Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Mol Med (Berl) ; 102(3): 365-377, 2024 03.
Article in English | MEDLINE | ID: mdl-38197965

ABSTRACT

The mechanisms underlying neurodegeneration in Parkinson's disease (PD) are still not fully understood. Glycosylation is an important post-translational modification that affects protein function, cell-cell contacts and inflammation and can be modified in pathologic conditions. Although the involvement of aberrant glycosylation has been proposed for PD, the knowledge of the diversity of glycans and their role in PD is still minimal. Sialyl Lewis X (sLeX) is a sialylated and fucosylated tetrasaccharide with essential roles in cell-to-cell recognition processes. Pathological conditions and pro-inflammatory mediators can up-regulate sLeX expression on cell surfaces, which has important consequences in intracellular signalling and immune function. Here, we investigated the expression of this glycan using in vivo and in vitro models of PD. We show the activation of deleterious glycation-related pathways in mouse striatum upon treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a toxin-based model of PD. Importantly, our results show that MPTP triggers the presentation of more proteins decorated with sLeX in mouse cortex and striatum in a time-dependent manner, as well as increased mRNA expression of its rate-limiting enzyme fucosyltransferase 7. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. Although the underlying mechanism that drives increased sLeX epitopes, the nature of the protein scaffolds and their functional importance in PD remain unknown, our data suggest for the first time that sLeX in the brain may have a role in neuronal signalling and immunomodulation in pathological conditions. KEY MESSAGES: MPTP triggers the presentation of proteins decorated with sLeX in mouse brain. MPTP triggers the expression of sLeX rate-limiting enzyme FUT 7 in striatum. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. sLeX in the brain may have a role in neuronal signalling and immunomodulation.


Subject(s)
Parkinson Disease , Animals , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Sialyl Lewis X Antigen , Inflammation , Brain/metabolism , Models, Theoretical , Disease Models, Animal , Mice, Inbred C57BL
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2171-2181, 2017 09.
Article in English | MEDLINE | ID: mdl-28583715

ABSTRACT

Impaired mitochondrial function and generation of reactive oxygen species are deeply implicated in Parkinson's disease progression. Indeed, mutations in genes that affect mitochondrial function account for most of the familial cases of the disease, and post mortem studies in sporadic PD patients brains revealed increased signs of oxidative stress. Moreover, exposure to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a mitochondrial complex I inhibitor, leads to clinical symptoms similar to sporadic PD. The bile acid tauroursodeoxycholic acid (TUDCA) is an anti-apoptotic molecule shown to protect against MPTP-induced neurodegeneration in mice, but the mechanisms involved are still incompletely identified. Herein we used MPTP-treated mice, as well as primary cultures of mice cortical neurons and SH-SY5Y cells treated with MPP+ to investigate the modulation of mitochondrial dysfunction by TUDCA in PD models. We show that TUDCA exerts its neuroprotective role in a parkin-dependent manner. Overall, our results point to the pharmacological up-regulation of mitochondrial turnover by TUDCA as a novel neuroprotective mechanism of this molecule, and contribute to the validation of TUDCA clinical application in PD.


Subject(s)
Antioxidants/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Taurochenodeoxycholic Acid/pharmacology , Animals , Male , Mice , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Ubiquitin-Protein Ligases/metabolism
3.
Front Cell Neurosci ; 10: 284, 2016.
Article in English | MEDLINE | ID: mdl-28018177

ABSTRACT

Neurogenesis in the subventricular zone (SVZ) is regulated by diffusible factors and cell-cell contacts. In vivo, SVZ stem cells are associated with the abluminal surface of blood vessels and such interactions are thought to regulate their neurogenic capacity. SVZ neural stem cells (NSCs) have been described to contact endothelial-derived laminin via α6ß1 integrin. To elucidate whether heterocellular contacts with brain endothelial cells (BEC) regulate SVZ cells neurogenic capacities, cocultures of SVZ neurospheres and primary BEC, both obtained from C57BL/6 mice, were performed. The involvement of laminin-integrin interactions in SVZ homeostasis was tested in three ways. Firstly, SVZ cells were analyzed following incubation of BEC with the protein synthesis inhibitor cycloheximide (CHX) prior to coculture, a treatment expected to decrease membrane proteins. Secondly, SVZ cells were cocultured with BEC in the presence of an anti-α6 integrin neutralizing antibody. Thirdly, BEC were cultured with ß1-/- SVZ cells. We showed that contact with BEC supports, at least in part, proliferation and stemness of SVZ cells, as evaluated by the number of BrdU positive (+) and Sox2+ cells in contact with BEC. These effects are dependent on BEC-derived laminin binding to α6ß1 integrin and are decreased in cocultures incubated with anti-α6 integrin neutralizing antibody and in cocultures with SVZ ß1-/- cells. Moreover, BEC-derived laminin sustains stemness in SVZ cell cultures via activation of the Notch and mTOR signaling pathways. Our results show that BEC/SVZ interactions involving α6ß1 integrin binding to laminin, contribute to SVZ cell proliferation and stemness.

4.
Stem Cells Dev ; 22(11): 1693-708, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23327619

ABSTRACT

Neural stem cells of the subventricular zone (SVZ) represent a potentially important source of surrogate cells for the treatment of brain damage. Proper use of these cells for neuronal replacement depends on the ability to drive neuronal differentiation. Several neuromodulators stimulate neurogenesis. Here we examined the effects of the neuropeptide galanin, on neuronal differentiation in murine SVZ cultures. SVZ neurospheres obtained from early postnatal mice were treated with 10 nM to 2 µM galanin. Galanin promoted neuronal differentiation, increasing numbers of NeuN-, vesicular GABA transporter- and tyrosine hydroxylase-expressing neurons. In contrast, galanin neither affected cell proliferation assessed by BrdU incorporation nor cell death evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Neuronal differentiation was further confirmed at the functional level by measuring [Ca(2+)]i variations in single SVZ cells after KCl and histamine stimulations to distinguish neurons from immature cells. Galanin treatment increased the numbers of neuronal-like responding cells compared to immature cells. Using selective agonists (M617, AR-M1896) and antagonists (galantide, M871) for galanin receptors 1 and 2, we showed that both galanin receptors mediated neuronal differentiation. Early proneuronal effects of galanin included positive regulation of the transcription factor neurogenin-1 (Ngn1). In addition, galanin promoted axonogenesis and dendritogenesis, increasing both the length of phosphorylated stress-activated protein kinase- and Tau-positive axons and the numbers of microtubule associated protein-2 (MAP-2)-labelled dendrites. Moreover, galanin inhibited SVZ cell migration in the transwell assay. Our results show a proneurogenic effect of galanin and open new perspectives for future applications in stem cell-based therapies for neuronal replacement.


Subject(s)
Cell Differentiation/drug effects , Galanin/pharmacology , Neural Stem Cells/metabolism , Receptor, Galanin, Type 1/metabolism , Receptor, Galanin, Type 2/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Culture Techniques , Cell Death/drug effects , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/drug effects , DNA-Binding Proteins , Galanin/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Nuclear Proteins/biosynthesis , Peptide Fragments/pharmacology , Tyrosine 3-Monooxygenase/biosynthesis , Vesicular Inhibitory Amino Acid Transport Proteins/biosynthesis
5.
J Neurosci ; 30(13): 4573-84, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20357108

ABSTRACT

In the adult mammalian brain, the subventricular zone (SVZ) hosts stem cells constantly generating new neurons. Angiopoietin-1 (Ang-1) is an endothelial growth factor with a critical role in division, survival, and adhesion of endothelial cells via Tie-2 receptor activity. Expression of Tie-2 in nonendothelial cells, especially neurons and stem cells, suggests that Ang-1 may be involved in neurogenesis. In the present work, we investigated the putative role of Ang-1 on SVZ neurogenesis. Immature cells from SVZ-derived neurospheres express Ang-1 and Tie-2 mRNA, suggesting a role for the Ang-1/Tie-2 system in the neurogenic niche. Moreover, we also found that Tie-2 protein expression is retained on differentiation in neurons and glial cells. Ang-1 triggered proliferation via activation of the ERK1/2 (extracellular signal-regulated kinase 1/2) mitogen-activated protein kinase (MAPK) kinase pathway but did not induce cell death. Accordingly, coincubation with an anti-Tie-2 neutralizing antibody prevented the pro-proliferative effect of Ang-1. Furthermore, Ang-1 increased the number of NeuN (neuronal nuclear protein)-positive neurons in cultures treated for 7 d, as well as the number of functional neurons, as assessed by monitoring [Ca(2+)](i) rises after application of specific stimuli for neurons and immature cells. The proneurogenic effect of Ang-1 is mediated by Tie-2 activation and subsequent mTOR (mammalian target of rapamycin kinase) mobilization. In agreement, neuronal differentiation significantly decreased after exposure to an anti-Tie-2 neutralizing antibody and to rapamycin. Moreover, Ang-1 elicited the activation of the SAPK (stress-activated protein kinase)/JNK (c-Jun N-terminal kinase) MAPK, involved in axonogenesis. Our work shows a proneurogenic effect of Ang-1, highlighting the relevance of blood vessel/stem cell cross talk in health and disease.


Subject(s)
Angiopoietin-1/physiology , Brain/cytology , Neurons/physiology , Stem Cells/physiology , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Animals , Axons/physiology , Brain/growth & development , Cell Death , Cell Differentiation , Cell Proliferation , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Neurogenesis , Neuroglia/cytology , Neuroglia/physiology , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/biosynthesis , Receptor, TIE-2/biosynthesis , Receptor, TIE-2/genetics , Stem Cells/cytology , TOR Serine-Threonine Kinases
6.
Neurobiol Dis ; 29(1): 30-40, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17869125

ABSTRACT

We investigated whether nerve cell damage by unconjugated bilirubin (UCB) is mediated by oxidative stress and ascertained the neuronal and astroglial susceptibility to injury. Several oxidative stress biomarkers and cell death were determined following incubation of neurons and astrocytes isolated from rat cortical cerebrum with UCB (0.01-1.0 microM). We show that UCB induces a dose-dependent increase in neuronal death in parallel with the oxidation of cell components and a decrease in the intracellular glutathione content. Comparison of the results obtained in both cell types demonstrates that neurons are more vulnerable than astrocytes to oxidative injury by UCB, for which accounts the lower glutathione stores in neuronal cells. Moreover, neuronal oxidative injury is prevented by supplementation with N-acetylcysteine, a glutathione precursor, whereas astroglial sensitivity to UCB is enhanced by inhibition of glutathione synthesis, using buthionine sulfoximine. Collectively, we demonstrate that oxidative stress is involved in UCB neurotoxicity and depict a new therapeutic approach for UCB-induced oxidative damage.


Subject(s)
Antioxidants/pharmacology , Astrocytes/drug effects , Bilirubin/pharmacology , Neurons/drug effects , Oxidation-Reduction/drug effects , Acetylcysteine/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Female , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...