Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
J Inflamm Res ; 17: 4549-4574, 2024.
Article in English | MEDLINE | ID: mdl-39011416

ABSTRACT

The prevalence of age-related neurodegenerative diseases, such as Parkinson's disease (PD) and related disorders continues to grow worldwide. Increasing evidence links intracellular inclusions of misfolded alpha-synuclein (α-syn) aggregates, so-called Lewy bodies (LB) and Lewy neuritis, to the progressive pathology of PD and other synucleinopathies. Our previous findings established that α-syn oligomers induce S-nitrosylation and deregulation of the E3-ubiquitin ligase Parkin, leading to mitochondrial disturbances in neuronal cells. The accumulation of damaged mitochondria as a consequence, together with the release of mitochondrial-derived damage-associated molecular patterns (mtDAMPs) could activate the innate immune response and induce neuroinflammation ("mito-inflammation"), eventually accelerating neurodegeneration. However, the molecular pathways that transmit pro-inflammatory signals from damaged mitochondria are not well understood. One of the proposed pathways could be the cyclic GMP-AMP synthase (cGAS) - stimulator of interferon genes (STING) (cGAS-STING) pathway, which plays a pivotal role in modulating the innate immune response. It has recently been suggested that cGAS-STING deregulation may contribute to the development of various pathological conditions. Especially, its excessive engagement may lead to neuroinflammation and appear to be essential for the development of neurodegenerative brain diseases, including PD. However, the precise molecular mechanisms underlying cGAS-STING pathway activation in PD and other synucleinopathies are not fully understood. This review focuses on linking mitochondrial dysfunction to neuroinflammation in these disorders, particularly emphasizing the role of the cGAS-STING signaling. We propose the cGAS-STING pathway as a critical driver of inflammation in α-syn-dependent neurodegeneration and hypothesize that cGAS-STING-driven "mito-inflammation" may be one of the key mechanisms promoting the neurodegeneration in PD. Understanding the molecular mechanisms of α-syn-induced cGAS-STING-associated "mito-inflammation" in PD and related synucleinopathies may contribute to the identification of new targets for the treatment of these disorders.

3.
Brain Sci ; 13(7)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37509018

ABSTRACT

Recent data suggest that defects in purinergic signalling are a common denominator of autism spectrum disorders (ASDs), though nothing is known about whether the disorder-related imbalance occurs at the receptor level. In this study, we investigated whether prenatal exposure to valproic acid (VPA) induces changes in purinergic receptor expression in adolescence and whether it corresponds to glial cell activation. Pregnant dams were subjected to an intraperitoneal injection of VPA at embryonic day 12.5. In the hippocampi of adolescent male VPA offspring, we observed an increase in the level of P2X1, with concomitant decreases in P2X7 and P2Y1 receptors. In contrast, in the cortex, the level of P2X1 was significantly reduced. Also, significant increases in cortical P2Y1 and P2Y12 receptors were detected. Additionally, we observed profound alterations in microglial cell numbers and morphology in the cortex of VPA animals, leading to the elevation of pro-inflammatory cytokine expression. The changes in glial cells were partially reduced via a single administration of a non-selective P2 receptor antagonist. These studies show the involvement of purinergic signalling imbalance in the modulation of brain inflammatory response induced via prenatal VPA exposure and may indicate that purinergic receptors are a novel target for pharmacological intervention in ASDs.

4.
Int J Mol Sci ; 24(7)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37047246

ABSTRACT

Neurons have long been central to the study of cellular networks in the nervous system [...].


Subject(s)
Nervous System Diseases , Neuroglia , Humans , Neuroglia/physiology , Neurons/physiology
5.
Int J Mol Sci ; 24(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37047292

ABSTRACT

Neurotrophic factors (NTFs) play an important role in maintaining homeostasis of the central nervous system (CNS) by regulating the survival, differentiation, maturation, and development of neurons and by participating in the regeneration of damaged tissues. Disturbances in the level and functioning of NTFs can lead to many diseases of the nervous system, including degenerative diseases, mental diseases, and neurodevelopmental disorders. Each CNS disease is characterized by a unique pathomechanism, however, the involvement of certain processes in its etiology is common, such as neuroinflammation, dysregulation of NTFs levels, or mitochondrial dysfunction. It has been shown that NTFs can control the activation of glial cells by directing them toward a neuroprotective and anti-inflammatory phenotype and activating signaling pathways responsible for neuronal survival. In this review, our goal is to outline the current state of knowledge about the processes affected by NTFs, the crosstalk between NTFs, mitochondria, and the nervous and immune systems, leading to the inhibition of neuroinflammation and oxidative stress, and thus the inhibition of the development and progression of CNS disorders.


Subject(s)
Brain Diseases , Central Nervous System Diseases , Humans , Neuroinflammatory Diseases , Nerve Growth Factors/metabolism , Neuroglia/metabolism , Neurons/metabolism , Brain Diseases/metabolism , Central Nervous System Diseases/etiology , Central Nervous System Diseases/metabolism , Transforming Growth Factor beta/metabolism
6.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108406

ABSTRACT

Maternal immune activation (MIA) is an important risk factor for neurodevelopmental disorders such as autism. The aim of the current study was to investigate the development-dependent changes in the mitochondrial function of MIA-exposed offspring, which may contribute to autism-like deficits. MIA was evoked by the single intraperitoneal administration of lipopolysaccharide to pregnant rats at gestation day 9.5, and several aspects of mitochondrial function in fetuses and in the brains of seven-day-old pups and adolescent offspring were analyzed along with oxidative stress parameters measurement. It was found that MIA significantly increased the activity of NADPH oxidase (NOX), an enzyme generating reactive oxygen species (ROS) in the fetuses and in the brain of seven-day-old pups, but not in the adolescent offspring. Although a lower mitochondrial membrane potential accompanied by a decreased ATP level was already observed in the fetuses and in the brain of seven-day-old pups, persistent alterations of ROS, mitochondrial membrane depolarization, and lower ATP generation with concomitant electron transport chain complexes downregulation were observed only in the adolescent offspring. We suggest that ROS observed in infancy are most likely of a NOX activity origin, whereas in adolescence, ROS are produced by damaged mitochondria. The accumulation of dysfunctional mitochondria leads to the intense release of free radicals that trigger oxidative stress and neuroinflammation, resulting in an interlinked vicious cascade.


Subject(s)
Prenatal Exposure Delayed Effects , Pregnancy , Humans , Female , Rats , Animals , Reactive Oxygen Species , Brain , Vitamins , Mitochondria , Adenosine Triphosphate , Behavior, Animal/physiology , Disease Models, Animal
7.
Int J Mol Sci ; 24(8)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37108467

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare genetic multisystem disorder caused by loss-of-function mutations in the tumour suppressors TSC1/TSC2, both of which are negative regulators of the mammalian target of rapamycin (mTOR) kinase. Importantly, mTOR hyperactivity seems to be linked with the pathobiology of autism spectrum disorders (ASD). Recent studies suggest the potential involvement of microtubule (MT) network dysfunction in the neuropathology of "mTORopathies", including ASD. Cytoskeletal reorganization could be responsible for neuroplasticity disturbances in ASD individuals. Thus, the aim of this work was to study the effect of Tsc2 haploinsufficiency on the cytoskeletal pathology and disturbances in the proteostasis of the key cytoskeletal proteins in the brain of a TSC mouse model of ASD. Western-blot analysis indicated significant brain-structure-dependent abnormalities in the microtubule-associated protein Tau (MAP-Tau), and reduced MAP1B and neurofilament light (NF-L) protein level in 2-month-old male B6;129S4-Tsc2tm1Djk/J mice. Alongside, pathological irregularities in the ultrastructure of both MT and neurofilament (NFL) networks as well as swelling of the nerve endings were demonstrated. These changes in the level of key cytoskeletal proteins in the brain of the autistic-like TSC mice suggest the possible molecular mechanisms responsible for neuroplasticity alterations in the ASD brain.


Subject(s)
Autism Spectrum Disorder , Tuberous Sclerosis , Mice , Animals , Male , Autism Spectrum Disorder/genetics , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Cytoskeletal Proteins/genetics , Microtubules/metabolism , Mammals/metabolism
8.
Biomedicines ; 10(12)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36551785

ABSTRACT

Autism spectrum disorders (ASD) are neurodevelopmental diseases characterised by deficits in social communication, restricted interests, and repetitive behaviours. The growing body of evidence points to a role for cerebellar changes in ASD pathology. Some of the findings suggest that not only motor problems but also social deficits, repetitive behaviours, and mental inflexibility associated with ASD are connected with damage to the cerebellum. However, the understanding of this brain structure's functions in ASD pathology needs future investigations. Therefore, in this study, we generated a rodent model of ASD through a single prenatal administration of valproic acid (VPA) into pregnant rats, followed by cerebellar morphological studies of the offspring, focusing on the alterations of key cytoskeletal elements. The expression (Western blot) of α/ß-tubulin and the major neuronal MT-associated proteins (MAP) such as MAP-Tau and MAP1B, MAP2, MAP6 (STOP) along with actin-crosslinking αII-spectrin and neurofilament light polypeptide (NF-L) was investigated. We found that maternal exposure to VPA induces a significant decrease in the protein levels of α/ß-tubulin, MAP-Tau, MAP1B, MAP2, and αII-spectrin. Moreover, excessive MAP-Tau phosphorylation at (Ser396) along with key Tau-kinases activation was indicated. Immunohistochemical staining showed chromatolysis in the cerebellum of autistic-like rats and loss of Purkinje cells shedding light on one of the possible molecular mechanisms underpinning neuroplasticity alterations in the ASD brain.

9.
Folia Neuropathol ; 60(4): 390-402, 2022.
Article in English | MEDLINE | ID: mdl-36734381

ABSTRACT

Purinergic signalling is involved in the control of several processes related to brain development, such as neurogenesis and gliogenesis, migration and differentiation of neuronal precursors, synaptogenesis and synaptic elimination to achieve a fully wired and efficient mature brain. Therefore, any deregulation of purine-dependent signalling mediated by stimulation of specific adenosine and purinergic receptor subtypes: P1, P2X, or P2Y, can lead to functional deficits and the development of neuropsychiatric disorders, including autism spectrum disorders (ASD). In this study, we investigated the changes in expression and activity of selected purinergic receptors during rat brain development in an animal model of ASD. Pregnant dams received an intraperitoneal injection of valproic acid (VPA; 450 mg/kg body weight) at embryonic day (ED) 12.5, around the time of neural tube closure. Subsequently, changes in the expression and activity of specific purinergic receptor subtypes were analysed at ED19, an important prenatal stage of brain development. Our results suggest that prenatal VPA exposure leads to a significant increase in the level and activity of adenosinergic receptors A1, A2b and A3, which are involved in the regulation of progenitor cell proliferation and nerve growth, and upregulation of purinergic P2X2/P2X3 receptors, which in turn may contribute to the postnatal neuroanatomical abnormalities and synaptic dysfunction. Conversely, the significant downregulation of P2Y1 and P2X7 receptors, together with their reduced activity in the embryonic VPA brain, may indicate disturbances in the processes of neuronal precursor migration and differentiation, dendritic and axonal formation, and glutamate/GABA imbalance, thereby altering neuronal excitability. In conclusion, defects in purinergic signalling induced by prenatal VPA exposure could have a profound impact on brain development during embryogenesis and on intellectual and behavioural functions after birth. These observations could provide clues for future implementation of potential therapeutic strategies for ASD.


Subject(s)
Prenatal Exposure Delayed Effects , Receptors, Purinergic P2 , Animals , Female , Rats , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Brain/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptors, Purinergic/metabolism , Receptors, Purinergic P2/metabolism , Valproic Acid/toxicity
10.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768946

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disease that is characterized by a deficit in social interactions and communication, as well as repetitive and restrictive behaviors. Increasing lines of evidence suggest an important role for immune dysregulation and/or inflammation in the development of ASD. Recently, a relationship between inflammation, oxidative stress, and mitochondrial dysfunction has been reported in the brain tissue of individuals with ASD. Some recent studies have also reported oxidative stress and mitochondrial abnormalities in animal models of maternal immune activation (MIA). This review is focused on the hypothesis that MIA induces microglial activation, oxidative stress, and mitochondrial dysfunction, a deleterious trio in the brain that can lead to neuroinflammation and neurodevelopmental pathologies in offspring. Infection during pregnancy activates the mother's immune system to release proinflammatory cytokines, such as IL-6, TNF-α, and others. Furthermore, these cytokines can directly cross the placenta and enter the fetal circulation, or activate resident immune cells, resulting in an increased production of proinflammatory cytokines, including IL-6. Proinflammatory cytokines that cross the blood-brain barrier (BBB) may initiate a neuroinflammation cascade, starting with the activation of the microglia. Inflammatory processes induce oxidative stress and mitochondrial dysfunction that, in turn, may exacerbate oxidative stress in a self-perpetuating vicious cycle that can lead to downstream abnormalities in brain development and behavior.


Subject(s)
Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Autism Spectrum Disorder/therapy , Cytokines/immunology , Disease Models, Animal , Female , Humans , Inflammation Mediators/immunology , Maternal-Fetal Exchange/immunology , Microglia/immunology , Mitochondria/immunology , Models, Immunological , Neuroimmunomodulation , Oxidative Stress/immunology , Pregnancy
11.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576223

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare, multi-system genetic disease with serious neurological and mental symptoms, including autism. Mutations in the TSC1/TSC2 genes lead to the overactivation of mTOR signalling, which is also linked to nonsyndromic autism. Our aim was to analyse synaptic pathology in a transgenic model of TSC: two-month-old male B6;129S4-Tsc2tm1Djk/J mice with Tsc2 haploinsufficiency. Significant brain-region-dependent alterations in the expression of several synaptic proteins were identified. The most prominent changes were observed in the immunoreactivity of presynaptic VAMP1/2 (ca. 50% increase) and phospho-synapsin-1 (Ser62/67) (ca. 80% increase). Transmission electron microscopy demonstrated serious ultrastructural abnormalities in synapses such as a blurred structure of synaptic density and a significantly increased number of synaptic vesicles. The impairment of synaptic mitochondrial ultrastructure was represented by excessive elongation, swelling, and blurred crista contours. Polyribosomes in the cytoplasm and swollen Golgi apparatus suggest possible impairment of protein metabolism. Moreover, the delamination of myelin and the presence of vacuolar structures in the cell nucleus were observed. We also report that Tsc2+/- mice displayed increased brain weights and sizes. The behavioural analysis demonstrated the impairment of memory function, as established in the novel object recognition test. To summarise, our data indicate serious synaptic impairment in the brains of male Tsc2+/- mice.


Subject(s)
Autism Spectrum Disorder/physiopathology , Synapses , Animals , Animals, Genetically Modified , Autism Spectrum Disorder/genetics , Behavior, Animal , Brain/physiology , Cell Nucleus/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Densitometry , Haploinsufficiency , Hippocampus/metabolism , Male , Mice , Microscopy, Electron, Transmission , Organ Size , Phosphorylation , RNA, Messenger/metabolism , Recognition, Psychology , Signal Transduction , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics
12.
Int J Mol Sci ; 22(6)2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33809910

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficient social interaction and communication besides repetitive, stereotyped behaviours. A characteristic feature of ASD is altered dendritic spine density and morphology associated with synaptic plasticity disturbances. Since microtubules (MTs) regulate dendritic spine morphology and play an important role in spine development and plasticity the aim of the present study was to investigate the alterations in the content of neuronal α/ß-tubulin and Tau protein level as well as phosphorylation state in the valproic acid (VPA)-induced rat model of autism. Our results indicated that maternal exposure to VPA induces: (1) decrease the level of α/ß-tubulin along with Tau accumulation in the hippocampus and cerebral cortex; (2) excessive Tau phosphorylation and activation of Tau-kinases: CDK5, ERK1/2, and p70S6K in the cerebral cortex; (3) up-regulation of mTOR kinase-dependent signalling in the hippocampus and cerebral cortex of adolescent rat offspring. Moreover, immunohistochemical staining showed histopathological changes in neurons (chromatolysis) in both analysed brain structures of rats prenatally exposed to VPA. The observed changes in Tau protein together with an excessive decrease in α/ß-tubulin level may suggest destabilization and thus dysfunction of the MT cytoskeleton network, which in consequence may lead to the disturbance in synaptic plasticity and the development of autistic-like behaviours.


Subject(s)
Autistic Disorder/etiology , Autistic Disorder/metabolism , Brain/metabolism , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects , Valproic Acid/adverse effects , tau Proteins/metabolism , Animals , Autistic Disorder/pathology , Biomarkers , Brain/pathology , Disease Susceptibility , Enzyme Activation , Female , Immunohistochemistry , Phosphorylation , Pregnancy , Rats , Signal Transduction , Tubulin/metabolism
13.
Front Aging Neurosci ; 13: 591475, 2021.
Article in English | MEDLINE | ID: mdl-33716707

ABSTRACT

Aberrant secretion and accumulation of α-synuclein (α-Syn) as well as the loss of parkin function are associated with the pathogenesis of Parkinson's disease (PD). Our previous study suggested a functional interaction between those two proteins, showing that the extracellular α-Syn evoked post-translational modifications of parkin, leading to its autoubiquitination and degradation. While parkin plays an important role in mitochondrial biogenesis and turnover, including mitochondrial fission/fusion as well as mitophagy, the involvement of parkin deregulation in α-Syn-induced mitochondrial damage is largely unknown. In the present study, we demonstrated that treatment with exogenous α-Syn triggers mitochondrial dysfunction, reflected by the depolarization of the mitochondrial membrane, elevated synthesis of the mitochondrial superoxide anion, and a decrease in cellular ATP level. At the same time, we observed a protective effect of parkin overexpression on α-Syn-induced mitochondrial dysfunction. α-Syn-dependent disturbances of mitophagy were also shown to be directly related to reduced parkin levels in mitochondria and decreased ubiquitination of mitochondrial proteins. Also, α-Syn impaired mitochondrial biosynthesis due to the parkin-dependent reduction of PGC-1α protein levels. Finally, loss of parkin function as a result of α-Syn treatment induced an overall breakdown of mitochondrial homeostasis that led to the accumulation of abnormal mitochondria. These findings may thus provide the first compelling evidence for the direct association of α-Syn-mediated parkin depletion to impaired mitochondrial function in PD. We suggest that improvement of parkin function may serve as a novel therapeutic strategy to prevent mitochondrial impairment and neurodegeneration in PD (thereby slowing the progression of the disease).

14.
Neuromolecular Med ; 23(1): 118-129, 2021 03.
Article in English | MEDLINE | ID: mdl-32926329

ABSTRACT

The abundance of docosahexaenoic acid (DHA) in phospholipids in the brain and retina has generated interest to search for its role in mediating neurological functions. Besides the source of many oxylipins with pro-resolving properties, DHA also undergoes peroxidation, producing 4-hydroxyhexenal (4-HHE), although its function remains elusive. Despite wide dietary consumption, whether supplementation of DHA may alter the peroxidation products and their relationship to phospholipid species in brain and other body organs have not been explored sufficiently. In this study, adult mice were administered a control or DHA-enriched diet for 3 weeks, and phospholipid species and peroxidation products were examined in brain, heart, and plasma. Results demonstrated that this dietary regimen increased (n-3) and decreased (n-6) species to different extent in all major phospholipid classes (PC, dPE, PE-pl, PI and PS) examined. Besides changes in phospholipid species, DHA-enriched diet also showed substantial increases in 4-HHE in brain, heart, and plasma. Among different brain regions, the hippocampus responded to the DHA-enriched diet showing significant increase in 4-HHE. Considering the pro- and anti-inflammatory pathways mediated by the (n-6) and (n-3) polyunsaturated fatty acids, unveiling the ability for DHA-enriched diet to alter phospholipid species and lipid peroxidation products in the brain and in different body organs may be an important step forward towards understanding the mechanism(s) for this (n-3) fatty acid on health and diseases.


Subject(s)
Brain/drug effects , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Heart/drug effects , Lipid Peroxidation/drug effects , Myocardium/metabolism , Phospholipids/metabolism , Aldehydes/metabolism , Animals , Brain/metabolism , Chromatography, Liquid , Docosahexaenoic Acids/administration & dosage , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Oxidation-Reduction , Phospholipids/analysis , Plasma , Random Allocation , Tandem Mass Spectrometry
15.
Int J Mol Sci ; 21(23)2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33291628

ABSTRACT

Copper is one of the most abundant basic transition metals in the human body. It takes part in oxygen metabolism, collagen synthesis, and skin pigmentation, maintaining the integrity of blood vessels, as well as in iron homeostasis, antioxidant defense, and neurotransmitter synthesis. It may also be involved in cell signaling and may participate in modulation of membrane receptor-ligand interactions, control of kinase and related phosphatase functions, as well as many cellular pathways. Its role is also important in controlling gene expression in the nucleus. In the nervous system in particular, copper is involved in myelination, and by modulating synaptic activity as well as excitotoxic cell death and signaling cascades induced by neurotrophic factors, copper is important for various neuronal functions. Current data suggest that both excess copper levels and copper deficiency can be harmful, and careful homeostatic control is important. This knowledge opens up an important new area for potential therapeutic interventions based on copper supplementation or removal in neurodegenerative diseases including Wilson's disease (WD), Menkes disease (MD), Alzheimer's disease (AD), Parkinson's disease (PD), and others. However, much remains to be discovered, in particular, how to regulate copper homeostasis to prevent neurodegeneration, when to chelate copper, and when to supplement it.


Subject(s)
Copper/metabolism , Disease Susceptibility , Hepatolenticular Degeneration/complications , Hepatolenticular Degeneration/metabolism , Neurodegenerative Diseases/etiology , Animals , Astrocytes/metabolism , Biological Transport , Biomarkers , Brain/metabolism , Brain/pathology , Copper/deficiency , Disease Management , Hepatolenticular Degeneration/genetics , Homeostasis , Humans , Metabolic Networks and Pathways , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Neurons/metabolism , Organ Specificity
16.
Neurochem Int ; 141: 104853, 2020 12.
Article in English | MEDLINE | ID: mdl-32980494

ABSTRACT

Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid ß (Aß), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aß precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , Humans , tau Proteins/metabolism
17.
Int J Mol Sci ; 21(11)2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32521803

ABSTRACT

Maternal immune activation (MIA), induced by infection during pregnancy, is an important risk factor for neuro-developmental disorders, such as autism. Abnormal maternal cytokine signaling may affect fetal brain development and contribute to neurobiological and behavioral changes in the offspring. Here, we examined the effect of lipopolysaccharide-induced MIA on neuro-inflammatory changes, as well as synaptic morphology and key synaptic protein level in cerebral cortex of adolescent male rat offspring. Adolescent MIA offspring showed elevated blood cytokine levels, microglial activation, increased pro-inflammatory cytokines expression and increased oxidative stress in the cerebral cortex. Moreover, pathological changes in synaptic ultrastructure of MIA offspring was detected, along with presynaptic protein deficits and down-regulation of postsynaptic scaffolding proteins. Consequently, ability to unveil MIA-induced long-term alterations in synapses structure and protein level may have consequences on postnatal behavioral changes, associated with, and predisposed to, the development of neuropsychiatric disorders.


Subject(s)
Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Encephalitis/etiology , Encephalitis/metabolism , Immunity , Maternal Exposure , Prenatal Exposure Delayed Effects , Synapses/metabolism , Age Factors , Animals , Autistic Disorder/etiology , Autistic Disorder/metabolism , Autistic Disorder/psychology , Behavior, Animal , Cerebral Cortex/pathology , Disease Models, Animal , Disease Susceptibility , Encephalitis/pathology , Female , Lipopolysaccharides/adverse effects , Maternal Exposure/adverse effects , Oxidative Stress , Phenotype , Pregnancy , Rats
18.
Int J Mol Sci ; 21(11)2020 May 31.
Article in English | MEDLINE | ID: mdl-32486485

ABSTRACT

The purinergic P2X7 receptor (P2X7R) belongs to a family of trimeric ion channels that are gated by extracellular adenosine 5'-triphosphate (ATP). Several studies have pointed to a role of P2X7R-dependent signalling in Parkinson's disease (PD)-related neurodegeneration. The pathology of (PD) is characterized by the formation of insoluble alpha-synuclein (α-Syn) aggregates-Lewy bodies, but the mechanisms underlying α-Syn-induced dopaminergic cell death are still partially unclear. Our previous studies indicate that extracellular α-Syn directly interact with neuronal P2X7R and induces intracellular free calcium mobilization in neuronal cells. The main objective of this study was to examine the involvement of P2X7R receptor in α-Syn-induced mitochondrial dysfunction and cell death. We found that P2X7R stimulation is responsible for α-Syn-induced oxidative stress and activation of the molecular pathways of programmed cell death. Exogenous α-Syn treatment led to P2X7R-dependent decrease in mitochondrial membrane potential as well as elevation of mitochondrial ROS production resulting in breakdown of cellular energy production. Moreover, P2X7R-dependent deregulation of AMP-activated protein kinase as well as decrease in parkin protein level could be responsible for α-Syn-induced mitophagy impairment and accumulation of dysfunctional mitochondria. P2X7R might be putative pharmacological targets in molecular mechanism of extracellular α-Syn toxicity.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mitochondria/pathology , Neuroblastoma/metabolism , Receptors, Purinergic P2X7/metabolism , alpha-Synuclein/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/chemistry , Cell Line, Tumor , Cell Survival , Free Radicals , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitophagy , Neurons/metabolism , Oxidation-Reduction , Oxidative Stress , Signal Transduction
19.
Am J Case Rep ; 21: e923273, 2020 May 12.
Article in English | MEDLINE | ID: mdl-32393730

ABSTRACT

BACKGROUND Central nervous system ischemia in acute pancreatitis is rare with only a handful of cases reported in the literature. We report a case of spinal cord ischemia due to microvascular thrombosis complicating acute on chronic pancreatitis. CASE REPORT A 37-year-old male was transferred to a university hospital intensive care unit with a diagnosis of acute onset chronic pancreatitis, paraplegia, and multi-organ failure. Laboratory studies showed elevated serum amylase activity and leukocytosis. The patient deteriorated quickly and anemia with thrombocytopenia and coagulation abnormalities developed. Computed tomography showed large pancreatic pseudocyst and ischemic lesions in abdominal organs. Symptoms of paraplegia preceded by the bilateral paresis were noted 7 days from the onset of his disease and magnetic resonance imaging showed ischemia involving the central part of the medullary cone resulting from microvascular thrombosis. The patient underwent endoscopic retrograde cholangiopancreatography and repeated surgery with a number of complications but 2 months later was discharged to rehabilitation center due to persistent neurologic deficit. CONCLUSIONS Patients with severe pancreatitis and multiorgan failure requiring intensive care should undergo routine neurological examination to identify and treat deficits early.


Subject(s)
Pancreatitis, Acute Necrotizing/complications , Spinal Cord Ischemia/etiology , Thrombotic Microangiopathies/complications , Adult , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/etiology , Humans , Male , Thrombocytopenia/etiology , Thrombotic Microangiopathies/etiology
20.
Int J Mol Sci ; 21(10)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443651

ABSTRACT

Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental conditions categorized as synaptopathies. Environmental risk factors contribute to ASD aetiology. In particular, prenatal exposure to the anti-epileptic drug valproic acid (VPA) may increase the risk of autism. In the present study, we investigated the effect of prenatal exposure to VPA on the synaptic morphology and expression of key synaptic proteins in the hippocampus and cerebral cortex of young-adult male offspring. To characterize the VPA-induced autism model, behavioural outcomes, microglia-related neuroinflammation, and oxidative stress were analysed. Our data showed that prenatal exposure to VPA impaired communication in neonatal rats, reduced their exploratory activity, and led to anxiety-like and repetitive behaviours in the young-adult animals. VPA-induced pathological alterations in the ultrastructures of synapses accompanied by deregulation of key pre- and postsynaptic structural and functional proteins. Moreover, VPA exposure altered the redox status and expression of proinflammatory genes in a brain region-specific manner. The disruption of synaptic structure and plasticity may be the primary insult responsible for autism-related behaviour in the offspring. The vulnerability of specific synaptic proteins to the epigenetic effects of VPA may highlight the potential mechanisms by which prenatal VPA exposure generates behavioural changes.


Subject(s)
Autism Spectrum Disorder/chemically induced , Microglia/drug effects , Prenatal Exposure Delayed Effects , Synapses/drug effects , Valproic Acid/adverse effects , Animals , Anticonvulsants/adverse effects , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Female , Inflammation , Male , Microglia/metabolism , Microglia/pathology , Oxidative Stress , Pregnancy , Rats , Synapses/pathology , Valproic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL