Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(10)2023 09 22.
Article in English | MEDLINE | ID: mdl-37892117

ABSTRACT

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.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , Autophagy/physiology , Dopaminergic Neurons/metabolism
2.
Cell Death Discov ; 7(1): 320, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34716312

ABSTRACT

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.

3.
ACR Open Rheumatol ; 2(9): 512-524, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32869536

ABSTRACT

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.

4.
Cell Death Dis ; 11(7): 578, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709881

ABSTRACT

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.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/metabolism , Mitochondrial Dynamics , Apolipoprotein E3/metabolism , Astrocytes/ultrastructure , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Line , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Sirolimus/pharmacology , Ubiquitination/drug effects
5.
Drug Dev Res ; 81(2): 184-193, 2020 04.
Article in English | MEDLINE | ID: mdl-31782539

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Gene Regulatory Networks/drug effects , Lysosomes/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy , Clinical Trials as Topic , Gene Expression Regulation/drug effects , Humans , Molecular Targeted Therapy
6.
J Alzheimers Dis ; 70(3): 861-875, 2019.
Article in English | MEDLINE | ID: mdl-31306119

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Apolipoprotein E3 , Apolipoprotein E4 , Hippocampus/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Autophagy/physiology , Immunohistochemistry , Mice , Mitophagy/physiology , Neurons/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
J Neurotrauma ; 35(21): 2581-2590, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29495919

ABSTRACT

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.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/pharmacology , Glutamic Acid/blood , Glutamic Acid/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/blood , Animals , Female , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology
8.
Cell Death Dis ; 9(2): 47, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352243

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzothiazoles/pharmacology , Breast Neoplasms/drug therapy , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Tyrphostins/pharmacology , Animals , Aptamers, Nucleotide , Benzothiazoles/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Oligodeoxyribonucleotides/administration & dosage , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Tyrphostins/administration & dosage , Xenograft Model Antitumor Assays , Nucleolin
9.
J Neurochem ; 143(5): 584-594, 2017 12.
Article in English | MEDLINE | ID: mdl-28921554

ABSTRACT

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.


Subject(s)
Autophagy/drug effects , Dopaminergic Neurons/drug effects , Microglia/drug effects , Phagocytosis/drug effects , Protein Deglycase DJ-1/metabolism , alpha-Synuclein/pharmacology , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Mice, Inbred BALB C , Oxidative Stress/drug effects , Protein Deglycase DJ-1/deficiency , alpha-Synuclein/metabolism
10.
PLoS One ; 12(2): e0171351, 2017.
Article in English | MEDLINE | ID: mdl-28151959

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Farnesol/analogs & derivatives , Salicylates/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Farnesol/pharmacology , Genes, ras/drug effects , HCT116 Cells/drug effects , Humans , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
11.
Oncotarget ; 7(40): 65320-65334, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27542246

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptor, ErbB-2/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Carcinogenesis , Cell Adhesion , Cell Growth Processes , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Molecular Targeted Therapy , Phosphoproteins/genetics , Protein Binding , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Receptor, ErbB-2/genetics , Signal Transduction , Survival Analysis , Nucleolin
12.
J Neurochem ; 138(6): 845-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27399849

ABSTRACT

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.


Subject(s)
ErbB Receptors/agonists , Neuroglia/pathology , Oligodeoxyribonucleotides/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Spinal Cord Injuries/pathology , Animals , Aptamers, Nucleotide , Astrocytes/drug effects , Behavior, Animal/drug effects , Cell Proliferation/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Humans , Immunohistochemistry , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/psychology , Nucleolin
13.
J Alzheimers Dis ; 51(3): 915-27, 2016.
Article in English | MEDLINE | ID: mdl-26923027

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia in elderly. Genetic studies revealed allelic segregation of the apolipoprotein E (ApoE) gene in sporadic AD and in families with higher risk of AD. The mechanisms underlying the pathological effects of ApoE4 are not yet entirely clear. Several studies indicate that autophagy, which plays an important role in degradation pathways of proteins, organelles and protein aggregates, may be impaired in AD. In the present study, we investigated the effects of ApoE4 versus the ApoE3 isoform on the process of autophagy in mouse-derived astrocytes. The results obtained reveal that under several autophagy-inducing conditions, astrocytes expressing ApoE4 exhibit lower autophagic flux compared to astrocytes expressing ApoE3. Using an in situ model, we examined the role of autophagy and the effects thereon of ApoE4 in the elimination of Aß plaques from isolated brain sections of transgenic 5xFAD mice. This revealed that ApoE4 astrocytes eliminate Aß plaques less effectively than the corresponding ApoE3 astrocytes. Additional experiments showed that the autophagy inducer, rapamycin, enhances Aß plaque degradation by ApoE4 astrocytes whereas the autophagy inhibitor, chloroquine, blocks Aß plaque degradation by ApoE3 astrocytes. Taken together, these findings show that ApoE4 impairs autophagy in astrocyte cultures and that this effect is associated with reduced capacity to clear Aß plaques. This suggests that impaired autophagy may play a role in mediating the pathological effects of ApoE4 in AD.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/physiology , Autophagy/physiology , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Astrocytes/drug effects , Astrocytes/pathology , Autophagy/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cells, Cultured , Central Nervous System Agents/pharmacology , Chloroquine/pharmacology , Disease Models, Animal , Humans , Mice, Transgenic , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Sirolimus/pharmacology , Time Factors
14.
Mol Cell Neurosci ; 68: 82-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25936601

ABSTRACT

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.


Subject(s)
Immunosuppressive Agents/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Sirolimus/therapeutic use , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Animals , Astrocytes/drug effects , Astrocytes/physiology , CD11b Antigen/metabolism , Cell Count , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , ELAV-Like Protein 3/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Time Factors
15.
Biol Cell ; 107(5): 130-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25735913

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Farnesol/analogs & derivatives , Neoplasms/drug therapy , Salicylates/pharmacology , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Farnesol/pharmacology , Humans
16.
Oncotarget ; 5(18): 8602-13, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25261371

ABSTRACT

The three oncogenes, ErbB receptors, Ras proteins and nucleolin may contribute to malignant transformation. Previously, we demonstrated that nucleolin could bind both Ras protein and ErbB receptors. We also showed that the crosstalk between the three proteins facilitates anchorage independent growth and tumor growth in nude mice, and that inhibition of this interaction in prostate and colon cancer cells reduces tumorigenicity. In the present study, we show that treatment with Ras and nucleolin inhibitors reduces the oncogenic effect induced by ErbB1 receptor in U87-MG cells. This combined treatment enhances cell death, reduces cell proliferation and cell migration. Moreover, we demonstrate a pivotal role of nucleolin in ErbB1 activation by its ligand. Nucleolin inhibitor prevents EGF-induced receptor activation and its downstream signaling followed by reduced proliferation. Furthermore, inhibition of Ras by Salirasib (FTS), mainly reduces cell viability and motility. The combined treatment, which targets both Ras and nucleolin, additively reduces tumorigenicity both in vitro and in vivo. These results suggest that targeting both nucleolin and Ras may represent an additional opportunity for inhibiting cancers, including glioblastoma, that are driven by these oncogenes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Animals , Aptamers, Nucleotide , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Farnesol/administration & dosage , Farnesol/analogs & derivatives , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice, Nude , Oligodeoxyribonucleotides/administration & dosage , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor Cross-Talk , Salicylates/administration & dosage , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , ras Proteins/metabolism , Nucleolin
17.
Growth Factors ; 32(2): 74-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24641597

ABSTRACT

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.


Subject(s)
ErbB Receptors/metabolism , ErbB Receptors/therapeutic use , Neoplasms/drug therapy , Animals , Humans , Membrane Proteins/metabolism , Mice , Protein Binding , Protein Conformation , Protein Transport , Receptor, ErbB-2/metabolism , Signal Transduction
18.
Oncotarget ; 5(3): 577-86, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24583697

ABSTRACT

Autophagy, a process of self-degradation and turnover of cellular components, plays a complex role in cancer. Evidence exists to show that autophagy may support tumor growth and cell survival, whereas it can also contribute to tumor suppression and have anti-survival characteristics in different cellular systems. Numerous studies have described the effects of various oncogenes and tumor suppressors on autophagy. The small GTPase Ras is an oncogene involved in the regulation of various cell-signaling pathways, and is mutated in 33% of human cancers. In the present review, we discuss the interplay between Ras and autophagy in relation to oncogenesis. It appears that Ras can upregulate or downregulate autophagy through several signaling pathways. In turn, autophagy can affect the tumorigenicity driven by Ras, resulting in either tumor progression or repression, depending on the cellular context. Furthermore, Ras inhibitors were shown to induce autophagy in several cancer cell lines.


Subject(s)
Autophagy/genetics , Genes, ras , Neoplasms/genetics , Neoplasms/therapy , Animals , Cell Transformation, Neoplastic/genetics , Humans , Neoplasms/pathology , Signal Transduction
19.
Oncotarget ; 5(1): 173-84, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24368422

ABSTRACT

The Ras family of small GTPases transmits extracellular signals that regulate cell growth, differentiation, motility and death. Ras signaling is constitutively active in a large number of human cancers. Ras can also regulate autophagy by affecting several signaling pathways including the mTOR pathway. Autophagy is a process that regulates the balance between protein synthesis and protein degradation. It is important for normal growth control, but may be defective in diseases. Previously, we have shown that Ras inhibition by FTS induces autophagy, which partially protects cancer cells and may limit the use of FTS as an anti-cancer drug. Since FTS is a non toxic drug we hypothesized that FTS and chloroquine (an autophagy inhibitor) will synergize in cell growth inhibition and cell death. Thus, in the present study, we explored the mechanism of each individual drug and their combined action. Our results demonstrate that in HCT-116 and in Panc-1 cells, FTS induces autophagy, which can be inhibited by chloroquine. Furthermore, the combined treatment synergistically decreased the number of viable cells. Interestingly, the combined treatment enhanced apoptotic cell death as indicated by increased sub-G1 cell population, increased Hoechst staining, activation of caspase 3, decrease in survivin expression and release of cytochrome c. Thus, chloroquine treatment may promote FTS-mediated inhibition of tumor cell growth and may stimulate apoptotic cell death.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chloroquine/pharmacology , Farnesol/analogs & derivatives , Salicylates/pharmacology , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Chloroquine/administration & dosage , Farnesol/administration & dosage , Farnesol/pharmacology , HCT116 Cells , Humans , Rats , Salicylates/administration & dosage , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism
20.
J Virol ; 87(24): 13397-408, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089565

ABSTRACT

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.


Subject(s)
Apoptosis , Autophagy , Cattle Diseases/physiopathology , Hemorrhagic Disease Virus, Epizootic/physiology , Reoviridae Infections/veterinary , Sheep Diseases/physiopathology , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/metabolism , Cattle Diseases/virology , Cell Line , Hemorrhagic Disease Virus, Epizootic/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Protein Biosynthesis , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reoviridae Infections/metabolism , Reoviridae Infections/physiopathology , Reoviridae Infections/virology , Sheep , Sheep Diseases/genetics , Sheep Diseases/metabolism , Sheep Diseases/virology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...