Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Biomedicines ; 10(2)2022 Jan 23.
Article in English | MEDLINE | ID: mdl-35203447

ABSTRACT

Microglia have been increasingly implicated in neurodegenerative diseases (NDs), and specific disease associated microglia (DAM) profiles have been defined for several of these NDs. Yet, the microglial profile in Machado-Joseph disease (MJD) remains unexplored. Here, we characterized the profile of microglia in the CMVMJD135 mouse model of MJD. This characterization was performed using primary microglial cultures and microglial cells obtained from disease-relevant brain regions of neonatal and adult CMVMJD135 mice, respectively. Machine learning models were implemented to identify potential clusters of microglia based on their morphological features, and an RNA-sequencing analysis was performed to identify molecular perturbations and potential therapeutic targets. Our findings reveal morphological alterations that point to an increased activation state of microglia in CMVMJD135 mice and a disease-specific transcriptional profile of MJD microglia, encompassing a total of 101 differentially expressed genes, with enrichment in molecular pathways related to oxidative stress, immune response, cell proliferation, cell death, and lipid metabolism. Overall, these results allowed us to define the cellular and molecular profile of MJD-associated microglia and to identify genes and pathways that might represent potential therapeutic targets for this disorder.

3.
Cells ; 11(3)2022 01 21.
Article in English | MEDLINE | ID: mdl-35159165

ABSTRACT

Exposure to methamphetamine (Meth) has been classically associated with damage to neuronal terminals. However, it is now becoming clear that addiction may also result from the interplay between glial cells and neurons. Recently, we demonstrated that binge Meth administration promotes microgliosis and microglia pro-inflammation via astrocytic glutamate release in a TNF/IP3R2-Ca2+-dependent manner. Here, we investigated the contribution of neuronal cells to this process. As the crosstalk between microglia and neurons may occur by contact-dependent and/or contact-independent mechanisms, we developed co-cultures of primary neurons and microglia in microfluidic devices to investigate how their interaction affects Meth-induced microglia activation. Our results show that neurons exposed to Meth do not activate microglia in a cell-autonomous way but require astrocyte mediation. Importantly, we found that neurons can partially prevent Meth-induced microglia activation via astrocytes, which seems to be achieved by increasing arginase 1 expression and strengthening the CD200/CD200r pathway. We also observed an increase in synaptic individual area, as determined by co-localization of pre- and post-synaptic markers. The present study provides evidence that contact-dependent mechanisms between neurons and microglia can attenuate pro-inflammatory events such as Meth-induced microglia activation.


Subject(s)
Methamphetamine , Methamphetamine/metabolism , Methamphetamine/pharmacology , Microglia/metabolism , Neuroglia/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism
4.
Neuropsychopharmacology ; 46(13): 2358-2370, 2021 12.
Article in English | MEDLINE | ID: mdl-34400780

ABSTRACT

Methamphetamine (Meth) is a powerful illicit psychostimulant, widely used for recreational purposes. Besides disrupting the monoaminergic system and promoting oxidative brain damage, Meth also causes neuroinflammation, contributing to synaptic dysfunction and behavioral deficits. Aberrant activation of microglia, the largest myeloid cell population in the brain, is a common feature in neurological disorders triggered by neuroinflammation. In this study, we investigated the mechanisms underlying the aberrant activation of microglia elicited by Meth in the adult mouse brain. We found that binge Meth exposure caused microgliosis and disrupted risk assessment behavior (a feature that usually occurs in individuals who abuse Meth), both of which required astrocyte-to-microglia crosstalk. Mechanistically, Meth triggered a detrimental increase of glutamate exocytosis from astrocytes (in a process dependent on TNF production and calcium mobilization), promoting microglial expansion and reactivity. Ablating TNF production, or suppressing astrocytic calcium mobilization, prevented Meth-elicited microglia reactivity and re-established risk assessment behavior as tested by elevated plus maze (EPM). Overall, our data indicate that glial crosstalk is critical to relay alterations caused by acute Meth exposure.


Subject(s)
Central Nervous System Stimulants , Methamphetamine , Tumor Necrosis Factor-alpha , Animals , Astrocytes , Central Nervous System Stimulants/toxicity , Glutamic Acid , Methamphetamine/toxicity , Mice , Microglia
5.
Neurosci Biobehav Rev ; 117: 98-109, 2020 10.
Article in English | MEDLINE | ID: mdl-31499082

ABSTRACT

Maternal separation (MS) is a widely-used paradigm to study the effect of early-life adversity on brain development and resilience to psychopathology. Most of the related literature focuses on MS impact on offspring, however, it should ideally also consider the impact of altered maternal behaviour caused by MS itself. This systematic review aimed at providing a comprehensive compilation of the effects of MS on key maternal behaviour aspects. We performed a keyword literature search using Boolean operators. Databases were searched between 2000-2018. Additional studies were included from manual search. Twenty-nine articles addressing the impact of MS on maternal behaviour and/or mothers' anxiety, depression-like behaviour, memory and consequences on underlying mechanisms. The methodological aspects and main conclusions were extracted from each study. This review shows that MS induces changes in dams. Results are particularly robust for increased anxiety and depressive-like symptoms, and altered maternal behaviours, predominantly for longer periods of MS. Finally, research in the field could strongly benefit from the establishment of guidelines to reduce the methodological inconsistencies here identified.


Subject(s)
Maternal Deprivation , Mothers , Animals , Anxiety , Female , Humans , Maternal Behavior , Rodentia , Stress, Psychological
6.
Front Neurosci ; 13: 453, 2019.
Article in English | MEDLINE | ID: mdl-31143097

ABSTRACT

Ascorbate, the reduced form of Vitamin C, is one of the most abundant and important low-molecular weight antioxidants in living tissues. Most animals synthesize vitamin C, but some primates, including humans, have lost this capacity due to disruption in L-gulono-gamma-lactone oxidase gene. Because of this incapacity, those animals must obtain Vitamin C from the diet. Ascorbate is highly concentrated in the central nervous system (CNS), including the retina, and plays essential roles in neuronal physiology. Ascorbate transport into cells is controlled by Sodium Vitamin C Co-Transporters (SVCTs). There are four SVCT isoforms and SVCT2 is the major isoform controlling ascorbate transport in the CNS. Regarding ascorbate release from retinal neurons, Glutamate, by activating its ionotropic receptors leads to ascorbate release via the reversion of SVCT2. Moreover, dopamine, via activation of D1 receptor/cyclic AMP/EPAC2 pathway, also induces ascorbate release via SVCT2 reversion. Because the dopaminergic and glutamatergic systems are interconnected in the CNS, we hypothesized that dopamine could regulate ascorbate release indirectly, via the glutamatergic system. Here we reveal that dopamine increases the release of D-Aspartate from retinal neurons in a way independent on calcium ions and dependent on excitatory amino acid transporters. In addition, dopamine-dependent SVCT2 reversion leading to ascorbate release occurs by activation of AMPA/Kainate receptors and downstream ERK/AKT pathways. Overall, our data reveal a dopamine-to-glutamate signaling that regulates the bioavailability of ascorbate in neuronal cells.

7.
Nat Commun ; 9(1): 3138, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087335

ABSTRACT

Group B streptococcal (GBS) meningitis remains a devastating disease. The absence of an animal model reproducing the natural infectious process has limited our understanding of the disease and, consequently, delayed the development of effective treatments. We describe here a mouse model in which bacteria are transmitted to the offspring from vaginally colonised pregnant females, the natural route of infection. We show that GBS strain BM110, belonging to the CC17 clonal complex, is more virulent in this vertical transmission model than the isogenic mutant BM110∆cylE, which is deprived of hemolysin/cytolysin. Pups exposed to the more virulent strain exhibit higher mortality rates and lung inflammation than those exposed to the attenuated strain. Moreover, pups that survive to BM110 infection present neurological developmental disability, revealed by impaired learning performance and memory in adulthood. The use of this new mouse model, that reproduces key steps of GBS infection in newborns, will promote a better understanding of the physiopathology of GBS-induced meningitis.


Subject(s)
Disease Models, Animal , Infectious Disease Transmission, Vertical , Streptococcal Infections/physiopathology , Animals , Animals, Newborn , Behavior, Animal , Body Weight , Female , Hemolysin Proteins/chemistry , Inflammation , Male , Maze Learning , Meningitis/microbiology , Meningitis, Bacterial , Mice , Mice, Inbred BALB C , Perforin/chemistry , Pregnancy , Pregnancy, Animal , Streptococcal Infections/transmission , Streptococcus agalactiae/pathogenicity , Vagina/microbiology
8.
Mol Neurobiol ; 55(10): 7858-7871, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29476450

ABSTRACT

Ascorbate, the reduced form of vitamin C, is highly concentrated in the central nervous system (CNS), including the retina, where it plays important physiological functions. In the CNS, the plasma membrane transporter sodium vitamin C co-transporter 2 (SVCT2) is responsible for ascorbate transport in neurons. The neurotransmitter dopamine (DA), acting through D1- and D2-like receptor subfamilies and classically coupled to adenylyl cyclase, is known to modulate synaptic transmission in the retina. Here, we reveal that DA controls the release of ascorbate from retinal neurons. Using primary retinal cultures, we show that this DA effect is dose-dependent, occurring by the reversal of the SVCT2, and could be elicited by brief and repetitive pulses of DA. The DA effect in inducing ascorbate release occurs by the activation of D1R and is independent of PKA. Moreover, the exchange protein directly activated by cAMP type 2 (EPAC2) is present in retinal neurons and its specific knockdown using shRNAs abrogates the D1R-induced ascorbate release. Confirming the physiological relevance of this pathway, activation of D1R or EPAC2 also triggered ascorbate release ex vivo in acute preparations of the intact retina. Overall, DA plays pivotal roles in regulating ascorbate homeostasis through an unanticipated signaling pathway involving D1R/adenylyl cyclase/cAMP/EPAC2, thereby suggesting that vitamin C might fine-tune dopaminergic neurotransmission in the retina.


Subject(s)
Ascorbic Acid/metabolism , Dopamine/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Dopamine D1/metabolism , Retinal Neurons/metabolism , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Chick Embryo , Retinal Neurons/drug effects , Signal Transduction/drug effects
9.
Mol Neurobiol ; 55(8): 6816-6833, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29349577

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor and still lacks effective therapeutic strategies. It has already been shown that old drugs like sulfasalazine (SAS) and valproic acid (VPA) present antitumoral activities in glioma cell lines. SAS has also been associated with a decrease of intracellular glutathione (GSH) levels through a potent inhibition of xc- glutamate/cystine exchanger leading to an antioxidant deprotection. In the same way, VPA was recently identified as a histone deacetylase (HDAT) inhibitor capable of activating tumor suppression genes. As both drugs are widely used in clinical practice and their profile of adverse effects is well known, the aim of our study was to investigate the effects of the combined treatment with SAS and VPA in GBM cell lines. We observed that both drugs were able to reduce cell viability in a dose-dependent manner and the combined treatment potentiated these effects. Combined treatment also increased cell death and inhibited proliferation of GBM cells, while having no effect on human and rat cultured astrocytes. Also, we observed high protein expression of the catalytic subunit of xc- in all the examined GBM cell lines, and treatment with SAS blocked its activity and decreased intracellular GSH levels. Noteworthy, SAS but not VPA was also able to reduce the [14C]-ascorbate uptake. Together, these data indicate that SAS and VPA exhibit a substantial effect on GBM cell's death related to an intracellular oxidative response imbalance, making this combination of drugs a promising therapeutic strategy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Intracellular Space/metabolism , Sulfasalazine/pharmacology , Valproic Acid/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Ascorbic Acid/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Therapy, Combination , Glutathione/metabolism , Humans , Mesoderm/drug effects , Mesoderm/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Oxidation-Reduction , Rats , Time Factors
10.
J Biol Chem ; 287(6): 3860-72, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22041898

ABSTRACT

Ascorbate is an important antioxidant, which also displays important functions in neuronal tissues, including the retina. The retina is responsible for the initial steps of visual processing, which is further refined in cerebral high-order centers. The retina is also a prototypical model for studying physiologic aspects of cells that comprise the nervous system. Of major importance also is the cellular messenger nitric oxide (NO). Previous studies have demonstrated the significance of NO for both survival and proliferation of cultured embryonic retinal cells. Cultured retinal cells express a high-affinity ascorbate transporter, and the release of ascorbate is delicately regulated by ionotropic glutamate receptors. Therefore, we proposed whether there is interplay between the ascorbate transport system and NO signaling pathway in retinal cells. Here we show compelling evidence that ascorbate uptake is tightly controlled by NO and its downstream signaling pathway in culture. NO also modulates the expression of SVCT-2, an effect mediated by cGMP and PKG. Kinetic studies suggest that NO increases the transport capacity for ascorbate, but not the affinity of SVCT-2 for its substrate. Interestingly, NO utilizes the NF-κB pathway, in a PKG-dependent manner, to modulate both SVCT-2 expression and ascorbate uptake. These results demonstrate that NO exerts a fine-tuned control of the availability of ascorbate to cultured retinal cells and strongly reinforces ascorbate as an important bioactive molecule in neuronal tissues.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/physiology , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Retina/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Animals , Ascorbic Acid/genetics , Ascorbic Acid/metabolism , Biological Transport, Active/physiology , Cell Proliferation , Chick Embryo , Chickens , Cyclic GMP-Dependent Protein Kinases/genetics , NF-kappa B/genetics , Nerve Tissue Proteins/genetics , Nitric Oxide/genetics , Retina/cytology , Retina/embryology , Signal Transduction/physiology , Sodium-Coupled Vitamin C Transporters/genetics
11.
J Neurochem ; 108(2): 507-20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19054286

ABSTRACT

Vitamin C is transported in the brain by sodium vitamin C co-transporter 2 (SVCT-2) for ascorbate and glucose transporters for dehydroascorbate. Here we have studied the expression of SVCT-2 and the uptake and release of [(14)C] ascorbate in chick retinal cells. SVCT-2 immunoreactivity was detected in rat and chick retina, specially in amacrine cells and in cells in the ganglion cell layer. Accordingly, SVCT-2 was expressed in cultured retinal neurons, but not in glial cells. [(14)C] ascorbate uptake was saturable and inhibited by sulfinpyrazone or sodium-free medium, but not by treatments that inhibit dehydroascorbate transport. Glutamate-stimulated vitamin C release was not inhibited by the glutamate transport inhibitor l-beta-threo-benzylaspartate, indicating that vitamin C release was not mediated by glutamate uptake. Also, ascorbate had no effect on [(3)H] D-aspartate release, ruling out a glutamate/ascorbate exchange mechanism. 2-Carboxy-3-carboxymethyl-4-isopropenylpyrrolidine (Kainate) or NMDA stimulated the release, effects blocked by their respective antagonists 6,7-initroquinoxaline-2,3-dione (DNQX) or (5R,2S)-(1)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate (MK-801). However, DNQX, but not MK-801 or 2-amino-5-phosphonopentanoic acid (APV), blocked the stimulation by glutamate. Interestingly, DNQX prevented the stimulation by NMDA, suggesting that the effect of NMDA was mediated by glutamate release and stimulation of non-NMDA receptors. The effect of glutamate was neither dependent on external calcium nor inhibited by 1,2-bis (2-aminophenoxy) ethane-N',N',N',N',-tetraacetic acid tetrakis (acetoxy-methyl ester) (BAPTA-AM), an internal calcium chelator, but was inhibited by sulfinpyrazone or by the absence of sodium. In conclusion, retinal cells take up and release vitamin C, probably through SVCT-2, and the release can be stimulated by NMDA or non-NMDA glutamate receptors.


Subject(s)
Ascorbic Acid/metabolism , Calcium/metabolism , Neurons/metabolism , Receptors, Glutamate/physiology , Retina/cytology , Sodium/metabolism , Animals , Animals, Newborn , Aspartic Acid/pharmacology , Biological Transport/drug effects , Chelating Agents/pharmacology , Chick Embryo , Cytoskeletal Proteins/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , N-Methylaspartate/pharmacology , Neurons/drug effects , Organic Anion Transporters, Sodium-Dependent/metabolism , Rats , Sodium-Coupled Vitamin C Transporters , Symporters/metabolism , Tissue Culture Techniques , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...