Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 14(1): 209, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796504

ABSTRACT

Stress affects the brain and alters its neuroarchitecture and function; these changes can be severe and lead to psychiatric disorders. Recent evidence suggests that astrocytes and microglia play an essential role in the stress response by contributing to the maintenance of cerebral homeostasis. These cells respond rapidly to all stimuli that reach the brain, including stressors. Here, we used a recently validated rodent model of post-traumatic stress disorder in which rats can be categorized as resilient or vulnerable after acute inescapable footshock stress. We then investigated the functional, molecular, and morphological determinants of stress resilience and vulnerability in the prefrontal cortex, focusing on glial and neuronal cells. In addition, we examined the effects of a single subanesthetic dose of ketamine, a fast-acting antidepressant recently approved for the treatment of resistant depression and proposed for other stress-related psychiatric disorders. The present results suggest a prompt glial cell response and activation of the NF-κB pathway after acute stress, leading to an increase in specific cytokines such as IL-18 and TNF-α. This response persists in vulnerable individuals and is accompanied by a significant change in the levels of critical glial proteins such as S100B, CD11b, and CX43, brain trophic factors such as BDNF and FGF2, and proteins related to dendritic arborization and synaptic architecture such as MAP2 and PSD95. Administration of ketamine 24 h after the acute stress event rescued many of the changes observed in vulnerable rats, possibly contributing to support brain homeostasis. Overall, our results suggest that pivotal events, including reactive astrogliosis, changes in brain trophic factors, and neuronal damage are critical determinants of vulnerability to acute traumatic stress and confirm the therapeutic effect of acute ketamine against the development of stress-related psychiatric disorders.


Subject(s)
Astrocytes , Disease Models, Animal , Ketamine , Microglia , Stress Disorders, Post-Traumatic , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microglia/drug effects , Microglia/metabolism , Male , Rats , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Stress, Psychological/metabolism , Rats, Sprague-Dawley , NF-kappa B/metabolism
2.
Brain Behav Immun ; 118: 380-397, 2024 May.
Article in English | MEDLINE | ID: mdl-38485064

ABSTRACT

Autoantibodies directed against the GluA3 subunit (anti-GluA3 hIgGs) of AMPA receptors have been identified in 20%-25% of patients with frontotemporal lobar degeneration (FTLD). Data from patients and in vitro/ex vivo pre-clinical studies indicate that anti-GluA3 hIgGs negatively affect glutamatergic neurotransmission. However, whether and how the chronic presence of anti-GluA3 hIgGs triggers synaptic dysfunctions and the appearance of FTLD-related neuropathological and behavioural signature has not been clarified yet. To address this question, we developed and characterized a pre-clinical mouse model of passive immunization with anti-GluA3 hIgGs purified from patients. In parallel, we clinically compared FTLD patients who were positive for anti-GluA3 hIgGs to negative ones. Clinical data showed that the presence of anti-GluA3 hIgGs defined a subgroup of patients with distinct clinical features. In the preclinical model, anti-GluA3 hIgGs administration led to accumulation of phospho-tau in the postsynaptic fraction and dendritic spine loss in the prefrontal cortex. Remarkably, the preclinical model exhibited behavioural disturbances that mostly reflected the deficits proper of patients positive for anti-GluA3 hIgGs. Of note, anti-GluA3 hIgGs-mediated alterations were rescued in the animal model by enhancing glutamatergic neurotransmission with a positive allosteric modulator of AMPA receptors. Overall, our study clarified the contribution of anti-GluA3 autoantibodies to central nervous system symptoms and pathology and identified a specific subgroup of FTLD patients. Our findings will be instrumental in the development of a therapeutic personalised medicine strategy for patients positive for anti-GluA3 hIgGs.


Subject(s)
Autoantibodies , Frontotemporal Lobar Degeneration , Animals , Humans , Mice , Autoantibodies/metabolism , Frontotemporal Dementia , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Receptors, AMPA , Synaptic Transmission , tau Proteins/metabolism
3.
Transl Psychiatry ; 13(1): 62, 2023 02 18.
Article in English | MEDLINE | ID: mdl-36806044

ABSTRACT

Stress represents a main risk factor for psychiatric disorders. Whereas it is known that even a single trauma may induce psychiatric disorders in humans, the mechanisms of vulnerability to acute stressors have been little investigated. In this study, we generated a new animal model of resilience/vulnerability to acute footshock (FS) stress in rats and analyzed early functional, molecular, and morphological determinants of stress vulnerability at tripartite glutamate synapses in the prefrontal cortex (PFC). We found that adult male rats subjected to FS can be deemed resilient (FS-R) or vulnerable (FS-V), based on their anhedonic phenotype 24 h after stress exposure, and that these two populations are phenotypically distinguishable up to two weeks afterwards. Basal presynaptic glutamate release was increased in the PFC of FS-V rats, while depolarization-evoked glutamate release and synapsin I phosphorylation at Ser9 were increased in both FS-R and FS-V. In FS-R and FS-V rats the synaptic expression of GluN2A and apical dendritic length of prelimbic PFC layers II-III pyramidal neurons were decreased, while BDNF expression was selectively reduced in FS-V. Depolarization-evoked (carrier-mediated) glutamate release from astroglia perisynaptic processes (gliosomes) was selectively increased in the PFC of FS-V rats, while GLT1 and xCt levels were higher and GS expression reduced in purified PFC gliosomes from FS-R. Overall, we show for the first time that the application of the sucrose intake test to rats exposed to acute FS led to the generation of a novel animal model of resilience/vulnerability to acute stress, which we used to identify early determinants of maladaptive response related to behavioral vulnerability to stress.


Subject(s)
Astrocytes , Glutamic Acid , Humans , Adult , Male , Animals , Rats , Models, Animal , Prefrontal Cortex , Synapses
4.
Biomolecules ; 12(9)2022 08 27.
Article in English | MEDLINE | ID: mdl-36139030

ABSTRACT

Palmitoylethanolamide (PEA), the naturally occurring amide of ethanolamine and palmitic acid, is an endogenous lipid compound endowed with a plethora of pharmacological functions, including analgesic, neuroprotective, immune-modulating, and anti-inflammatory effects. Although the properties of PEA were first characterized nearly 65 years ago, the identity of the receptor mediating these actions has long remained elusive, causing a period of research stasis. In the last two decades, a renewal of interest in PEA occurred, and a series of interesting studies have demonstrated the pharmacological properties of PEA and clarified its mechanisms of action. Recent findings showed the ability of formulations containing PEA in promoting oligodendrocyte differentiation, which represents the first step for the proper formation of myelin. This evidence opens new and promising research opportunities. White matter defects have been detected in a vast and heterogeneous group of diseases, including age-related neurodegenerative disorders. Here, we summarize the history and pharmacology of PEA and discuss its therapeutic potential in restoring white matter defects.


Subject(s)
Palmitic Acid , White Matter , Amides , Analgesics , Anti-Inflammatory Agents/pharmacology , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Palmitic Acids/therapeutic use
5.
Pharmaceutics ; 14(6)2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35745837

ABSTRACT

A combination of anticancer drugs and chemosensitizing agents has been approached as a promising strategy to potentiate chemotherapy and reduce toxicity in aggressive and chemoresistant cancers, like hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and pancreatic ductal adenocarcinoma (PDAC). In the present study, the ability of caryophyllane sesquiterpenes to potentiate sorafenib efficacy was studied in HCC, CCA, and PDAC cell models, focusing on the modulation of STAT3 signaling and ABC transporters; tolerability studies in normal cells were also performed. Results showed that the combination of sorafenib and caryophyllane sesquiterpenes synergized the anticancer drug, especially in pancreatic Bx-PC3 adenocarcinoma cells; a similar trend, although with lower efficacy, was found for the standard ABC transporter inhibitors. Synergistic effects were associated with a modulation of MDR1 (or Pgp) and MRP transporters, both at gene and protein level; moreover, activation of STAT3 cascade and cell migration appeared significantly affected, suggesting that the STAT3/ABC-transporters axis finely regulated efficacy and chemoresistance to sorafenib, thus appearing as a suitable target to overcome drawbacks of sorafenib-based chemotherapy in hepato-biliary-pancreatic cancers. Present findings strengthen the interest in caryophyllane sesquiterpenes as chemosensitizing and chemopreventive agents and contribute to clarifying drug resistance mechanisms in HCC, CCA, and PDAC cancers and to developing possible novel therapeutic strategies.

6.
Biomedicines ; 10(6)2022 May 26.
Article in English | MEDLINE | ID: mdl-35740258

ABSTRACT

Oligodendrocytes are cells fundamental for brain functions as they form the myelin sheath and feed axons. They perform these critical functions thanks to the cooperation with other glial cells, mainly astrocytes. The astrocyte/oligodendrocyte crosstalk needs numerous mediators and receptors, such as peroxisome proliferator-activated receptors (PPARs). PPAR agonists promote oligodendrocyte precursor cells (OPCs) maturation in myelinating oligodendrocytes. In the Alzheimer's disease brain, deposition of beta-amyloid (Aß) has been linked to several alterations, including astrogliosis and changes in OPCs maturation. However, very little is known about the molecular mechanisms. Here, we investigated for the first time the maturation of OPCs co-cultured with astrocytes in an in vitro model of Aß1-42 toxicity. We also tested the potential beneficial effect of the anti-inflammatory and neuroprotective composite palmitoylethanolamide and luteolin (co-ultra PEALut), which is known to engage the isoform alfa of the PPARs. Our results show that Aß1-42 triggers astrocyte reactivity and inflammation and reduces the levels of growth factors important for OPCs maturation. Oligodendrocytes indeed show low cell surface area and few arborizations. Co-ultra PEALut counteracts the Aß1-42-induced inflammation and astrocyte reactivity preserving the morphology of co-cultured oligodendrocytes through a mechanism that in some cases involves PPAR-α. This is the first evidence of the negative effects exerted by Aß1-42 on astrocyte/oligodendrocyte crosstalk and discloses a never-explored co-ultra PEALut ability in restoring oligodendrocyte homeostasis.

7.
Front Pharmacol ; 13: 759626, 2022.
Article in English | MEDLINE | ID: mdl-35370690

ABSTRACT

Stress represents a major risk factor for psychiatric disorders, including post-traumatic stress disorder (PTSD). Recently, we dissected the destabilizing effects of acute stress on the excitatory glutamate system in the prefrontal cortex (PFC). Here, we assessed the effects of single subanesthetic administration of ketamine (10 mg/kg) on glutamate transmission and dendritic arborization in the PFC of footshock (FS)-stressed rats, along with changes in depressive, anxious, and fear extinction behaviors. We found that ketamine, while inducing a mild increase of glutamate release in the PFC of naïve rats, blocked the acute stress-induced enhancement of glutamate release when administered 24 or 72 h before or 6 h after FS. Accordingly, the treatment with ketamine 6 h after FS also reduced the stress-dependent increase of spontaneous excitatory postsynaptic current (sEPSC) amplitude in prelimbic (PL)-PFC. At the same time, ketamine injection 6 h after FS was found to rescue apical dendritic retraction of pyramidal neurons induced by acute stress in PL-PFC and facilitated contextual fear extinction. These results show rapid effects of ketamine in animals subjected to acute FS, in line with previous studies suggesting a therapeutic action of the drug in PTSD models. Our data are consistent with a mechanism of ketamine involving re-establishment of synaptic homeostasis, through restoration of glutamate release, and structural remodeling of dendrites.

8.
Biomolecules ; 11(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921556

ABSTRACT

The available treatments for patients affected by Alzheimer's disease (AD) are not curative. Numerous clinical trials have failed during the past decades. Therefore, scientists need to explore new avenues to tackle this disease. In the present review, we briefly summarize the pathological mechanisms of AD known so far, based on which different therapeutic tools have been designed. Then, we focus on a specific approach that is targeting astrocytes. Indeed, these non-neuronal brain cells respond to any insult, injury, or disease of the brain, including AD. The study of astrocytes is complicated by the fact that they exert a plethora of homeostatic functions, and their disease-induced changes could be context-, time-, and disease specific. However, this complex but fervent area of research has produced a large amount of data targeting different astrocytic functions using pharmacological approaches. Here, we review the most recent literature findings that have been published in the last five years to stimulate new hypotheses and ideas to work on, highlighting the peculiar ability of palmitoylethanolamide to modulate astrocytes according to their morpho-functional state, which ultimately suggests a possible potential disease-modifying therapeutic approach for AD.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Gliosis/metabolism , Neuroprotective Agents/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Animals , Astrocytes/drug effects , Gliosis/drug therapy , Gliosis/etiology , Humans , Neuroprotective Agents/therapeutic use
9.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471239

ABSTRACT

BACKGROUND: At the earliest stage of Alzheimer's disease (AD), although patients are still asymptomatic, cerebral alterations have already been triggered. In addition to beta amyloid (Aß) accumulation, both glial alterations and neuroinflammation have been documented at this stage. Starting treatment at this prodromal AD stage could be a valuable therapeutic strategy. AD requires long-term care; therefore, only compounds with a high safety profile can be used, such as the new formulation containing palmitoylethanolamide and luteolin (co-ultra PEALut) already approved for human use. Therefore, we investigated it in an in vivo pharmacological study that focused on the prodromal stage of AD. METHODS: We tested the anti-inflammatory and neuroprotective effects of co-ultra PEALut (5 mg/Kg) administered for 14 days in rats that received once, 5 µg Aß(1-42) into the hippocampus. RESULTS: Glial activation and elevated levels of proinflammatory mediators were observed in Aß-infused rats. Early administration of co-ultra PEALut prevented the Aß-induced astrogliosis and microgliosis, the upregulation in gene expression of pro-inflammatory cytokines and enzymes, as well as the reduction of mRNA levels BDNF and GDNF. Our findings also highlight an important neuroprotective effect of co-ultra PEALut treatment, which promoted neuronal survival. CONCLUSIONS: Our results reveal the presence of cellular and molecular modifications in the prodromal stage of AD. Moreover, the data presented here demonstrate the ability of co-ultra PEALut to normalize such Aß-induced alterations, suggesting it as a valuable therapeutic strategy.


Subject(s)
Alzheimer Disease/drug therapy , Ethanolamines/therapeutic use , Gliosis/drug therapy , Luteolin/therapeutic use , Neuroprotective Agents/therapeutic use , Alzheimer Disease/pathology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cytokines/genetics , Cytokines/metabolism , Drug Combinations , Ethanolamines/administration & dosage , Ethanolamines/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Gliosis/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Luteolin/administration & dosage , Luteolin/pharmacology , Male , Neuroglia/drug effects , Neuroglia/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Prodromal Symptoms , Rats , Rats, Sprague-Dawley
10.
Front Pharmacol ; 10: 644, 2019.
Article in English | MEDLINE | ID: mdl-31244658

ABSTRACT

Old age is a risk factor for Alzheimer's disease (AD), which is characterized by hippocampal impairment together with substantial changes in glial cell functions. Are these alterations due to the disease progression or are they a consequence of aging? To start addressing this issue, we studied the expression of specific astrocytic and microglial structural and functional proteins in a validated transgenic model of AD (3×Tg-AD). These mice develop both amyloid plaques and neurofibrillary tangles, and initial signs of the AD-like pathology have been documented as early as three months of age. We compared male 3×Tg-AD mice at 6 and 12 months of age with their wild-type age-matched counterparts. We also investigated neurons by examining the expression of both the microtubule-associated protein 2 (MAP2), a neuronal structural protein, and the brain-derived neurotrophic factor (BDNF). The latter is indeed a crucial indicator for synaptic plasticity and neurogenesis/neurodegeneration. Our results show that astrocytes are more susceptible to aging than microglia, regardless of mouse genotype. Moreover, we discovered significant age-dependent alterations in the expression of proteins responsible for astrocyte-astrocyte and astrocyte-neuron communication, as well as a significant age-dependent decline in BDNF expression. Our data promote further research on the unexplored role of astroglia in both physiological and pathological aging.

11.
Front Pharmacol ; 10: 1656, 2019.
Article in English | MEDLINE | ID: mdl-32063858

ABSTRACT

Among the diverse cell types included in the general population named glia, astrocytes emerge as being the focus of a growing body of research aimed at characterizing their heterogeneous and complex functions. Alterations of both their morphology and activities have been linked to a variety of neurological diseases. One crucial physiological need satisfied by astrocytes is the cleansing of the cerebral tissue from waste molecules. Several data demonstrate that aquaporin-4 (AQP-4), a protein expressed by astrocytes, is crucially important for facilitating the removal of waste products from the brain. Aquaporins are water channels found in all district of the human organism and the most abundant isoform in the brain is AQP-4. This protein is involved in a myriad of astrocytic activities, including calcium signal transduction, potassium buffering, synaptic plasticity, astrocyte migration, glial scar formation and neuroinflammation. The highest density of AQP-4 is found at the astrocytic domains closest to blood vessels, the endfeet that envelop brain vessels, with low to zero expression in other astrocytic membrane regions. Increased AQP-4 expression and loss of polarization have recently been documented in altered physiological conditions. Here we review the latest findings related to aging and Alzheimer's disease (AD) on this topic, as well as the available knowledge on pharmacological tools to target AQP-4.

12.
Oxid Med Cell Longev ; 2018: 4720532, 2018.
Article in English | MEDLINE | ID: mdl-29576849

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder responsible for the majority of dementia cases in elderly people. It is widely accepted that the main hallmarks of AD are not only senile plaques and neurofibrillary tangles but also reactive astrogliosis, which often precedes detrimental deposits and neuronal atrophy. Such phenomenon facilitates the regeneration of neural networks; however, under some circumstances, like in AD, reactive astrogliosis is detrimental, depriving neurons of the homeostatic support, thus contributing to neuronal loss. We investigated the presence of reactive astrogliosis in 3×Tg-AD mice and the effects of palmitoylethanolamide (PEA), a well-documented anti-inflammatory molecule, by in vitro and in vivo studies. In vitro results revealed a basal reactive state in primary cortical 3×Tg-AD-derived astrocytes and the ability of PEA to counteract such phenomenon and improve viability of 3×Tg-AD-derived neurons. In vivo observations, performed using ultramicronized- (um-) PEA, a formulation endowed with best bioavailability, confirmed the efficacy of this compound. Moreover, the schedule of treatment, mimicking the clinic use (chronic daily administration), revealed its beneficial pharmacological properties in dampening reactive astrogliosis and promoting the glial neurosupportive function. Collectively, our results encourage further investigation on PEA effects, suggesting it as an alternative or adjunct treatment approach for innovative AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Disease Models, Animal , Ethanolamines/pharmacology , Gliosis/drug therapy , Palmitic Acids/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amides , Animals , Astrocytes/metabolism , Astrocytes/pathology , Gliosis/genetics , Gliosis/metabolism , Gliosis/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Transl Psychiatry ; 8(1): 32, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29382825

ABSTRACT

In an aging society, Alzheimer's disease (AD) exerts an increasingly serious health and economic burden. Current treatments provide inadequate symptomatic relief as several distinct pathological processes are thought to underlie the decline of cognitive and neural function seen in AD. This suggests that the efficacy of treatment requires a multitargeted approach. In this context, palmitoylethanolamide (PEA) provides a novel potential adjunct therapy that can be incorporated into a multitargeted treatment strategy. We used young (6-month-old) and adult (12-month-old) 3×Tg-AD mice that received ultramicronized PEA (um-PEA) for 3 months via a subcutaneous delivery system. Mice were tested with a range of cognitive and noncognitive tasks, scanned with magnetic resonance imaging/magnetic resonance spectroscopy (MRI/MRS), and neurochemical release was assessed by microdialysis. Potential neuropathological mechanisms were assessed postmortem by western blot, reverse transcription-polymerase chain reaction (RT-PCR), and immunofluorescence. Our data demonstrate that um-PEA improves learning and memory, and ameliorates both the depressive and anhedonia-like phenotype of 3×Tg-AD mice. Moreover, it reduces Aß formation, the phosphorylation of tau proteins, and promotes neuronal survival in the CA1 subregion of the hippocampus. Finally, um-PEA normalizes astrocytic function, rebalances glutamatergic transmission, and restrains neuroinflammation. The efficacy of um-PEA is particularly potent in younger mice, suggesting its potential as an early treatment. These data demonstrate that um-PEA is a novel and effective promising treatment for AD with the potential to be integrated into a multitargeted treatment strategy in combination with other drugs. Um-PEA is already registered for human use. This, in combination with our data, suggests the potential to rapidly proceed to clinical use.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/deficiency , Anti-Inflammatory Agents/pharmacology , CA1 Region, Hippocampal/drug effects , Cognitive Dysfunction/drug therapy , Ethanolamines/pharmacology , Inflammation/drug therapy , Learning/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Palmitic Acids/pharmacology , tau Proteins/drug effects , Age Factors , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amides , Animals , Anti-Inflammatory Agents/administration & dosage , Behavior, Animal/drug effects , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Ethanolamines/administration & dosage , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Memory Disorders/immunology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Neuroprotective Agents/administration & dosage , Palmitic Acids/administration & dosage
14.
Mol Autism ; 9: 66, 2018.
Article in English | MEDLINE | ID: mdl-30603062

ABSTRACT

Background: Neuroglial cells that provide homeostatic support and form defence of the nervous system contribute to all neurological disorders. We analyzed three major types of neuroglia, astrocytes, oligodendrocytes, and microglia in the brains of an animal model of autism spectrum disorder, in which rats were exposed prenatally to antiepileptic and mood stabilizer drug valproic acid; this model being of acknowledged clinical relevance. Methods: We tested the autistic-like behaviors of valproic acid-prenatally exposed male rats by performing isolation-induced ultrasonic vocalizations, the three-chamber test, and the hole board test. To account for human infancy, adolescence, and adulthood, such tasks were performed at postnatal day 13, postnatal day 35, and postnatal day 90, respectively. After sacrifice, we examined gene and protein expression of specific markers of neuroglia in hippocampus, prefrontal cortex, and cerebellum, these brain regions being associated with autism spectrum disorder pathogenesis. Results: Infant offspring of VPA-exposed dams emitted less ultrasonic vocalizations when isolated from their mothers and siblings and, in adolescence and adulthood, they showed altered sociability in the three chamber test and increased stereotypic behavior in the hole board test. Molecular analyses indicate that prenatal valproic acid exposure affects all types of neuroglia, mainly causing transcriptional modifications. The most prominent changes occur in prefrontal cortex and in the hippocampus of autistic-like animals; these changes are particularly evident during infancy and adolescence, while they appear to be mitigated in adulthood. Conclusions: Neuroglial pathological phenotype in autism spectrum disorder rat model appears to be rather mild with little signs of widespread and chronic neuroinflammation.


Subject(s)
Autistic Disorder/pathology , Brain/pathology , Neuroglia/pathology , Animals , Autistic Disorder/etiology , Autistic Disorder/physiopathology , Brain/drug effects , Female , Male , Neuroglia/drug effects , Rats , Rats, Wistar , Stereotyped Behavior , Valproic Acid/pharmacology , Valproic Acid/toxicity , Vocalization, Animal
15.
Methods Mol Biol ; 1727: 333-341, 2018.
Article in English | MEDLINE | ID: mdl-29222792

ABSTRACT

Hippocampal organotypic cultures constitute a very easy but delicate method widely used to study amyloid ß-peptide toxicity. This ex vivo technique is performed on tissues isolated from newborn rats. Here, we describe a protocol for the preparation and culture of hippocampal organotypic slices that can be maintained for 14-21 days and their application to the study of amyloid ß-peptide toxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Hippocampus/cytology , Organ Culture Techniques/methods , Animals , Animals, Newborn , Hippocampus/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar
16.
Methods Mol Biol ; 1727: 343-352, 2018.
Article in English | MEDLINE | ID: mdl-29222793

ABSTRACT

The intrahippocampal injection of amyloid beta peptide (1-42) (Aß(1-42)) represents one of the most useful animal models of Alzheimer disease. Since none of these available models fully represents the main pathological hallmarks of Alzheimer disease, stereotaxic Aß(1-42) infusion provides researchers with an in vivo alternative paradigm. When performed by well-trained individuals, this model is the best-suited one for short-term studies focusing on the effects of Aß(1-42) on a specific brain region or circuitry. Here, we describe all methodological phases of such a model.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Disease Models, Animal , Alzheimer Disease/etiology , Amyloid beta-Peptides/toxicity , Animals , Injections , Mice , Rats , Stereotaxic Techniques
17.
Curr Pharm Des ; 23(33): 4979-4989, 2017.
Article in English | MEDLINE | ID: mdl-28699521

ABSTRACT

Alzheimer's disease (AD) is a devastating neurological illness with a heavy economic impact. Further comorbidities in combination with the social impact of this disorder increase the urgency of a clearer comprehension of its etiopathogenesis, allowing the execution of novel therapeutic strategies. Despite astrocytes have been widely described as active participant in the regulation of cerebral circuits, available data are still poor. Even less information is available about their precise role in the pathogenesis of illness. Moreover, the scant knowledge about the astrocyte-neuron interplay in health and disease still impedes pioneering discoveries. The focus of this review is to look for new and innovative pharmacological approaches against AD. In order to perform this, we used the following keywords in PubMed search engine: astrocytes, therapy, Alzheimer's disease, and glia in different combinations. With this review, we collected data available in literature describing how also astrocytes besides neurons might be new potential targets for drug discovery. Different approaches currently being studied include modulation of glutamate transporters expression, astroglial genetic manipulation, free radicals inhibition, up-regulation of neurotrophins, and regulation of astrogliosis and neuroinflammation. Since several studies already demonstrated that astrocytes are definitely involved in AD pathogenesis, these cells can represent a promising new therapeutic target.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Astrocytes/metabolism , Free Radical Scavengers/metabolism , Nerve Growth Factors/biosynthesis , Therapies, Investigational/trends , Animals , Humans , Therapies, Investigational/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...