Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 24(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36982965

ABSTRACT

Mitochondria-nucleus communication during stress dictates cellular fate with consequences on the etiopathology of multiple age-related diseases. Impaired mitochondrial quality control through loss of function of the mitochondrial protease HtrA2 associates with accumulation of damaged mitochondria and triggers the integrated stress response, implicating the transcription factor CHOP. Here we have employed a combined model of impaired mitochondria quality control, namely HtrA2 loss of function, and/or integrated stress response, namely CHOP loss of function, and genotoxicity to address the distinctive roles of these cellular components in modulating intracellular and intercellular responses. The genotoxic agents employed were cancer therapeutic agents such as irradiation with X-ray and protons or treatment with the radiomimetic bleomycin. The irradiation had an enhanced effect in inducing DNA damage in cells with CHOP loss of function, while the bleomycin treatment induced more DNA damage in all the transgenic cells as compared to the control. The genetic modifications impaired the transmission of DNA damage signalling intercellularly. Furthermore, we have dissected the signalling pathways modulated by irradiation in selected genotypes with RNA sequencing analysis. We identified that loss of HtrA2 and CHOP function, respectively, lowers the threshold where irradiation may induce the activation of innate immune responses via cGAS-STING; this may have a significant impact on decisions for combined therapeutic approaches for various diseases.


Subject(s)
Mitochondria , Signal Transduction , Mitochondria/metabolism , Cell Nucleus/metabolism , Membrane Proteins/metabolism , DNA Damage , DNA, Mitochondrial/metabolism
3.
Neuro Oncol ; 22(4): 493-504, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31883012

ABSTRACT

BACKGROUND: Malignant gliomas are the most frequent primary brain tumors and remain among the most incurable cancers. Although the role of the gap junction protein, connexin43 (Cx43), has been deeply investigated in malignant gliomas, no compounds have been reported with the ability to recapitulate the tumor suppressor properties of this protein in in vivo glioma models. METHODS: TAT-Cx43266-283 a cell-penetrating peptide which mimics the effect of Cx43 on c-Src inhibition, was studied in orthotopic immunocompetent and immunosuppressed models of glioma. The effects of this peptide in brain cells were also analyzed. RESULTS: While glioma stem cell malignant features were strongly affected by TAT-Cx43266-283, these properties were not significantly modified in neurons and astrocytes. Intraperitoneally administered TAT-Cx43266-283 decreased the invasion of intracranial tumors generated by GL261 mouse glioma cells in immunocompetent mice. When human glioma stem cells were intracranially injected with TAT-Cx43266-283 into immunodeficient mice, there was reduced expression of the stemness markers nestin and Sox2 in human glioma cells at 7 days post-implantation. Consistent with the role of Sox2 as a transcription factor required for tumorigenicity, TAT-Cx43266-283 reduced the number and stemness of human glioma cells at 30 days post-implantation. Furthermore, TAT-Cx43266-283 enhanced the survival of immunocompetent mice bearing gliomas derived from murine glioma stem cells. CONCLUSION: TAT-Cx43266-283 reduces the growth, invasion, and progression of malignant gliomas and enhances the survival of glioma-bearing mice without exerting toxicity in endogenous brain cells, which suggests that this peptide could be considered as a new clinical therapy for high-grade gliomas.


Subject(s)
Brain Neoplasms , Glioma , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Connexin 43 , Disease Models, Animal , Glioma/drug therapy , Mice , Peptides
4.
J Alzheimers Dis ; 65(4): 1109-1124, 2018.
Article in English | MEDLINE | ID: mdl-30103329

ABSTRACT

Amyloid-ß (Aß) peptides, Aß40, Aß42, and recently Aß25 - 35, have been directly implicated in the pathogenesis of Alzheimer's disease (AD). We have previously shown that all three peptides decrease neuronal viability, but Aß40 also promotes synaptic disassembling. In this work, we have studied the effects of these peptides on astrocytes in primary culture and found that the three Aß peptides were internalized by astrocytes and significantly decreased astrocyte viability, while increasing ROS production. Aß peptide internalization is temperature-dependent, a fact that supports the idea that Aß peptides are actively endocytosed by astrocytes. However, inhibiting caveolae formation by methyl-beta-cyclodextrin or by silencing caveolin-1 with RNA interference did not prevent Aß endocytosis, which suggests that Aß peptides do not use caveolae to enter astrocytes. Conversely, inhibition of clathrin-coated vesicle formation by chlorpromazine or by silencing clathrin with RNA interference significantly decreased Aß internalization and partially reverted the decrease of astrocyte viability caused by the presence of Aß. These results suggest that Aß is endocytosed by clathrin-coated vesicles in astrocytes. Aß-loaded astrocytes, when co-incubated with non-treated astrocytes in separate wells but with the same incubation medium, promoted cell death in non-treated astrocytes; a fact that was associated with the presence of Aß inside previously unloaded astrocytes. This phenomenon was inhibited by the presence of chlorpromazine in the co-incubation medium. These results suggest that astrocyte may perform Aß transcytosis, a process that could play a role in the clearance of Aß peptides from the brain to cerebrospinal fluid.


Subject(s)
Amyloid beta-Peptides/pharmacology , Astrocytes/drug effects , Endocytosis/drug effects , Transcytosis/drug effects , Amyloid beta-Peptides/metabolism , Animals , Animals, Newborn , Antipsychotic Agents/pharmacology , Brain/cytology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Survival/drug effects , Cells, Cultured , Chlorpromazine/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Peptides, Cyclic/genetics , Peptides, Cyclic/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Temperature , Transfection , beta-Cyclodextrins/pharmacology
5.
Front Mol Neurosci ; 10: 418, 2017.
Article in English | MEDLINE | ID: mdl-29326548

ABSTRACT

The non-receptor tyrosine kinase c-Src is an important mediator in several signaling pathways related to neuroinflammation. Our previous study showed that cortical injection of kainic acid (KA) promoted a transient increase in c-Src activity in reactive astrocytes surrounding the neuronal lesion. As a cell-penetrating peptide based on connexin43 (Cx43), specifically TAT-Cx43266-283, inhibits Src activity, we investigated the effect of TAT-Cx43266-283 on neuronal death promoted by cortical KA injections in adult mice. As expected, KA promoted neuronal death, estimated by the reduction in NeuN-positive cells and reactive gliosis, characterized by the increase in glial fibrillary acidic protein (GFAP) expression. Interestingly, TAT-Cx43266-283 injected with KA diminished neuronal death and reactive gliosis compared to KA or KA+TAT injections. In order to gain insight into the neuroprotective mechanism, we used in vitro models. In primary cultured neurons, TAT-Cx43266-283 did not prevent neuronal death promoted by KA, but when neurons were grown on top of astrocytes, TAT-Cx43266-283 prevented neuronal death promoted by KA. These observations demonstrate the participation of astrocytes in the neuroprotective effect of TAT-Cx43266-283. Furthermore, the neuroprotective effect was also present in non-contact co-cultures, suggesting the contribution of soluble factors released by astrocytes. As glial hemichannel activity is associated with the release of several factors, such as ATP and glutamate, that cause neuronal death, we explored the participation of these channels on the neuroprotective effect of TAT-Cx43266-283. Our results confirmed that inhibitors of ATP and NMDA receptors prevented neuronal death in co-cultures treated with KA, suggesting the participation of astrocyte hemichannels in neurotoxicity. Furthermore, TAT-Cx43266-283 reduced hemichannel activity promoted by KA in neuron-astrocyte co-cultures as assessed by ethidium bromide (EtBr) uptake assay. In fact, TAT-Cx43266-283 and dasatinib, a potent c-Src inhibitor, strongly reduced the activation of astrocyte hemichannels. In conclusion, our results suggest that TAT-Cx43266-283 exerts a neuroprotective effect through the reduction of hemichannel activity likely mediated by c-Src in astrocytes. These data unveil a new role of c-Src in the regulation of Cx43-hemichannel activity that could be part of the mechanism by which astroglial c-Src participates in neuroinflammation.

6.
J Alzheimers Dis ; 55(1): 171-182, 2017.
Article in English | MEDLINE | ID: mdl-27662292

ABSTRACT

Amyloid-ß (Aß), Aß40, Aß42, and, recently, Aß25-35 have been directly implicated in the pathogenesis of Alzheimer's disease. We have studied the effects of Aß on neuronal death, reactive oxygen species (ROS) production, and synaptic assembling in neurons in primary culture. Aß25-35, Aß40, and Aß42 significantly decreased neuronal viability, although Aß25-35 showed a higher effect. Aß25-35 showed a more penetrating ability to reach mitochondria while Aß40 did not enter the neuronal cytosol and Aß42 was scarcely internalized. We did not observe a direct correlation between ROS production and cell death because both Aß40 and Aß42 decreased neuronal viability but Aß40 did not change ROS production. Rather, ROS production seems to correlate with the penetrating ability of each Aß. No significant differences were found between Aß40 and Aß42 regarding the extent of the deleterious effects of both peptides on neuronal viability or synaptophysin expression. However, Aß40 elicited a clear delocalization of PSD-95 and synaptotagmin from prospective synapsis to the neuronal soma, suggesting the occurrence of a crucial effect of Aß40 on synaptic disassembling. The formation of Aß40- or Aß42-serum albumin complexes avoided the effects of these peptides on neuronal viability, synaptophysin expression, and PSD-95/synaptotagmin disarrangement suggesting that sequestration of Aß by albumin prevents deleterious effects of these peptides. We can conclude that Aß borne by albumin can be safely transported through body fluids, a fact that may be compulsory for Aß disposal by peripheral tissues.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Serum Albumin, Human/metabolism , Alzheimer Disease/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Cells, Cultured , Cytosol/metabolism , Cytosol/pathology , Disks Large Homolog 4 Protein/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Neurons/pathology , Rats, Wistar , Reactive Oxygen Species/metabolism , Synaptophysin/metabolism , Synaptotagmins/metabolism
7.
Oncotarget ; 7(31): 49819-49833, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27391443

ABSTRACT

Connexin43 (Cx43), the major protein forming gap junctions in astrocytes, is reduced in high-grade gliomas, where its ectopic expression exerts important effects, including the inhibition of the proto-oncogene tyrosine-protein kinase Src (c-Src). In this work we aimed to investigate the mechanism responsible for this effect. The inhibition of c-Src requires phosphorylation at tyrosine 527 mediated by C-terminal Src kinase (Csk) and dephosphorylation at tyrosine 416 mediated by phosphatases, such as phosphatase and tensin homolog (PTEN). Our results showed that the antiproliferative effect of Cx43 is reduced when Csk and PTEN are silenced in glioma cells, suggesting the involvement of both enzymes. Confocal microscopy and immunoprecipitation assays confirmed that Cx43, in addition to c-Src, binds to PTEN and Csk in glioma cells transfected with Cx43 and in astrocytes. Pull-down assays showed that region 266-283 in Cx43 is sufficient to recruit c-Src, PTEN and Csk and to inhibit the oncogenic activity of c-Src. As a result of c-Src inhibition, PTEN was increased with subsequent inactivation of Akt and reduction of proliferation of human glioblastoma stem cells. We conclude that the recruitment of Csk and PTEN to the region between residues 266 and 283 within the C-terminus of Cx43 leads to c-Src inhibition.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Connexin 43/metabolism , Glioma/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/cytology , Phosphorylation , Prosencephalon/cytology , Protein Domains , Proto-Oncogene Mas , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Wistar , Tyrosine/chemistry
8.
Brain Res ; 1570: 13-25, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24836198

ABSTRACT

Our previous work has shown that oleic acid synthesized by astrocytes in response to serum albumin behaves as a neurotrophic factor in neurons, upregulating the expression of GAP-43 and MAP-2 proteins, which are respectively markers of axonal and dendrite growth. In addition, oleic acid promoted neuron migration and aggregation, resulting in clusters of neurons connected each other by the newly formed neurites. In this work we show that the presence of albumin or albumin plus oleic acid increases neuron migration in cultured explants of the lateral periventricular zone, resulting in an increase in the number of GAP-43-positive neurons leaving the explant. Upon silencing stearoyl-CoA desaturase-1 (SCD-1), a key enzyme in oleic acid synthesis by RNA of interference mostly prevented the effect of albumin but not that of albumin plus oleic acid, suggesting that the oleic acid synthesized due to the effect of albumin would be responsible for the increase in neuron migration. Oleic acid increased doublecortin (DCX) expression in cultured neurons, explants and organotypic slices, suggesting that DCX may mediate in the effect of oleic acid on neuron migration. The effect of oleic acid on neuron migration may be destined for the formation of synapses because the presence of oleic acid increased the expression of synaptotagmin and that of postsynaptic density protein (PDS-95), respectively markers of the pre- and postsynaptic compartments. In addition, confocal microscopy revealed the occurrence of points of colocalization between synaptotagmin and PDS-95, which is consistent with the idea that oleic acid promotes synapse arrangement.


Subject(s)
Brain/growth & development , Brain/physiology , Neurons/physiology , Oleic Acid/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Astrocytes/physiology , Axons/physiology , Cell Movement/physiology , Cells, Cultured , Disks Large Homolog 4 Protein , Doublecortin Domain Proteins , Doublecortin Protein , GAP-43 Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neurites/physiology , Neuropeptides/metabolism , Rats, Wistar , Serum Albumin/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Synapses/physiology , Synaptotagmins/metabolism , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...