Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Glia ; 72(8): 1501-1517, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780232

ABSTRACT

Methamphetamine (Meth) use is known to induce complex neuroinflammatory responses, particularly involving astrocytes and microglia. Building upon our previous research, which demonstrated that Meth stimulates astrocytes to release tumor necrosis factor (TNF) and glutamate, leading to microglial activation, this study investigates the role of the anti-inflammatory cytokine interleukin-10 (IL-10) in this process. Our findings reveal that the presence of recombinant IL-10 (rIL-10) counteracts Meth-induced excessive glutamate release in astrocyte cultures, which significantly reduces microglial activation. This reduction is associated with the modulation of astrocytic intracellular calcium (Ca2+) dynamics, particularly by restricting the release of Ca2+ from the endoplasmic reticulum to the cytoplasm. Furthermore, we identify the small Rho GTPase Cdc42 as a crucial intermediary in the astrocyte-to-microglia communication pathway under Meth exposure. By employing a transgenic mouse model that overexpresses IL-10 (pMT-10), we also demonstrate in vivo that IL-10 prevents Meth-induced neuroinflammation. These findings not only enhance our understanding of Meth-related neuroinflammatory mechanisms, but also suggest IL-10 and Cdc42 as putative therapeutic targets for treating Meth-induced neuroinflammation.


Subject(s)
Astrocytes , Interleukin-10 , Methamphetamine , Mice, Transgenic , Microglia , cdc42 GTP-Binding Protein , Animals , Methamphetamine/toxicity , Methamphetamine/pharmacology , Interleukin-10/metabolism , Interleukin-10/pharmacology , Astrocytes/metabolism , Astrocytes/drug effects , cdc42 GTP-Binding Protein/metabolism , Microglia/drug effects , Microglia/metabolism , Mice , Mice, Inbred C57BL , Central Nervous System Stimulants/toxicity , Central Nervous System Stimulants/pharmacology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Cells, Cultured , Glutamic Acid/metabolism , Glutamic Acid/toxicity
2.
Cell Rep ; 42(12): 113447, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37980559

ABSTRACT

Microglia, the largest population of brain immune cells, continuously interact with synapses to maintain brain homeostasis. In this study, we use conditional cell-specific gene targeting in mice with multi-omics approaches and demonstrate that the RhoGTPase Rac1 is an essential requirement for microglia to sense and interpret the brain microenvironment. This is crucial for microglia-synapse crosstalk that drives experience-dependent plasticity, a fundamental brain property impaired in several neuropsychiatric disorders. Phosphoproteomics profiling detects a large modulation of RhoGTPase signaling, predominantly of Rac1, in microglia of mice exposed to an environmental enrichment protocol known to induce experience-dependent brain plasticity and cognitive performance. Ablation of microglial Rac1 affects pathways involved in microglia-synapse communication, disrupts experience-dependent synaptic remodeling, and blocks the gains in learning, memory, and sociability induced by environmental enrichment. Our results reveal microglial Rac1 as a central regulator of pathways involved in the microglia-synapse crosstalk required for experience-dependent synaptic plasticity and cognitive performance.


Subject(s)
Brain , Cognition , Microglia , Neuronal Plasticity , Neuropeptides , rac1 GTP-Binding Protein , Microglia/metabolism , Cognition/physiology , Animals , Mice , Neuropeptides/genetics , Neuropeptides/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology , Male , Female , Mice, Mutant Strains , Synapses/physiology , Brain/physiology , Gene Knockdown Techniques
3.
Front Cell Neurosci ; 17: 1109611, 2023.
Article in English | MEDLINE | ID: mdl-37305435

ABSTRACT

It is now well-accepted that psychostimulants act on glial cells causing neuroinflammation and adding to the neurotoxic effects of such substances. Neuroinflammation can be described as an inflammatory response, within the CNS, mediated through several cytokines, reactive oxygen species, chemokines and other inflammatory markers. These inflammatory players, in particular cytokines, play important roles. Several studies have demonstrated that psychostimulants impact on cytokine production and release, both centrally and at the peripheral level. Nevertheless, the available data is often contradictory. Because understanding how cytokines are modulated by psychoactive substances seems crucial to perspective successful therapeutic interventions, here, we conducted a scoping review of the available literature. We have focused on how different psychostimulants impact on the cytokine profile. Publications were grouped according to the substance addressed (methamphetamine, cocaine, methylphenidate, MDMA or other amphetamines), the type of exposure and period of evaluation (acute, short- or long-term exposure, withdrawal, and reinstatement). Studies were further divided in those addressing central cytokines, circulating (peripheral) levels, or both. Our analysis showed that the classical pro-inflammatory cytokines TNF-α, IL-6, and IL-1ß were those more investigated. The majority of studies have reported increased levels of these cytokines in the central nervous system after acute or repeated drug. However, studies investigating cytokine levels during withdrawal or reinstatement have shown higher variability in their findings. Although we have identified fewer studies addressing circulating cytokines in humans, the available data suggest that the results may be more robust in animal models than in patients with problematic drug use. As a major conclusion, an extensive use of arrays for relevant cytokines should be considered to better determine which cytokines, upon the classical ones, may be involved in the progression from episodic use to the development of addiction. A concerted effort is still necessary to address the link between peripheral and central immune players, including from a longitudinal perspective. Until there, the identification of new biomarkers and therapeutic targets to envision personalized immune-based therapeutics will continue to be unlikely.

4.
Transl Psychiatry ; 12(1): 463, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333302

ABSTRACT

Depressive mothers often find mother-child interaction to be challenging. Maternal stress may further impair mother-child attachment, which may increase the risk of negative developmental consequences. We used rats with different vulnerability to depressive-like behavior (Wistar and Kyoto) to investigate the impact of stress (maternal separation-MS) on maternal behavior and adolescent offspring cognition. MS in Kyoto dams increased pup-contact, resulting in higher oxytocin levels and lower anxiety-like behavior after weaning, while worsening their adolescent offspring cognitive behavior. Whereas MS in Wistar dams elicited higher quality of pup-directed behavior, increasing brain-derived neurotrophic factor (BDNF) in the offspring, which seems to have prevented a negative impact on cognition. Hypothalamic oxytocin seems to affect the salience of the social environment cues (negatively for Kyoto) leading to different coping strategies. Our findings highlight the importance of contextual and individual factors in the understanding of the oxytocin role in modulating maternal behavior and stress regulatory processes.


Subject(s)
Maternal Deprivation , Oxytocin , Female , Humans , Animals , Rats , Depression , Rats, Wistar , Maternal Behavior , Adaptation, Psychological , Anxiety/psychology , Stress, Psychological , Behavior, Animal
5.
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.

6.
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
7.
Mol Neurobiol ; 59(2): 872-889, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34796462

ABSTRACT

Microglia, the 'resident immunocompetent cells' of the central nervous system (CNS), are key players in innate immunity, synaptic refinement and homeostasis. Dysfunctional microglia contribute heavily to creating a toxic inflammatory milieu, a driving factor in the pathophysiology of several CNS disorders. Therefore, strategies to modulate the microglial function are required to tackle exacerbated tissue inflammation. Carbon monoxide (CO), an endogenous gaseous molecule produced by the degradation of haem, has anti-inflammatory, anti-apoptotic, and pro-homeostatic and cytoprotective roles, among others. ALF-826A, a novel molybdenum-based CO-releasing molecule, was used for the assessment of neuron-microglia remote communication. Primary cultures of rat microglia and neurons, or the BV-2 microglial and CAD neuronal murine cell lines, were used to study the microglia-neuron interaction. An approach based on microglial-derived conditioned media in neuronal culture was applied. Medium derived from CO-treated microglia provided indirect neuroprotection against inflammation by limiting the lipopolysaccharide (LPS)-induced expression of reactivity markers (CD11b), the production of reactive oxygen species (ROS) and the secretion of inflammatory factors (TNF-α, nitrites). This consequently prevented neuronal cell death and maintained neuronal morphology. In contrast, in the absence of inflammatory stimulus, conditioned media from CO-treated microglia improved neuronal morphological complexity, which is an indirect manner of assessing neuronal function. Likewise, the microglial medium also prevented neuronal cell death induced by pro-oxidant tert-Butyl hydroperoxide (t-BHP). ALF-826 treatment reinforced microglia secretion of Interleukin-10 (IL-10) and adenosine, mediators that may protect against t-BHP stress in this remote communication model. Chemical inhibition of the adenosine receptors A2A and A1 reverted the CO-derived neuroprotective effect, further highlighting a role for CO in regulating neuron-microglia communication via purinergic signalling. Our findings indicate that CO has a modulatory role on microglia-to-neuron communication, promoting neuroprotection in a non-cell autonomous manner. CO enhances the microglial release of neurotrophic factors and blocks exacerbated microglial inflammation. CO improvement of microglial neurotrophism under non-inflammatory conditions is here described for the first time.


Subject(s)
Microglia , Neuroprotective Agents , Animals , Carbon Monoxide/metabolism , Carbon Monoxide/pharmacology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats
8.
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
9.
Brain Commun ; 2(2): fcaa135, 2020.
Article in English | MEDLINE | ID: mdl-33225275

ABSTRACT

Donnai-Barrow syndrome, a genetic disorder associated to LRP2 (low-density lipoprotein receptor 2/megalin) mutations, is characterized by unexplained neurological symptoms and intellectual deficits. Megalin is a multifunctional endocytic clearance cell-surface receptor, mostly described in epithelial cells. This receptor is also expressed in the CNS, mainly in neurons, being involved in neurite outgrowth and neuroprotective mechanisms. Yet, the mechanisms involved in the regulation of megalin in the CNS are poorly understood. Using transthyretin knockout mice, a megalin ligand, we found that transthyretin positively regulates neuronal megalin levels in different CNS areas, particularly in the hippocampus. Transthyretin is even able to rescue megalin downregulation in transthyretin knockout hippocampal neuronal cultures, in a positive feedback mechanism via megalin. Importantly, transthyretin activates a regulated intracellular proteolysis mechanism of neuronal megalin, producing an intracellular domain, which is translocated to the nucleus, unveiling megalin C-terminal as a potential transcription factor, able to regulate gene expression. We unveil that neuronal megalin reduction affects physiological neuronal activity, leading to decreased neurite number, length and branching, and increasing neuronal susceptibility to a toxic insult. Finally, we unravel a new unexpected role of megalin in synaptic plasticity, by promoting the formation and maturation of dendritic spines, and contributing for the establishment of active synapses, both in in vitro and in vivo hippocampal neurons. Moreover, these structural and synaptic roles of megalin impact on learning and memory mechanisms, since megalin heterozygous mice show hippocampal-related memory and learning deficits in several behaviour tests. Altogether, we unveil a complete novel role of megalin in the physiological neuronal activity, mainly in synaptic plasticity with impact in learning and memory. Importantly, we contribute to disclose the molecular mechanisms underlying the cognitive and intellectual disabilities related to megalin gene pathologies.

10.
Sci Signal ; 13(650)2020 09 22.
Article in English | MEDLINE | ID: mdl-32963013

ABSTRACT

Alcohol abuse adversely affects the lives of millions of people worldwide. Deficits in synaptic transmission and in microglial function are commonly found in human alcohol abusers and in animal models of alcohol intoxication. Here, we found that a protocol simulating chronic binge drinking in male mice resulted in aberrant synaptic pruning and substantial loss of excitatory synapses in the prefrontal cortex, which resulted in increased anxiety-like behavior. Mechanistically, alcohol intake increased the engulfment capacity of microglia in a manner dependent on the kinase Src, the subsequent activation of the transcription factor NF-κB, and the consequent production of the proinflammatory cytokine TNF. Pharmacological blockade of Src activation or of TNF production in microglia, genetic ablation of Tnf, or conditional ablation of microglia attenuated aberrant synaptic pruning, thereby preventing the neuronal and behavioral effects of the alcohol. Our data suggest that aberrant pruning of excitatory synapses by microglia may disrupt synaptic transmission in response to alcohol abuse.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/drug effects , Ethanol/administration & dosage , Neuronal Plasticity/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Animals , Anxiety/psychology , Behavior, Animal/physiology , Cells, Cultured , Central Nervous System Depressants/administration & dosage , Ethanol/blood , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Neuronal Plasticity/physiology , Synapses/physiology , Synaptic Transmission/physiology , Tumor Necrosis Factor-alpha/metabolism
11.
Sci Rep ; 10(1): 13326, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32769999

ABSTRACT

The development of substance abuse problems occurs due to a diverse combination of risk factors. Among these risks, studies have reported depression and early-life stress as of importance. These two factors often occur simultaneously, however, there is a lack of understanding of how their combined effect may impact vulnerability to drug abuse in adolescence. The present study used rats with different vulnerability to depression (Wistar and Wistar-Kyoto) to investigate the impact of maternal separation (MS) on emotional state and drug addiction vulnerability during the adolescence period. Mothers and their litters were subjected to MS (180 min/day) from postnatal day 2 to 14. The offspring emotional state was assessed by observing their exploratory behavior. Drug abuse vulnerability was assessed through conditioning to cocaine. MS impacted the emotional state in both strains. Wistar responded with increased exploration, while Wistar-Kyoto increased anxiety-like behaviours. Despite the different coping strategies displayed by the two strains when challenged with the behavioural tests, drug conditioning was equally impacted by MS in both strains. Early-life stress appears to affect drug abuse vulnerability in adolescence independently of a depression background, suggesting emotional state as the main driving risk factor.


Subject(s)
Adverse Childhood Experiences/psychology , Stress, Psychological/psychology , Substance-Related Disorders/etiology , Animals , Animals, Newborn/psychology , Anxiety/complications , Anxiety/psychology , Cocaine/adverse effects , Depression/complications , Depression/psychology , Exploratory Behavior/physiology , Female , Humans , Male , Maternal Deprivation , Rats , Rats, Inbred WKY , Risk Factors , Substance-Related Disorders/psychology
12.
Cell Death Dis ; 11(6): 415, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488063

ABSTRACT

Growing evidences suggest that sustained neuroinflammation, caused by microglia overactivation, is implicated in the development and aggravation of several neurological and psychiatric disorders. In some pathological conditions, microglia produce increased levels of cytotoxic and inflammatory mediators, such as tumor necrosis factor alpha (TNF-α), which can reactivate microglia in a positive feedback mechanism. However, specific molecular mediators that can be effectively targeted to control TNF-α-mediated microglia overactivation, are yet to be uncovered. In this context, we aim to identify novel TNF-α-mediated micro(mi)RNAs and to dissect their roles in microglia activation, as well as to explore their impact on the cellular communication with neurons. A miRNA microarray, followed by RT-qPCR validation, was performed on TNF-α-stimulated primary rat microglia. Gain- and loss-of-function in vitro assays and proteomic analysis were used to dissect the role of miR-342 in microglia activation. Co-cultures of microglia with hippocampal neurons, using a microfluidic system, were performed to understand the impact on neurotoxicity. Stimulation of primary rat microglia with TNF-α led to an upregulation of Nos2, Tnf, and Il1b mRNAs. In addition, ph-NF-kB p65 levels were also increased. miRNA microarray analysis followed by RT-qPCR validation revealed that TNF-α stimulation induced the upregulation of miR-342. Interestingly, miR-342 overexpression in N9 microglia was sufficient to activate the NF-kB pathway by inhibiting BAG-1, leading to increased secretion of TNF-α and IL-1ß. Conversely, miR-342 inhibition led to a strong decrease in the levels of these cytokines after TNF-α activation. In fact, both TNF-α-stimulated and miR-342-overexpressing microglia drastically affected neuron viability. Remarkably, increased levels of nitrites were detected in the supernatants of these co-cultures. Globally, our findings show that miR-342 is a crucial mediator of TNF-α-mediated microglia activation and a potential target to tackle microglia-driven neuroinflammation.


Subject(s)
MicroRNAs/metabolism , Microglia/pathology , NF-kappa B/metabolism , Neurotoxins/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Animals , Animals, Newborn , Cell Line , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Inflammation/genetics , Inflammation/pathology , Mice, Inbred C57BL , MicroRNAs/genetics , Microglia/drug effects , Microglia/metabolism , Models, Biological , Rats, Wistar , Transcription Factors/metabolism
13.
Cell Rep ; 31(12): 107796, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579923

ABSTRACT

Nervous tissue homeostasis requires the regulation of microglia activity. Using conditional gene targeting in mice, we demonstrate that genetic ablation of the small GTPase Rhoa in adult microglia is sufficient to trigger spontaneous microglia activation, producing a neurological phenotype (including synapse and neuron loss, impairment of long-term potentiation [LTP], formation of ß-amyloid plaques, and memory deficits). Mechanistically, loss of Rhoa in microglia triggers Src activation and Src-mediated tumor necrosis factor (TNF) production, leading to excitotoxic glutamate secretion. Inhibiting Src in microglia Rhoa-deficient mice attenuates microglia dysregulation and the ensuing neurological phenotype. We also find that the Rhoa/Src signaling pathway is disrupted in microglia of the APP/PS1 mouse model of Alzheimer disease and that low doses of Aß oligomers trigger microglia neurotoxic polarization through the disruption of Rhoa-to-Src signaling. Overall, our results indicate that disturbing Rho GTPase signaling in microglia can directly cause neurodegeneration.


Subject(s)
Aging/pathology , Microglia/pathology , Nerve Degeneration/pathology , Neurons/metabolism , rhoA GTP-Binding Protein/deficiency , Aging/metabolism , Amyloid beta-Peptides/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Cell Polarity , Cell Survival , Mice, Inbred C57BL , Microglia/metabolism , Phenotype , Synapses/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
14.
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
15.
J Comp Psychol ; 133(4): 452-462, 2019 11.
Article in English | MEDLINE | ID: mdl-30896232

ABSTRACT

Prosocial behavior in rats is known to occur in response to a familiar rat's distress, but the motivations underlying prosocial behavior remain elusive. In this study, we adapted the experimental setting of Ben-Ami Bartal, Decety, and Mason (2011) to explore different motivations behind helping behavior in adolescent rats. In the original setting, a free rat is placed in an arena where a cagemate is trapped inside a restrainer that can only be opened from the outside by the free rat. Here we added a dark compartment to the experimental setting that allowed the free rat to escape the arena and the distress evoked by the trapped cagemate, based on rodents' aversion to bright areas. As a control, we tested rats in the same arena but with the door to the dark area closed. Our results showed that all free rats, except one in the escape condition, learned to open the restrainer's door. However, in the escape condition, rats took significantly longer to open the restrainer to the cagemates when compared with rats that could not escape. To further explore the motivations underlying these group differences in door-opening latencies, we measured both rats' behavior. We found that struggling behavior (i.e., distress) in the trapped rat did not affect door-opening, whereas exploratory behavior (i.e., proactive/positive behavior) in both rats contributed to shorter times. Our results highlight that adolescent rats show prosocial behavior even when they can escape without helping and contribute to demonstrate the role of positive emotional states in prosocial behavior. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Subject(s)
Behavior, Animal/physiology , Helping Behavior , Psychological Distress , Psychomotor Performance/physiology , Animals , Male , Rats , Rats, Wistar
16.
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
17.
Toxicol Lett ; 283: 21-31, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29129797

ABSTRACT

Alcohol use disorders affect millions of people worldwide causing huge social and economic burden on modern society. Excessive alcohol consumption or intoxication provokes severe damage to the body inducing immune suppression, liver damage and neurological disorder. In the central nervous system (CNS), alcohol exposure can lead to neuronal loss, cognitive decline, motor dysfunction, inflammation and impairment of neuroimmune responses. Glial cells, from which microglia represent roughly 10-15%, are primary modulators of the neuroimmune responses and inflammation in the CNS. Here we overview literature relating alcohol exposure with microglia activation and brain inflammation, highlighting that microglia are critical regulators of alcohol responses in the CNS. Different studies indicate that alcohol intake alters the microglial activation spectrum, with the microglial response varying according to the dose, duration, and pattern of alcohol administration. Presently, further investigation is required to establish whether microglia dysfunction initiates or simply amplifies the neurotoxicity of alcohol in the brain. Such knowledge can be greatly facilitated by the use of microglia-specific genetic targeting in animal models and will be critical for the development of better therapeutics for mitigating the neurotoxicity induced by alcohol.


Subject(s)
Alcoholism/immunology , Alcoholism/pathology , Microglia/immunology , Microglia/pathology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Animals , Central Nervous System Depressants/toxicity , Encephalitis/pathology , Ethanol/toxicity , Humans
18.
Biochim Biophys Acta Gene Regul Mech ; 1860(6): 685-694, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28274785

ABSTRACT

The differential expression of mRNAs containing tandem alternative 3' UTRs, achieved by mechanisms of alternative polyadenylation and post-transcriptional regulation, has been correlated with a variety of cellular states. In differentiated cells and brain tissues there is a general use of distal polyadenylation signals, originating mRNAs with longer 3' UTRs, in contrast with proliferating cells and other tissues such as testis, where most mRNAs contain shorter 3' UTRs. Although cell type and state are relevant in many biological processes, how these mechanisms occur in specific brain cell types is still poorly understood. Rac1 is a member of the Rho family of small GTPases with essential roles in multiple cellular processes, including cell differentiation and axonal growth. Here we used different brain cell types and tissues, including oligodendrocytes, microglia, astrocytes, cortical and hippocampal neurons, and optical nerve, to show that classical Rho GTPases express mRNAs with alternative 3' UTRs differently, by gene- and cell- specific mechanisms. In particular, we show that Rac1 originate mRNA isoforms with longer 3' UTRs specifically during neurite growth of cortical, but not hippocampal neurons. Furthermore, we demonstrate that the longest Rac1 3' UTR is necessary for driving the mRNA to the neurites, and also for neurite outgrowth in cortical neurons. Our results indicate that the expression of Rac1 longer 3' UTR is a gene and cell-type specific mechanism in the brain, with a new physiological function in cortical neuron differentiation.


Subject(s)
3' Untranslated Regions/physiology , Cerebral Cortex/enzymology , Gene Expression Regulation, Enzymologic/physiology , Neurites/enzymology , rac1 GTP-Binding Protein/biosynthesis , Animals , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Humans , Rats , Rats, Wistar , rac1 GTP-Binding Protein/genetics
19.
Lab Anim ; 51(2): 147-159, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27252027

ABSTRACT

Ketamine administration has been associated with controversial behavioural impairments and psychotic episodes. Even though ketamine alone and in combination with midazolam or dexmedetomidine are frequently used in laboratory animals, the side-effects of such protocols are not well known. Therefore, our aim was to evaluate the effects of ketamine alone and in combination with midazolam or dexmedetomidine on emotional reactivity, as well as the effects on learning and memory in adult rats at least 48 h after anaesthesia. The evaluation of the potential influence of 100 mg/kg ketamine administered alone and in combination with midazolam (5 mg/kg), or dexmedetomidine (0.25 mg/kg) on spatial learning and recognition memory was studied in adult Wistar rats using the radial maze as well as object recognition and location tests. The influence of these combinations on emotional reactivity was investigated using the new exploration test and the elevated plus maze. Results showed that ketamine alone or in combination with midazolam or dexmedetomidine affected neither spatial and recognition memory, nor emotional reactivity. These results reinforce the safe clinical use of ketamine and its combinations in rats in a research context since the administration of these anaesthetic combinations did not produce significant changes with regard to spatial and recognition memory or emotional reactivity. Furthermore, these results indicate that the quality of scientific data produced in adult rat neurobehavioural research is not jeopardized by the use of these anaesthetic protocols.


Subject(s)
Anesthetics, Dissociative/adverse effects , Emotions/drug effects , Hypnotics and Sedatives/adverse effects , Learning/drug effects , Memory/drug effects , Rats/physiology , Animals , Dexmedetomidine/adverse effects , Drug Combinations , Ketamine/adverse effects , Male , Midazolam/adverse effects , Rats, Wistar
20.
PLoS One ; 11(11): e0165465, 2016.
Article in English | MEDLINE | ID: mdl-27802308

ABSTRACT

Bone repair is a specialized type of wound repair controlled by complex multi-factorial events. The nervous system is recognized as one of the key regulators of bone mass, thereby suggesting a role for neuronal pathways in bone homeostasis. However, in the context of bone injury and repair, little is known on the interplay between the nervous system and bone. Here, we addressed the neuropeptide Y (NPY) neuronal arm during the initial stages of bone repair encompassing the inflammatory response and ossification phases in femoral-defect mouse model. Spatial and temporal analysis of transcriptional and protein levels of NPY and its receptors, Y1R and Y2R, reported to be involved in bone homeostasis, was performed in bone, dorsal root ganglia (DRG) and hypothalamus after femoral injury. The results showed that NPY system activity is increased in a time- and space-dependent manner during bone repair. Y1R expression was trigged in both bone and DRG throughout the inflammatory phase, while a Y2R response was restricted to the hypothalamus and at a later stage, during the ossification step. Our results provide new insights into the involvement of NPY neuronal pathways in bone repair.


Subject(s)
Femur/injuries , Hypothalamus/physiology , Neural Pathways/physiology , Neuropeptide Y/metabolism , Wound Healing , Animals , Femur/innervation , Femur/pathology , Femur/physiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Neuropeptide Y/analysis , Neuropeptide Y/genetics , RNA, Messenger/genetics , Receptors, Neuropeptide Y/analysis , Receptors, Neuropeptide Y/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...