Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Neurogenet ; 37(1-2): 10-19, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36000467

RESUMEN

Tau is a microtubule-associated protein that forms insoluble filaments that accumulate as neurofibrillary tangles in neurodegenerative diseases such as Alzheimer's disease and other related tauopathies. A relationship between abnormal Tau accumulation and ubiquitin-proteasome system impairment has been reported. However, the molecular mechanism linking Tau accumulation and ubiquitin proteasome system (UPS) dysfunction remains unclear. Here, we show that overexpression of wild-type or mutant (P301L) Tau increases the abundance of polyubiquitinated proteins and activates the autophagy-lysosome pathway in mammalian neuronal cells. Previous studies found that PTK2 inhibition mitigates toxicity induced by UPS impairment. Thus, we investigated whether PTK2 inhibition can attenuate Tau-induced UPS impairment and cell toxicity. We found that PTK2 inhibition significantly reduces Tau-induced death in mammalian neuronal cells. Moreover, overexpression of WT or mutant Tau increased the phosphorylation levels of PTK2 and p62. We also confirmed that PTK2 inhibition suppresses Tau-induced phosphorylation of PTK2 and p62. Furthermore, PTK2 inhibition significantly attenuated the climbing defect and shortened the lifespan in the Drosophila model of tauopathy. In addition, we observed that phosphorylation of p62 is markedly increased in Alzheimer's disease patients with tauopathies. Taken together, our results indicate that the UPS dysfunction induced by Tau accumulation might contribute directly to neurodegeneration in tauopathies and that PTK2 could be a promising therapeutic target for tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Animales , Enfermedad de Alzheimer/metabolismo , Proteínas tau/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Tauopatías/metabolismo , Ubiquitinas/metabolismo , Mamíferos/metabolismo
2.
J Neuroinflammation ; 17(1): 299, 2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33054766

RESUMEN

BACKGROUND: Cytoplasmic inclusions of transactive response DNA binding protein of 43 kDa (TDP-43) in neurons and astrocytes are a feature of some neurodegenerative diseases, such as frontotemporal lobar degeneration with TDP-43 (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). However, the role of TDP-43 in astrocyte pathology remains largely unknown. METHODS: To investigate whether TDP-43 overexpression in primary astrocytes could induce inflammation, we transfected primary astrocytes with plasmids encoding Gfp or TDP-43-Gfp. The inflammatory response and upregulation of PTP1B in transfected cells were examined using quantitative RT-PCR and immunoblot analysis. Neurotoxicity was analysed in a transwell coculture system of primary cortical neurons with astrocytes and cultured neurons treated with astrocyte-conditioned medium (ACM). We also examined the lifespan, performed climbing assays and analysed immunohistochemical data in pan-glial TDP-43-expressing flies in the presence or absence of a Ptp61f RNAi transgene. RESULTS: PTP1B inhibition suppressed TDP-43-induced secretion of inflammatory cytokines (interleukin 1 beta (IL-1ß), interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-α)) in primary astrocytes. Using a neuron-astrocyte coculture system and astrocyte-conditioned media treatment, we demonstrated that PTP1B inhibition attenuated neuronal death and mitochondrial dysfunction caused by overexpression of TDP-43 in astrocytes. In addition, neuromuscular junction (NMJ) defects, a shortened lifespan, inflammation and climbing defects caused by pan-glial overexpression of TDP-43 were significantly rescued by downregulation of ptp61f (the Drosophila homologue of PTP1B) in flies. CONCLUSIONS: These results indicate that PTP1B inhibition mitigates the neuronal toxicity caused by TDP-43-induced inflammation in mammalian astrocytes and Drosophila glial cells.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Unión al ADN/biosíntesis , Mediadores de Inflamación/metabolismo , Degeneración Nerviosa/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/biosíntesis , Animales , Animales Modificados Genéticamente , Astrocitos/patología , Células Cultivadas , Proteínas de Unión al ADN/genética , Drosophila , Expresión Génica , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética
3.
Biochem Biophys Res Commun ; 513(4): 925-932, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31005259

RESUMEN

The most prominent hallmarks of many neurodegenerative diseases are the accumulation of misfolded protein aggregates and the death of certain neuronal populations. Autophagy is the major intracellular mechanism that degrades protein aggregates and damaged cellular components. Many studies have reported that the dysfunction of autophagy is associated with several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), and Parkinson's disease. Here, we identified a novel mechanism of autophagy regulation. Inhibition of MEK5 reduced the level of p62 and increased the ratio of LC3-II to LC3-I, which is a marker for the activation of the autophagy-lysosome pathway (ALP). One of the most well-known regulators of the ALP is mTOR, and previous studies have reported that the major substrate of MEK5 is ERK5. However, we found that MEK5 modulates the autophagy-lysosome pathway in an mTOR- and ERK5-independent manner. Moreover, MEK5 inhibition alleviated the mislocalization of TDP-43 (an ALS-associated protein) and cell death in TDP-43-GFP-expressing neuronal cells. Taken together, these findings suggest that MEK5 is a novel autophagy modulator and that this kinase could be a therapeutic target for neurodegenerative diseases such as amyotrophic lateral sclerosis.


Asunto(s)
Autofagia , Proteínas de Unión al ADN/toxicidad , Lisosomas/metabolismo , MAP Quinasa Quinasa 5/antagonistas & inhibidores , Redes y Vías Metabólicas/fisiología , Neuronas/citología , Animales , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Proteínas de Unión al ARN/metabolismo , Serina-Treonina Quinasas TOR/fisiología
4.
Int J Mol Sci ; 20(3)2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-30678217

RESUMEN

Chronic neuroinflammation is a common feature of the aged brain, and its association with the major neurodegenerative changes involved in cognitive impairment and motor dysfunction is well established. One of the most potent antiaging interventions tested so far is dietary restriction (DR), which extends the lifespan in various organisms. Microglia and astrocytes are two major types of glial cells involved in the regulation of neuroinflammation. Accumulating evidence suggests that the age-related proinflammatory activation of astrocytes and microglia is attenuated under DR. However, the molecular mechanisms underlying DR-mediated regulation of neuroinflammation are not well understood. Here, we review the current understanding of the effects of DR on neuroinflammation and suggest an underlying mechanistic link between DR and neuroinflammation that may provide novel insights into the role of DR in aging and age-associated brain disorders.


Asunto(s)
Astrocitos/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Animales , Astrocitos/inmunología , Encefalopatías/inmunología , Encefalopatías/metabolismo , Restricción Calórica , Humanos , Inflamación/inmunología , Microglía/inmunología
5.
Crit Rev Immunol ; 35(1): 77-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26111426

RESUMEN

Lipocalin-2 (LCN2), a secretory protein, regulates diverse cellular processes such as cell death/survival, cell migration/invasion, cell differentiation, iron delivery, inflammation, insulin resistance, and tissue regeneration. Recently, we reported that LCN2 is secreted by brain astrocytes under inflammatory conditions and that it promotes apoptosis, morphological changes, and migration in astrocytes both in vitro and in vivo. Activated astrocytes release LCN2 not only to induce the morphological transformation associated with reactive astrocytosis, but also to promote their own death. Under inflammatory conditions, activated astrocytes also show functional dichotomy similar to the M1/M2 phenotypes of microglia and macrophages. LCN2 is thought to be a chemokine inducer and an autocrine promoter of the classical proinflammatory activation of astrocytes. This article summarizes the current knowledge regarding the role of astrocyte-derived LCN2 as a proinflammatory mediator in the central nervous system and discusses LCN2's role in neuroinflammatory disorders.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Astrocitos/inmunología , Encéfalo/inmunología , Lipocalinas/metabolismo , Enfermedades Neurodegenerativas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Fase Aguda/inmunología , Animales , Apoptosis , Comunicación Autocrina , Humanos , Lipocalina 2 , Lipocalinas/inmunología , Inflamación Neurogénica , Proteínas Proto-Oncogénicas/inmunología
6.
J Immunol ; 191(10): 5204-19, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24089194

RESUMEN

Astrocytes provide structural and functional support for neurons, as well as display neurotoxic or neuroprotective phenotypes depending upon the presence of an immune or inflammatory microenvironment. This study was undertaken to characterize multiple phenotypes of activated astrocytes and to investigate the regulatory mechanisms involved. We report that activated astrocytes in culture exhibit two functional phenotypes with respect to pro- or anti-inflammatory gene expression, glial fibrillary acidic protein expression, and neurotoxic or neuroprotective activities. The two distinct functional phenotypes of astrocytes were also demonstrated in a mouse neuroinflammation model, which showed pro- or anti-inflammatory gene expression in astrocytes following challenge with classical or alternative activation stimuli; similar results were obtained in the absence of microglia. Subsequent studies involving recombinant lipocalin-2 (LCN2) protein treatment or Lcn2-deficient mice indicated that the pro- or anti-inflammatory functionally polarized phenotypes of astrocytes and their intracellular signaling pathway were critically regulated by LCN2 under in vitro and in vivo conditions. Astrocyte-derived LCN2 promoted classical proinflammatory activation of astrocytes but inhibited IL-4-STAT6 signaling, a canonical pathway involved in alternative anti-inflammatory activation. Our results suggest that the secreted protein LCN2 is an autocrine modulator of the functional polarization of astrocytes in the presence of immune or inflammatory stimuli and that LCN2 could be targeted therapeutically to dampen proinflammatory astrocytic activation and related pathologies in the CNS.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Astrocitos/metabolismo , Encéfalo/inmunología , Lipocalinas/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Fase Aguda/deficiencia , Proteínas de Fase Aguda/genética , Animales , Astrocitos/citología , Astrocitos/inmunología , Polaridad Celular , Células Cultivadas , Inflamación/inmunología , Interleucina-4/metabolismo , Lipocalina 2 , Lipocalinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/inmunología
7.
FASEB J ; 27(3): 1176-90, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23207546

RESUMEN

Activated macrophages are classified into two different forms: classically activated (M1) or alternatively activated (M2) macrophages. The presence of M1/M2 phenotypic polarization has also been suggested for microglia. Here, we report that the secreted protein lipocalin 2 (LCN2) amplifies M1 polarization of activated microglia. LCN2 protein (EC 1 µg/ml), but not glutathione S-transferase used as a control, increased the M1-related gene expression in cultured mouse microglial cells after 8-24 h. LCN2 was secreted from M1-polarized, but not M2-polarized, microglia. LCN2 inhibited phosphorylation of STAT6 in IL-4-stimulated microglia, suggesting LCN2 suppression of the canonical M2 signaling. In the lipopolysaccharide (LPS)-induced mouse neuroinflammation model, the expression of LCN2 was notably increased in microglia. Primary microglial cultures derived from LCN2-deficient mice showed a suppressed M1 response and enhanced M2 response. Mice lacking LCN2 showed a markedly reduced M1-related gene expression in microglia after LPS injection, which was consistent with the results of histological analysis. Neuroinflammation-associated impairment in motor behavior and cognitive function was also attenuated in the LCN2-deficient mice, as determined by the rotarod performance test, fatigue test, open-field test, and object recognition task. These findings suggest that LCN2 is an M1-amplifier in brain microglial cells.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Polaridad Celular/fisiología , Lipocalinas/metabolismo , Microglía/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Fase Aguda/genética , Animales , Polaridad Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Células Jurkat , Lipocalina 2 , Lipocalinas/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Mutantes , Microglía/citología , Proteínas Oncogénicas/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas/genética , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo
8.
J Neurol Sci ; 457: 122885, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38278691

RESUMEN

Amyotrophic Lateral Sclerosis (ALS) is the most common type of motor neuron disease characterized by progressive motor neuron degeneration in brain and spinal cord. Most cases are sporadic in ALS and 5-10% of cases are familiar. >50 genes are known to be associated with ALS and one of them is ERBB4. In this paper, we report the case of a 53-year-old ALS patient with progressive muscle weakness and fasciculation, but he had no cognitive decline. We performed the next generation sequencing (NGS) and in silico analysis, it predicted a highly pathogenic variant, c.2116 A > G, p.Asn706Asp (N706D) in the ERBB4 gene. The amino acid residue is highly conserved among species. ERBB4 is a member of the ERBB family of receptor tyrosine kinases. ERBB4 has multiple tyrosine phosphorylation sites, including an autophosphorylation site at tyrosine 1284 residue. Autophosphorylation of ERBB4 promotes biological activity and it associated with NRG-1/ERBB4 pathway. It is already known that tyrosine 128 phosphorylation of ERBB4 is decreased in patients who have ALS-associated ERBB4 mutations. We generated ERBB4 N706D construct using site-directed mutagenesis and checked the phosphorylation level of ERBB4 N706D in NSC-34 cells. We found that the phosphorylation of ERBB4 N706D was decreased compared to ERBB4 wild-type, indicating a loss of function mutation in ERBB4. We report a novel variant in ERBB4 gene leading to ALS through dysfunction of ERBB4.


Asunto(s)
Esclerosis Amiotrófica Lateral , Masculino , Humanos , Persona de Mediana Edad , Esclerosis Amiotrófica Lateral/metabolismo , Mutación/genética , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo , Tirosina
9.
J Biol Chem ; 287(12): 9414-28, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22275363

RESUMEN

Prostaglandin D synthase (PGDS) is responsible for the conversion of PGH(2) to PGD(2). Two distinct types of PGDS have been identified: hematopoietic-type PGDS (H-PGDS) and lipocalin-type PGDS (L-PGDS). L-PGDS acts as both a PGD(2)-synthesizing enzyme and as an extracellular transporter of various lipophilic small molecules. Although L-PGDS is one of the most abundant proteins in the cerebrospinal fluid, little is known about the function of L-PGDS in the central nervous system (CNS). To better understand the role of L-PGDS in the CNS, effects of L-PGDS on the migration and morphology of glial cells were investigated. The L-PGDS protein accelerated the migration of cultured glial cells. Expression of the L-pgds gene was detected in glial cells and neurons. L-PGDS protein also induced morphological changes in glia similar to the characteristic phenotypic changes in reactive gliosis. L-PGDS-induced cell migration was associated with augmented formation of actin filaments and focal adhesion, which was accompanied by activation of AKT, RhoA, and JNK pathways. L-PGDS protein injected into the mouse brain promoted migration and accumulation of astrocytes in vivo. Furthermore, the cell migration-promoting effect of L-PGDS on glial cells was independent of the PGD(2) products. The L-PGDS protein interacted with myristoylated alanine-rich protein kinase C substrate (MARCKS) to promote cell migration. These results demonstrate the critical role of L-PGDS as a secreted lipocalin in the regulation of glial cell migration and morphology. The results also indicate that L-PGDS may participate in reactive gliosis in an autocrine or paracrine manner, and may have pathological implications in neuroinflammatory diseases.


Asunto(s)
Movimiento Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Proteínas de la Membrana/metabolismo , Neuroglía/citología , Neuroglía/enzimología , Prostaglandina D2/metabolismo , Animales , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular/genética , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Células 3T3 NIH , Neuroglía/metabolismo
10.
Front Aging Neurosci ; 15: 1047897, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875699

RESUMEN

The endoplasmic reticulum (ER) is a major organelle involved in protein quality control and cellular homeostasis. ER stress results from structural and functional dysfunction of the organelle, along with the accumulation of misfolded proteins and changes in calcium homeostasis, it leads to ER stress response pathway such as unfolded protein response (UPR). Neurons are particularly sensitive to the accumulation of misfolded proteins. Thus, the ER stress is involved in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, prion disease and motor neuron disease (MND). Recently, the complex involvement of ER stress pathways has been demonstrated in experimental models of amyotrophic lateral sclerosis (ALS)/MND using pharmacological and genetic manipulation of the unfolded protein response (UPR), an adaptive response to ER stress. Here, we aim to provide recent evidence demonstrating that the ER stress pathway is an essential pathological mechanism of ALS. In addition, we also provide therapeutic strategies that can help treat diseases by targeting the ER stress pathway.

11.
J Biol Chem ; 286(51): 43855-43870, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22030398

RESUMEN

The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.


Asunto(s)
Proteínas de Fase Aguda/química , Proteínas de Fase Aguda/metabolismo , Quimiocina CXCL10/química , Regulación de la Expresión Génica , Lipocalinas/química , Lipocalinas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Animales , Astrocitos/citología , Encéfalo/metabolismo , Movimiento Celular , Quimiocinas/metabolismo , Medios de Cultivo Condicionados/farmacología , Humanos , Inflamación , Lipocalina 2 , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas/química , Factor de Transcripción STAT3/metabolismo
12.
J Neurosci Res ; 90(3): 540-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22038922

RESUMEN

A secreted protein, lipocalin-2 (LCN2), has been previously shown to regulate a variety of cellular phenotypes such as cell death, migration, and morphology. The role of LCN2, however, appears to be different depending on the cellular context. Here, we investigated how LCN2 influences neuronal phenotypes by using primary cortical neuronal cell cultures and neuroblastoma cell lines as a model. When exposed to LCN2 protein, neurons and neuroblastoma cells were sensitized to cell death evoked by nitric oxide, oxidative stress, and tumor necrosis factor-α (TNF-α). A forced expression of lcn2 in glia enhanced neuronal cell death in cocultures of glia and neurons, indicating that both exogenous protein addition and endogenous expression of lcn2 give rise to similar results. Iron and BCL2-interacting mediator of cell death (BIM) protein were involved in LCN2-induced cell death sensitization, based on the studies using iron donor, chelator, siderophore, and short hairpin RNA (shRNA)-mediated knockdown of bim expression. Furthermore, cell migration assay and immunofluorescence microscopic observation revealed that LCN2 accelerated neuronal motility and process extension, suggesting multiple roles for LCN2 in the regulation of neuronal cell death, migration, and morphology.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Muerte Celular/fisiología , Movimiento Celular/fisiología , Forma de la Célula/fisiología , Lipocalinas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Técnicas de Cocultivo , Lipocalina 2 , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/farmacología
13.
Exp Cell Res ; 317(14): 2007-18, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21645515

RESUMEN

Small G protein superfamily consists of more than 150 members, and is classified into six families: the Ras, Rho, Rab, Arf, Ran, and RGK families. They regulate a wide variety of cell functions such as cell proliferation/differentiation, cytoskeletal reorganization, vesicle trafficking, nucleocytoplasmic transport and microtubule organization. The small G proteins have also been shown to regulate cell death/survival and cell shape. In this study, we compared the role of representative members of the six families of small G proteins in cell migration and cell death/survival, two cellular phenotypes that are associated with inflammation, tumorigenesis, and metastasis. Our results show that small G proteins of the six families differentially regulate cell death and cell cycle distribution. In particular, our results indicate that Rho family of small G proteins is antiapoptotic. Ras, Rho, and Ran families promoted cell migration. There was no significant correlation between the cell death- and cell migration-regulating activities of the small G proteins. Nevertheless, RalA was not only cytoprotective against multiple chemotherapeutic drugs, but also promigratory inducing stress fiber formation, which was accompanied by the activation of Akt and Erk pathways. Our study provides a framework for further systematic investigation of small G proteins in the perspectives of cell death/survival and motility in inflammation and cancer.


Asunto(s)
Movimiento Celular , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Animales , Western Blotting , Células COS , Muerte Celular , Supervivencia Celular , Células Cultivadas , Chlorocebus aethiops , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HeLa , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión al GTP ral/genética
14.
Dev Cell ; 57(6): 783-798.e8, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35320731

RESUMEN

Fused in sarcoma (FUS) is a DNA/RNA-binding protein that is involved in DNA repair and RNA processing. FUS is associated with neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, the molecular mechanisms underlying FUS-mediated neurodegeneration are largely unknown. Here, using a Drosophila model, we showed that the overexpression of glutathione transferase omega 2 (GstO2) reduces cytoplasmic FUS aggregates and prevents neurodegenerative phenotypes, including neurotoxicity and mitochondrial dysfunction. We found a FUS glutathionylation site at the 447th cysteine residue in the RanBP2-type ZnF domain. The glutathionylation of FUS induces FUS aggregation by promoting phase separation. GstO2 reduced cytoplasmic FUS aggregation by deglutathionylation in Drosophila brains. Moreover, we demonstrated that the overexpression of human GSTO1, the homolog of Drosophila GstO2, attenuates FUS-induced neurotoxicity and cytoplasmic FUS accumulation in mouse neuronal cells. Thus, the modulation of FUS glutathionylation might be a promising therapeutic strategy for FUS-associated neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Drosophila/metabolismo , Ratones , Mutación/genética , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo
15.
Br J Pharmacol ; 179(5): 998-1016, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34524687

RESUMEN

BACKGROUND AND PURPOSE: There is a scarcity of information regarding the role of prothrombin kringle-2 (pKr-2), which can be generated by active thrombin, in hippocampal neurodegeneration and Alzheimer's disease (AD). EXPERIMENTAL APPROACH: To assess the role of pKr-2 in association with the neurotoxic symptoms of AD, we determined pKr-2 protein levels in post-mortem hippocampal tissues of patients with AD and the hippocampi of five familial AD (5XFAD) mice compared with those of age-matched controls and wild-type (WT) mice, respectively. In addition, we investigated whether the hippocampal neurodegeneration and object memory impairments shown in 5XFAD mice were mediated by changes to pKr-2 up-regulation. KEY RESULTS: Our results demonstrated that pKr-2 was up-regulated in the hippocampi of patients with AD and 5XFAD mice, but was not associated with amyloid-ß aggregation in 5XFAD mice. The up-regulation of pKr-2 expression was inhibited by preservation of the blood-brain barrier (BBB) via addition of caffeine to their water supply or by treatment with rivaroxaban, an inhibitor of factor Xa that is associated with thrombin production. Moreover, the prevention of up-regulation of pKr-2 expression reduced neurotoxic symptoms, such as hippocampal neurodegeneration and object recognition decline due to neurotoxic inflammatory responses in 5XFAD mice. CONCLUSION AND IMPLICATIONS: We identified a novel pathological mechanism of AD mediated by abnormal accumulation of pKr-2, which functions as an important pathogenic factor in the adult brain via blood brain barrier (BBB) breakdown. Thus, pKr-2 represents a novel target for AD therapeutic strategies and those for related conditions.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Kringles , Ratones , Ratones Transgénicos , Protrombina/metabolismo , Protrombina/uso terapéutico , Trombina
16.
Exp Neurobiol ; 30(5): 341-355, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34737239

RESUMEN

Sirtuin 3 (SIRT3), a well-known mitochondrial deacetylase, is involved in mitochondrial function and metabolism under various stress conditions. In this study, we found that the expression of SIRT3 was markedly increased by oxidative stress in dopaminergic neuronal cells. In addition, SIRT3 overexpression enhanced mitochondrial activity in differentiated SH-SY5Y cells. We also showed that SIRT3 overexpression attenuated rotenoneor H2O2-induced toxicity in differentiated SH-SY5Y cells (human dopaminergic cell line). We further found that knockdown of SIRT3 enhanced rotenone- or H2O2-induced toxicity in differentiated SH-SY5Y cells. Moreover, overexpression of SIRT3 mitigated cell death caused by LPS/IFN-γ stimulation in astrocytes. We also found that the rotenone treatment increases the level of SIRT3 in Drosophila brain. We observed that downregulation of sirt2 (Drosophila homologue of SIRT3) significantly accelerated the rotenone-induced toxicity in flies. Taken together, these findings suggest that the overexpression of SIRT3 mitigates oxidative stress-induced cell death and mitochondrial dysfunction in dopaminergic neurons and astrocytes.

17.
Antioxidants (Basel) ; 11(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35052586

RESUMEN

TAR DNA-binding protein 43 (TDP-43) is a member of an evolutionarily conserved family of heterogeneous nuclear ribonucleoproteins that modulate multiple steps in RNA metabolic processes. Cytoplasmic aggregation of TDP-43 in affected neurons is a pathological hallmark of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), and limbic predominant age-related TDP-43 encephalopathy (LATE). Mislocalized and accumulated TDP-43 in the cytoplasm induces mitochondrial dysfunction and reactive oxidative species (ROS) production. Here, we show that TDP-43- and rotenone-induced neurotoxicity in the human neuronal cell line SH-SY5Y were attenuated by hydroxocobalamin (Hb, vitamin B12 analog) treatment. Although Hb did not affect the cytoplasmic accumulation of TDP-43, Hb attenuated TDP-43-induced toxicity by reducing oxidative stress and mitochondrial dysfunction. Moreover, a shortened lifespan and motility defects in TDP-43-expressing Drosophila were significantly mitigated by dietary treatment with hydroxocobalamin. Taken together, these findings suggest that oral intake of hydroxocobalamin may be a potential therapeutic intervention for TDP-43-associated proteinopathies.

18.
Front Pharmacol ; 12: 747975, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925009

RESUMEN

The autophagy-lysosomal pathway is an essential cellular mechanism that degrades aggregated proteins and damaged cellular components to maintain cellular homeostasis. Here, we identified HEXA-018, a novel compound containing a catechol derivative structure, as a novel inducer of autophagy. HEXA-018 increased the LC3-I/II ratio, which indicates activation of autophagy. Consistent with this result, HEXA-018 effectively increased the numbers of autophagosomes and autolysosomes in neuronal cells. We also found that the activation of autophagy by HEXA-018 is mediated by the AMPK-ULK1 pathway in an mTOR-independent manner. We further showed that ubiquitin proteasome system impairment- or oxidative stress-induced neurotoxicity was significantly reduced by HEXA-018 treatment. Moreover, oxidative stress-induced mitochondrial dysfunction was strongly ameliorated by HEXA-018 treatment. In addition, we investigated the efficacy of HEXA-018 in models of TDP-43 proteinopathy. HEXA-018 treatment mitigated TDP-43 toxicity in cultured neuronal cell lines and Drosophila. Our data indicate that HEXA-018 could be a new drug candidate for TDP-43-associated neurodegenerative diseases.

19.
J Neurosci ; 29(1): 234-49, 2009 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-19129400

RESUMEN

Astrocytes, the most abundant glial cell type in the brain, provide metabolic and trophic support to neurons and modulate synaptic activity. In response to a brain injury, astrocytes proliferate and become hypertrophic with an increased expression of intermediate filament proteins. This process is collectively referred to as reactive astrocytosis. Lipocalin 2 (lcn2) is a member of the lipocalin family that binds to small hydrophobic molecules. We propose that lcn2 is an autocrine mediator of reactive astrocytosis based on the multiple roles of lcn2 in the regulation of cell death, morphology, and migration of astrocytes. lcn2 expression and secretion increased after inflammatory stimulation in cultured astrocytes. Forced expression of lcn2 or treatment with LCN2 protein increased the sensitivity of astrocytes to cytotoxic stimuli. Iron and BIM (Bcl-2-interacting mediator of cell death) proteins were involved in the cytotoxic sensitization process. LCN2 protein induced upregulation of glial fibrillary acidic protein (GFAP), cell migration, and morphological changes similar to characteristic phenotypic changes termed reactive astrocytosis. The lcn2-induced phenotypic changes of astrocytes occurred through a Rho-ROCK (Rho kinase)-GFAP pathway, which was positively regulated by nitric oxide and cGMP. In zebrafishes, forced expression of rat lcn2 gene increased the number and thickness of cellular processes in GFAP-expressing radial glia cells, suggesting that lcn2 expression in glia cells plays an important role in vivo. Our results suggest that lcn2 acts in an autocrine manner to induce cell death sensitization and morphological changes in astrocytes under inflammatory conditions and that these phenotypic changes may be the basis of reactive astrocytosis in vivo.


Asunto(s)
Anexina A2/metabolismo , Astrocitos/efectos de los fármacos , Comunicación Autocrina/fisiología , Amidas/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Comunicación Autocrina/efectos de los fármacos , Encéfalo/citología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Embrión no Mamífero , Citometría de Flujo/métodos , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos ICR , Nitritos/metabolismo , Nitroprusiato/farmacología , Penicilamina/análogos & derivados , Penicilamina/farmacología , Fagocitos/efectos de los fármacos , Fagocitos/fisiología , Propidio , Piridinas/farmacología , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Sales de Tetrazolio , Tiazoles , Transfección/métodos , Factor de Necrosis Tumoral alfa/farmacología , Pez Cebra , Proteínas de Unión al GTP rho/metabolismo
20.
J Neurosci Res ; 88(10): 2188-96, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20162721

RESUMEN

Glucocorticoid-induced tumor necrosis factor receptor family-related protein ligand (GITRL) is a member of the tumor necrosis factor superfamily (TNFSF) and is known to act as a costimulator in the immune system by binding to GITR. GITRL is expressed in endothelial cells, dendritic cells, macrophages, and B cells, but it is not known whether GITRL is expressed in brain microglia cells. Here, we investigated the expression of GITR and GITRL and their potential role in microglia cells. Using BV-2 mouse microglia cells and mouse primary microglia cultures, we have demonstrated that 1) both GITR and GITRL are expressed in microglia cells; 2) stimulation of GITRL induces inflammatory activation of microglia on the basis of production of nitric oxide (NO) and expression of inducible nitric oxide synthase, cyclooxygenase-2, CD40, and matrix metalloproteinase-9; 3) GITRL-mediated microglial NO production partially depends on p38 MAPK, JNK, and nuclear factor-kappaB pathways; and 4) GITRL stimulation also induces microglia cell death. These results indicate that GITR and GITRL are functionally expressed on brain microglia and that the stimulation of GITRL can induce inflammatory activation of microglia. The GITR/GITRL system may play an important role in neuroinflammation.


Asunto(s)
Microglía/inmunología , Microglía/fisiología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Muerte Celular/fisiología , Línea Celular , Células Cultivadas , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Ratones , Microglía/enzimología , Óxido Nítrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA