Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurochem ; 157(4): 1284-1299, 2021 05.
Article in English | MEDLINE | ID: mdl-33180957

ABSTRACT

Diminished glutamate (Glu) uptake via the excitatory amino acid transporter EAAT2, which normally accounts for ~90% of total forebrain EAAT activity, may contribute to neurodegeneration via Glu-mediated excitotoxicity. C-terminal cleavage by caspase-3 (C3) was reported to mediate EAAT2 inactivation and down-regulation in the context of neurodegeneration. For a detailed analysis of C3-dependent EAAT2 degradation, we employed A172 glioblastoma as well as hippocampal HT22 cells and murine astrocytes over-expressing VSV-G-tagged EAAT2 constructs. C3 activation was induced by staurosporine (STR). In HT22 cells, STR-induced C3 activation-induced rapid EAAT2 protein degradation. The mutation of asparagine 504 to aspartate (D504N), which should inactivate the putative C3 cleavage site, increased EAAT2 activity in A172 cells. In contrast, the D504N mutation did not protect EAAT2 protein against STR-induced degradation in HT22 cells, whereas inhibition of caspases, ubiquitination and the proteasome did. Similar results were obtained in astrocytes. Phylogenetic analysis showed that C-terminal ubiquitin acceptor sites-but not the putative C3 cleavage site-exhibit a high degree of conservation. Moreover, C-terminal truncation mimicking C3 cleavage increased rather than decreased EAAT2 activity and stability as well as protected EAAT2 against STR-induced ubiquitination-dependent degradation. We conclude that cellular stress associated with endogenous C3 activation degrades EAAT2 via a pathway involving ubiquitination and the proteasome but not direct C3-mediated cleavage. In addition, C3 cleavage of EAAT2, described to occur in other models, is unlikely to inactivate EAAT2. However, mutation of the highly conserved D504 within the putative C3 cleavage site increases EAAT2 activity via an unknown mechanism.


Subject(s)
Caspase 3/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Nerve Degeneration/metabolism , Proteasome Endopeptidase Complex/metabolism , Stress, Physiological/physiology , Animals , Cells, Cultured , Enzyme Inhibitors/toxicity , Humans , Mice , Staurosporine/toxicity , Ubiquitination
2.
Cereb Cortex ; 29(6): 2701-2715, 2019 06 01.
Article in English | MEDLINE | ID: mdl-29982364

ABSTRACT

Excessive excitation has been hypothesized to subsume a significant part of the acute damage occurring after traumatic brain injury (TBI). However, reduced neuronal excitability, loss of neuronal firing, and a disturbed excitation/inhibition balance have been detected. Parvalbumin (PV) interneurons are major regulators of perisomatic inhibition, principal neurons firing, and overall cortical excitability. However, their role in acute TBI pathogenic cascades is unclear. We exploited the chemogenetic Pharmacologically Selective Activation Module and Pharmacologically Selective Effector Module control of PV-Cre+ neurons and the Designer Receptors Exclusively Activated by Designer Drug (DREADD) control of principal neurons in a blunt model of TBI to explore the role of inhibition in shaping neuronal vulnerability to TBI. We demonstrated that inactivation of PV interneurons at the instance or soon after trauma enhances survival of principal neurons and reduces gliosis at 7 dpi whereas, activation of PV interneurons decreased neuronal survival. The protective effect of PV inactivation was suppressed by expressing the nuclear calcium buffer PV-nuclear localisation sequence in principal neurons, implying an activity-dependent neuroprotective signal. In fact, protective effects were obtained by increasing the excitability of principal neurons directly using DREADDs. Thus, we show that sustaining neuronal excitation in the early phases of TBI may reduce neuronal vulnerability by increasing activity-dependent survival, while excess activation of perisomatic inhibition is detrimental to neuronal integrity.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Interneurons/physiology , Animals , Mice , Neurons/physiology , Parvalbumins/metabolism
3.
J Neurotrauma ; 35(22): 2718-2735, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29774782

ABSTRACT

Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.


Subject(s)
Alcoholic Intoxication/metabolism , Brain Injuries, Traumatic/metabolism , ErbB Receptors/drug effects , Ethanol/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/physiopathology , ErbB Receptors/metabolism , Interneurons/drug effects , Interneurons/metabolism , Mice , Signal Transduction/drug effects
4.
Exp Neurol ; 299(Pt A): 15-25, 2018 01.
Article in English | MEDLINE | ID: mdl-28941811

ABSTRACT

Traumatic brain injury (TBI) has been proposed as a risk factor for neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). To determine whether TBI might trigger or exacerbate ALS-relevant pathology, we delivered a mild stab-wound injury to the motor cortex of three different ALS mouse models expressing mutations in SOD1, TDP-43 or FUS and scrutinized the effects on the formation of phospho-TDP-43 (pTDP-43) cytoplasmic granules. Stab-injury induced the formation of cytoplasmic TDP-43 granules in wt animals, peaking at 3dpi; a much larger response was seen in mutant TDP-43 mice, whose response peaked at 7dpi. The pTDP-43 granules did not colocalize with the stress markers TIAR-1 and FUS but colocalized with FMRP (35%) and with p62 (65%), suggesting their involvement in transport granules and their clearance by autophagy. A similar, albeit smaller effect, was seen in mutant FUS mice. In the SOD1G93A mouse model, neither increase in pTDP-43 granules nor in SOD1 aggregates were detected. In all cases, pTDP-43 granules were cleared and the number of pTDP-43-positive neurons returned to baseline by 40dpi. Neither injury-related neuronal loss nor motor performance or survival was significantly different in transgenic mice receiving injury vs sham mice. Thus, trauma can trigger ALS-related TDP-43 pathology, the extent of which is modulated by ALS-related mutations. However, the pathological findings prove reversible and do not affect disease progression and neuronal vulnerability.


Subject(s)
Brain Injuries, Traumatic/pathology , DNA-Binding Proteins/metabolism , Motor Cortex/pathology , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy/genetics , Behavior, Animal , Brain Injuries, Traumatic/psychology , Cytoplasmic Granules/pathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Transgenic , Motor Cortex/injuries , Motor Neurons/pathology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
5.
Int Immunopharmacol ; 51: 66-75, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28806641

ABSTRACT

Ethanol intoxication is a common comorbidity in traumatic brain injury. To date, the effect of ethanol on TBI pathogenic cascades and resulting outcomes remains debated. A closed blunt weight-drop murine TBI model has been implemented to investigate behavioral (by sensorimotor and neurological tests), and neuro-immunological (by tissue cytokine arrays and immuno-histology) effects of ethanol intoxication on TBI. The effect of the occurrence of traumatic intracerebral hemorrhage was also studied. The results indicate that ethanol pretreatment results in a faster and better recovery after TBI with reduced infiltration of leukocytes and reduced microglia activation. These outcomes correspond to reduced parenchymal levels of GM-CSF, IL-6 and IL-3 and to the transient upregulation of IL-13 and VEGF, indicating an early shift in the cytokine profile towards reduced inflammation. A significant difference in the cytokine profile was still observed 24h post injury in the ethanol pretreated mice, as shown by the delayed peak in IL-6 and by the suppression of GM-CSF, IFN-γ, and IL-3. Seven days post-injury, ethanol-pretreated mice displayed a significant decrease both in CD45+ cells infiltration and in microglial activation. On the other hand, in the case of traumatic intracerebral hemorrhage, the cytokine profile was dominated by KC, CCL5, M-CSF and several interleukins and ethanol pretreatment did not produce any modification. We can thus conclude that ethanol intoxication suppresses the acute neuro-inflammatory response to TBI, an effect which is correlated with a faster and complete neurological recovery, whereas, the presence of traumatic intracerebral hemorrhage overrides the effects of ethanol.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Injuries, Traumatic/drug therapy , Cerebral Hemorrhage, Traumatic/drug therapy , Ethanol/therapeutic use , Leukocytes/immunology , Microglia/immunology , Neurogenic Inflammation/drug therapy , Administration, Oral , Animals , Cell Movement , Cytokines/metabolism , Disease Models, Animal , Feedback, Sensory , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Tissue Array Analysis
6.
Aging (Albany NY) ; 8(2): 382-93, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26978042

ABSTRACT

Telomere shortening has been linked to a variety of neurodegenerative diseases. Recent evidence suggests that reduced telomerase expression results in shorter telomeres in leukocytes from sporadic patients with amyotrophic lateral sclerosis (ALS) compared with healthy controls. Here, we have characterized telomere length in microglia, astroglia and neurons in human post mortem brain tissue from ALS patients and healthy controls. Moreover, we studied the consequences of telomerase deletion in a genetic mouse model for ALS. We found a trend towards longer telomeres in microglia in the brains of ALS patients compared to non-neurologic controls. Knockout of telomerase leading to telomere shortening accelerated the ALS phenotype inSOD1G93A-transgenic mice. Our results suggest that telomerase dysfunction might contribute to the age-related risk for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , Telomere Shortening/physiology , Telomere/pathology , Age of Onset , Animals , Astrocytes/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Knockout , Mice, Transgenic , Microglia/pathology , Neurons/pathology
7.
Hum Mol Genet ; 24(8): 2228-40, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25552654

ABSTRACT

Mutations in components of the molecular motor dynein/dynactin lead to neurodegenerative diseases of the motor system or atypical parkinsonism. These mutations are associated with prominent accumulation of vesicles involved in autophagy and lysosomal pathways, and with protein inclusions. Whether alleviating these defects would affect motor symptoms remain unknown. Here, we show that a mouse model expressing low levels of disease linked-G59S mutant dynactin p150(Glued) develops motor dysfunction >8 months before loss of motor neurons or dopaminergic degeneration is observed. Abnormal accumulation of autophagosomes and protein inclusions were efficiently corrected by lowering dietary protein content, and this was associated with transcriptional upregulations of key players in autophagy. Most importantly this dietary modification partially rescued overall neurological symptoms in these mice after onset. Similar observations were made in another mouse strain carrying a point mutation in the dynein heavy chain gene. Collectively, our data suggest that stimulating the autophagy/lysosomal system through appropriate nutritional intervention has significant beneficial effects on motor symptoms of dynein/dynactin diseases even after symptom onset.


Subject(s)
Dietary Proteins/metabolism , Dyneins/genetics , Microtubule-Associated Proteins/genetics , Mutation, Missense , Nerve Degeneration/diet therapy , Nerve Degeneration/metabolism , Animals , Autophagy , Disease Models, Animal , Dynactin Complex , Dyneins/metabolism , Female , Humans , Male , Mice , Mice, Inbred C3H , Microtubule-Associated Proteins/metabolism , Motor Activity , Motor Neurons/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...