Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Drug Dev Res ; 81(2): 184-193, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31782539

RESUMEN

A growing body of evidence indicates that autophagy, an intracellular degradation pathway, profoundly affects Alzheimer's disease (AD) pathogenesis. Autophagy mediates the degradation of neurotoxic material and damaged organelles, allowing their clearance by glial and neuronal cells, while impaired autophagy may account for the accumulation of protein aggregates. Accordingly, dysfunctional autophagy is one of AD hallmarks; it occurs early in the disease development, which makes it an attractive therapeutic intervention target. Therefore, in recent years, the potential of autophagy induction as a treatment for AD has been studied extensively using various autophagy inducers, most of which are already in clinical practice for other medical conditions. Albeit promising results, including in AD clinical trials, this therapeutic strategy still requires careful consideration in order to fully understand the role of autophagy in AD pathogenesis and to further improve the outcomes. This review summarizes the current findings in this field and raises open questions and new prospects.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Redes Reguladoras de Genes/efectos de los fármacos , Lisosomas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Autofagia , Ensayos Clínicos como Asunto , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Terapia Molecular Dirigida
2.
J Neurochem ; 143(5): 584-594, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28921554

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disorder, of which 1% of the hereditary cases are linked to mutations in DJ-1, an oxidative stress sensor. The pathological hallmark of PD is intercellular inclusions termed Lewy Bodies, composed mainly of α-Synuclein (α-Syn) protein. Recent findings have shown that α-Syn can be transmitted from cell to cell, suggesting an important role of microglia, as the main scavenger cells of the brain, in clearing α-Syn. We previously reported that the knock down (KD) of DJ-1 in microglia increased cells' neurotoxicity to dopaminergic neurons. Here, we discovered that α-Syn significantly induced elevated secretion of the proinflammatory cytokines IL-6 and IL-1ß and a significant dose-dependent elevation in the production of nitric oxide in DJ-1 KD microglia, compared to control microglia. We further investigated the ability of DJ-1 KD microglia to uptake and degrade soluble α-Syn, and discovered that DJ-1 KD reduces cell-surface lipid raft expression in microglia and impairs their ability to uptake soluble α-Syn. Autophagy is an important mechanism for degradation of intracellular proteins and organelles. We discovered that DJ-1 KD microglia exhibit an impaired autophagy-dependent degradation of p62 and LC3 proteins, and that manipulation of autophagy had less effect on α-Syn uptake and clearance in DJ-1 KD microglia, compared to control microglia. Further studies of the link between DJ-1, α-Syn uptake and autophagy may provide useful insights into the role of microglia in the etiology of the PD.


Asunto(s)
Autofagia/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Proteína Desglicasa DJ-1/metabolismo , alfa-Sinucleína/farmacología , Animales , Células Cultivadas , Citocinas/metabolismo , Humanos , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Proteína Desglicasa DJ-1/deficiencia , alfa-Sinucleína/metabolismo
3.
J Neurochem ; 138(6): 845-58, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27399849

RESUMEN

Glial scarring, formed by reactive astrocytes, is one of the major impediments for regeneration after spinal cord injury (SCI). Reactive astrocytes become hypertrophic, proliferate and secrete chondroitin sulphate proteoglycans into the extracellular matrix (ECM). Many studies have demonstrated that epidermal growth factor receptors (EGFR) can mediate astrocyte reactivity after neurotrauma. Previously we showed that there is crosstalk between nucleolin and EGFR that leads to increased EGFR activation followed by increased cell proliferation. Treatment with the nucleolin inhibitor GroA (AS1411) prevented these effects in vitro and in vivo. In this study, we hypothesized that similar interactions may mediate astrogliosis after SCI. Our results demonstrate that nucleolin and EGFR interaction may play a pivotal role in mediating astrocyte proliferation and reactivity after SCI. Moreover, we demonstrate that treatment with GroA reduces EGFR activation, astrocyte proliferation and chondroitin sulphate proteoglycans secretion, therefore promoting axonal regeneration and sprouting into the lesion site. Our results identify, for the first time, a role for the interaction between nucleolin and EGFR in astrocytes after SCI, indicating that nucleolin inhibitor GroA may be used as a novel treatment after neurotrauma. A major barrier for axonal regeneration after spinal cord injury is glial scar created by reactive and proliferating astrocytes. EGFR mediate astrocyte reactivity. We showed that inhibition of nucleolin by GroA, reduces EGFR activation, which results in attenuation of astrocyte reactivity and proliferation in vivo and in vitro. EGFR, epidermal growth factor receptor.


Asunto(s)
Receptores ErbB/agonistas , Neuroglía/patología , Oligodesoxirribonucleótidos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Traumatismos de la Médula Espinal/patología , Animales , Aptámeros de Nucleótidos , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Humanos , Inmunohistoquímica , Locomoción/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/psicología , Nucleolina
4.
Biol Cell ; 107(5): 130-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25735913

RESUMEN

The Ras oncogene transmits signals, which regulate various cellular processes including cell motility, differentiation, growth and death. Since Ras signalling is abnormally activated in more than 30% of human cancers, Ras and its downstream signalling pathways are considered good targets for therapeutic interference. Ras is post-translationally modified by the addition of a farnesyl group, which permits its attachment to the plasma membrane. Exploiting this knowledge, a synthetic Ras inhibitor, S-trans, trans-farnesylthiosalicylic acid (FTS; Salirasib), was developed. FTS resembles the farnesylcysteine group of Ras, and acts as an effective Ras antagonist. In the present review, the effect of FTS in combination with various other drugs, as tested in vitro and in vivo, and its therapeutic potential are discussed. As reviewed, FTS cooperates with diverse therapeutic agents, which significantly improves treatment outcome. Therefore, combinations of FTS with other agents have a potential to serve as anti-cancer or anti-inflammatory therapies.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Farnesol/análogos & derivados , Neoplasias/tratamiento farmacológico , Salicilatos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Farnesol/farmacología , Humanos
5.
Mol Cell Neurosci ; 68: 82-91, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25936601

RESUMEN

Spinal cord injury (SCI) frequently leads to a permanent functional impairment as a result of the initial injury followed by secondary injury mechanism, which is characterised by increased inflammation, glial scarring and neuronal cell death. Finding drugs that may reduce inflammatory cell invasion and activation to reduce glial scarring and increase neuronal survival is of major importance for improving the outcome after SCI. In the present study, we examined the effect of rapamycin, an mTORC1 inhibitor and an inducer of autophagy, on recovery from spinal cord injury. Autophagy, a process that facilitates the degradation of cytoplasmic proteins, is also important for maintenance of neuronal homeostasis and plays a major role in neurodegeneration after neurotrauma. We examined rapamycin effects on the inflammatory response, glial scar formation, neuronal survival and regeneration in vivo using spinal cord hemisection model in mice, and in vitro using primary cortical neurons and human astrocytes. We show that a single injection of rapamycin, inhibited p62/SQSTM1, a marker of autophagy, inhibited mTORC1 downstream effector p70S6K, reduced macrophage/neutrophil infiltration into the lesion site, microglia activation and secretion of TNFα. Rapamycin inhibited astrocyte proliferation and reduced the number of GFAP expressing cells at the lesion site. Finally, it increased neuronal survival and axonogenesis towards the lesion site. Our study shows that rapamycin treatment increased significantly p-Akt levels at the lesion site following SCI. Similarly, rapamycin treatment of neurons and astrocytes induced p-Akt elevation under stress conditions. Together, these findings indicate that rapamycin is a promising candidate for treatment of acute SCI condition and may be a useful therapeutic agent.


Asunto(s)
Inmunosupresores/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Sirolimus/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Antígeno CD11b/metabolismo , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 3 Similar a ELAV/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Ratas , Factores de Tiempo
6.
Growth Factors ; 32(2): 74-81, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24641597

RESUMEN

The epidermal growth factor receptor (EGFR) undergoes a conformational change in response to ligand binding. The ligand-induced changes in cell surface aggregation and mobility have a profound effect on the function of all the family members. Ligand also activates the EGFR intracellular kinase, stimulating proliferation and cell survival. The EGFR family are often activated, overexpressed or mutated in cancer cells and therapeutic drugs (including antibodies) can slow the progress of some cancers. This article provides a brief, annotated summary of the presentations and discussion which occurred at the Epidermal Growth Factor Receptor - Future Directions Conference held in Jerusalem in November 2013.


Asunto(s)
Receptores ErbB/metabolismo , Receptores ErbB/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Receptor ErbB-2/metabolismo , Transducción de Señal
7.
J Virol ; 87(24): 13397-408, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24089565

RESUMEN

The mode and timing of virally induced cell death hold the potential of regulating viral yield, viral transmission, and the severity of virally induced disease. Orbiviruses such as the epizootic hemorrhagic disease virus (EHDV) are nonenveloped and cytolytic. To date, the death of cells infected with EHDV, the signal transduction pathways involved in this process, and the consequence of their inhibition have yet to be characterized. Here, we report that the Ibaraki strain of EHDV2 (EHDV2-IBA) induces apoptosis, autophagy, a decrease in cellular protein synthesis, the activation of c-Jun N-terminal kinase (JNK), and the phosphorylation of the JNK substrate c-Jun. The production of infectious virions decreased upon inhibition of apoptosis with the pan-caspase inhibitor Q-VD-OPH (quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone), upon inhibition of autophagy with 3-methyladenine or via the knockout of the autophagy regulator Atg5, or upon treatment of infected cells with the JNK inhibitor SP600125 or the cyclin-dependent kinase (CDK) inhibitor roscovitine, which also inhibited c-Jun phosphorylation. Moreover, Q-VD-OPH, SP600125, and roscovitine partially reduced EHDV2-IBA-induced cell death, and roscovitine diminished the induction of autophagy by EHDV2-IBA. Taken together, our results imply that EHDV induces and benefits from the activation of signaling pathways involved in cell stress and death.


Asunto(s)
Apoptosis , Autofagia , Enfermedades de los Bovinos/fisiopatología , Virus de la Enfermedad Hemorrágica Epizoótica/fisiología , Infecciones por Reoviridae/veterinaria , Enfermedades de las Ovejas/fisiopatología , Animales , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/virología , Línea Celular , Virus de la Enfermedad Hemorrágica Epizoótica/genética , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/fisiopatología , Infecciones por Reoviridae/virología , Ovinos , Enfermedades de las Ovejas/genética , Enfermedades de las Ovejas/metabolismo , Enfermedades de las Ovejas/virología , Transducción de Señal
8.
Biomolecules ; 13(10)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37892117

RESUMEN

Parkinson's disease (PD) is a devastating disease associated with accumulation of α-synuclein (α-Syn) within dopaminergic neurons, leading to neuronal death. PD is characterized by both motor and non-motor clinical symptoms. Several studies indicate that autophagy, an important intracellular degradation pathway, may be involved in different neurodegenerative diseases including PD. The autophagic process mediates the degradation of protein aggregates, damaged and unneeded proteins, and organelles, allowing their clearance, and thereby maintaining cell homeostasis. Impaired autophagy may cause the accumulation of abnormal proteins. Incomplete or impaired autophagy may explain the neurotoxic accumulation of protein aggregates in several neurodegenerative diseases including PD. Indeed, studies have suggested the contribution of impaired autophagy to α-Syn accumulation, the death of dopaminergic neurons, and neuroinflammation. In this review, we summarize the recent literature on the involvement of autophagy in PD pathogenesis.


Asunto(s)
Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , Agregado de Proteínas , alfa-Sinucleína/metabolismo , Autofagia/fisiología , Neuronas Dopaminérgicas/metabolismo
9.
EMBO Rep ; 10(3): 285-92, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19180116

RESUMEN

Autophagy, an evolutionarily conserved process, has functions both in cytoprotective and programmed cell death mechanisms. Beclin 1, an essential autophagic protein, was recently identified as a BH3-domain-only protein that binds to Bcl-2 anti-apoptotic family members. The dissociation of beclin 1 from its Bcl-2 inhibitors is essential for its autophagic activity, and therefore should be tightly controlled. Here, we show that death-associated protein kinase (DAPK) regulates this process. The activated form of DAPK triggers autophagy in a beclin-1-dependent manner. DAPK phosphorylates beclin 1 on Thr 119 located at a crucial position within its BH3 domain, and thus promotes the dissociation of beclin 1 from Bcl-XL and the induction of autophagy. These results reveal a substrate for DAPK that acts as one of the core proteins of the autophagic machinery, and they provide a new phosphorylation-based mechanism that reduces the interaction of beclin 1 with its inhibitors to activate the autophagic machinery.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de la Membrana/metabolismo , Proteína bcl-X/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Beclina-1 , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Línea Celular , Proteínas Quinasas Asociadas a Muerte Celular , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Moleculares , Fosforilación , Conformación Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína bcl-X/química , Proteína bcl-X/genética
10.
Cell Death Discov ; 7(1): 320, 2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34716312

RESUMEN

Perturbations to cellular homeostasis, including reduction of the cholesterol level, induce autophagy, a self-digestion process of cellular constituents through an autophagosomal-lysosomal pathway. In accord with its function as a membrane organizer and metabolic sentinel, the cellular response to cholesterol depletion comprises multiple phenomena, including the activation of transcriptional responses, accumulation of reactive oxygen species (ROS), and activation of stress-related signaling pathways. However, the molecular mechanisms by which cholesterol depletion regulates autophagy and the putative involvement of transcriptional responses, ROS and/or stress-related signaling in autophagy regulation in this biological context are not fully understood. Here, we find that cholesterol depletion regulates autophagy at three different levels. First, employing RNA-seq, we show that cholesterol depletion increases the expression of autophagy-related genes independent of ROS or JNK activity. Second, analysis of LC3 lipidation and intracellular localization, and of p62 levels and degradation kinetics, reveals that cholesterol depletion mediates autophagy induction while interfering with autophagic flux. Of note, only the latter depends on ROS accumulation and JNK activity. In view of the common use of cholesterol-reducing drugs as therapeutic agents, our findings have important implications for multiple cellular settings in which autophagy plays a prominent role.

11.
J Cell Biochem ; 110(5): 1262-71, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564222

RESUMEN

Autophagy, a process of self-digestion of cellular constituents, regulates the balance between protein synthesis and protein degradation. Beclin 1 represents an important component of the autophagic machinery. It interacts with proteins that positively regulate autophagy, such as Vps34, UVRAG, and Ambra1, as well as with anti-apoptotic proteins such as Bcl-2 via its BH3-like domain to negatively regulate autophagy. Thus, Beclin 1 interactions with several proteins may regulate autophagy. To identify novel Beclin 1 interacting proteins, we utilized a GST-Beclin 1 fusion protein. Using mass spectroscopic analysis, we identified Beclin 1 as a protein that interacts with GST-Beclin 1. Further examination by cross linking and co-immunoprecipitation experiments confirmed that Beclin 1 self-interacts and that the coiled coil and the N-terminal region of Beclin 1 contribute to its oligomerization. Importantly, overexpression of vps34, UVRAG, or Bcl-x(L), had no effect on Beclin 1 self-interaction. Moreover, this self-interaction was independent of autophagy induction by amino acid deprivation or rapamycin treatment. These results suggest that full-length Beclin 1 is a stable oligomer under various conditions. Such an oligomer may provide a platform for further protein-protein interactions.


Asunto(s)
Aminoácidos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Sirolimus/farmacología , Aminoácidos/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Beclina-1 , Sitios de Unión/genética , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo , Proteína bcl-X/metabolismo
12.
ACR Open Rheumatol ; 2(9): 512-524, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32869536

RESUMEN

OBJECTIVE: RasGTPases are master regulators of multiple intracellular signaling cascades. Perturbation of this pathway has been implicated in the pathogenesis of rheumatoid arthritis (RA). In this study we aimed to define the therapeutic potential of a novel RasGTPases inhibitor, farnesylthiosalicylate (FTS), in the preclinical mouse model of collagen-induced arthritis (CIA) and better delineate its immunomodulatory effects both ex vivo and in the mouse. METHODS: We analyzed in vitro the immunomodulatory effects of FTS on various CD4+ T-cell functions such as activation, proliferation, T-helper polarization, and production of proinflammatory cytokines. Using the CIA model, we further determined the efficacy of FTS to inhibit clinical, histopathologic, and diverse immunological outcomes of arthritis. RESULTS: FTS treatment of CD4+ T cells in vitro effectively targeted distinct kinases (extracellular signal-regulated kinase 1/2, p38, protein kinase B/AKT, and mammalian target of rapamycin), the production of interleukin (IL)-17A, IL-22, and granulocyte-macrophage colony-stimulating factor, and Th17 polarization. FTS therapy in the mouse CIA model significantly reduced clinical disease severity and joint inflammation/damage by histology. Importantly, FTS suppressed the in vivo induction of splenic IL-17+ IL-22+ Th17 cells and the secretion of proinflammatory cytokines. The production of pathogenic autoantibodies and their abnormal hyposialylation was significantly attenuated by FTS therapy. Importantly, in vivo generation of collagen type-II specific effector CD4+ T cells was likewise repressed by FTS therapy. CONCLUSION: The RasGTPases inhibitor FTS attenuates the production of proinflammatory cytokines by in vitro-activated T cells and is a potent immunomodulatory compound in the CIA model, primarily targeting the generation of autoreactive Th17 cells and the production of autoantibodies and their subsequent pathogenic hyposialylation.

13.
Cell Death Dis ; 11(7): 578, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709881

RESUMEN

APOE4 is a major risk factor for sporadic Alzheimer's disease; however, it is unclear how it exerts its pathological effects. Others and we have previously shown that autophagy is impaired in APOE4 compared to APOE3 astrocytes, and demonstrated differences in the expression of mitochondrial dynamics proteins in brains of APOE3 and APOE4 transgenic mice. Here, we investigated the effect of APOE4 expression on several aspects of mitochondrial function and network dynamics, including fusion, fission, and mitophagy, specifically in astrocytes. We found that APOE3 and APOE4 astrocytes differ in their mitochondrial dynamics, suggesting that the mitochondria of APOE4 astrocytes exhibit reduced fission and mitophagy. APOE4 astrocytes also show impaired mitochondrial function. Importantly, the autophagy inducer rapamycin enhanced mitophagy and improved mitochondrial functioning in APOE4 astrocytes. Collectively, the results demonstrate that APOE4 expression is associated with altered mitochondrial dynamics, which might lead to impaired mitochondrial function in astrocytes. This, in turn, may contribute to the pathological effects of APOE4 in Alzheimer's disease.


Asunto(s)
Apolipoproteína E4/metabolismo , Astrocitos/metabolismo , Dinámicas Mitocondriales , Apolipoproteína E3/metabolismo , Astrocitos/ultraestructura , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Línea Celular , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Sirolimus/farmacología , Ubiquitinación/efectos de los fármacos
14.
J Alzheimers Dis ; 70(3): 861-875, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31306119

RESUMEN

This study examined the effects of apolipoprotein E4 (APOE4), the most prevalent genetic risk factor for Alzheimer's disease (AD), on proteins involved in mitochondrial dynamics and autophagy, in the hippocampus of targeted replacement mice. Immunohistochemical measurements revealed that the levels of the mitochondrial fusion-mediating protein, MFN1, were higher, whereas those of corresponding fission-regulating protein, DRP-1, were lower in the hippocampus of ApoE4 mice than in the corresponding ApoE3 mice, indicating that APOE4 is associated with increased mitochondrial fusion and decreased fission. A similar ApoE4-driven decrease in DRP-1 was also observed in AD brains. The levels of the mitochondrial proteins COX1 and Tom40, were higher in the ApoE4 mice, which is consistent with the increased fusion. Measurements of the levels of cleaved PINK1 and parkin, which mark and target mitochondria for mitophagic degradation, revealed lower levels of cleaved PINK1, suggesting reduced mitochondrial membrane potential, and higher levels of parkin in the hippocampus of ApoE4 compared with the ApoE3 mice, indicating altered mitophagy. The levels of the ubiquitin-binding scaffold protein, p62/SQSTM1, which directs selected cargo to the autophagosomes, were also higher in the ApoE4 mice. These findings suggest that APOE4 is associated with enhanced mitochondrial fusion and decreased fission. Additionally, the results indicate that mitophagy/autophagy is reduced in ApoE4 mice, resulting in higher levels of proteins such as parkin and p62, which are normally degraded during this process. Taken together, these results suggest a novel mechanism that may underlie the pathological effects of APOE4 and indicate that use of APOE4 genotyping could pave the way for identification of novel APOE4-related therapeutic targets.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E3 , Apolipoproteína E4 , Hipocampo/metabolismo , Dinámicas Mitocondriales/fisiología , Proteínas Mitocondriales/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Autofagia/fisiología , Inmunohistoquímica , Ratones , Mitofagia/fisiología , Neuronas/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
J Neurotrauma ; 35(21): 2581-2590, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29495919

RESUMEN

Neurotrauma causes immediate elevation of extracellular glutamate (Glu) levels, which creates excitotoxicity and facilitates inflammation, glial scar formation, and consequently neuronal death. Finding factors that reduce the inflammatory response and glial scar formation, and increase neuronal survival and neurite outgrowth, are of major importance for improving the outcome after spinal cord injury (SCI). In the present study, we evaluated a new treatment aiming to remove central nervous system (CNS) Glu into the systemic blood circulation by intravenous (IV) administration of blood Glu scavengers (BGS) such as the enzyme recombinant glutamate-oxaloacetate transaminase 1 (rGOT1) and its co-substrate. In this study we induced in mice an SCI (hemisection), and 1 h post-injury started administering BGS treatment for 5 consecutive days. The treatment reduced the expression levels of p-p38, which regulates apoptosis and increased the expression of p-Akt, which mediates cell survival. Moreover, this treatment decreased pro-inflammatory cytokine expression and microglia activation, reduced astrocytes' reactivity, and facilitated expression of radial glia markers such as Pax6 and nestin. BGS treatment increased the survival of neurons at lesion site and enabled axonal regeneration into the injury site. These effects were correlated with improved functional recovery of the left paretic hindlimb. Thus, early pharmacological intervention with BGS following SCI may be neuroprotective and create a pro-regenerative environment by modulating glia cell response. In light of our results, the availability of the method to remove excess Glu from CNS without the need to deliver drugs across the blood-brain barrier (BBB) and with minimal or no adverse effects may provide a major therapeutic asset.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/farmacología , Ácido Glutámico/sangre , Ácido Glutámico/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/sangre , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología
16.
Cell Death Dis ; 9(2): 47, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29352243

RESUMEN

ErbB2, a member of the ErbB family of receptor tyrosine kinases, is an essential player in the cell's growth and proliferation signaling pathways. Amplification or overexpression of ErbB2 is observed in ∼30% of breast cancer patients, and often drives cellular transformation and cancer development. Recently, we have shown that ErbB2 interacts with the nuclear-cytoplasmic shuttling protein nucleolin, an interaction which enhances cell transformation in vitro, and increases mortality risk and disease progression rate in human breast cancer patients. Given these results, and since acquired resistance to anti-ErbB2-targeted therapy is a major obstacle in treatment of breast cancer, we have examined the therapeutic potential of targeting the ErbB2-nucleolin complex. The effect of the nucleolin-specific inhibitor GroA (AS1411) on ErbB2-positive breast cancer was tested in vivo, in a mouse xenograft model for breast cancer; as well as in vitro, alone and in combination with the ErbB2 kinase-inhibitor tyrphostin AG-825. Here, we show that in vivo treatment of ErbB2-positive breast tumor xenografts with GroA reduces tumor size and leads to decreased ErbB2-mediated signaling. Moreover, we found that co-treatment of breast cancer cell lines with GroA and the ErbB2 kinase-inhibitor tyrphostin AG-825 enhances the anti-cancer effects exerted by GroA alone in terms of cell viability, mortality, migration, and invasiveness. We, therefore, suggest a novel therapeutic approach, consisting of combined inhibition of ErbB2 and nucleolin, which has the potential to improve breast cancer treatment efficacy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benzotiazoles/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Oligodesoxirribonucleótidos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Tirfostinos/farmacología , Animales , Aptámeros de Nucleótidos , Benzotiazoles/administración & dosificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Oligodesoxirribonucleótidos/administración & dosificación , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Tirfostinos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Nucleolina
17.
PLoS One ; 12(2): e0171351, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28151959

RESUMEN

High percentage of human cancers involves alteration or mutation in Ras proteins, including the most aggressive malignancies, such as lung, colon and pancreatic cancers. FTS (Salirasib) is a farnesylcysteine mimetic, which acts as a functional Ras inhibitor, and was shown to exert anti-tumorigenic effects in vitro and in vivo. Previously, we have demonstrated that short-term treatment with FTS also induces protective autophagy in several cancer cell lines. Drug resistance is frequently observed in cancer cells exposed to prolonged treatment, and is considered a major cause for therapy inefficiency. Therefore, in the present study, we examined the effect of a prolonged treatment with FTS on drug resistance of HCT-116 human colon cancer cells, and the involvement of autophagy in this process. We found that cells grown in the presence of FTS for 6 months have become resistant to FTS-induced cell growth inhibition and cell death. Furthermore, we discovered that the resistant cells exhibit altered autophagy, reduced apoptosis and changes in Ras-related signaling pathways following treatment with FTS. Moreover we found that while FTS induces an apoptosis-related cleavage of p62, the FTS-resistant cells were more resistant to apoptosis and p62 cleavage.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Farnesol/análogos & derivados , Salicilatos/farmacología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Farnesol/farmacología , Genes ras/efectos de los fármacos , Células HCT116/efectos de los fármacos , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos
18.
Biochem Pharmacol ; 72(4): 427-36, 2006 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-16780807

RESUMEN

Prostate cancer is one of the most frequently diagnosed cancers in human males. Progression of these tumors is facilitated by autocrine/paracrine growth factors which activate critical signaling cascades that promote prostate cancer cell growth, survival and migration. Among these, Ras pathways have a major role. Here we examined the effect of the Ras inhibitor S-trans, trans-farnesylthiosalicylic acid (FTS), on growth and viability of androgen-dependent and androgen-independent prostate cancer cells. FTS downregulated Ras, inhibited signaling to Akt and reduced the levels of cell-cycle regulatory proteins including cyclin D1, p-RB, E2F-1 and cdc42 in LNCaP and PC3 cells. Consequently the anchorage-dependent and anchorage-independent growth of LNCaP and PC3 cells were inhibited. FTS also induced apoptotic cell death which was inhibited by the broad-spectrum caspases inhibitor, Boc-asp-FMK. Our study demonstrated that androgen-dependent and androgen-independent prostate cancer cells require active Ras for growth and survival. Ras inhibition by FTS results in growth arrest and cell death. FTS may be qualified as a potential agent for the treatment of prostate cancer.


Asunto(s)
Andrógenos/fisiología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Farnesol/análogos & derivados , Salicilatos/farmacología , Proteínas ras/antagonistas & inhibidores , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Factor de Transcripción E2F1/metabolismo , Farnesol/farmacología , Humanos , Masculino , Proteína Quinasa de Distrofia Miotónica , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo , Proteínas ras/fisiología
19.
J Mol Neurosci ; 29(3): 257-67, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17085783

RESUMEN

The neuroprotective effects of neuregulin (NRG), a polypeptide growth factor, on 1-methyl-4-phenylpyridinium ion (MPP+)-induced cell death and oxidative stress in PC12-ErbB4 cells were investigated. Treatment of PC12-ErbB4 cells with MPP+ induced cell death that was markedly attenuated by NRG. The PI3K/PKB/Akt and Ras/MapK signaling pathways probably mediate the survival effect of NRG. NRG induces prolonged activation of PKB/Akt and Erk. Moreover, inhibition of the PI3K and MEK activities prevented the NRG-induced survival effect. Overexpression of constitutively active PI3K or H-Ras (12V) inhibited MPP+-mediated cell death. In addition, MPP+- mediated reactive oxygen species (ROS) elevation was also inhibited by NRG. The effect of NRG on ROS levels was blocked by PI3K and MEK inhibitors, indicating that both signaling pathways can regulate the toxic ROS levels induced by MPP+. Taken together, these results indicate that in PC12-ErbB4 cells, the NRG-induced neuroprotective effect from MPP+ treatment, requires PI3K/PKB/Akt and Ras/MapK signaling networks.


Asunto(s)
1-Metil-4-fenilpiridinio/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Receptores ErbB/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Neurregulina-1/farmacología , Células PC12/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/fisiología , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Cromonas/farmacología , Receptores ErbB/efectos de los fármacos , Flavonoides/farmacología , Genes Reporteros , Genes ras , Imidazoles/farmacología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Células PC12/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos adversos , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Piridinas/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptor ErbB-4 , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transfección
20.
Oncotarget ; 7(40): 65320-65334, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27542246

RESUMEN

ErbB2 is an important member of the ErbB family, which activates growth and proliferation signaling pathways. ErbB2 is often overexpressed in various malignancies, especially in breast cancer, and is a common target for anti-cancer drugs. Breast cancer is currently one of the leading mortality causes in women, and acquired resistance to ErbB2-targeted therapies is a major obstacle in its treatment. Thus, understanding ErbB2-mediated signaling is crucial for further development of anti-cancer therapeutics and disease treatment. Previously, we have reported that the ErbB receptors interact with the major nucleolar protein nucleolin. In addition to its function in the nucleoli of cells, nucleolin participates in various cellular processes at the cytoplasm and cell-surface. Deregulated nucleolin is frequently overexpressed on the membrane of cancer cells. Here, we show that nucleolin increases colony formation and anchorage-independent growth of ErbB2-overexpressing cells. Importantly, this enhanced tumorigenicity also occurs in human ErbB2-positive breast cancer patients; namely, nucleolin overexpression in these patients is associated with reduced patient survival rates and increased disease-risk. ErbB2-nucleolin complexes are formed endogenously in both normal and cancer cells, and their effect on tumorigenicity is mediated through activation of ErbB2 signaling. Accordingly, nucleolin inhibition reduces cell viability and ErbB2 activation in ErbB2-positive cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptor ErbB-2/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Carcinogénesis , Adhesión Celular , Procesos de Crecimiento Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Terapia Molecular Dirigida , Fosfoproteínas/genética , Unión Proteica , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Receptor ErbB-2/genética , Transducción de Señal , Análisis de Supervivencia , Nucleolina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA