation of ERK and STAT3 in BV2 microglia cells. In addition, CA140-injected wild-type mice exhibited significantly decreased LPS-induced microglial and astrocyte activation. Moreover, CA140-injected 5xFAD mice exhibited significantly reduced microglial and astrocyte activation. CONCLUSIONS: CA140 may be beneficial for preventing and treating neuroinflammatory-related diseases, including AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Antiinflamatorios/uso terapéutico , Dopamina/análogos & derivados , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Dopamina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Polisacáridos/farmacología , Presenilina-1/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Int J Mol Sci ; 19(11)2018 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-30423807

RESUMEN

We demonstrated that capsaicin (CAP), an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), inhibits microglia activation and microglia-derived oxidative stress in the substantia nigra (SN) of MPP⁺-lesioned rat. However, the detailed mechanisms how microglia-derived oxidative stress is regulated by CAP remain to be determined. Here we report that ciliary neurotrophic factor (CNTF) endogenously produced by CAP-activated astrocytes through TRPV1, but not microglia, inhibits microglial activation and microglia-derived oxidative stress, as assessed by OX-6 and OX-42 immunostaining and hydroethidine staining, respectively, resulting in neuroprotection. The significant increase in levels of CNTF receptor alpha (CNTFRα) expression was evident on microglia in the MPP⁺-lesioned rat SN and the observed beneficial effects of CNTF was abolished by treatment with CNTF receptor neutralizing antibody. It is therefore likely that CNTF can exert its effect via CNTFRα on microglia, which rescues dopamine neurons in the SN of MPP⁺-lesioned rats and ameliorates amphetamine-induced rotations. Immunohistochemical analysis revealed also a significantly increased expression of CNTFRα on microglia in the SN from human Parkinson's disease patients compared with age-matched controls, indicating that these findings may have relevance to the disease. These data suggest that CNTF originated from TRPV1 activated astrocytes may be beneficial to treat neurodegenerative disease associated with neuro-inflammation such as Parkinson's disease.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Factor Neurotrófico Ciliar/farmacología , Neuronas Dopaminérgicas/patología , Microglía/patología , Neuroprotección/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo , Anciano , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Capsaicina/farmacología , Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Biológicos , Degeneración Nerviosa/patología , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptor de Factor Neurotrófico Ciliar/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Canales Catiónicos TRPV/metabolismo
6.
J Cell Biochem ; 118(12): 4672-4685, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28498494

RESUMEN

Histone deacetylase inhibitors (HDACIs) have emerged as potential anticancer agents for the treatment of solid and hematopoietic cancers. Several HDACIs delay cell growth, induce differentiation, or activate apoptosis in multiple types of tumors, including glioblastomas. In the present study, we showed that the mercaptoacetamide-based HDACI W2 inhibits cell migration and invasion in monomorphic malignant human glioma cells. W2 treatment significantly decreased the activity and expression levels of matrix metalloprotease-2 in malignant A172 cells but not in U373MG cells. Key signaling pathways involved in cell migration and invasion, including PI3K-AKT, ERK-JNK-P38, and FAK/STAT3, were examined to identify the mechanism of action of W2. W2 increased the phosphorylation of AKT and altered cell migration and invasion in an AKT-independent manner. W2 inhibited the phosphorylation of FAK/STAT3, and treatment with a FAK/STAT3 inhibitor significantly suppressed cancer cell migration and MMP-2 activity in the presence of W2. In addition, W2 significantly inhibited the nuclear translocation of phospho-STAT3. Taken together, our results suggest that W2 suppresses cancer cell migration and invasion by inhibiting FAK/STAT3 signaling and STAT3 translocation to the nucleus in monomorphic malignant human glioma cells. J. Cell. Biochem. 118: 4672-4685, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Tioacetamida/análogos & derivados , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/genética , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Factor de Transcripción STAT3/genética , Tioacetamida/farmacología
7.
Mol Ther ; 23(3): 445-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25502903

RESUMEN

Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Región CA1 Hipocampal/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Neuronas/metabolismo , Neuropéptidos/genética , Transducción Genética/métodos , Animales , Anticuerpos Neutralizantes/farmacología , Factor Neurotrófico Derivado del Encéfalo/agonistas , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/genética , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Dependovirus/genética , Dependovirus/metabolismo , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/agonistas , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Trombina/antagonistas & inhibidores , Trombina/toxicidad
8.
Free Radic Biol Med ; 160: 575-595, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-32896600

RESUMEN

Regulating amyloid beta (Aß) pathology and neuroinflammatory responses holds promise for the treatment of Alzheimer's disease (AD) and other neurodegenerative and/or neuroinflammation-related diseases. In this study, the effects of KVN93, an inhibitor of dual-specificity tyrosine phosphorylation-regulated kinase-1A (DYRK1A), on cognitive function and Aß plaque levels and the underlying mechanism of action were evaluated in 5x FAD mice (a mouse model of AD). KVN93 treatment significantly improved long-term memory by enhancing dendritic synaptic function. In addition, KVN93 significantly reduced Aß plaque levels in 5x FAD mice by regulating levels of the Aß degradation enzymes neprilysin (NEP) and insulin-degrading enzyme (IDE). Moreover, Aß-induced microglial and astrocyte activation were significantly suppressed in the KVN-treated 5xFAD mice. KVN93 altered neuroinflammation induced by LPS in microglial cells but not primary astrocytes by regulating TLR4/AKT/STAT3 signaling, and in wild-type mice injected with LPS, KVN93 treatment reduced microglial and astrocyte activation. Overall, these results suggest that the novel DYRK1A inhibitor KVN93 is a potential therapeutic drug for regulating cognitive/synaptic function, Aß plaque load, and neuroinflammatory responses in the brain.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Cognición , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Microglía , Placa Amiloide/tratamiento farmacológico , Quinasas DyrK
9.
Exp Neurobiol ; 28(2): 289-299, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31138996

RESUMEN

Transient receptor potential vanilloid subtype 1 (TRPV1) on astrocytes prevents ongoing degeneration of nigrostriatal dopamine (DA) neurons in MPP+-lesioned rats via ciliary neurotrophic factor (CNTF). The present study determined whether such a beneficial effect of astrocytic TRPV1 could be achieved after completion of injury of DA neurons, rather than ongoing injury, which seems more relevant to therapeutics. To test this, the MPP+-lesioned rat model utilized here exhibited approximately 70~80% degeneration of nigrostriatal DA neurons that was completed at 2 weeks post medial forebrain bundle injection of MPP+. TRPV1 agonist, capsaicin (CAP), was intraperitoneally administered. CNTF receptor alpha neutralizing antibody (CNTFRαNAb) was nigral injected to evaluate the role of CNTF endogenously produced by astrocyte through TRPV1 activation on DA neurons. Delayed treatment of CAP produced a significant reduction in amphetamine-induced rotational asymmetry. Accompanying this behavioral recovery, CAP treatment increased CNTF levels and tyrosine hydroxylase (TH) activity in the substantia nigra pars compacta (SNpc), and levels of DA and its metabolites in the striatum compared to controls. Interestingly, behavioral recovery and increases in biochemical indices were not reflected in trophic changes of the DA system. Instead, behavioral recovery was temporal and dependent on the continuous presence of CAP treatment. The results suggest that delayed treatment of CAP increases nigral TH enzyme activity and striatal levels of DA and its metabolites by CNTF endogenously derived from CAP-activated astrocytes through TRPV1, leading to functional recovery. Consequently, these findings may be useful in the treatment of DA imbalances associated with Parkinson's disease.

10.
Front Aging Neurosci ; 10: 269, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319390

RESUMEN

Recent studies have shown that Liuwei Dihuang pills (LWPs) can positively affect learning, memory and neurogenesis. However, the underlying molecular mechanisms are not understood. In the present study, we developed ALWPs, a mixture of Antler and LWPs, and investigated whether ALWPs can affect neuroinflammatory responses. We found that ALWPs (500 mg/ml) inhibited lipopolysaccharide (LPS)-induced proinflammatory cytokine IL-1ß mRNA levels in BV2 microglial cells but not primary astrocytes. ALWPs significantly reduced LPS-induced cell-surface levels of TLR4 to alter neuroinflammation. An examination of the molecular mechanisms by which ALWPs regulate the LPS-induced proinflammatory response revealed that ALWPs significantly downregulated LPS-induced levels of FAK phosphorylation, suggesting that ALWPs modulate FAK signaling to alter LPS-induced IL-1ß levels. In addition, treatment with ALWPs followed by LPS resulted in decreased levels of the transcription factor NF-κB in the nucleus compared with LPS alone. Moreover, ALWPs significantly suppressed LPS-induced BV2 microglial cell migration. To examine whether ALWPs modulate learning and memory in vivo, wild-type C57BL/6J mice were orally administered ALWPs (200 mg/kg) or PBS daily for 3 days, intraperitoneally injected (i.p.) with LPS (250 µg/kg) or PBS, and assessed in Y maze and NOR tests. We observed that oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly rescued short- and long-term memory. More importantly, oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly reduced microglial activation in the hippocampus and cortex. Taken together, our results suggest that ALWPs can suppress neuroinflammation-associated cognitive deficits and that ALWPs have potential as a drug for neuroinflammation/neurodegeneration-related diseases, including Alzheimer's disease (AD).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA