Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Aug 14.
Article in English | MEDLINE | ID: mdl-37645937

ABSTRACT

Career athletes, active military, and head trauma victims are at increased risk for mild repetitive traumatic brain injury (rTBI), a condition that contributes to the development of epilepsy and neurodegenerative diseases. Standard clinical imaging fails to identify rTBI-induced lesions, and novel non-invasive methods are needed. Here, we evaluated if hyperpolarized 13C magnetic resonance spectroscopic imaging (HP 13C MRSI) could detect long-lasting changes in brain metabolism 3.5 months post-injury in a rTBI mouse model. Our results show that this metabolic imaging approach can detect changes in cortical metabolism at that timepoint, whereas multimodal MR imaging did not detect any structural or contrast alterations. Using Machine Learning, we further show that HP 13C MRSI parameters can help classify rTBI vs. Sham and predict long-term rTBI-induced behavioral outcomes. Altogether, our study demonstrates the potential of metabolic imaging to improve detection, classification and outcome prediction of previously undetected rTBI.

2.
Sci Rep ; 13(1): 1749, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720960

ABSTRACT

Interplanetary space travel poses many hazards to the human body. To protect astronaut health and performance on critical missions, there is first a need to understand the effects of deep space hazards, including ionizing radiation, confinement, and altered gravity. Previous studies of rodents exposed to a single such stressor document significant deficits, but our study is the first to investigate possible cumulative and synergistic impacts of simultaneous ionizing radiation, confinement, and altered gravity on behavior and cognition. Our cohort was divided between 6-month-old female and male mice in group, social isolation, or hindlimb unloading housing, exposed to 0 or 50 cGy of 5 ion simplified simulated galactic cosmic radiation (GCRsim). We report interactions and independent effects of GCRsim exposure and housing conditions on behavioral and cognitive performance. Exposure to GCRsim drove changes in immune cell populations in peripheral blood collected early after irradiation, while housing conditions drove changes in blood collected at a later point. Female mice were largely resilient to deficits observed in male mice. Finally, we used principal component analysis to represent total deficits as principal component scores, which were predicted by general linear models using GCR exposure, housing condition, and early blood biomarkers.


Subject(s)
Cosmic Radiation , Monocytes , Humans , Female , Male , Animals , Mice , Infant , Cognition , Social Isolation , Astronauts
3.
Proc Natl Acad Sci U S A ; 119(42): e2209427119, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36227915

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of long-term neurological disability in the world and the strongest environmental risk factor for the development of dementia. Even mild TBI (resulting from concussive injuries) is associated with a greater than twofold increase in the risk of dementia onset. Little is known about the cellular mechanisms responsible for the progression of long-lasting cognitive deficits. The integrated stress response (ISR), a phylogenetically conserved pathway involved in the cellular response to stress, is activated after TBI, and inhibition of the ISR-even weeks after injury-can reverse behavioral and cognitive deficits. However, the cellular mechanisms by which ISR inhibition restores cognition are unknown. Here, we used longitudinal two-photon imaging in vivo after concussive injury in mice to study dendritic spine dynamics in the parietal cortex, a brain region involved in working memory. Concussive injury profoundly altered spine dynamics measured up to a month after injury. Strikingly, brief pharmacological treatment with the drug-like small-molecule ISR inhibitor ISRIB entirely reversed structural changes measured in the parietal cortex and the associated working memory deficits. Thus, both neural and cognitive consequences of concussive injury are mediated in part by activation of the ISR and can be corrected by its inhibition. These findings suggest that targeting ISR activation could serve as a promising approach to the clinical treatment of chronic cognitive deficits after TBI.


Subject(s)
Brain Concussion , Brain Injuries, Traumatic , Cognitive Dysfunction , Dementia , Animals , Brain Concussion/complications , Brain Injuries, Traumatic/complications , Cognitive Dysfunction/etiology , Memory Disorders , Mice
4.
Int J Mol Sci ; 23(20)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36293308

ABSTRACT

Neuroinflammation has emerged as an important factor in the molecular underpinnings of major depressive disorder (MDD) pathophysiology and in the mechanism of action of antidepressants. Among the inflammatory mediators dysregulated in depressed patients, interleukin (IL)-6 has recently been proposed to play a crucial role. IL-6 activates a signaling pathway comprising the JAK/STAT proteins and characterized by a specific negative feedback loop exerted by the cytoplasmic protein suppressor of cytokine signalling-3 (SOCS3). On these bases, here, we explored the potential involvement of IL-6 signaling in the ability of the antidepressant drug agomelatine to normalize the anhedonic-like phenotype induced in the rat by chronic stress exposure. To this aim, adult male Wistar rats were subjected to the chronic mild stress (CMS) paradigm and chronically treated with vehicle or agomelatine. The behavioral evaluation was assessed by the sucrose consumption test, whereas molecular analyses were performed in the prefrontal cortex. We found that CMS was able to stimulate IL-6 production and signaling, including SOCS3 gene and protein expression, but the SOCS3-mediated feedback-loop inhibition failed to suppress the IL-6 cascade in stressed animals. Conversely, agomelatine treatment normalized the stress-induced decrease in sucrose consumption and restored the negative modulation of the IL-6 signaling via SOCS3 expression and activity. Our results provide additional information about the pleiotropic mechanisms that contribute to agomelatine's therapeutic effects.


Subject(s)
Depressive Disorder, Major , Interleukin-6 , Animals , Rats , Male , Interleukin-6/genetics , Interleukin-6/metabolism , Depression/drug therapy , Depression/etiology , Depression/metabolism , Rats, Wistar , Depressive Disorder, Major/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Signal Transduction , Inflammation Mediators/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Sucrose
5.
Psychopharmacology (Berl) ; 239(8): 2547-2557, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35459959

ABSTRACT

RATIONALE: Although the occurrence of stressful events is very common during life, their impact may be different depending on the experience severity and duration. Specifically, acute challenges may trigger adaptive responses and even improve the individual's performance. However, such a physiological positive coping can only take place if the underlying molecular mechanisms are properly functioning. Indeed, if these systems are compromised by genetic factors or previous adverse conditions, the response set in motion by an acute challenge may be maladaptive and even cause the insurgence or the relapse of stress-related psychiatric disorders. OBJECTIVES: On these bases, we evaluated in the rat brain the role of the antioxidant component of the redox machinery on the acute stress responsiveness and its modulation by potential detrimental or beneficial events. METHODS: The expression of several antioxidant enzymes was assessed in different brain areas of adult male rats exposed to acute stress 3 weeks after a chronic immobilization paradigm with or without a concomitant treatment with the antipsychotic lurasidone. RESULTS: The acute challenge was able to trigger a marked antioxidant response that, despite the washout period, was impaired by the previous adverse experience and restored by lurasidone in an anatomical-specific manner. CONCLUSIONS: We found that a working antioxidant machinery takes part in acute stress response and may be differentially affected by other experiences. Given the essential role of stress responsiveness in almost every life process, the identification of the underlying mechanisms and their potential pharmacological modulation add further translational value to our data.


Subject(s)
Antipsychotic Agents , Lurasidone Hydrochloride , Animals , Antioxidants/pharmacology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Humans , Lurasidone Hydrochloride/pharmacology , Male , Rats
6.
Cell Mol Neurobiol ; 42(4): 1225-1240, 2022 May.
Article in English | MEDLINE | ID: mdl-33259004

ABSTRACT

One of the most substantial and established environmental risk factors for neurological and psychiatric disorders is stress exposure, whose detrimental consequences hinge on several variables including time. In this regard the gestational period is known to present an intrinsic vulnerability to environmental insults and thus stressful events during pregnancy can lead to severe consequences on the offspring's brain development with long-term repercussions throughout adulthood. On this basis, we investigated the long-lasting impact of prenatal stress exposure on the susceptibility to the experimental autoimmune encephalomyelitis (EAE), a well-established murine model of multiple sclerosis. Although stress is considered a triggering factor for this chronic, progressive, autoimmune disease, little is known about the underlying mechanisms. To this end, EAE was induced by immunization with MOG35-55/CFA and pertussis toxin administration in adult female C57BL/6 mice born from control or stressed dams exposed to restraint stress during the last days of gestation. Our results demonstrate that gestational stress induces a marked increase in the severity of EAE symptoms in adulthood. Further, we highlight an altered maturation of oligodendrocytes in the spinal cord of prenatally stressed EAE mice, as indicated by the higher levels of GPR17, a marker of immature oligodendrocyte precursor cells. These behavioral and molecular alterations are paralleled by changes in the expression and signaling of the neurotrophin BDNF, an important mediator of neural plasticity that may contribute to stress-induced impaired remyelination. Since several already marketed drugs are able to modulate BDNF levels, these results pave the way to the possibility of repositioning these drugs in multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Brain-Derived Neurotrophic Factor/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/metabolism
7.
Neurosci Biobehav Rev ; 126: 509-514, 2021 07.
Article in English | MEDLINE | ID: mdl-33862064

ABSTRACT

In 2024 the first female astronaut will land on the moon, advancing our preparations for human missions to Mars. While on Earth we are protected from space radiation by our planet's magnetic field, on such deep space voyages astronauts will be exposed to high energy particles from solar flares and galactic cosmic rays (GCR). This exposure carries risks to the central nervous system (CNS) that could jeopardize the mission and astronaut health. Earth-bound studies have employed a variety of single-beam and sequential radiation exposures to simulate the effects of GCR exposure in rodents. Multiple studies have shown that GCR simulation induces a maladaptive activation of microglia - the brain-resident immune cells. GCR simulation also induced synaptic changes resulting in lasting cognitive and behavioral defects. Female and male mice show different susceptibilities to GCR exposure, and evidence suggests this sexually dimorphic response is linked to microglia. Manipulating microglia can prevent the development of cognitive deficits in male mice exposed to components of GCR. This discovery may provide clues towards how to protect astronauts' cognitive and behavioral health both during deep space missions and upon return to Earth.


Subject(s)
Cosmic Radiation , Space Flight , Animals , Astronauts , Female , Humans , Male , Mice , Microglia
8.
Neurosci Lett ; 741: 135462, 2021 01 10.
Article in English | MEDLINE | ID: mdl-33259927

ABSTRACT

Microglia are the resident immune cells of the central nervous system (CNS). In physiological conditions, microglia contribute to maintaining brain homeostasis by scanning the surrounding parenchyma and acting as scavenger cells. Following different insults to the CNS, microglia turn into a "reactive" state characterized by the production of inflammatory mediators that promote tissue repair to restore homeostasis. Brain insults such as traumatic brain injury, therapeutic brain irradiation and galactic cosmic ray exposure are associated with chronic microglia activation. Chronic microglia activation contributes to injury-related impairments in cognitive functions. Microglia depletion achieved either by pharmacological or genetic techniques represents not only a useful tool for more extensive investigations of microglia roles, but also a potential therapeutic approach to ameliorate or prevent cognitive dysfunctions following brain injury.


Subject(s)
Brain Injuries/immunology , Brain Injuries/psychology , Cognition/physiology , Cosmic Radiation/adverse effects , Microglia/immunology , Microglia/radiation effects , Animals , Brain Injuries/etiology , Cognition/radiation effects , Encephalitis/etiology , Encephalitis/immunology , Humans
9.
Pharmacol Res ; 163: 105330, 2021 01.
Article in English | MEDLINE | ID: mdl-33276101

ABSTRACT

Psychiatric disorders represent a critical challenge to our society, given their high global prevalence, complex symptomatology, elusive etiology and the variable effectiveness of pharmacological therapies. Recently, there has been a shift in investigating and redefining these diseases by integrating behavioral observations and multilevel neurobiological measures. Accordingly, endophenotype-oriented studies are needed to develop new therapeutic strategies, with the idea of targeting shared symptoms instead of one defined disease. With these premises, here we investigated the therapeutic properties of chronic treatment with the second-generation antipsychotic blonanserin in counteracting the alterations caused by 7 weeks of Chronic Mild Stress (CMS) in the rat. CMS is a well-established preclinical model able to induce depressive and anxiety-like alterations, which are shared by different psychiatric disorders. Our results demonstrated that the antipsychotic treatment normalizes the CMS-induced emotionality deficits, an effect that may be due to its ability in modulating, within the prefrontal cortex, redox mechanisms, a molecular dysfunction associated with several psychiatric disorders. These evidences provide new insights into the therapeutic properties and potential use of blonanserin as well as in its mechanisms of action and provide further support for the role of oxidative stress in the pathophysiology of psychiatric disorders.


Subject(s)
Antipsychotic Agents/therapeutic use , Piperazines/therapeutic use , Piperidines/therapeutic use , Stress, Psychological/drug therapy , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cytoskeletal Proteins/genetics , Disease Models, Animal , Male , Maze Learning/drug effects , Nerve Tissue Proteins/genetics , Oxidoreductases/genetics , Piperazines/pharmacology , Piperidines/pharmacology , Rats, Wistar , Stress, Psychological/genetics
10.
Elife ; 92020 12 01.
Article in English | MEDLINE | ID: mdl-33258451

ABSTRACT

With increased life expectancy, age-associated cognitive decline becomes a growing concern, even in the absence of recognizable neurodegenerative disease. The integrated stress response (ISR) is activated during aging and contributes to age-related brain phenotypes. We demonstrate that treatment with the drug-like small-molecule ISR inhibitor ISRIB reverses ISR activation in the brain, as indicated by decreased levels of activating transcription factor 4 (ATF4) and phosphorylated eukaryotic translation initiation factor eIF2. Furthermore, ISRIB treatment reverses spatial memory deficits and ameliorates working memory in old mice. At the cellular level in the hippocampus, ISR inhibition (i) rescues intrinsic neuronal electrophysiological properties, (ii) restores spine density and (iii) reduces immune profiles, specifically interferon and T cell-mediated responses. Thus, pharmacological interference with the ISR emerges as a promising intervention strategy for combating age-related cognitive decline in otherwise healthy individuals.


Subject(s)
Acetamides/pharmacology , Cyclohexylamines/pharmacology , Memory/drug effects , Nootropic Agents/pharmacology , Activating Transcription Factor 4/metabolism , Aging/drug effects , Animals , Brain/drug effects , Cognitive Dysfunction/drug therapy , Dendritic Spines/drug effects , Female , Hippocampus/cytology , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Spatial Learning/drug effects , Stress, Physiological
11.
Int J Mol Sci ; 21(17)2020 Aug 29.
Article in English | MEDLINE | ID: mdl-32872446

ABSTRACT

Depression is a recurrent disorder, with about 50% of patients experiencing relapse. Exposure to stressful events may have an adverse impact on the long-term course of the disorder and may alter the response to a subsequent stressor. Indeed, not all the systems impaired by stress may normalize during symptoms remission, facilitating the relapse to the pathology. Hence, we investigated the long-lasting effects of chronic restraint stress (CRS) and its influence on the modifications induced by the exposure to a second hit on brain-derived neurotrophic factor (BDNF) signaling in the prefrontal cortex (PFC). We exposed adult male Sprague Dawley rats to 4 weeks of CRS, we left them undisturbed for the subsequent 3 weeks, and then we exposed animals to one hour of acute restraint stress (ARS). We found that CRS influenced the release of corticosterone induced by ARS and inhibited the ability of ARS to activate mature BDNF, its receptor Tropomyosin receptor kinase B (TRKB), and their associated intracellular cascades: the TRKB-PI3K-AKT), the MEK-MAPK/ERK, and the Phospholipase C γ (PLCγ) pathways, positively modulated by ARS in non-stressed animals. These results suggest that CRS induces protracted and detrimental consequences that interfere with the ability of PFC to cope with a challenging situation.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Corticosterone/metabolism , Prefrontal Cortex/metabolism , Restraint, Physical/psychology , Stress, Psychological/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction , Stress, Psychological/etiology , Stress, Psychological/genetics
12.
Psychopharmacology (Berl) ; 237(6): 1783-1793, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32296859

ABSTRACT

RATIONALE: Patients diagnosed with schizophrenia typically receive life-long treatments with antipsychotic drugs (APDs). However, the impact of chronic APDs treatment on neuroplastic mechanisms in the brain remains largely elusive. OBJECTIVE: Here, we focused on blonanserin, a second-generation antipsychotic (SGA) that acts as an antagonist at dopamine D2, D3, and serotonin 5-HT2A receptors, and represents an important tool for the treatment of schizophrenia. METHODS: We used rats to investigate the ability of chronic treatment blonanserin to modulate the activity of brain structures relevant for schizophrenia, under baseline conditions or in response to an acute forced swim session (FSS). We measured the expression of different immediate early genes (IEGs), including c-Fos, Arc/Arg 3.1, Zif268 and Npas4. RESULTS: Blonanserin per se produced limited changes in the expression of these genes under basal conditions, while, as expected, FSS produced a significant elevation of IEGs transcription in different brain regions. The response of blonanserin-treated rats to FSS show anatomical and gene-selective differences. Indeed, the upregulation of IEGs was greatly reduced in the striatum, a brain structure enriched in dopamine receptors, whereas the upregulation of some genes (Zif268, Npas4) was largely preserved in other regions, such as the prefrontal cortex and the ventral hippocampus. CONCLUSIONS: Taken together, our findings show that chronic exposure to blonanserin modulates selective IEGs with a specific anatomical profile. Moreover, the differential activation of specific brain regions under challenging conditions may contribute to specific clinical features of the drug.


Subject(s)
Antipsychotic Agents/administration & dosage , Brain/drug effects , Genes, Immediate-Early/drug effects , Piperazines/administration & dosage , Piperidines/administration & dosage , Stress, Psychological/drug therapy , Animals , Brain/physiology , Drug Administration Schedule , Genes, Immediate-Early/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Schizophrenia/drug therapy , Schizophrenia/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology
13.
Pharmacol Ther ; 210: 107520, 2020 06.
Article in English | MEDLINE | ID: mdl-32165136

ABSTRACT

While neurotransmitter dysfunction represents a key component in mental illnesses, there is now a wide agreement for a central pathophysiological hub that includes hormones, neuroinflammation, redox mechanisms as well as oxidative stress. With respect to oxidation-reduction (redox) mechanisms, preclinical and clinical evidence suggests that an imbalance in the pro/anti-oxidative homeostasis toward the increased production of substances with oxidizing potential may contribute to the etiology and manifestation of different psychiatric disorders. The substantial and continous demand for energy renders the brain highly susceptible to disturbances in its energy supply, especially following exposure to stressful events, which may lead to overproduction of reactive oxygen and nitrogen species under conditions of perturbed antioxidant defenses. This will eventually induce different molecular alterations, including extensive protein and lipid peroxidation, increased blood-brain barrier permeability and neuroinflammation, which may contribute to the changes in brain function and morphology observed in mental illnesses. This view may also reconcile different key concepts for psychiatric disorders, such as the neurodevelopmental origin of these diseases, as well as the vulnerability of selective cellular populations that are critical for specific functional abnormalities. The possibility to pharmacologically modulate the redox system is receiving increasing interest as a novel therapeutic strategy to counteract the detrimental effects of the unbalance in brain oxidative mechanisms. This review will describe the main mechanisms and mediators of the redox system and will examine the alterations of oxidative stress found in animal models of psychiatric disorders as well as in patients suffering from mental illnesses, such as schizophrenia and major depressive disorder. In addition, it will discuss studies that examined the effects of psychotropic drugs, including antipsychotics and antidepressants, on the oxidative balance as well as studies that investigated the effectiveness of a direct modulation of oxidative mechanisms in counteracting the behavioral and functional alterations associated with psychiatric disorders, which supports the promising role of the redox system as a novel therapeutic target for the improved treatment of brain disorders.


Subject(s)
Antioxidants/therapeutic use , Central Nervous System Agents/therapeutic use , Central Nervous System/drug effects , Mental Disorders/drug therapy , Oxidative Stress/drug effects , Animals , Central Nervous System/metabolism , Central Nervous System/physiopathology , Humans , Mental Disorders/metabolism , Mental Disorders/physiopathology , Mental Disorders/psychology , Oxidation-Reduction
14.
Brain Behav Immun ; 82: 422-431, 2019 11.
Article in English | MEDLINE | ID: mdl-31525509

ABSTRACT

Vincristine (VCR) treatment is often associated to painful neuropathy. Its development is independent from antitumoral mechanism and involves neuroinflammation. We investigated the role of the chemokine prokineticin (PK)2 in a mouse model of VCR induced neuropathy using a PK-receptors (PK-R) antagonist to counteract its development. We also evaluated emotional like deficits in VCR mice. VCR (0,1 mg/kg) was i.p. injected in C57BL/6J male mice once a day for 14 consecutive days. Pain, anxiety and depressive like behaviors were assessed in animals. PK2, PK-Rs, cytokines, neuroinflammatory markers (CD68, CD11b, GFAP, TLR4) and ATF3 were evaluated in DRG, spinal cord, prefrontal cortex and hippocampus. The PK-Rs antagonist PC1, was s.c. injected (150 µg/kg) twice a day from day 7 (hypersensitivity state) until day 14. Its effect on pain and neuroinflammation was evaluated. VCR mice developed neuropathic pain but not mood alterations. After 7 days of VCR treatment we observed a neuroinflammatory condition in DRG with high levels of PK-Rs, TLR4, CD68, ATF3 and IL-1ß without relevant alterations in spinal cord. At day 14, an upregulation of PK system and a marked neuroinflammation was evident also in spinal cord. Moreover, at the same time, we observed initial alterations in supraspinal brain areas. PC1 treatment significantly counteracted neuropathic pain and blunted neuroinflammation.


Subject(s)
Gastrointestinal Hormones/metabolism , Neuralgia/chemically induced , Neuralgia/metabolism , Neuropeptides/metabolism , Vincristine/toxicity , Animals , Anxiety/chemically induced , Anxiety/metabolism , Behavior, Animal/drug effects , Cytokines/metabolism , Depression/chemically induced , Depression/metabolism , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/drug effects , Random Allocation , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism
15.
Front Mol Neurosci ; 12: 166, 2019.
Article in English | MEDLINE | ID: mdl-31379496

ABSTRACT

A growing body of evidence supports the close relationship between major depressive disorder (MDD), a severe psychiatric disease more common among women than men, and alterations of the immune/inflammatory system. However, despite the large number of studies aimed at understanding the molecular bases of this association, a lack of information exists on the potential cross-talk between systems known to be involved in depression and components of the inflammatory response, especially with respect to sex differences. Brain-derived neurotrophic factor (BDNF) is a neurotrophin with a well-established role in MDD etiopathology: it is altered in depressed patients as well as in animal models of the disease and its changes are restored by antidepressant drugs. Interestingly, this neurotrophin is also involved in the inflammatory response. Indeed, it can be secreted by microglia, the primary innate immune cells in the central nervous system whose functions may be in turn regulated by BDNF. With these premises, in this study, we investigated the reciprocal impact of BDNF and the immune system by evaluating the neuroinflammatory response in male and female BDNF-heterozygous mutant mice acutely treated with the cytokine-inducer lipopolysaccharide (LPS). Specifically, we assessed the potential onset of an LPS-induced sickness behavior as well as changes of inflammatory mediators in the mouse hippocampus and frontal cortex, with respect to both genotype and sex. We found that the increased inflammatory response induced by LPS in the brain of male mice was independent of the genotype, whereas in the female, it was restricted to the heterozygous mice with no changes in the wild-type group, suggestive of a role for BDNF in the sex-dependent effect of the inflammatory challenge. Considering the involvement of both BDNF and neuroinflammation in several psychiatric diseases and the diverse incidence of such pathologies in males and females, a deeper investigation of the mechanisms underlying their interaction may have a critical translational relevance.

16.
Curr Med Chem ; 26(20): 3685-3701, 2019.
Article in English | MEDLINE | ID: mdl-31333079

ABSTRACT

Population aging is accelerating rapidly worldwide, from 461 million people older than 65 years in 2004 to an estimated 2 billion people by 2050, leading to critical implications for the planning and delivery of health and social care. The most problematic expression of population aging is the clinical condition of frailty, which is a state of increased vulnerability that develops as a consequence of the accumulation of microscopic damages in many physiological systems that lead to a striking and disproportionate change in health state, even after an apparently small insult. Since little is known about the biology of frailty, an important perspective to understand this phenomenon is to establish how the alterations that physiologically occur during a condition of healthy aging may instead promote cumulative decline with subsequent depletion of homoeostatic reserve and increase the vulnerability also after minor stressor events. In this context, the present review aims to provide a description of the molecular mechanisms that, by having a critical impact on behavior and neuronal function in aging, might be relevant for the development of frailty. Moreover, since these biological systems are also involved in the coping strategies set in motion to respond to environmental challenges, we propose a role for lifestyle stress as an important player to drive frailty in aging.


Subject(s)
Aging/physiology , Frailty/physiopathology , Healthy Aging , Humans
17.
Front Pharmacol ; 10: 658, 2019.
Article in English | MEDLINE | ID: mdl-31244664

ABSTRACT

Alzheimer's disease (AD) is the most frequent type of dementia in older people. The complex nature of AD calls for the development of multitarget agents addressing key pathogenic processes. Donepezil, an acetylcholinesterase inhibitor, is a first-line acetylcholinesterase inhibitor used for the treatment of AD. Although several studies have demonstrated the symptomatic efficacy of donepezil treatment in AD patients, the possible effects of donepezil on the AD process are not yet known. In this study, a novel feruloyl-donepezil hybrid compound (PQM130) was synthesized and evaluated as a multitarget drug candidate against the neurotoxicity induced by Aß1-42 oligomer (AßO) injection in mice. Interestingly, PQM130 had already shown anti-inflammatory activity in different in vivo models and neuroprotective activity in human neuronal cells. The intracerebroventricular (i.c.v.) injection of AßO in mice caused the increase of memory impairment, oxidative stress, neurodegeneration, and neuroinflammation. Instead, PQM130 (0.5-1 mg/kg) treatment after the i.c.v. AßO injection reduced oxidative damage and neuroinflammation and induced cell survival and protein synthesis through the modulation of glycogen synthase kinase 3ß (GSK3ß) and extracellular signal-regulated kinases (ERK1/2). Moreover, PQM130 increased brain plasticity and protected mice against the decline in spatial cognition. Even more interesting is that PQM130 modulated different pathways compared to donepezil, and it is much more effective in counteracting AßO damage. Therefore, our findings highlighted that PQM130 is a potent multi-functional agent against AD and could act as a promising neuroprotective compound for anti-AD drug development.

18.
Int J Mol Sci ; 19(12)2018 Nov 26.
Article in English | MEDLINE | ID: mdl-30486287

ABSTRACT

Traumatic brain injury (TBI) is of particular concern for the aging community since there is both increased incidence of TBI and decreased functional recovery in this population. In addition, TBI is the strongest environmental risk factor for development of Alzheimer's disease and other dementia-related neurodegenerative disorders. Critical changes that affect cognition take place over time following the initial insult. Our previous work identified immune system activation as a key contributor to cognitive deficits observed in aged animals. Using a focal contusion model in the current study, we demonstrate a brain lesion and cavitation formation, as well as prolonged blood⁻brain barrier breakdown. These changes were associated with a prolonged inflammatory response, characterized by increased microglial cell number and phagocytic activity 30 days post injury, corresponding to significant memory deficits. We next aimed to identify the injury-induced cellular and molecular changes that lead to chronic cognitive deficits in aged animals, and measured increases in complement initiation components C1q, C3, and CR3, which are known to regulate microglial⁻synapse interactions. Specifically, we found significant accumulation of C1q on synapses within the hippocampus, which was paralleled by synapse loss 30 days post injury. We used genetic and pharmacological approaches to determine the mechanistic role of complement initiation on cognitive loss in aging animals after TBI. Notably, both genetic and pharmacological blockade of the complement pathway prevented memory deficits in aged injured animals. Thus, therapeutically targeting early components of the complement cascade represents a significant avenue for possible clinical intervention following TBI in the aging population.


Subject(s)
Aging/pathology , Brain Injuries, Traumatic/complications , Complement System Proteins/metabolism , Memory Disorders/etiology , Microglia/pathology , Synapses/pathology , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Brain Injuries, Traumatic/pathology , Cell Count , Chronic Disease , Contusions , Disease Progression , Female , Magnetic Resonance Imaging , Male , Memory Disorders/pathology , Mice, Inbred C57BL , Microglia/metabolism , Models, Biological , Phagocytosis , Synapses/metabolism
19.
Sci Rep ; 8(1): 10297, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29967490

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

20.
Int J Neuropsychopharmacol ; 21(9): 883-893, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29788232

ABSTRACT

Background: Psychiatric disorders are associated with altered function of inhibitory neurotransmission within the limbic system, which may be due to the vulnerability of selective neuronal subtypes to challenging environmental conditions, such as stress. In this context, parvalbumin-positive GABAergic interneurons, which are critically involved in processing complex cognitive tasks, are particularly vulnerable to stress exposure, an effect that may be the consequence of dysregulated redox mechanisms. Methods: Adult Male Wistar rats were subjected to the chronic mild stress procedure for 7 weeks. After 2 weeks, both control and stress groups were further divided into matched subgroups to receive chronic administration of vehicle or lurasidone (3 mg/kg/d) for the subsequent 5 weeks. Using real-time RT-PCR and western blot, we investigated the expression of GABAergic interneuron markers and the levels of key mediators of the oxidative balance in the dorsal and ventral hippocampus. Results: Chronic mild stress induced a specific decrease of parvalbumin expression in the dorsal hippocampus, an effect normalized by lurasidone treatment. Interestingly, the regulation of parvalbumin levels was correlated to the modulation of the antioxidant master regulator NRF2 and its chaperon protein KEAP1, which were also modulated by pharmacological intervention. Conclusions: Our findings suggest that the susceptibility of parvalbumin neurons to stress may represent a key mechanism contributing to functional and structural impairments in specific brain regions relevant for psychiatric disorders. Moreover, we provide new insights on the mechanism of action of lurasidone, demonstrating that its chronic treatment normalizes chronic mild stress-induced parvalbumin alterations, possibly by potentiating antioxidant mechanisms, which may ameliorate specific functions that are deteriorated in psychiatric patients.


Subject(s)
Antipsychotic Agents/pharmacology , Hippocampus/metabolism , Lurasidone Hydrochloride/pharmacology , Parvalbumins/metabolism , Stress, Psychological/metabolism , Animals , Chronic Disease , Disease Models, Animal , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Gene Expression/drug effects , Hippocampus/drug effects , Interneurons/drug effects , Interneurons/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Male , NADPH Oxidase 2/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , RNA, Messenger/metabolism , Rats, Wistar , Stress, Psychological/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...