Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 119: 431-453, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636566

RESUMEN

Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.


Asunto(s)
Transportador 1 de Casete de Unión a ATP , Macrófagos , Traumatismos de la Médula Espinal , Animales , Transportador 1 de Casete de Unión a ATP/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Ratones , Masculino , Macrófagos/metabolismo , Células Espumosas/metabolismo , Ratones Endogámicos C57BL , Médula Espinal/metabolismo , Ratones Noqueados , Modelos Animales de Enfermedad
2.
iScience ; 27(2): 108895, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38318363

RESUMEN

Spinal cord injury (SCI) often leads to neuronal loss, axonal degeneration, and behavioral dysfunction. We recently show that in vivo reprogramming of NG2 glia produces new neurons, reduces glial scaring, and ultimately leads to improved function after SCI. By examining endogenous neurons, we here unexpectedly uncover that NG2 glia reprogramming also induces robust axonal regeneration of the corticospinal tract and serotonergic neurons. Such reprogramming-induced axonal regeneration may contribute to the reconstruction of neural networks essential for behavioral recovery.

3.
Cell Death Dis ; 15(1): 4, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38177100

RESUMEN

Effective therapeutics is much needed for amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease mainly affecting motor neurons. By screening chemical compounds in human patient-derived and aging-relevant motor neurons, we identify a neuroprotective compound and show that MAP4Ks may serve as therapeutic targets for treating ALS. The lead compound broadly improves survival and function of motor neurons directly converted from human ALS patients. Mechanistically, it works as an inhibitor of MAP4Ks, regulates the MAP4Ks-HDAC6-TUBA4A-RANGAP1 pathway, and normalizes subcellular distribution of RANGAP1 and TDP-43. Finally, in an ALS mouse model we show that inhibiting MAP4Ks preserves motor neurons and significantly extends animal lifespan.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Ratones , Animales , Adulto , Humanos , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas Motoras/metabolismo , Envejecimiento , Modelos Animales de Enfermedad , Ratones Transgénicos
4.
bioRxiv ; 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37398355

RESUMEN

Spinal cord injury (SCI) often leads to neuronal loss, axonal degeneration and behavioral dysfunction. We recently show that in vivo reprogramming of NG2 glia produces new neurons, reduces glial scaring, and ultimately leads to improved function after SCI. By examining endogenous neurons, we here unexpectedly uncover that NG2 glia reprogramming also induces robust axonal regeneration of the corticospinal tract and serotonergic neurons. Such reprogramming-induced axonal regeneration may contribute to the reconstruction of neural networks essential for behavioral recovery.

5.
Curr Opin Genet Dev ; 82: 102090, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37506560

RESUMEN

Spinal cord injury (SCI) can lead to the loss of motor, sensory, or autonomic function due to neuronal death. Unfortunately, the adult mammalian spinal cord has limited intrinsic regenerative capacity, making it difficult to rebuild the neural circuits necessary for functional recovery. However, recent evidence suggests that in vivo fate reprogramming of resident cells that are normally non-neurogenic can generate new neurons. This process also improves the pathological microenvironment, and the new neurons can integrate into the local neural network, resulting in better functional outcomes in SCI animal models. In this concise review, we focus on recent advances while also discussing the challenges, pitfalls, and opportunities in the field of in vivo cell fate reprogramming for spinal cord repair.


Asunto(s)
Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Animales , Neuronas/metabolismo , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/terapia , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Mamíferos
6.
bioRxiv ; 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37162962

RESUMEN

Effective therapeutics is much needed for amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease mainly affecting motor neurons. By screening chemical compounds in human patient-derived and aging-relevant motor neurons, we identify a neuroprotective compound and show that MAP4Ks may serve as therapeutic targets for treating ALS. The lead compound broadly improves survival and function of motor neurons directly converted from human ALS patients. Mechanistically, it works as an inhibitor of MAP4Ks, regulates the MAP4Ks-HDAC6-TUBA4A-RANGAP1 pathway, and normalizes subcellular distribution of RANGAP1 and TDP-43. Finally, in an ALS mouse model we show that inhibiting MAP4Ks preserves motor neurons and significantly extends animal lifespan.

7.
Glia ; 70(2): 273-286, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34643969

RESUMEN

In response to brain injury, resident astrocytes become reactive and play dynamic roles in neural repair and regeneration. The signaling pathways underlying such reactive astrogliosis remain largely unclear. We here show that NEK6, a NIMA-related serine/threonine protein kinase, is rapidly induced following pathological stimulations and plays critical roles in reactive astrogliosis. Enhanced NEK6 expression promotes reactive astrogliosis and exacerbates brain lesions; and conversely, NEK6 downregulation dampens injury-induced astrocyte reactivity and reduces lesion size. Mechanistically, NEK6 associates with and phosphorylates STAT3. Kinase activity of NEK6 is required for induction of GFAP and PCNA, markers of reactive astrogliosis. Interestingly, NEK6 is also localized in the nucleus and binds to STAT3-responsive genomic elements in astrocytes. These results indicate that NEK6 constitutes a molecular target for the regulation of reactive astrogliosis.


Asunto(s)
Astrocitos , Gliosis , Quinasas Relacionadas con NIMA , Factor de Transcripción STAT3 , Astrocitos/metabolismo , Lesiones Encefálicas , Proteína Ácida Fibrilar de la Glía , Gliosis/patología , Humanos , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Fosforilación , Antígeno Nuclear de Célula en Proliferación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
8.
Cell Stem Cell ; 28(5): 923-937.e4, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675690

RESUMEN

Adult neurogenesis plays critical roles in maintaining brain homeostasis and responding to neurogenic insults. However, the adult mammalian spinal cord lacks an intrinsic capacity for neurogenesis. Here we show that spinal cord injury (SCI) unveils a latent neurogenic potential of NG2+ glial cells, which can be exploited to produce new neurons and promote functional recovery after SCI. Although endogenous SOX2 is required for SCI-induced transient reprogramming, ectopic SOX2 expression is necessary and sufficient to unleash the full neurogenic potential of NG2 glia. Ectopic SOX2-induced neurogenesis proceeds through an expandable ASCL1+ progenitor stage and generates excitatory and inhibitory propriospinal neurons, which make synaptic connections with ascending and descending spinal pathways. Importantly, SOX2-mediated reprogramming of NG2 glia reduces glial scarring and promotes functional recovery after SCI. These results reveal a latent neurogenic potential of somatic glial cells, which can be leveraged for regenerative medicine.


Asunto(s)
Neuroglía , Traumatismos de la Médula Espinal , Animales , Neurogénesis , Recuperación de la Función , Médula Espinal
9.
Front Cell Neurosci ; 14: 107, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32390804

RESUMEN

The adult mammalian central nervous system (CNS) has very limited regenerative capacity upon neural injuries or under degenerative conditions. In recent years, however, significant progress has been made on in vivo cell fate reprogramming for neural regeneration. Resident glial cells can be reprogrammed into neuronal progenitors and mature neurons in the CNS of adult mammals. In this review article, we briefly summarize the current knowledge on innate adult neurogenesis under pathological conditions and then focus on induced neurogenesis through cell fate reprogramming. We discuss how the reprogramming process can be regulated and raise critical issues requiring careful considerations to move the field forward. With emerging evidence, we envision that fate reprogramming-based regenerative medicine will have a great potential for treating neurological conditions such as brain injury, spinal cord injury (SCI), Alzheimer's disease (AD), Parkinson's disease (PD), and retinopathy.

10.
Cereb Cortex ; 29(1): 54-69, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29161339

RESUMEN

Injury to the adult brain induces activation of local astrocytes, which serves as a compensatory response that modulates tissue damage and recovery. However, the mechanism governing astrocyte activation during brain injury remains largely unknown. Here we provide in vivo evidence that SOX2, a transcription factor critical for stem cells and brain development, is also required for injury-induced activation of adult cortical astrocytes. Genome-wide chromatin immunoprecipitation-seq analysis of mouse cortical tissues reveals that SOX2 binds to regulatory regions of genes associated with signaling pathways that control glial cell activation, such as Nr2e1, Mmd2, Wnt7a, and Akt2. Astrocyte-specific deletion of Sox2 in adult mice greatly diminishes glial response to controlled cortical impact injury and, most unexpectedly, dampens injury-induced cortical loss and benefits behavioral recovery of mice after injury. Together, these results uncover an essential role of SOX2 in somatic cells under pathological conditions and indicate that SOX2-dependent astrocyte activation could be targeted for functional recovery after traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Eliminación de Gen , Recuperación de la Función/fisiología , Factores de Transcripción SOXB1/deficiencia , Animales , Astrocitos/patología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Células Cultivadas , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales , Factores de Transcripción SOXB1/genética
11.
Cell Transplant ; 26(3): 469-482, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-27737726

RESUMEN

Spinal cord injury (SCI) causes functional impairment as a result of the initial injury followed by secondary injury mechanism. SCI provokes an inflammatory response that causes secondary tissue damage and neurodegeneration. While the use of neural stem cell (NSC) engraftment to mitigate secondary injury has been of interest to many researchers, it still faces several limitations. As such, we investigated if NSC-conditioned medium (NSC-M) possesses therapeutic potential for the treatment of SCI. It has been proposed that many of the beneficial effects attributed to stem cell therapies are due to secreted factors. Utilizing primary cell culture and murine models of SCI, we determined that systemic treatment with NSC-M was able to significantly improve motor function and lesion healing. In addition, NSC-M demonstrated significant anti-inflammatory potential in vitro and in vivo, reducing inflammatory cytokine expression in both activated macrophages and injured spinal cord tissues. NSC-M was also able to reduce the expression of inducible nitric oxide synthase (iNOS) within the spleen of injured animals, indicating an ability to reduce systemic inflammation. Thus, we believe that NSC-M offers a possible alternative to direct stem cell engraftment for the treatment of SCI.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Inflamación/tratamiento farmacológico , Células-Madre Neurales/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fagocitosis/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Traumatismos de la Médula Espinal/metabolismo
12.
Cell Rep ; 17(3): 891-903, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27732862

RESUMEN

Although the adult mammalian spinal cord lacks intrinsic neurogenic capacity, glial cells can be reprogrammed in vivo to generate neurons after spinal cord injury (SCI). How this reprogramming process is molecularly regulated, however, is not clear. Through a series of in vivo screens, we show here that the p53-dependent pathway constitutes a critical checkpoint for SOX2-mediated reprogramming of resident glial cells in the adult mouse spinal cord. While it has no effect on the reprogramming efficiency, the p53 pathway promotes cell-cycle exit of SOX2-induced adult neuroblasts (iANBs). As such, silencing of either p53 or p21 markedly boosts the overall production of iANBs. A neurotrophic milieu supported by BDNF and NOG can robustly enhance maturation of these iANBs into diverse but predominantly glutamatergic neurons. Together, these findings have uncovered critical molecular and cellular checkpoints that may be manipulated to boost neuron regeneration after SCI.


Asunto(s)
Envejecimiento/metabolismo , Reprogramación Celular , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Médula Espinal/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Microambiente Celular , Ratones Endogámicos C57BL , Neuronas/metabolismo
13.
Mol Brain ; 9(1): 48, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27153974

RESUMEN

BACKGROUND: Macrophages play an important role in the inflammatory responses involved with spinal cord injury (SCI). We have previously demonstrated that infiltrated bone marrow-derived macrophages (BMDMs) engulf myelin debris, forming myelin-laden macrophages (mye-Mϕ). These mye-Mϕ promote disease progression through their pro-inflammatory phenotype, enhanced neurotoxicity, and impaired phagocytic capacity for apoptotic cells. We thus hypothesize that the excessive accumulation of mye-Mϕ is the root of secondary injury, and that targeting mye-Mϕ represents an efficient strategy to improve the local inflammatory microenvironment in injured spinal cords and to further motor neuron function recovery. In this study, we administer murine embryonic stem cell conditioned media (ESC-M) as a cell-free stem cell based therapy to treat a mouse model of SCI. RESULTS: We showed that BMDMs, but not microglial cells, engulf myelin debris generated at the injury site. Phagocytosis of myelin debris leads to the formation of mye-Mϕ in the injured spinal cord, which are surrounded by activated microglia cells. These mye-Mϕ are pro-inflammatory and lose the normal macrophage phagocytic capacity for apoptotic cells. We therefore focus on how to trigger lipid efflux from mye-Mϕ and thus restore their function. Using ESC-M as an immune modulating treatment for inflammatory damage after SCI, we rescued mye-Mϕ function and improved functional locomotor recovery. ESC-M treatment on mye-Mϕ resulted in improved exocytosis of internalized lipids and a normal capacity for apoptotic cell phagocytosis. Furthermore, when ESC-M was administered intraperitoneally after SCI, animals exhibited significant improvements in locomotor recovery. Examination of spinal cords of the ESC-M treated mice revealed similar improvements in macrophage function as well as a shift towards a more anti-inflammatory environment at the lesion and parenchyma. CONCLUSIONS: The embryonic stem cell conditioned media can be used as an effective treatment for SCI to resolve inflammation and improve functional recovery while circumventing the complications involved in whole cell transplantation.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Macrófagos/patología , Células Madre Embrionarias de Ratones/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Inflamación/complicaciones , Inflamación/patología , Lípidos/química , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/patología
14.
Front Immunol ; 5: 275, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25071759

RESUMEN

Stem cell therapies have had tremendous potential application for many diseases in recent years. However, the tumorigenic properties of stem cells restrict their potential clinical application; therefore, strategies for reducing the tumorigenic potential of stem cells must be established prior to transplantation. We have demonstrated that syngeneic transplantation of embryonic stem cells (ESCs) provokes an inflammatory response that involves the rapid recruitment of bone marrow-derived macrophages (BMDMs). ESCs are able to prevent mature macrophages from macrophage colony-stimulating factor (M-CSF) withdrawal-induced apoptosis, and thus prolong macrophage lifespan significantly by blocking various apoptotic pathways in an M-CSF-independent manner. ESCs express and secrete IL-34, which may be responsible for ESC-promoted macrophage survival. This anti-apoptotic effect of ESCs involves activation of extracellular signal-regulated kinase (ERK)1/2 and PI3K/Akt pathways and thus, inhibition of ERK1/2 and PI3K/AKT activation decreases ESC-induced macrophage survival. Functionally, ESC-treated macrophages also showed a higher level of phagocytic activity. ESCs further serve to polarize BMDMs into M2-like macrophages that exhibit most tumor-associated macrophage phenotypic and functional features. ESC-educated macrophages produce high levels of arginase-1, Tie-2, and TNF-α, which participate in angiogenesis and contribute to teratoma progression. Our study suggests that induction of M2-like macrophage activation is an important mechanism for teratoma development. Strategies targeting macrophages to inhibit teratoma development would increase the safety of ESC-based therapies, inasmuch as the depletion of macrophages completely inhibits ESC-induced angiogenesis and teratoma development.

15.
Naunyn Schmiedebergs Arch Pharmacol ; 387(1): 75-85, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24071813

RESUMEN

Amyloid precursor protein (APP) metabolism is a key factor in the pathogenesis of Alzheimer's disease (AD). Amyloid-beta (Aß) in mitochondria comes from APP mitochondrial metabolism or from the uptake Aß from outside of mitochondria. It has been recently proposed that mitochondria are involved in the biochemical pathways through which Aß causes neuronal dysfunction. The accumulated Aß in mitochondria decreases the level of cytochrome c oxidase (COX IV) and attenuates the ATP production consequently. FLZ is a synthetic cyclic derivative of squamosamide from Annona glabra. In this study, the effect of FLZ on APP processing in mitochondria was investigated in SH-SY5Y cells over-expressing APP695 (wt/Swe). FLZ treatment attenuated APP processing and decreased Aß production in mitochondria. The mitochondrial function was increased with the upregulation of COX IV both at protein and activity levels. ATP production was also increased after FLZ treatment. The mechanistic study showed that FLZ inhibited γ-secretase activity by decreasing C-terminal fragment protein level of presenilin, the active center of γ-secretase. The effect of FLZ differs from DAPT (a non-selective γ-secretase inhibitor), suggesting FLZ is a selective γ-secretase inhibitor. FLZ selectively inhibited γ-secretase in the cleavage of recombinant C terminus of APP in vitro, without specifically modulating the processing of recombinant Notch intracellular domain. These results indicate that FLZ decreases Aß accumulation in mitochondria by selectively inhibiting γ-secretase. We propose that FLZ is a potential anti-AD drug candidate, and its mechanism may be improving mitochondrial function by reducing the Aß burden in mitochondria.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/biosíntesis , Bencenoacetamidas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Mitocondrias/metabolismo , Fenoles/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/biosíntesis , Línea Celular Tumoral , Células HeLa , Humanos , Mitocondrias/efectos de los fármacos
16.
PLoS One ; 8(11): e78033, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223757

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia worldwide and mainly characterized by the aggregated ß-amyloid (Aß) and hyperphosphorylated tau. FLZ is a novel synthetic derivative of natural squamosamide and has been proved to improve memory deficits in dementia animal models. In this study, we aimed to investigate the mechanisms of FLZ's neuroprotective effect in APP/PS1 double transgenic mice and SH-SY5Y (APPwt/swe) cells. The results showed that treatment with FLZ significantly improved the memory deficits of APP/PS1 transgenic mice and decreased apoptosis of SH-SY5Y (APPwt/swe) cells. FLZ markedly attenuated Aß accumulation and tau phosphorylation both in vivo and in vitro. Mechanistic study showed that FLZ interfered APP processing, i.e., FLZ decreased ß-amyloid precursor protein (APP) phosphorylation, APP-carboxy-terminal fragment (APP-CTF) production and ß-amyloid precursor protein cleaving enzyme 1 (BACE1) expression. These results indicated that FLZ reduced Aß production through inhibiting amyloidogenic pathway. The mechanistic study about FLZ's inhibitory effect on tau phosphorylation revealed t the involvement of Akt/glycogen synthase kinase 3ß (GSK3ß) pathway. FLZ treatment increased Akt activity and inhibited GSK3ß activity both in vivo and in vitro. The inhibitory effect of FLZ on GSK3ß activity and tau phosphorylation was suppressed by inhibiting Akt activity, indicating that Akt/GSK3ß pathway might be the possible mechanism involved in the inhibitory effect of FLZ on tau hyperphosphorylation. These results suggested FLZ might be a potential anti-AD drug as it not only reduced Aß production via inhibition amyloidogenic APP processing pathway, but also attenuated tau hyperphosphoylation mediated by Akt/GSK3ß.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Bencenoacetamidas/farmacología , Fármacos Neuroprotectores/farmacología , Fenoles/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis , Ácido Aspártico Endopeptidasas/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Reacción de Fuga , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
17.
Neuropharmacology ; 75: 201-12, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23916477

RESUMEN

The participation of neuroinflammation in the pathogenesis of Parkinson's disease (PD) has long been validated. Excessive activated microglia release a large number of pro-inflammatory factors, damage surrounding neurons and eventually induce neurodegeneration. Inhibition of microglial over-activation might be a promising strategy for PD treatment. FLZ (formulated as: N-(2-(4-hydroxy-phenyl)-ethyl)-2-(2, 5-dimethoxy-phenyl)-3-(3-methoxy-4-hydroxy-phenyl)-acrylamide, the code name: FLZ), a natural squamosamide derivative from a Chinese herb, has been shown to inhibit over-activated microglia and protect dopaminergic neurons in previous studies, but the mechanism remains unclear. In the present study, we further investigated the mechanism in lipopolysaccharide (LPS)-induced in vivo and in vitro PD models. FLZ treatment significantly improved the motor dysfunction of PD model rats induced by intra-nigral injection of LPS and this beneficial effect of FLZ attributed to the inhibition of microglial over-activation and the protection on dopaminergic neurons in the substantia nigra (SN). In vitro mechanistic study revealed that the inhibitive effect of FLZ on microglia was mediated by suppressing Src kinase related inflammatory signaling pathway activation and subsequent NF-κBp65 nuclear translocation, inhibiting nitric oxide (NO) and reactive oxygen species (ROS) production, decreasing nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation. In conclusion, the present study supports that FLZ exerts neuroprotection against LPS-induced dopaminergic neurodegeneration through its anti-inflammatory effect, which is mediated by suppressing Src tyrosine kinase and the downstream inflammatory signaling pathway. Furthermore, this study defines a critical role of Src tyrosine kinase in neuroinflammation, and suggests that particular tyrosine kinase inhibition may be a potential anti-inflammatory approach for PD treatment.


Asunto(s)
Bencenoacetamidas/farmacología , Encefalitis/tratamiento farmacológico , Activadores de Enzimas/farmacología , Enfermedad de Parkinson/complicaciones , Fenoles/farmacología , Familia-src Quinasas/metabolismo , Animales , Apomorfina/farmacología , Bencenoacetamidas/química , Línea Celular , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Encefalitis/etiología , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/química , Conducta Exploratoria/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson/etiología , Fenoles/química , Ratas , Ratas Wistar
18.
Yao Xue Xue Bao ; 47(3): 346-53, 2012 Mar.
Artículo en Chino | MEDLINE | ID: mdl-22645758

RESUMEN

Microglia are the principal immune effectors in brain and participate in a series ofneurodegenerative diseases. The microglial shapes are highly plastic. The morphology is closely related with their activation status and biological functions. Cerebral ischemia could induce microglial activation, and microglial activation is subjected to precise regulation. Microglia could play either protective or neurotoxic roles in cerebral ischemia. Therefore, regulating the expression of receptors or protein molecules on microglia, inhibiting the excessive activation of microglia and production of pro-inflammatory factors, promoting the release of neuroprotective substances might be beneficial to the treatment of cerebral ischemia. The study about relationship between microglia and cerebral ischemia will shed a light on the treatment of cerebral ischemia. This paper is a review of microglial activation and regulation during cerebral ischemia as well as related therapeutic methods.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Microglía/metabolismo , Microglía/fisiología , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Humanos , Inflamación/metabolismo , Microglía/citología , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Regeneración , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores Toll-Like/metabolismo
19.
Neurobiol Aging ; 33(6): 1122.e1-10, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22212405

RESUMEN

Although there are numerous studies regarding Alzheimer's disease (AD), the cause and progression of AD are still not well understood. The researches in the past decade implicated amyloid-beta (Aß) overproduction as a causative event in disease pathogenesis, but still failed to clarify the mechanism of pathology from Aß production to central neural system defects in AD. The present review raises the hypothesis that the onset of AD pathology is closely related with mitochondrial dysfunction induced by Aß and brain-derived neurotrophic factor (BDNF) axonal transport deficits. It is well-known that axonal transport defect and attenuation of BDNF-neurotrophic tyrosine receptor kinase 2 (TrkB) signal are fatal to neuronal function and survival. We hypothesized that abnormal amyloid precursor protein (APP) processing and Aß production in mitochondria disturb the axonal transport by impairing mitochondrial function and attenuate BDNF-neurotrophic tyrosine receptor kinase 2 signal subsequently. For this hypothesis, the factors related with the initiation of AD pathology are not only limited to the neurons per se but also expanded to the microenvironment around neurons, such as the secretion of BDNF from astrocytes. The modification of the origin in this pathway may contribute to slow down the disease progression of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Transporte Axonal/genética , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Mitocondrias/patología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/fisiología , Línea Celular Tumoral , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Inhibición Neural/genética
20.
Chem Biol Interact ; 188(1): 161-70, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20674558

RESUMEN

1-(3',4',5'-Trimethoxyphenyl)-3-(3'',4''-dimethoxy-2''-hydroxyphenyl)-propane (DP), a novel synthesized 1,3-diarylpropanes compound, showed growth inhibitory effect on human hepatoma HepG2 cells in a concentration-dependent manner. The growth inhibitory effect of DP on HepG2 cells was associated with microtubule depolymerization, G2/M phase arrest and apoptosis induction. The G2/M phase arrest induced by DP resulted from its microtubule-depolymerizing ability, and DP-treated HepG2 cells finally underwent caspase-dependent apoptosis. DP increased the levels of death receptor 4 (DR4), death receptor 5 (DR5) and pro-apoptotic protein Bax, but decreased the levels of anti-apoptotic protein Bcl-2. Meanwhile, the decrease in the mitochondrial membrane potential (MMP) and the release of cytochrome c from mitochondria were observed in DP-treated HepG2 cells. DP increased the levels of reactive oxygen species (ROS) in HepG2 cells, and antioxidant N-acetylcysteine (NAC) completely blocked DP-induced ROS accumulation and the disruption of the balance between Bax and Bcl-2 proteins, and effectively blocked the decreased MMP and apoptosis, but had no effect on the activation of caspase-8 and the up-regulations of DR4 and DR5 induced by DP. These results suggest that DP induces G2/M phase arrest through interruption of microtubule network followed by the death receptor- and ROS-mediated apoptosis in HepG2 cells.


Asunto(s)
Anisoles/farmacología , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Fase G2/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Línea Celular Tumoral , Electroforesis en Gel de Agar , Citometría de Flujo , Humanos , Inmunohistoquímica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA