Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Neurosci Lett ; 764: 136235, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34508846

ABSTRACT

The mechanisms involved in the maintenance of cigarette smoking and nicotine reward remain unclear. Immune response might play an important role in this context. Nicotine may induce both central and systemic inflammatory responses as well as changes in the regulation of brain-derived neurotrophic factor (BDNF). The conditioned place preference (CPP) is a method used for the evaluation of nicotine-induced reward, reproducing nicotine-seeking behavior in humans. So far, there are no studies investigating the relationship between neuroinflammation and nicotine-induced CPP. This study aimed to evaluate the levels of inflammatory mediators and neurotrophic factors in key areas of the central nervous system (CNS) of mice subject to nicotine-induced CPP. CPP was induced with an intraperitoneal administration of 0.5 mg/kg of nicotine in male Swiss mice, using an unbiased protocol. Control group received vehicle by the same route. The levels of cytokines, chemokines, and neurotrophic factors were measured using Enzyme-Linked Immunosorbent Assay (ELISA) in the brain after CPP test. As expected, nicotine induced place preference behavior. In parallel, we observed increased peripheral levels of IL-6 and IL-10 alongside increased hippocampal levels of NGF but decreased GDNF in mice treated with nicotine compared to controls. In the striatum, nicotine promoted decrease of IL-1ß, IL-10 and GDNF levels, while the levels of all the mediators were similar between groups in the pre-frontal cortex. Our results provide evidence on the role of cytokines and neurotrophic factors in nicotine-induced CPP in mice.


Subject(s)
Conditioning, Psychological/drug effects , Neuroinflammatory Diseases/psychology , Nicotine/administration & dosage , Reward , Tobacco Use Disorder/psychology , Animals , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/immunology , Hippocampus/pathology , Humans , Injections, Intraperitoneal , Interleukin-10/analysis , Interleukin-10/metabolism , Interleukin-1beta/analysis , Interleukin-1beta/metabolism , Male , Mice , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Nicotine/adverse effects , Prefrontal Cortex/immunology , Prefrontal Cortex/pathology , Tobacco Use Disorder/immunology , Tobacco Use Disorder/pathology
2.
Front Immunol ; 12: 644294, 2021.
Article in English | MEDLINE | ID: mdl-33953715

ABSTRACT

The basal ganglia network is represented by an interconnected group of subcortical nuclei traditionally thought to play a crucial role in motor learning and movement execution. During the last decades, knowledge about basal ganglia physiology significantly evolved and this network is now considered as a key regulator of important cognitive and emotional processes. Accordingly, the disruption of basal ganglia network dynamics represents a crucial pathogenic factor in many neurological and psychiatric disorders. The striatum is the input station of the circuit. Thanks to the synaptic properties of striatal medium spiny neurons (MSNs) and their ability to express synaptic plasticity, the striatum exerts a fundamental integrative and filtering role in the basal ganglia network, influencing the functional output of the whole circuit. Although it is currently established that the immune system is able to regulate neuronal transmission and plasticity in specific cortical areas, the role played by immune molecules and immune/glial cells in the modulation of intra-striatal connections and basal ganglia activity still needs to be clarified. In this manuscript, we review the available evidence of immune-based regulation of synaptic activity in the striatum, also discussing how an abnormal immune activation in this region could be involved in the pathogenesis of inflammatory and degenerative central nervous system (CNS) diseases.


Subject(s)
Basal Ganglia/immunology , Corpus Striatum/immunology , Neurodegenerative Diseases/immunology , Neuroimmunomodulation , Synaptic Transmission/immunology , Animals , Basal Ganglia/pathology , Corpus Striatum/pathology , Humans , Neurodegenerative Diseases/pathology
3.
Neurorehabil Neural Repair ; 35(6): 501-512, 2021 06.
Article in English | MEDLINE | ID: mdl-33825570

ABSTRACT

BACKGROUND: Very early exercise has been reported to exacerbate motor dysfunction; however, its mechanism is largely unknown. OBJECTIVE: This study examined the effect of very early exercise on motor recovery and associated brain damage following intracerebral hemorrhage (ICH) in rats. METHODS: Collagenase solution was injected into the left striatum to induce ICH. Rats were randomly assigned to receive placebo surgery without exercise (SHAM) or ICH without (ICH) or with very early exercise within 24 hours of surgery (ICH+VET). We observed sensorimotor behaviors before surgery, and after surgery preexercise and postexercise. Postexercise brain tissue was collected 27 hours after surgery to investigate the hematoma area, brain edema, and Il1b, Tgfb1, and Igf1 mRNA levels in the striatum and sensorimotor cortex using real-time reverse transcription polymerase chain reaction. NeuN, PSD95, and GFAP protein expression was analyzed by Western blotting. RESULTS: We observed significantly increased skillful sensorimotor impairment in the horizontal ladder test and significantly higher Il1b mRNA levels in the striatum of the ICH+VET group compared with the ICH group. NeuN protein expression was significantly reduced in both brain regions of the ICH+VET group compared with the SHAM group. CONCLUSION: Our results suggest that very early exercise may be associated with an exacerbation of motor dysfunction because of increased neuronal death and region-specific changes in inflammatory factors. These results indicate that implementing exercise within 24 hours after ICH should be performed with caution.


Subject(s)
Cerebral Hemorrhage , Exercise Therapy/adverse effects , Motor Activity/physiology , Neuroinflammatory Diseases , Neurological Rehabilitation , Physical Conditioning, Animal/physiology , Animals , Behavior, Animal/physiology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/physiopathology , Cerebral Hemorrhage/rehabilitation , Corpus Striatum/immunology , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/physiopathology , Random Allocation , Rats , Rats, Wistar , Sensorimotor Cortex/immunology , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/physiopathology
4.
Immunity ; 54(2): 225-234.e6, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33476547

ABSTRACT

Microglia are activated in many neurological diseases and have been suggested to play an important role in the development of affective disorders including major depression. To investigate how microglial signaling regulates mood, we used bidirectional chemogenetic manipulations of microglial activity in mice. Activation of microglia in the dorsal striatum induced local cytokine expression and a negative affective state characterized by anhedonia and aversion, whereas inactivation of microglia blocked aversion induced by systemic inflammation. Interleukin-6 signaling and cyclooxygenase-1 mediated prostaglandin synthesis in the microglia were critical for the inflammation-induced aversion. Correspondingly, microglial activation led to a prostaglandin-dependent reduction of the excitability of striatal neurons. These findings demonstrate a mechanism by which microglial activation causes negative affect through prostaglandin-dependent modulation of striatal neurons and indicate that interference with this mechanism could milden the depressive symptoms in somatic and psychiatric diseases involving microglial activation.


Subject(s)
Anhedonia/physiology , Corpus Striatum/immunology , Depression/immunology , Microglia/immunology , Neurons/physiology , Animals , Animals, Genetically Modified , Behavior, Animal , Cells, Cultured , Disease Models, Animal , Humans , Inflammation , Interleukin-6/metabolism , Macrophage Activation , Mice , Neurogenic Inflammation , Prostaglandins/metabolism
5.
Neurobiol Dis ; 149: 105229, 2021 02.
Article in English | MEDLINE | ID: mdl-33352233

ABSTRACT

Alpha-synuclein (a-syn) can aggregate and form toxic oligomers and insoluble fibrils which are the main component of Lewy bodies. Intra-neuronal Lewy bodies are a major pathological characteristic of Parkinson's disease (PD). These fibrillar structures can act as seeds and accelerate the aggregation of monomeric a-syn. Indeed, recent studies show that injection of preformed a-syn fibrils (PFF) into the rodent brain can induce aggregation of the endogenous monomeric a-syn resulting in neuronal dysfunction and eventual cell death. We injected 8 µg of murine a-syn PFF, or soluble monomeric a-syn into the right striatum of rats. The animals were monitored behaviourally using the cylinder test, which measures paw asymmetry, and the corridor task that measures lateralized sensorimotor response to sugar treats. In vivo PET imaging was performed after 6, 13 and 22 weeks using [11C]DTBZ, a marker of the vesicular monoamine 2 transporter (VMAT2), and after 15 and 22 weeks using [11C]UCB-J, a marker of synaptic SV2A protein in nerve terminals. Histology was performed at the three time points using antibodies against dopaminergic markers, aggregated a-syn, and MHCII to evaluate the immune response. While the a-syn PFF injection caused only mild behavioural changes, [11C]DTBZ PET showed a significant and progressive decrease of VMAT2 binding in the ipsilateral striatum. This was accompanied by a small progressive decrease in [11C]UCB-J binding in the same area. In addition, our histological analysis revealed a gradual spread of misfolded a-syn pathology in areas anatomically connected to striatum that became bilateral with time. The striatal a-syn PFF injection resulted in a progressive unilateral degeneration of dopamine terminals, and an early and sustained presence of MHCII positive ramified microglia in the ipsilateral striatum and substantia nigra. Our study shows that striatal injections of a-syn fibrils induce progressive pathological synaptic dysfunction prior to cell death that can be detected in vivo with PET. We confirm that intrastriatal injection of a-syn PFFs provides a model of progressive a-syn pathology with loss of dopaminergic and synaptic function accompanied by neuroinflammation, as found in human PD.


Subject(s)
Corpus Striatum/metabolism , Disease Progression , Dopaminergic Neurons/metabolism , Positron-Emission Tomography/methods , Protein Aggregates/physiology , alpha-Synuclein/toxicity , Animals , Corpus Striatum/immunology , Corpus Striatum/pathology , Dopaminergic Neurons/immunology , Dopaminergic Neurons/pathology , Female , Injections, Intraventricular , Rats , Rats, Sprague-Dawley , alpha-Synuclein/administration & dosage , alpha-Synuclein/immunology
6.
Am J Psychiatry ; 178(1): 48-64, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32539528

ABSTRACT

OBJECTIVE: Pediatric obsessive-compulsive disorder (OCD) sometimes appears rapidly, even overnight, often after an infection. Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections, or PANDAS, describes such a situation after infection with Streptococcus pyogenes. PANDAS may result from induced autoimmunity against brain antigens, although this remains unproven. Pilot work suggests that IgG antibodies from children with PANDAS bind to cholinergic interneurons (CINs) in the striatum. CIN deficiency has been independently associated with tics in humans and with repetitive behavioral pathology in mice, making it a plausible locus of pathology. The authors sought to replicate and extend earlier work and to investigate the cellular effects of PANDAS antibodies on cholinergic interneurons. METHODS: Binding of IgG to specific neurons in human and mouse brain slices was evaluated ex vivo after incubation with serum from 27 children with rigorously characterized PANDAS, both at baseline and after intravenous immunoglobulin (IVIG) treatment, and 23 matched control subjects. Binding was correlated with symptom measures. Neural activity after serum incubation was assessed in mouse slices using molecular markers and electrophysiological recording. RESULTS: IgG from children with PANDAS bound to CINs, but not to several other neuron types, more than IgG from control subjects, in three independent cohorts of patients. Post-IVIG serum had reduced IgG binding to CINs, and this reduction correlated with symptom improvement. Baseline PANDAS sera decreased activity of striatal CINs, but not of parvalbumin-expressing GABAergic interneurons, and altered their electrophysiological responses, in acute mouse brain slices. Post-IVIG PANDAS sera and IgG-depleted baseline sera did not alter the activity of striatal CINs. CONCLUSIONS: These findings provide strong evidence for striatal CINs as a critical cellular target that may contribute to pathophysiology in children with rapid-onset OCD symptoms, and perhaps in other conditions.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Cholinergic Neurons/immunology , Corpus Striatum/immunology , Obsessive-Compulsive Disorder/immunology , Streptococcal Infections/immunology , Animals , Autoimmune Diseases/complications , Case-Control Studies , Child , Child, Preschool , Cholinergic Neurons/physiology , Corpus Striatum/physiopathology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obsessive-Compulsive Disorder/complications , Obsessive-Compulsive Disorder/etiology , Streptococcal Infections/complications
7.
Sci Rep ; 10(1): 16647, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024242

ABSTRACT

Nonhuman primates (NHPs) are an essential research model for gaining a comprehensive understanding of the neural mechanisms of neurocognitive aging in our own species. In the present study, we used resting state functional connectivity (rsFC) to investigate the relationship between prefrontal cortical and striatal neural interactions, and cognitive flexibility, in unanaesthetized common marmosets (Callithrix jacchus) at two time points during late middle age (8 months apart, similar to a span of 5-6 years in humans). Based on our previous findings, we also determine the reproducibility of connectivity measures over the course of 8 months, particularly previously observed sex differences in rsFC. Male marmosets exhibited remarkably similar patterns of stronger functional connectivity relative to females and greater cognitive flexibility between the two imaging time points. Network analysis revealed that the consistent sex differences in connectivity and related cognitive associations were characterized by greater node strength and/or degree values in several prefrontal, premotor and temporal regions, as well as stronger intra PFC connectivity, in males compared to females. The current study supports the existence of robust sex differences in prefrontal and striatal resting state networks that may contribute to differences in cognitive function and offers insight on the neural systems that may be compromised in cognitive aging and age-related conditions such as mild cognitive impairment and Alzheimer's disease.


Subject(s)
Aging/psychology , Callithrix/psychology , Cognition/physiology , Corpus Striatum/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Sex Characteristics , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Corpus Striatum/immunology , Female , Male
8.
Biosci Trends ; 14(4): 271-278, 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32741856

ABSTRACT

Tourette's syndrome (TS) is an inherited neurologic disorder characterized by involuntary stereotyped motor and vocal tics. Its pathogenesis is still unclear and its treatment remains limited. Recent research has suggested the involvement of immune mechanisms in the pathophysiology of TS. Microglia are the brain's resident innate immune cells. They can mediate neuroinflammation and regulate brain development and homeostasis. A traditional Chinese medicine (TCM), Ningdong granule (NDG), has been found to be efficacious in the treatment of TS while causing few adverse reactions. In the current study, a rat model of 3,3'-iminodipropionitrile (IDPN)-induced TS was used to explore the regulating effects and mechanisms of NDG on microglia-mediated neuroinflammation. IDNP led to robust pathological changes and neurobehavioral complications, with activation of microglia in the striatum of rats with TS. After activation by IDNP, microglia strongly responded to this specific injury, and TNF-α, IL-6, and MCP-1 were released in the striatum and/or serum of rats with TS. Interestingly, NDG inhibited the activation of microglia and decreased the abnormal expression of TNF-α, IL-6, and MCP-1 in the striatum and/or serum of rats with TS, thus controlling tics. However, there were no significant changes in the striatum and/or serum of rats with TS after treatment with haloperidol. The anti-TS action of haloperidol might occur not through microglial activation and neuroinflammation but through the DAT system, thus controlling tics. In conclusion, microglia might play key roles in mediating neuroinflammatory responses in TS, triggering the release of TNF-α, IL-6, and MCP-1.NDG inhibited tics in rats with TS, and this mechanism may be associated with a reduction in the increased number of activated microglia and a decrease in the expression of pro-inflammatory cytokines and chemokines in the striatum and/or serum.


Subject(s)
Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Microglia/drug effects , Tourette Syndrome/drug therapy , Animals , Chemokine CCL2/blood , Chemokine CCL2/metabolism , Corpus Striatum/cytology , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Haloperidol/pharmacology , Haloperidol/therapeutic use , Humans , Interleukin-6/blood , Interleukin-6/metabolism , Male , Microglia/immunology , Microglia/metabolism , Nitriles/toxicity , Rats , Rats, Wistar , Tourette Syndrome/chemically induced , Tourette Syndrome/immunology , Tourette Syndrome/pathology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
9.
J Neuroimmune Pharmacol ; 15(2): 224-237, 2020 06.
Article in English | MEDLINE | ID: mdl-31802419

ABSTRACT

Regulatory T cells (Tregs), which secrete transforming growth factor (TGF)-ß and interleukin (IL)-10, have essential role in anti-inflammatory and neurotrophic functions. Herein, we explore the neuroprotection of Tregs in Parkinson's disease (PD) by adoptive transfer of Tregs. Tregs, isolated by magnetic sorting, were activated in vitro and then were adoptively transferred to 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP)-treated mice. Neuroinflammation, dopaminergic neuronal loss and behavioral changes of PD mice were evaluated. Live cell imaging system detected a dynamic contact of Tregs with MN9D cells that were stained with CD45 and galectin-1, respectively. Tregs prevented MPTP-induced dopaminergic neuronal loss, behavioral changes, and attenuated the inflammatory reaction in the brain. When blockade the LFA-1 activity in Tregs or the ICAM-1 activity in endothelial cells, the percentage of Tregs in substantia nigra (SN) decreased. CD45 and galectin-1 were expressed by Tregs and MN9D cells, respectively. CD45-labeled Tregs dynamically contacted with galectin-1-labeled MN9D cells. Inhibiting CD45 in Tregs impaired the ability of Tregs to protect dopaminergic neurons against MPP+ toxicity. Similarly, galectin-1 knockdown in MN9D cells reduced the ability of Tregs neuroprotection. Adoptive transfer of Tregs protects dopaminergic neurons in PD mice by a cell-to-cell contact mechanism underlying CD45-galectin-1 interaction. Graphical Abstract.


Subject(s)
Adoptive Transfer/methods , Dopaminergic Neurons/immunology , Inflammation Mediators/immunology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Corpus Striatum/immunology , Corpus Striatum/pathology , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/pathology , T-Lymphocytes, Regulatory/transplantation
10.
Brain Behav Immun ; 81: 630-645, 2019 10.
Article in English | MEDLINE | ID: mdl-31351185

ABSTRACT

Neuroinflammation has been involved in pathogenesis of Parkinson's disease (PD), a chronic neurodegenerative disease characterized neuropathologically by progressive dopaminergic neuronal loss in the substantia nigra (SN). We recently have shown that helper T (Th)17 cells facilitate dopaminergic neuronal loss in vitro. Herein, we demonstrated that interleukin (IL)-17A, a proinflammatory cytokine produced mainly by Th17 cells, contributed to PD pathogenesis depending on microglia. Mouse and rat models for PD were prepared by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or striatal injection of 1-methyl-4-phenylpyridinium (MPP+), respectively. Both in MPTP-treated mice and MPP+-treated rats, blood-brain barrier (BBB) was disrupted and IL-17A level increased in the SN but not in cortex. Effector T (Teff) cells that were adoptively transferred via tail veins infiltrated into the brain of PD mice but not into that of normal mice. The Teff cell transfer aggravated nigrostriatal dopaminergic neurodegeneration, microglial activation and motor impairment. Contrarily, IL-17A deficiency alleviated BBB disruption, dopaminergic neurodegeneration, microglial activation and motor impairment. Anti-IL-17A-neutralizing antibody that was injected into lateral cerebral ventricle in PD rats ameliorated the manifestations mentioned above. IL-17A activated microglia but did not directly affect dopaminergic neuronal survival in vitro. IL-17A exacerbated dopaminergic neuronal loss only in the presence of microglia, and silencing IL-17A receptor gene in microglia abolished the IL-17A effect. IL-17A-treated microglial medium that contained higher concentration of tumor necrosis factor (TNF)-α facilitated dopaminergic neuronal death. Further, TNF-α-neutralizing antibody attenuated MPP+-induced neurotoxicity. The findings suggest that IL-17A accelerates neurodegeneration in PD depending on microglial activation and at least partly TNF-α release.


Subject(s)
Interleukin-17/immunology , Microglia/immunology , Parkinson Disease/immunology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cell Death/immunology , Corpus Striatum/immunology , Disease Models, Animal , Dopamine/immunology , Dopaminergic Neurons/immunology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neuroimmunomodulation/immunology , Rats , Rats, Sprague-Dawley , Substantia Nigra/immunology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/metabolism
11.
Environ Toxicol ; 34(10): 1114-1120, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31231976

ABSTRACT

The aim of this study was designed to investigate the effects of rhynchophyllin (RH) on neuroinflammation in Tourette syndrome (TS) rats. TS model was established in rats by the injection of selective 5-HT2A/2C agonist 1-(2, 5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Behavior in DOI-induced rats was tested. Inflammatory cytokines levels such as interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in serum and striatum were detected. The expression levels of janus kinase 2 (JAK2)/signal transducer and transcription activator 3 (STAT3) and nuclear factor (NF)-κB pathways in striatum were measured by Western blot. Data indicated that RH can significantly reduce the numbers of nodding experiment of TS rats. RH significantly decreased IL-6, IL-1ß, and TNF-α in serum and striatum of TS rats, with altered expression of P-JAK2, P-STAT3, P-NF-κBp65, and P-IκBα in TS rats, as evidenced by Western blot analysis and immunohistochemistry, suggesting that the regulation of JAK2/STAT3 and NF-κB pathways might be involved in the mechanism of RH on TS.


Subject(s)
Corpus Striatum/immunology , Drugs, Chinese Herbal/administration & dosage , Janus Kinase 2/immunology , Oxindoles/administration & dosage , Tourette Syndrome/drug therapy , Uncaria/chemistry , Animals , Corpus Striatum/drug effects , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Janus Kinase 2/genetics , Male , NF-kappa B/genetics , NF-kappa B/immunology , Propane/adverse effects , Propane/analogs & derivatives , Rats , Rats, Wistar , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Tourette Syndrome/chemically induced , Tourette Syndrome/genetics , Tourette Syndrome/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
12.
Transl Psychiatry ; 9(1): 151, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31123247

ABSTRACT

Psychiatric disorders such as schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD) arise from complex interactions between genetic and environmental factors. Common genetic variants associated with multiple psychiatric disorders suggest that shared genetic architecture could contribute to divergent clinical syndromes. To evaluate shared transcriptional alterations across connected brain regions, Affymetrix microarrays were used to profile postmortem dorsolateral prefrontal cortex (DLPFC), hippocampus, and associative striatum from 19 well-matched tetrads of subjects with SCZ, BD, MDD, or unaffected controls. SCZ subjects showed a substantial burden of differentially expressed genes across all examined brain regions with the greatest effects in hippocampus, whereas BD and MDD showed less robust alterations. Pathway analysis of transcriptional profiles compared across diagnoses demonstrated commonly enriched pathways between all three disorders in hippocampus, significant overlap between SCZ and BD in DLPFC, but no significant overlap of enriched pathways between disorders in striatum. SCZ samples showed increased expression of transcripts associated with inflammation across all brain regions examined, which was not evident in BD or MDD, or in rat brain following chronic dosing with antipsychotic drugs. Several markers of inflammation were confirmed by RT-PCR in hippocampus, including S100A8/9, IL-6, MAFF, APOLD1, IFITM3, and BAG3. A cytokine ELISA panel showed significant increases in IL-2 and IL-12p70 protein content in hippocampal tissue collected from same SCZ subjects when compared to matched control subjects. These data suggest an overlapping subset of dysregulated pathways across psychiatric disorders; however, a widespread increase in inflammation appears to be a specific feature of the SCZ brain and is not likely to be attributable to chronic antipsychotic drug treatment.


Subject(s)
Bipolar Disorder , Corpus Striatum , Depressive Disorder, Major , Gene Expression Profiling , Hippocampus , Inflammation , Prefrontal Cortex , Schizophrenia , Animals , Autopsy , Bipolar Disorder/genetics , Bipolar Disorder/immunology , Bipolar Disorder/metabolism , Corpus Striatum/immunology , Corpus Striatum/metabolism , Depressive Disorder, Major/genetics , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Hippocampus/immunology , Hippocampus/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Male , Prefrontal Cortex/immunology , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Schizophrenia/genetics , Schizophrenia/immunology , Schizophrenia/metabolism
13.
J Neuroinflammation ; 16(1): 91, 2019 Apr 17.
Article in English | MEDLINE | ID: mdl-30995916

ABSTRACT

BACKGROUND: During excitotoxic damage, neuronal death results from the increase in intracellular calcium, the induction of oxidative stress, and a subsequent inflammatory response. NADPH oxidases (NOX) are relevant sources of reactive oxygen species (ROS) during excitotoxic damage. NADPH oxidase-2 (NOX-2) has been particularly related to neuronal damage and death, as well as to the resolution of the subsequent inflammatory response. As ROS are crucial components of the regulation of inflammatory response, in this work, we evaluated the role of NOX-2 in the progression of inflammation resulting from glutamate-induced excitotoxic damage of the striatum in an in vivo model. METHODS: The striata of wild-type C57BL/6 J and NOX-2 KO mice (gp91Cybbtm1Din/J) were stereotactically injected with monosodium glutamate either alone or in combination with IL-4 or IL-10. The damage was evaluated in histological sections stained with cresyl violet and Fluoro-Jade B. The enzymatic activity of caspase-3 and NOX were also measured. Additionally, the cytokine profile was identified by ELISA and motor activity was verified by the tests of the cylinder, the adhesive tape removal, and the inverted grid. RESULTS: Our results show a neuroprotective effect in mice with a genetic inhibition of NOX-2, which is partially due to a differential response to excitotoxic damage, characterized by the production of anti-inflammatory cytokines. In NOX-2 KO animals, the excitotoxic condition increased the production of interleukin-4, which could contribute to the production of interleukin-10 that decreased neuronal apoptotic death and the magnitude of striatal injury. Treatment with interleukin-4 and interleukin-10 protected from excitotoxic damage in wild-type animals. CONCLUSIONS: The release of proinflammatory cytokines during the excitotoxic event promotes an additional apoptotic death of neurons that survived the initial damage. During the subsequent inflammatory response to excitotoxic damage, ROS generated by NOX-2 play a decisive role in the extension of the lesion and consequently in the severity of the functional compromise, probably by regulating the anti-inflammatory cytokines production.


Subject(s)
Corpus Striatum/enzymology , Corpus Striatum/pathology , Inflammation/enzymology , Inflammation/pathology , NADPH Oxidase 2/metabolism , Animals , Corpus Striatum/immunology , Disease Progression , Glutamic Acid/toxicity , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Brain Behav Immun ; 80: 146-162, 2019 08.
Article in English | MEDLINE | ID: mdl-30853569

ABSTRACT

Gintonin (GT), a ginseng-derived lysophosphatidic acid receptor ligand, regulates various cellular effects and represses inflammation. However, little is known about the potential value of GT regarding inflammation in the neurodegenerative diseases, such as Huntington's disease (HD). In this study, we investigated whether GT could ameliorate the neurological impairment and striatal toxicity in cellular or animal model of HD. Pre-, co-, and onset-treatment with GT (25, 50, or 100 mg/kg/day, p.o.) alleviated the severity of neurological impairment and lethality following 3-nitropropionic acid (3-NPA). Pretreatment with GT also attenuated mitochondrial dysfunction i.e. succinate dehydrogenase and MitoSOX activities, apoptosis, microglial activation, and mRNA expression of inflammatory mediators i.e. IL-1ß, IL-6, TNF-α, COX-2, and iNOS in the striatum after 3-NPA-intoxication. Its action mechanism was associated with lysophosphatidic acid receptors (LPARs) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway activations and the inhibition of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) signaling pathways. These beneficial effects of GT were neutralized by pre-inhibiting LPARs with Ki16425 (a LPAR1/3 antagonist). Interestingly, GT reduced cell death and mutant huntingtin (HTT) aggregates in STHdh cells. It also mitigated neurological impairment in mice with adeno-associated viral (AAV) vector serotype DJ-mediated overexpression of N171-82Q-mutant HTT in the striatum. Taken together, our findings firstly suggested that GT has beneficial effects with a wide therapeutic time-window in 3-NPA-induced striatal toxicity by antioxidant and anti-inflammatory activities through LPA. In addition, GT exerts neuroprotective effects in STHdh cells and AAV vector-infected model of HD. Thus GT might be an innovative therapeutic candidate to treat HD-like syndromes.


Subject(s)
NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Death/drug effects , Corpus Striatum/immunology , Corpus Striatum/metabolism , Disease Models, Animal , Huntington Disease/drug therapy , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Panax , Plant Extracts/metabolism , Receptors, Lysophosphatidic Acid/drug effects , Receptors, Lysophosphatidic Acid/physiology , Signal Transduction/drug effects
15.
PLoS One ; 13(11): e0207027, 2018.
Article in English | MEDLINE | ID: mdl-30403753

ABSTRACT

Opioids are widely used for treating different types of pains, but overuse and abuse of prescription opioids have led to opioid epidemic in the United States. Besides analgesic effects, chronic use of opioid can also cause tolerance, dependence, and even addiction. Effective treatment of opioid addiction remains a big challenge today. Studies on addictive effects of opioids focus on striatum, a main component in the brain responsible for drug dependence and addiction. Some transcription regulators have been associated with opioid addiction, but relationship between analgesic effects of opioids and dependence behaviors mediated by them at the molecular level has not been thoroughly investigated. In this paper, we developed a new computational strategy that identifies novel targets and potential therapeutic molecular compounds for opioid dependence and addiction. We employed several statistical and machine learning techniques and identified differentially expressed genes over time which were associated with dependence-related behaviors after exposure to either morphine or heroin, as well as potential transcription regulators that regulate these genes, using time course gene expression data from mouse striatum. Moreover, our findings revealed that some of these dependence-associated genes and transcription regulators are known to play key roles in opioid-mediated analgesia and tolerance, suggesting that an intricate relationship between opioid-induce pain-related pathways and dependence may develop at an early stage during opioid exposure. Finally, we determined small compounds that can potentially target the dependence-associated genes and transcription regulators. These compounds may facilitate development of effective therapy for opioid dependence and addiction. We also built a database (http://daportals.org) for all opioid-induced dependence-associated genes and transcription regulators that we discovered, as well as the small compounds that target those genes and transcription regulators.


Subject(s)
Analgesics, Opioid/adverse effects , Machine Learning , Opioid-Related Disorders/etiology , Analgesics, Opioid/pharmacology , Animals , Corpus Striatum/drug effects , Corpus Striatum/immunology , Corpus Striatum/metabolism , Gene Expression Regulation/drug effects , Gene Ontology , Heroin/toxicity , Mice , Morphine/adverse effects , Morphine/pharmacology , Neurons/drug effects , Neurons/metabolism , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/therapy , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
16.
AJNR Am J Neuroradiol ; 39(12): 2263-2269, 2018 12.
Article in English | MEDLINE | ID: mdl-30467216

ABSTRACT

Systemic lupus erythematosus is a chronic autoimmune disease characterized by the production of autoantibodies resulting in tissue injury across multiple organs; up to 50% of patients develop neurologic involvement, collectively referred to as neuropsychiatric systemic lupus erythematosus. The cases in this clinical report will highlight a subtype of neuropsychiatric systemic lupus erythematosus demonstrating imaging findings of striatal inflammation responsive to plasmapheresis similar to those in the subset of N-methyl-D-aspartate receptor autoimmune encephalitis that involves the striatum. Although the cause for this striking imaging appearance is not definitely known, literature will be presented supporting the hypothesis that it is due to peripheral anti-double-stranded DNA antibodies entering the central nervous system to cross-react with N-methyl-D-aspartate receptor antigens.


Subject(s)
Corpus Striatum/immunology , Encephalitis/immunology , Encephalitis/pathology , Lupus Vasculitis, Central Nervous System/immunology , Lupus Vasculitis, Central Nervous System/pathology , Adult , Aged , Antibodies, Antinuclear/immunology , Autoantibodies/immunology , Corpus Striatum/diagnostic imaging , Corpus Striatum/pathology , Cross Reactions , Encephalitis/therapy , Female , Humans , Lupus Vasculitis, Central Nervous System/therapy , Male , Plasmapheresis , Receptors, N-Methyl-D-Aspartate/immunology , Young Adult
17.
Sci Rep ; 8(1): 10188, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976948

ABSTRACT

Increased glucocorticoid concentrations have been shown to favor resilience towards autoimmune phenomena. Here, we addressed whether experimentally induced elevations in circulating glucocorticoids mitigate the abnormalities exhibited by an experimental model of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus (PANDAS). This is a pathogenic hypothesis linking repeated exposures to Group-A-beta-hemolytic streptococcus (GAS), autoantibodies targeting selected brain nuclei and neurobehavioral abnormalities. To persistently elevate glucocorticoid concentrations, we supplemented lactating SJL/J mice with corticosterone (CORT; 80 mg/L) in the drinking water. Starting in adolescence (postnatal day 28), developing offspring were exposed to four injections - at bi-weekly intervals - of a GAS homogenate and tested for behavioral, immunological, neurochemical and molecular alterations. GAS mice showed increased perseverative behavior, impaired sensorimotor gating, reduced reactivity to a serotonergic agonist and inflammatory infiltrates in the anterior diencephalon. Neonatal CORT persistently increased circulating glucocorticoids concentrations and counteracted these alterations. Additionally, neonatal CORT increased peripheral and CNS concentrations of the anti-inflammatory cytokine IL-9. Further, upstream regulator analysis of differentially expressed genes in the striatum showed that the regulatory effect of estradiol is inhibited in GAS-treated mice and activated in GAS-treated mice exposed to CORT. These data support the hypothesis that elevations in glucocorticoids may promote central immunomodulatory processes.


Subject(s)
Autoimmune Diseases/immunology , Corpus Striatum/immunology , Corticosterone/immunology , Obsessive-Compulsive Disorder/immunology , Streptococcal Infections/immunology , Stress, Psychological/immunology , Animals , Animals, Newborn , Autoimmune Diseases/blood , Autoimmune Diseases/diagnosis , Autoimmune Diseases/microbiology , Behavior Observation Techniques , Behavior, Animal , Corpus Striatum/metabolism , Corticosterone/administration & dosage , Corticosterone/blood , Disease Models, Animal , Female , Gene Expression Profiling , Interleukin-9/immunology , Interleukin-9/metabolism , Lactation , Male , Mice , Mice, Inbred Strains , Obsessive-Compulsive Disorder/blood , Obsessive-Compulsive Disorder/diagnosis , Obsessive-Compulsive Disorder/microbiology , Streptococcal Infections/blood , Streptococcal Infections/diagnosis , Streptococcal Infections/microbiology , Streptococcus/pathogenicity , Stress, Psychological/blood
18.
Sci Rep ; 8(1): 10722, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30013201

ABSTRACT

We previously demonstrated that pretreatment with Exendin-4, a glucagon-like peptide-1 (GLP-1) receptor agonist, reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) -mediated dopaminergic neurodegeneration. The use of GLP-1 or Exendin-4 for Parkinson's disease (PD) patients is limited by their short half-lives. The purpose of this study was to evaluate a new extended release Exendin-4 formulation, PT302, in a rat model of PD. Subcutaneous administration of PT302 resulted in sustained elevations of Exendin-4 in plasma for >20 days in adult rats. To define an efficacious dose within this range, rats were administered PT302 once every 2 weeks either before or following the unilaterally 6-hydroxydopamine lesioning. Pre- and post-treatment with PT302 significantly reduced methamphetamine-induced rotation after lesioning. For animals given PT302 post lesion, blood and brain samples were collected on day 47 for measurements of plasma Exendin-4 levels and brain tyrosine hydroxylase immunoreactivity (TH-IR). PT302 significantly increased TH-IR in the lesioned substantia nigra and striatum. There was a significant correlation between plasma Exendin-4 levels and TH-IR in the substantia nigra and striatum on the lesioned side. Our data suggest that post-treatment with PT302 provides long-lasting Exendin-4 release and reduces neurodegeneration of nigrostriatal dopaminergic neurons in a 6-hydroxydopamine rat model of PD at a clinically relevant dose.


Subject(s)
Dopaminergic Neurons/drug effects , Exenatide/administration & dosage , Incretins/administration & dosage , Parkinson Disease, Secondary/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/immunology , Corpus Striatum/pathology , Delayed-Action Preparations/administration & dosage , Disease Models, Animal , Dopaminergic Neurons/immunology , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Male , Oxidopamine/administration & dosage , Oxidopamine/toxicity , Parkinson Disease, Secondary/etiology , Parkinson Disease, Secondary/immunology , Parkinson Disease, Secondary/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/immunology , Substantia Nigra/pathology , Treatment Outcome , Tyrosine 3-Monooxygenase/immunology
20.
Toxicol Lett ; 294: 73-86, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29775722

ABSTRACT

Bifenthrin (BF) is a synthetic pyrethroid pesticide widely used in several countries to manage insect pests on diverse agricultural crops. Growing evidence indicates that BF exposure is associated with an increased risk of developing neurodegenerative disorders. However, the mechanisms by which BF induces neurological and anxiety alterations in the frontal cortex and striatum are not well known. The present in vivo study was carried out to determine whether reactive oxygen species (ROS)-mediated oxidative stress (OS) and neuroinflammation are involved in such alterations. Thirty-six Wistar rats were thus randomly divided into three groups and were orally administered with BF (0.6 and 2.1 mg/kg body weight, respectively) or the vehicle (corn oil), on a daily basis for 60 days. Results revealed that BF exposure in rats enhanced anxiety-like behavior after 60 days of treatment, as assessed with the elevated plus-maze test by decreases in the percentage of time spent in open arms and frequency of entries into these arms. BF-treated rats also exhibited increased oxidation of lipids and carbonylated proteins in the frontal cortex and striatum, and decreased glutathione levels and antioxidant enzyme activities including superoxide dismutase, catalase and glutathione peroxidase. Treatment with BF also increased protein synthesis and mRNA expression of the inflammatory mediators cyclooxygenase-2 (COX-2), microsomal prostaglandin synthase-1 (mPGES-1) and nuclear factor-kappaBp65 (NF-kBp65), as well as the production of tumor necrosis factor-α (TNF-α) and ROS. Moreover, BF exposure significantly decreased protein synthesis and mRNA expression of nuclear factor erythroid-2 (Nrf2) and acetylcholinesterase (AChE), as well as gene expression of muscarinic-cholinergic receptors (mAchR) and choline acetyltransferase (ChAT) in the frontal cortex and striatum. These data suggest that BF induced neurological alterations in the frontal cortex and striatum of rats, and that this may be associated with neuroinflammation and oxidative stress via the activation of Nrf2/NF-kBp65 pathways, which might promote anxiety-like behavior.


Subject(s)
Anxiety/etiology , Insecticides/toxicity , Neuritis/chemically induced , Neurotoxicity Syndromes/physiopathology , Oxidative Stress/drug effects , Pyrethrins/toxicity , Tremor/etiology , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/immunology , Cholinergic Neurons/metabolism , Corpus Striatum/drug effects , Corpus Striatum/immunology , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/metabolism , Gene Expression Regulation/drug effects , Insecticides/administration & dosage , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuritis/immunology , Neuritis/metabolism , Neuritis/physiopathology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Pyrethrins/administration & dosage , Random Allocation , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...