Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 13(4): 381, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444186

ABSTRACT

The pathogenic mechanisms that underlie the progression of remote degeneration after spinal cord injury (SCI) are not fully understood. In this study, we examined the relationship between endoplasmic reticulum (ER) stress and macroautophagy, hereafter autophagy, and its contribution to the secondary damage and outcomes that are associated with remote degeneration after SCI. Using a rat model of spinal cord hemisection at the cervical level, we measured ER stress and autophagy markers in the axotomized neurons of the red nucleus (RN). In SCI animals, mRNA and protein levels of markers of ER stress, such as GRP78, CHOP, and GADD34, increased 1 day after the injury, peaking on Day 5. Notably, in SCI animals, the increase of ER stress markers correlated with a blockade in autophagic flux, as evidenced by the increase in microtubule-associated protein 2 light chain 3 (LC3-II) and p62/SQSTM1 (p62) and the decline in LAMP1 and LAMP2 levels. After injury, treatment with guanabenz protected neurons from UPR failure and increased lysosomes biogenesis, unblocking autophagic flux. These effects correlated with greater activation of TFEB and improved neuronal survival and functional recovery-effects that persisted after suspension of the treatment. Collectively, our results demonstrate that in remote secondary damage, impairments in autophagic flux are intertwined with ER stress, an association that contributes to the apoptotic cell death and functional damage that are observed after SCI.


Subject(s)
Autophagosomes , Spinal Cord Injuries , Animals , Apoptosis , Autophagosomes/metabolism , Autophagy , Endoplasmic Reticulum Stress , Proteostasis , Rats , Spinal Cord/pathology , Spinal Cord Injuries/pathology
2.
Mol Neurobiol ; 54(7): 4896-4907, 2017 09.
Article in English | MEDLINE | ID: mdl-27514755

ABSTRACT

Spinal cord injuries (SCIs) are devastating conditions of the central nervous system (CNS) for which there are no restorative therapies. Neuronal death at the primary lesion site and in remote regions that are functionally connected to it is one of the major contributors to neurological deficits following SCI.Disruption of autophagic flux induces neuronal death in many CNS injuries, but its mechanism and relationship with remote cell death after SCI are unknown. We examined the function and effects of the modulation of autophagy on the fate of axotomized rubrospinal neurons in a rat model of spinal cord dorsal hemisection (SCH) at the cervical level. Following SCH, we observed an accumulation of LC3-positive autophagosomes (APs) in the axotomized neurons 1 and 5 days after injury. Furthermore, this accumulation was not attributed to greater initiation of autophagy but was caused by a decrease in AP clearance, as demonstrated by the build-up of p62, a widely used marker of the induction of autophagy. In axotomized rubrospinal neurons, the disruption of autophagic flux correlated strongly with remote neuronal death and worse functional recovery. Inhibition of AP biogenesis by 3-methyladenine (3-MA) significantly attenuated remote degeneration and improved spontaneous functional recovery, consistent with the detrimental effects of autophagy in remote damage after SCH. Collectively, our results demonstrate that autophagic flux is blocked in axotomized neurons on SCI and that the inhibition of AP formation improves their survival. Thus, autophagy is a promising target for the development of therapeutic interventions in the treatment of SCIs.


Subject(s)
Autophagy , Neurons , Spinal Cord Injuries/pathology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Disease Models, Animal , Lysosomes/drug effects , Lysosomes/metabolism , Male , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Wistar , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy
3.
Exp Neurol ; 287(Pt 1): 21-33, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27771352

ABSTRACT

The presence of α-synuclein (α-syn) in Lewy bodies and Lewy neurites is an important characteristic of the neurodegenerative processes of substantia nigra pars compacta (SNpc) dopaminergic (DAergic) neurons in Parkinson's disease (PD) and other synucleinopathies. Here we report that Berlin-Druckrey rats carrying a spontaneous mutation in the 3' untranslated region of α-syn mRNA (m/m rats) display a marked accumulation of α-syn in the mesencephalic area, striatum and frontal cortex, accompanied to severe dysfunctions in the dorsolateral striatum. Despite a small reduction in the number of SNpc and ventral tegmental area DAergic cells, the surviving dopaminergic neurons of the m/m rats do not show clear-cut alterations of the spontaneous and evoked firing activity, DA responses and somatic amphetamine-induced firing inhibition. Interestingly, mutant DAergic neurons display diminished whole-cell Ih conductance and a reduced frequency of spontaneous excitatory synaptic currents. By contrast, m/m rats show a severe impairment of DA and glutamate release in the dorsolateral striatum, as revealed by amperometric measure of DA currents and by electrophysiological recordings of glutamatergic synaptic events in striatal medium spiny neurons. These functional impairments are paralleled by a decreased expression of the DA transporter and VGluT1 proteins in the same area. Thus, together with α-syn overload in the mesencephalic region, striatum and frontal cortex, the main functional alterations occur in the DAergic and glutamatergic terminals in the dorsal striatum of the m/m rats.


Subject(s)
Dopaminergic Neurons/physiology , Glutamic Acid/metabolism , Membrane Potentials/physiology , Mesencephalon/cytology , alpha-Synuclein/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Cell Count , Dopamine/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABA-B Receptor Agonists/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Rats , Synaptic Potentials/drug effects , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , alpha-Synuclein/genetics
4.
J Neuroinflammation ; 13(1): 150, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27301743

ABSTRACT

BACKGROUND: After focal brain injuries occur, in addition to the effects that are attributable to the primary site of damage, the resulting functional impairments depend highly on changes that occur in regions that are remote but functionally connected to the site of injury. Such effects are associated with apoptotic and inflammatory cascades and are considered to be important predictors of outcome. Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive technique that is used to treat various central nervous system (CNS) pathologies and enhance functional recovery after brain damage. OBJECTIVE: This study examined the efficacy of rTMS in mitigating remote degeneration and inflammation and in improving functional recovery in a model of focal brain damage. METHODS: Rats that were undergoing hemicerebellectomy (HCb) were treated with an rTMS protocol for 7 days, and neuronal death indices, glial activation, and functional recovery were assessed. RESULTS: rTMS significantly reduced neuronal death and glial activation in remote regions and improved functional recovery. CONCLUSIONS: Our finding opens up a completely new scenario for exploiting the potential of rTMS as an anti-apoptotic and anti-inflammatory treatment.


Subject(s)
Apoptosis/radiation effects , Brain Injuries/complications , Inflammation/etiology , Inflammation/therapy , Transcranial Magnetic Stimulation , Animals , Brain Injuries/pathology , Calcium-Binding Proteins/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Gene Expression Regulation/radiation effects , Glial Fibrillary Acidic Protein/metabolism , Male , Microfilament Proteins/metabolism , Neuroglia/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Messenger , Rats , Rats, Wistar , Recovery of Function/radiation effects
5.
Cerebellum ; 14(1): 15-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25253422

ABSTRACT

When CNS lesions develop, neuronal degeneration occurs locally but in regions that are remote, yet functionally connected, to the primary lesion site. This process, known as "remote damage," significantly affects long-term outcomes in many CNS pathologies, such as stroke, multiple sclerosis, and traumatic brain and spinal cord injuries. Remote damage can last several days or months after the primary lesion, providing a window during which therapeutic approaches can be implemented to effect neuroprotection. The recognition of the importance of remote damage in determining disease outcomes has prompted considerable interest in examining remote damage-associated mechanisms, most of which is derived from the potential of this research to develop innovative pharmacological approaches for preserving neurons and improving functional outcomes. To this end, the hemicerebellectomy (HCb) experimental paradigm has been instrumental in highlighting the complexity and variety of the systems that are involved, identifying mechanisms of life/death decisions, and providing a testing ground for novel neuroprotective approaches. Inflammation, oxidative stress, apoptosis, autophagy, and neuronal changes in receptor mosaics are several remote damage mechanisms that have been identified and examined using the HCb model. In this review, we discuss our current understanding of remote degeneration mechanisms and their potential for exploitation with regard to neuroprotective approaches, focusing on HCb studies.


Subject(s)
Cerebellum/physiopathology , Nerve Degeneration/physiopathology , Animals , Cell Death/physiology , Cerebellum/pathology , Disease Models, Animal , Nerve Degeneration/pathology , Neurons/physiology
6.
Neurotoxicology ; 45: 67-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25305366

ABSTRACT

Genetic risk factors acting during pregnancy or early after birth have been proposed to account for the exponential increase of autism diagnoses in the past 20 years. In particular, a potential link with exposure to environmental mercury has been suggested. Male sex constitutes a second risk factor for autism. A third potential genetic risk factor is decreased Reelin expression. Male heterozygous reeler (rl(+/-)) mice show an autism-like phenotype, including Purkinje cells (PCs) loss and behavioral rigidity. We evaluated the complex interactions between 3 risk factors, i.e. genetic status, sex, and exposure to methylmercury (MeHg), in rl(+/-) mice. Mice were exposed to MeHg during the prenatal and early postnatal period, either at a subtoxic dose (2 ppm in Dams' drinking water), or at a toxic dose (6 ppm Dams' drinking water), based on observations in other rodent species and mice strains. We show that: (a) 2 ppm MeHg does not cause PCs loss in the different animal groups, and does not enhance PCs loss in rl(+/-) males; consistent with a lack of overt neurotoxicity, 2 ppm MeHg per se does not cause behavioral alterations (separation-induced ultrasonic calls in newborns, or sociability and social preference in adults); (b) in stark contrast, 6 ppm MeHg causes a dramatic reduction of PCs number in all groups, irrespective of genotype and sex. Cytochrome C release from mitochondria of PCs is enhanced in 6 ppm MeHg-exposed groups, with a concomitant increase of µ-calpain active subunit. At the behavioral level, 6 ppm MeHg exposure strongly increases ultrasonic vocalizations in all animal groups. Notably, 6 ppm MeHg significantly decreases sociability in rl(+/-) male mice, while the 2 ppm group does not show such as decrease. At a subtoxic dose, MeHg does not enhance the autism-like phenotype of male rl(+/-) mice. At the higher MeHg dose, the scenario is more complex, with some "autism-like" features (loss of sociability, preference for sameness) being evidently affected only in rl(+/-) males, while other neuropathological and behavioral parameters being altered in all groups, independently from genotype and sex. Mitochondrial abnormalities appear to play a crucial role in the observed effects.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cerebellum/drug effects , Child Development Disorders, Pervasive/chemically induced , Extracellular Matrix Proteins/genetics , Methylmercury Compounds/toxicity , Nerve Tissue Proteins/genetics , Prenatal Exposure Delayed Effects , Serine Endopeptidases/genetics , Animals , Apoptosis/drug effects , Cell Count , Cerebellum/metabolism , Child Development Disorders, Pervasive/genetics , Disease Models, Animal , Female , Heterozygote , Male , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/analysis , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , Pregnancy , Purkinje Cells/drug effects , Purkinje Cells/ultrastructure , Reelin Protein , Risk Factors , Sex Factors , Social Behavior , Vocalization, Animal/drug effects
7.
Cell Mol Life Sci ; 70(12): 2191-204, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23296125

ABSTRACT

Endocannabinoids (eCBs) and glucocorticoids (GCs) are two distinct classes of signaling lipids that exert both neuroprotective and immunosuppressive effects; however, the possibility of an actual interaction of their receptors [i.e., type-2 cannabinoid (CB2) and glucocorticoid receptor α (GRα), respectively] remains unexplored. Here, we demonstrate that the concomitant activation of CB2 and GRα abolishes the neuroprotective effects induced by each receptor on central neurons and on glial cells in animal models of remote cell death. We also show that the ability of eCBs and GCs, used individually, to inhibit tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) production from activated human T lymphocytes is lost when CB2 and GRα are activated simultaneously. In addition, signal transduction pathways triggered by concomitant activation of both receptors led to increased levels of GRß, heat-shock proteins-70 and -90, and p-JNK, as well as to reduced levels of p-STAT6. These effects were reversed only by selectively antagonizing CB2, but not GRα. Overall, our study demonstrates for the first time the existence of a CB2-driven negative cross-talk between eCB and GC signaling in both rats and humans, thus paving the way to the possible therapeutic exploitation of CB2 as a new target for chronic inflammatory and neurodegenerative diseases.


Subject(s)
Neuroglia/metabolism , Neurons/metabolism , Receptor Cross-Talk/physiology , Receptor, Cannabinoid, CB2/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Blotting, Western , Endocannabinoids/metabolism , Flow Cytometry , Glucocorticoids/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Interferon-gamma/metabolism , Male , Rats , Rats, Wistar , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Autophagy ; 8(2): 222-35, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22248716

ABSTRACT

Autophagy is the evolutionarily conserved degradation and recycling of cellular constituents. In mammals, autophagy is implicated in the pathogenesis of many neurodegenerative diseases. However, its involvement in acute brain damage is unknown. This study addresses the function of autophagy in neurodegeneration that has been induced by acute focal cerebellar lesions. We provide morphological, ultrastructural, and biochemical evidence that lesions in a cerebellar hemisphere activate autophagy in axotomized precerebellar neurons. Through time course analyses of the apoptotic cascade, we determined mitochondrial dysfunction to be the early trigger of degeneration. Further, the stimulation of autophagy by rapamycin and the employment of mice with impaired autophagic responses allowed us to demonstrate that autophagy protects from damage promoting functional recovery. These findings have therapeutic significance, demonstrating the potential of pro-autophagy treatments for acute brain pathologies, such as stroke and brain trauma.


Subject(s)
Autophagy/drug effects , Brain Injuries/complications , Cytoprotection/drug effects , Nerve Degeneration/prevention & control , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sirolimus/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Axotomy , Beclin-1 , Brain Injuries/drug therapy , Brain Injuries/pathology , Cerebellum/drug effects , Cerebellum/surgery , Chloroquine/pharmacology , Cytochromes c/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/ultrastructure , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Neurons/cytology , Neurons/pathology , Neurons/ultrastructure , Neuroprotective Agents/therapeutic use , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Sirolimus/therapeutic use
9.
J Neurosci Res ; 88(3): 500-9, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19795376

ABSTRACT

Nitric oxide (NO) and purinergic ionotropic receptors (P2X) mediate cellular events in the central nervous system (CNS) under physiological conditions as well as during pathological events, and they have been recently proposed to interact in mediating CNS response to injury (Viscomi et al. [2004] Neuroscience 123:393-404; Florenzano et al. [2008] Pflugers Arch. 452:622-644). Trimethyltin (TMT) is an organotin compound that generates neurotoxic effects, and it has been used in a model of neurodegenerative disease and memory dysfunction. TMT causes neuronal death and reactive gliosis primarily in the hippocampus and other limbic regions. In the present study, we examined the degenerative events and the expression of nitric oxide synthase (NOS) and P2X receptor subtypes (P2X(1,2,4,7)Rs) that were induced by TMT administration at different time points (3, 7, 14, and 21 days) by conventional and confocal microscopy and Western blotting. Massive glial activation and neuronal death in the CA1 and CA3 regions were observed after TMT treatment. In these areas, astrocytic P2X(2)R and neuronal NOS were temporarily enhanced in association with the progression of neuronal death. In the hippocampus, the physiological expression of P2X(1)R, P2X(4)R, and P2X(7)R was not modified by TMT. The present data demonstrate that, as in other neurodegenerative models, TMT-induced hippocampal degeneration is associated with nitrergic and purinergic activations. Nevertheless, at odds with previous data, in this model the two systems are active in segregated cell populations, namely, P2XR in astrocytes and NOS in neurons. Finally, the temporal relations between P2XR and NOS expression and neuronal degeneration suggest interactions between P2XR/NO signaling and cell survival.


Subject(s)
Central Nervous System Agents/toxicity , Hippocampus/drug effects , Nitric Oxide Synthase/metabolism , Receptors, Purinergic P2/metabolism , Trimethyltin Compounds/toxicity , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/physiology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/enzymology , CA3 Region, Hippocampal/physiology , Cell Death/drug effects , Female , Hippocampus/enzymology , Hippocampus/physiology , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Nerve Degeneration/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/physiology , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Wistar , Receptors, Purinergic P2X , Time Factors , Up-Regulation/drug effects
10.
J Neurosci ; 29(14): 4564-70, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19357281

ABSTRACT

Endocannabinoids are neuroprotective in vivo and in vitro, but the mechanisms by which they act are largely unknown. The present study addressed the role of cannabinoid receptors during remote cell death of central neurons in a model that is based on cerebellar lesions. A lesion in one cerebellar hemisphere induced remote cell death and type 2 cannabinoid receptor (CB2R) expression in contralateral precerebellar neurons. Of the selective agonists and antagonists that modulated cannabinoid receptor activity, we found that the CB2R agonist JWH-015 reduced neuronal loss and cytochrome-c release, leading to neurological recovery; these effects were reversed by the selective CB2R antagonist SR144528. Analysis of CB2R-triggered signal transduction demonstrated that in axotomized neurons, CB2R regulated Akt and JNK phosphorylation through a PI3K-dependent pathway, whereas other major signaling routes that are dependent on CB2R, such as ERK1/2 and p38, were not involved. This result was corroborated by the observation that the selective PI3K inhibitor LY294002 blocked the CB2R stimulation effects on neuronal survival as well as Akt and JNK phosphorylation levels. Together, these data demonstrate that axonal damage induces CB2R expression in central neurons and that stimulation of this receptor has a neuroprotective effect that is achieved through PI3K/Akt signaling.


Subject(s)
Apoptosis/physiology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptor, Cannabinoid, CB2/agonists , Signal Transduction/physiology , Animals , Axotomy , Cell Survival/drug effects , Cell Survival/physiology , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/pathology , Indoles/pharmacology , Male , Neurons/enzymology , Neurons/pathology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB2/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...