Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Transl Psychiatry ; 14(1): 289, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39009558

ABSTRACT

Prenatal exposure to infections is a risk factor for neurodevelopmental disorders in offspring, and alterations in mitochondrial function are discussed as a potential underlying factor. Here, using a mouse model of viral-like maternal immune activation (MIA) based on poly(I:C) (POL) treatment at gestational day (GD) 12, we show that adult offspring exhibit behavioral deficits, such as reduced levels of social interaction. In addition, we found increased nicotinamidadenindinucleotid (NADH)- and succinate-linked mitochondrial respiration and maximal electron transfer capacity in the prefrontal cortex (PFC) and in the amygdala (AMY) of males and females. The increase in respiratory capacity resulted from an increase in mitochondrial mass in neurons (as measured by complex IV activity and transcript expression), presumably to compensate for a reduction in mitochondrion-specific respiration. Moreover, in the PFC of control (CON) male offspring a higher excess capacity compared to females was observed, which was significantly reduced in the POL-exposed male offspring, and, along with a higher leak respiration, resulted in a lower mitochondrial coupling efficiency. Transcript expression of the uncoupling proteins (UCP4 and UCP5) showed a reduction in the PFC of POL male mice, suggesting mitochondrial dysfunction. In addition, in the PFC of CON females, a higher expression of the antioxidant enzyme superoxide dismutase (SOD1) was observed, suggesting a higher antioxidant capacity as compared to males. Finally, transcripts analysis of genes involved in mitochondrial biogenesis and dynamics showed reduced expression of fission/fusion transcripts in PFC of POL offspring of both sexes. In conclusion, we show that MIA causes alterations in neuronal mitochondrial function and mass in the PFC and AMY of adult offspring with some effects differing between males and females.


Subject(s)
Mitochondria , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Animals , Female , Prenatal Exposure Delayed Effects/immunology , Pregnancy , Mitochondria/metabolism , Mice , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/immunology , Poly I-C/pharmacology , Disease Models, Animal , Brain/immunology , Brain/metabolism , Amygdala/metabolism , Amygdala/immunology , Behavior, Animal , Mice, Inbred C57BL , Neurons/metabolism , Neurons/immunology
2.
Sci Rep ; 12(1): 2165, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140249

ABSTRACT

Aging and reduced exposure to environmental microbes can both potentiate neuroinflammatory responses. Prior studies indicate that immunization with the immunoregulatory and anti-inflammatory bacterium, Mycobacterium vaccae (M. vaccae), in aged rats limits neuroimmune activation and cognitive impairments. However, the mechanisms by which M. vaccae immunization ameliorates age-associated neuroinflammatory "priming" and whether microglia are a primary target remain unclear. Here, we investigated whether M. vaccae immunization protects against microglia morphological changes in response to aging. Adult (3 mos) and aged (24 mos) Fisher 344 × Brown Norway rats were immunized with either M. vaccae or vehicle once every week for 3 weeks. Aging led to elevated Iba1 immunoreactivity, microglial density, and deramification of microglia processes in the hippocampus and amygdala but not other brain regions. Additionally, aged rats exhibited larger microglial somas in the dorsal hippocampus, suggestive of a more activated phenotype. Notably, M. vaccae treatment ameliorated indicators of microglia activation in both the amygdala and hippocampus. While changes in morphology appeared to be region-specific, gene markers indicative of microglia activation were upregulated by age and lowered in response to M. vaccae in all brain regions evaluated. Taken together, these data suggest that peripheral immunization with M. vaccae quells markers of age-associated microglia activation.


Subject(s)
Aging , Amygdala/cytology , Hippocampus/cytology , Microglia/immunology , Microglia/ultrastructure , Mycobacteriaceae/immunology , Amygdala/immunology , Animals , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/immunology , Hippocampus/immunology , Immunization , Male , Microfilament Proteins/analysis , Microfilament Proteins/immunology , Rats
3.
Behav Brain Res ; 414: 113503, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34331970

ABSTRACT

Maternal infection during pregnancy is an environmental risk factor for neurodevelopmental dysfunction, such as autism spectrum disorder (ASD). This study investigated the effect of maternal immune activation (MIA) on the behavior profile of prepubertal offspring and whether MIA alters the neuronal activation pattern of brain areas related to social play behavior. Pregnant Wistar rats received 500 µg/kg of lipopolysaccharide or saline solution on gestational day 16. Their offspring were tested using behavioral tasks to capture some of the core and associated ASD-like symptoms. Neuronal activation, indexed via c-fos expression after social play behavior, was evaluated in several brain areas. MIA had a number of adverse effects on dams and reduced the number of successful births and litter size. MIA induced sex-specific autistic-like features by a reduction in ultrasonic vocalizations in response to separation from the mother and nest, reduction in discrimination between neutral odors and their nest odor, moderate effect in stereotypies in the hole-board test, impaired risk assessment phenotype, and reduction in social play behavior without changes in locomotor activity only in prepubertal male offspring. A decrease in social play behavior may be associated with a decrease in the number of c-fos-positive cells in the prefrontal cortex and striatum, but hyperactivation of the basolateral and basomedial amygdala. Prenatal immune challenge results in ASD-like symptoms such as impaired risk assessment behavior, communication, and social interactions in male prepubertal offspring. Impaired social play behavior is correlated with neuronal hyperactivation in the amygdala.


Subject(s)
Amygdala , Autism Spectrum Disorder/physiopathology , Behavior, Animal/physiology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Sex Characteristics , Social Behavior , Age Factors , Amygdala/immunology , Amygdala/metabolism , Amygdala/physiopathology , Animals , Disease Models, Animal , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar
4.
Behav Brain Res ; 411: 113371, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34019914

ABSTRACT

Maternal high-fat diets (HFD) can generate inflammation in the offspring's amygdala, which can lead to anxiety-like behaviors. Conversely, lipopolysaccharide (LPS) tolerance can reduce neuroinflammation in the offspring caused by maternal high-fat diets. This study evaluated the combination of LPS tolerance and high-fat maternal diet on amygdala's inflammatory parameters and the anxiety-like behavior in adolescent offspring. Female pregnant Wistar rats received randomly a standard diet or a high-fat diet during gestation and lactation. On gestation days 8, 10, and 12, half of the females in each group were intraperitonially injected with LPS (0.1 mg.kg-1). After weaning, the male offspring (n = 96) were placed in individual boxes in standard conditions, and when 6 weeks-old, the animals underwent: Open-Field, Light/Dark Box, Elevated Plus-Maze, and Rotarod tests. When 50 days-old the offspring were euthanized and the amygdala removed for cytokine and redox status analysis. The offspring in the HFD group showed lower amygdala IL-10 levels, high IL-6/IL-10 ratio, and anxiety-like behaviors. These effects were attenuated in the HFD offspring submitted to LPS tolerance, which showed an anti-inflammatory compensatory response in the amygdala. Also, this group showed a higher activity of the enzyme catalase in the amygdala. In addition, receiving the combination of LPS tolerance and maternal HFD did not lead to anxiety-like behavior in the offspring. The results suggest that LPS tolerance attenuated amygdala inflammation through an anti-inflammatory compensatory response besides preventing anxiety-like behavior caused by the high-fat maternal diet.


Subject(s)
Amygdala/metabolism , Behavior, Animal/drug effects , Drug Tolerance/physiology , Amygdala/immunology , Animals , Anxiety/chemically induced , Anxiety/metabolism , Behavior, Animal/physiology , Brain/metabolism , Diet, High-Fat/adverse effects , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Male , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar
5.
J Integr Neurosci ; 20(4): 933-943, 2021 Dec 30.
Article in English | MEDLINE | ID: mdl-34997716

ABSTRACT

Alcoholism causes various maladaptations in the central nervous system, including the neuroimmune system. Studies of alcohol-induced dysregulation of the neuroimmune system generally focus on alcohol dependence and brain damage, but our previous research indicates that repetitive binge-like consumption perturbs cytokines independent of cell death. This paper extends that research by examining the impact of binge-like consumption on microglia in the hippocampus and the amygdala. Microglia were assessed using immunohistochemistry following binge-like ethanol consumption based on Drinking-in-the-Dark model. Immunohistochemistry results showed that binge-like ethanol consumption caused an increase in Iba-1 immunoreactivity and the number of Iba-1+ cells after one Drinking-in-the-Dark cycle. However, after three Drinking-in-the-Dark cycles, the number of microglia decreased in the hippocampus. We showed that in the dentate gyrus, the average immunoreactivity/cell was increased following ethanol exposure despite the decrease in number after three cycles. Likewise, Ox-42, an indicator of microglia activation, was upregulated after ethanol consumption. No significant effects on microglia number or immunoreactivity (Iba-1 nor Ox-42) were observed in the amygdala. Finally, ethanol caused an increase in the expression of the microglial gene Aif-1 during intoxication and ten days into abstinence, suggesting persistence of ethanol-induced upregulation of microglial genes. Altogether, these findings indicate that repetitive binge-like ethanol is sufficient to elicit changes in microglial reactivity. This altered neuroimmune state may contribute to the development of alcohol use disorders.


Subject(s)
Alcoholism , Binge Drinking , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hippocampus , Microglia , Alcoholism/immunology , Alcoholism/metabolism , Amygdala/drug effects , Amygdala/immunology , Amygdala/metabolism , Animals , Behavior, Animal/physiology , Binge Drinking/immunology , Binge Drinking/metabolism , Central Nervous System Depressants/administration & dosage , Disease Models, Animal , Ethanol/administration & dosage , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism
6.
Article in English | MEDLINE | ID: mdl-32849310

ABSTRACT

G protein-coupled estrogen receptor (GPER) in the amygdala and the dorsal hippocampus mediates actions of estradiol on anxiety, social recognition and spatial memory. In addition, GPER participates in the estrogenic regulation of synaptic function in the amygdala and in the process of adult neurogenesis in the dentate gyrus. While the distribution of the canonical estrogen receptors α and ß in the amygdala and dorsal hippocampus are well characterized, little is known about the regional distribution of GPER in these brain regions and whether this distribution is affected by sex or the stages of the estrous cycle. In this study we performed a morphometric analysis of GPER immunoreactivity in the posterodorsal medial, anteroventral medial, basolateral, basomedial and central subdivisions of the amygdala and in all the histological layers of CA1 and the dentate gyrus of the dorsal hippocampal formation. The number of GPER immunoreactive cells was estimated in these different structures. GPER immunoreactivity was detected in all the assessed subdivisions of the amygdaloid nucleus and dorsal hippocampal formation. The number of GPER immunoreactive cells was higher in males than in estrus females in the central (P = 0.001) and the posterodorsal medial amygdala (P < 0.05); higher in males than in diestrus females in the strata orients (P < 0.01) and radiatum-lacunosum-moleculare (P < 0.05) of CA1-CA3 and in the molecular layer of the dentate gyrus (P < 0.01); higher in diestrus females than in males in the basolateral amygdala (P < 0.05); higher in diestrus females than in estrus females in the central (P < 0.01), posterodorsal medial (P < 0.01) and basolateral amygdala (P < 0.01) and higher in estrus females than in diestrus females in the strata oriens (P < 0.05) and radiatum-lacunosum-moleculare (P < 0.05) of CA1-CA3 and in the molecular layer (P < 0.05) and the hilus of the dentate gyrus (P < 0.05). The findings suggest that estrogenic regulation of the amygdala and hippocampus through GPER may be different in males and in females and may fluctuate during the estrous cycle.


Subject(s)
Amygdala/metabolism , Estrus/physiology , Hippocampus/metabolism , Receptors, G-Protein-Coupled/metabolism , Amygdala/immunology , Animals , Female , Hippocampus/immunology , Male , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/immunology , Sex Factors
7.
Proc Natl Acad Sci U S A ; 117(28): 16475-16480, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601180

ABSTRACT

Autism spectrum disorder (ASD) is characterized by impaired social interactions and communication. The pathogenesis of ASD is not known, but it involves activation of microglia. We had shown that the peptide neurotensin (NT) is increased in the serum of children with ASD and stimulates cultured adult human microglia to secrete the proinflammatory molecules IL-1ß and CXCL8. This process is inhibited by the cytokine IL-37. Another cytokine, IL-38, has been reported to have antiinflammatory actions. In this report, we show that pretreatment of cultured adult human microglia with recombinant IL-38 (aa3-152, 1-100 ng/mL) inhibits (P < 0.0001) NT-stimulated (10 nM) secretion of IL-1ß (at 1 ng/mL) and CXCL8 (at 100 ng/mL). In fact, IL-38 (aa3-152, 1 ng/mL) is more potent than IL-37 (100 ng/mL). Here, we report that pretreatment with IL-38 (100 ng/mL) of embryonic microglia (HMC3), in which secretion of IL-1ß was undetectable, inhibits secretion of CXCL8 (P = 0.004). Gene expression of IL-38 and its receptor IL-36R are decreased (P = 0.001 and P = 0.04, respectively) in amygdala from patients with ASD (n = 8) compared to non-ASD controls (n = 8), obtained from the University of Maryland NeuroBioBank. IL-38 is increased (P = 0.03) in the serum of children with ASD. These findings indicate an important role for IL-38 in the inhibition of activation of human microglia, thus supporting its development as a treatment approach for ASD.


Subject(s)
Amygdala/immunology , Autism Spectrum Disorder/immunology , Interleukins/immunology , Microglia/immunology , Adolescent , Autism Spectrum Disorder/blood , Cells, Cultured , Child , Child, Preschool , Humans , Interleukin-16/blood , Interleukin-16/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Interleukin-8/immunology , Interleukins/blood , Male , Neurotensin/blood , Neurotensin/immunology
8.
Exp Biol Med (Maywood) ; 244(5): 362-371, 2019 04.
Article in English | MEDLINE | ID: mdl-30808184

ABSTRACT

IMPACT STATEMENT: A combined odor and taste cue was paired with a binge-like ethanol exposure (4 g/kg intraperitoneal) using a single-trial learning paradigm. Re-exposure to the CS alone was sufficient to evoke a conditioned Interleukin (IL)-6 elevation in the amygdala in adolescents, an effect that was not observed in young adults. This demonstrates a particular sensitivity of adolescents to alcohol-associated cues and neuroimmune learning, whereas prior work indicated that adults require multiple pairings of ethanol to the CS in order to achieve a conditioned amygdala IL-6 response. While the role of immune conditioning has been studied in other drugs of abuse, these findings highlight a previously unknown aspect of alcohol-related learning. Given the emergent importance of the neuroimmune system in alcohol abuse, these findings may be important for understanding cue-induced reinstatement of alcohol intake among problem drinkers.


Subject(s)
Alcoholism , Conditioning, Classical/physiology , Neuroimmunomodulation/physiology , Age Factors , Alcohol Drinking/immunology , Alcohol Drinking/metabolism , Amygdala/immunology , Amygdala/metabolism , Animals , Cues , Interleukin-6/biosynthesis , Interleukin-6/immunology , Male , Rats , Rats, Sprague-Dawley
9.
Cell Physiol Biochem ; 45(6): 2529-2539, 2018.
Article in English | MEDLINE | ID: mdl-29558743

ABSTRACT

BACKGROUND/AIMS: Impaired fear memory extinction is widely considered a key mechanism of post-traumatic stress disorder (PTSD). Recent studies have suggested that neuroinflammation after a single prolonged stress (SPS) exposure may play a critical role in the impaired fear memory extinction. Studies have shown that high mobility group box chromosomal protein 1 (HMGB-1) is critically involved in neuroinflammation. However, the role of HMGB-1 underlying the development of impairment of fear memory extinction is still not known. METHODS: Thus, we examined the levels of HMGB-1 in the basolateral amygdala (BLA) following SPS using Western blot and evaluated the levels of microglia and astrocytes activation in the BLA after SPS using immunohistochemical staining. We then examined the effects of pre-SPS intra-BLA administration of glycyrrhizin, an HMGB1 inhibitor, or LPS-RS, a competitive TLR4 antagonist, on subsequent post-SPS fear extinction. RESULTS: We found that SPS treatment prolonged the extinction of contextual fear memory after the SPS. The impairment of SPS-induced extinction of contextual fear memory was associated with increased HMGB1 and Toll-like receptor 4 (TLR4) levels in the BLA. Additionally, the impairment of SPS-induced extinction of contextual fear memory was associated with increased activation of microglia and astrocyte in the BLA. Intra-BLA administrations of glycyrrhizin (HMGB-1 inhibitor) or LPS-RS (TLR4 antagonist) can prevent the development of SPS-induced fear extinction impairment. CONCLUSION: Taken together, these results suggested that SPS treatment may not only produce short term effects on the HMGB1/TLR4-mediated pro-inflammation, but alter the response of microglia and astrocytes to the exposure to fear associated contextual stimuli.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Fear/drug effects , Glycyrrhizic Acid/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Stress, Psychological/drug therapy , Amygdala/drug effects , Amygdala/immunology , Amygdala/pathology , Animals , HMGB1 Protein/analysis , HMGB1 Protein/immunology , Male , Memory/drug effects , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/immunology , Stress Disorders, Post-Traumatic/pathology , Stress, Psychological/immunology , Stress, Psychological/pathology , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/immunology
10.
J Neurosci ; 38(13): 3358-3372, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29491010

ABSTRACT

Inflammatory processes may be involved in the pathophysiology of neuropsychiatric illnesses including autism spectrum disorder (ASD). Evidence from studies in rodents indicates that immune activation during early development can produce core features of ASD (social interaction deficits, dysregulation of communication, increases in stereotyped behaviors, and anxiety), although the neural mechanisms of these effects are not thoroughly understood. We treated timed-pregnant mice with polyinosinic:polycytidylic acid (Poly I:C), which simulates a viral infection, or vehicle on gestational day 12.5 to produce maternal immune activation (MIA). Male offspring received either vehicle or lipopolysaccharide, which simulates a bacterial infection, on postnatal day 9 to produce postnatal immune activation (PIA). We then used optogenetics to address the possibility that early developmental immune activation causes persistent alterations in the flow of signals within the mPFC to basolateral amygdala (BLA) pathway, a circuit implicated in ASD. We found that our MIA regimen produced increases in synaptic strength in glutamatergic projections from the mPFC to the BLA. In contrast, our PIA regimen produced decreases in feedforward GABAergic inhibitory postsynaptic responses resulting from activation of local circuit interneurons in the BLA by mPFC-originating fibers. Both effects were seen together when the regimens were combined. Changes in the balance between excitation and inhibition were differentially translated into the modified spike output of BLA neurons. Our findings raise the possibility that prenatal and postnatal immune activation may affect different cellular targets within brain circuits that regulate some of the core behavioral signs of conditions such as ASD.SIGNIFICANCE STATEMENT Immune system activation during prenatal and early postnatal development may contribute to the development of autism spectrum disorder (ASD). Combining optogenetic approaches and behavioral assays that reflect core features of ASD (anxiety, decreased social interactions), we uncovered mechanisms by which the ASD-associated behavioral impairments induced by immune activation could be mediated at the level of interactions within brain circuits implicated in control of emotion and motivation (mPFC and BLA, specifically). Here, we present evidence that prenatal and postnatal immune activation can have different cellular targets in the brain, providing support to the notion that the etiology of ASD may be linked to the excitation/inhibition imbalance in the brain affecting the signal flow within relevant behavior-driving neural microcircuits.


Subject(s)
Amygdala/physiopathology , Autism Spectrum Disorder/immunology , Prefrontal Cortex/physiopathology , Prenatal Exposure Delayed Effects/immunology , Synaptic Transmission , Amygdala/immunology , Animals , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/physiopathology , Female , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Interneurons/metabolism , Interneurons/physiology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/immunology , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/physiopathology
11.
Addict Biol ; 23(3): 889-903, 2018 05.
Article in English | MEDLINE | ID: mdl-28840972

ABSTRACT

Chronic ethanol consumption stimulates neuroimmune signaling in the brain, and Toll-like receptor (TLR) activation plays a key role in ethanol-induced inflammation. However, it is unknown which of the TLR signaling pathways, the myeloid differentiation primary response gene 88 (MyD88) dependent or the TIR-domain-containing adapter-inducing interferon-ß (TRIF) dependent, is activated in response to chronic ethanol. We used voluntary (every-other-day) chronic ethanol consumption in adult C57BL/6J mice and measured expression of TLRs and their signaling molecules immediately following consumption and 24 hours after removing alcohol. We focused on the prefrontal cortex where neuroimmune changes are the most robust and also investigated the nucleus accumbens and amygdala. Tlr mRNA and components of the TRIF-dependent pathway (mRNA and protein) were increased in the prefrontal cortex 24 hours after ethanol and Cxcl10 expression increased 0 hour after ethanol. Expression of Tlr3 and TRIF-related components increased in the nucleus accumbens, but slightly decreased in the amygdala. In addition, we demonstrate that the IKKε/TBK1 inhibitor Amlexanox decreases immune activation of TRIF-dependent pathway in the brain and reduces ethanol consumption, suggesting the TRIF-dependent pathway regulates drinking. Our results support the importance of TLR3 and the TRIF-dependent pathway in ethanol-induced neuroimmune signaling and suggest that this pathway could be a target in the treatment of alcohol use disorders.


Subject(s)
Adaptor Proteins, Vesicular Transport/drug effects , Brain/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Neuroimmunomodulation/drug effects , Toll-Like Receptor 3/drug effects , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Aminopyridines/pharmacology , Amygdala/drug effects , Amygdala/immunology , Animals , Brain/immunology , Chemokine CXCL10/drug effects , Chemokine CXCL10/immunology , I-kappa B Kinase/antagonists & inhibitors , Lipopolysaccharide Receptors/drug effects , Lipopolysaccharide Receptors/immunology , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/immunology , Nucleus Accumbens/drug effects , Nucleus Accumbens/immunology , Prefrontal Cortex/drug effects , Prefrontal Cortex/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 2/immunology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/immunology
12.
Brain Behav Immun ; 66: 210-220, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28698116

ABSTRACT

The contribution of the early postnatal environment to the pervasive effects of prenatal alcohol exposure (PAE) is poorly understood. Moreover, PAE often carries increased risk of exposure to adversity/stress during early life. Dysregulation of immune function may play a role in how pre- and/or postnatal adversity/stress alters brain development. Here, we combine two animal models to examine whether PAE differentially increases vulnerability to immune dysregulation in response to early-life adversity. PAE and control litters were exposed to either limited bedding (postnatal day [PN] 8-12) to model early-life adversity or normal bedding, and maternal behavior and pup vocalizations were recorded. Peripheral (serum) and central (amygdala) immune (cytokines and C-reactive protein - CRP) responses of PAE animals to early-life adversity were evaluated at PN12. Insufficient bedding increased negative maternal behavior in both groups. Early-life adversity increased vocalization in all animals; however, PAE pups vocalized less than controls. Early-life adversity reduced serum TNF-α, KC/GRO, and IL-10 levels in control but not PAE animals. PAE increased serum CRP, and levels were even higher in pups exposed to adversity. Finally, PAE reduced KC/GRO and increased IL-10 levels in the amygdala. Our results indicate that PAE alters immune system development and both behavioral and immune responses to early-life adversity, which could have subsequent consequences for brain development and later life health.


Subject(s)
Ethanol/administration & dosage , Maternal Behavior , Prenatal Exposure Delayed Effects/immunology , Amygdala/immunology , Amygdala/metabolism , Animals , C-Reactive Protein/metabolism , Cytokines/blood , Female , Inflammation/immunology , Inflammation/metabolism , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Rats, Sprague-Dawley , Stress, Psychological/immunology , Stress, Psychological/metabolism , Vocalization, Animal
13.
Brain Res ; 1657: 262-268, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28017669

ABSTRACT

The critical factor considered in a depression induced by diabetes is the inflammation eliciting hippocampal, amygdala and thalamic neuronal injury. Therefore, inhibiting inflammatory reactions in the brain and reducing neuronal injury can alleviate depression in rodents suffering from diabetes mellitus. The oral administration of astaxanthin has been employed in emotional disorders and diabetic complications due to its anti-depressant, anti-inflammatory and anti-apoptotic functions. However, it has not been reported whether astaxanthin can improve diabetes-related depression-like behavior, and its potential mechanisms have not been elucidated. The objective of the present study is to elucidate the effect of astaxanthin on depression in diabetic mice and to understand the underlying molecular mechanisms. In this study, experimental diabetic mice were given a single intraperitoneal injection of streptozotocin (STZ, 150mg/kg, dissolved in citrate buffer) after fasting for 12h. The diabetic model was assessed 72h after STZ injection, and mice with a fasting blood glucose level more than or equal to 16.7mmol/L were used in this study, and oral astaxanthin (25mg/kg) was provided uninterrupted for ten weeks. Depression-like behavior was evaluated by the tail suspension test (TST) and forced swimming test (FST). The glial fibrillary acidic protein (GFAP) and cleaved caspase-3-positive cells were measured by immunohistochemistry, and the western blotting was used to test the protein levels of interleukin-6 (IL-6), interleukin-1ß (IL-1ß) and cyclooxygenase (COX-2). The results showed that astaxanthin had an anti-depressant effect on diabetic mice. Furthermore, we observed that astaxanthin significantly reduced the number of GFAP-positive cells in the hippocampus and hypothalamus, and also the expression of cleaved caspase-3 in the hippocampus, amygdala and hypothalamus was decreased as well. Moreover, astaxanthin could down-regulate the expression of IL-6, IL-1ß and COX-2 in the hippocampus. These findings suggest that the mechanism of astaxanthin in preventing depression in diabetic mice involves the inhibition of inflammation/inflammation inhibition, thereby protecting neurons in hippocampus, amygdala and hypothalamus against hyperglycemic damage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Depression/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Hippocampus/drug effects , Inflammation/drug therapy , Amygdala/drug effects , Amygdala/immunology , Amygdala/pathology , Animals , Antidepressive Agents/pharmacology , Caspase 3/metabolism , Cyclooxygenase 2/metabolism , Depression/immunology , Depression/pathology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Drug Evaluation, Preclinical , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/immunology , Hippocampus/pathology , Hypothalamus/drug effects , Hypothalamus/immunology , Hypothalamus/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/psychology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred ICR , Random Allocation , Xanthophylls/pharmacology
14.
Brain Res ; 1646: 62-72, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27208497

ABSTRACT

Acute ethanol intoxication is associated with Rapid Alterations in Neuroimmune Gene Expression (RANGE), including increased Interleukin (IL)-6 and Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα), and suppressed IL-1ß and Tumor necrosis factor (TNF) α, yet little is known about adaptations in cytokines across the first few ethanol exposures. Thus, the present studies examined central cytokines during intoxication (3h post-ethanol) following 2, 4 or 6 intragastric ethanol challenges (4g/kg) delivered either daily or every-other-day (EOD). Subsequent analyses of blood ethanol concentrations (BECs) and corticosterone were performed to determine whether the schedule of ethanol delivery would alter the pharmacokinetics of, or general sensitivity to, subacute ethanol exposure. As expected, ethanol led to robust increases in IL-6 and IκBα gene expression in hippocampus, amygdala and bed nucleus of the stria terminalis (BNST), whereas IL-1ß and TNFα were suppressed, thereby replicating our prior work. Ethanol-dependent increases in IL-6 and IκBα remained significant in all structures - even after 6 days of ethanol. When these doses were administered EOD, modest IL-6 increases in BNST were observed, with TNFα and IL-1ß suppressed exclusively in the hippocampus. Analysis of BECs revealed a small but significant reduction in ethanol after 4 EOD exposures - an effect which was not observed when ethanol was delivered after 6 daily intubations. These findings suggest that ethanol-induced RANGE effects are not simply a function of ethanol load per se, and underscore the critical role that ethanol dosing interval plays in determining the neuroimmune consequences of alcohol.


Subject(s)
Brain/drug effects , Brain/metabolism , Cytokines/metabolism , Ethanol/administration & dosage , Gene Expression/drug effects , Neuroimmunomodulation/drug effects , Amygdala/drug effects , Amygdala/immunology , Amygdala/metabolism , Animals , Brain/immunology , Corticosterone/blood , Ethanol/blood , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Septal Nuclei/drug effects , Septal Nuclei/immunology , Septal Nuclei/metabolism
15.
Brain Behav Immun ; 52: 40-48, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26386321

ABSTRACT

When memories are recalled, they enter a transient labile phase in which they can be impaired or enhanced followed by a new stabilization process termed reconsolidation. It is unknown, however, whether reconsolidation is restricted to neurocognitive processes such as fear memories or can be extended to peripheral physiological functions as well. Here, we show in a paradigm of behaviorally conditioned taste aversion in rats memory-updating in learned immunosuppression. The administration of sub-therapeutic doses of the immunosuppressant cyclosporin A together with the conditioned stimulus (CS/saccharin) during retrieval blocked extinction of conditioned taste aversion and learned suppression of T cell cytokine (interleukin-2; interferon-γ) production. This conditioned immunosuppression is of clinical relevance since it significantly prolonged the survival time of heterotopically transplanted heart allografts in rats. Collectively, these findings demonstrate that memories can be updated on both neural and behavioral levels as well as on the level of peripheral physiological systems such as immune functioning.


Subject(s)
Avoidance Learning/physiology , Extinction, Psychological/physiology , Mental Recall/physiology , Amygdala/immunology , Amygdala/physiology , Animals , Avoidance Learning/drug effects , Conditioning, Classical/physiology , Cyclosporine/pharmacology , Extinction, Psychological/drug effects , Fear/physiology , Immune Tolerance/physiology , Immunosuppressive Agents/pharmacology , Interferon-gamma/immunology , Interleukin-2/immunology , Male , Mental Recall/drug effects , Rats , Rats, Inbred Strains , Taste/physiology
16.
Toxicology ; 338: 95-103, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26524701

ABSTRACT

Chlorpyrifos (CPF), one of organophosphorus pesticides (OPs), is associated with developmental neurotoxicity. Inflammatory response is closely related with CPF-induced neurotoxicity. The present study aimed at exploring whether sub-toxic CPF exposure on neonatal rats results in neuroinflammation that mediated by HMGB1/TLR4/NF-κB signaling pathway in the amygdala. The neonatal rats were subcutaneously injected with 5mg/kg CPF for 4 consecutive days (postnatal day 11-14) with or without HMGB1 inhibitor, glycyrrhizin. We assessed the levels of pro-inflammatory cytokines at 12, 24, and 72 h after CPF exposure. The role of HMGB1 on neuroinflammation in sub-toxic exposure during brain development was studied. CPF-treated neonatal rats exhibited a significant increase in the expression of pro-inflammatory cytokines, such as IL-6, TNF-α and HMGB1, and a significant increase in the activation of NF-κB in the amygdala after CPF exposure. Inhibited HMGB1 reduced the release of IL-6 and TNF-α, and inhibited activation of NF-κB. Our findings indicate that CPF exposure on developmental brain might induce the activation of neuroinflammation mediated by HMGB1/TLR4/NF-κB pathway in the amygdala.


Subject(s)
Amygdala/drug effects , Chlorpyrifos/toxicity , Encephalitis/prevention & control , HMGB1 Protein/metabolism , Pesticides/toxicity , Amygdala/growth & development , Amygdala/immunology , Amygdala/metabolism , Animals , Animals, Newborn , Cytokines/metabolism , Encephalitis/chemically induced , Encephalitis/immunology , Encephalitis/metabolism , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/antagonists & inhibitors , Inflammation Mediators/metabolism , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , NF-kappa B/metabolism , Protein Transport , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 4/metabolism
17.
eNeuro ; 2(5)2015.
Article in English | MEDLINE | ID: mdl-26730400

ABSTRACT

Epilepsy is a common neurological disorder with many causes. For temporal lobe epilepsy, antecedent insults are typically found. These risk factors include trauma or history of long fever-associated seizures (febrile status epilepticus) in childhood. Whereas the mechanisms by which such insults promote temporal lobe epilepsy are unknown, an extensive body of work has implicated inflammation and inflammatory mediators in both human and animal models of the disorder. However, direct evidence for an epileptogenic role for inflammation is lacking. Here we capitalized on a model where only a subgroup of insult-experiencing rodents develops epilepsy. We reasoned that if inflammation was important for generating epilepsy, then early inflammation should be more prominent in individuals destined to become epileptic compared with those that will not become epileptic. In addition, the molecular and temporal profile of inflammatory mediators would provide insights into which inflammatory pathways might be involved in the disease process. We examined inflammatory profiles in hippocampus and amygdala of individual rats and correlated them with a concurrent noninvasive, amygdalar magnetic resonance imaging epilepsy-predictive marker. We found significant individual variability in the expression of several important inflammatory mediators, but not in others. Of interest, a higher expression of a subset of hippocampal and amygdalar inflammatory markers within the first few hours following an insult correlated with the epilepsy-predictive signal. These findings suggest that some components of the inflammatory gene network might contribute to the process by which insults promote the development of temporal lobe epilepsy.


Subject(s)
Amygdala/immunology , Hippocampus/immunology , Seizures, Febrile/immunology , Status Epilepticus/immunology , Amygdala/pathology , Animals , Astrocytes/immunology , Astrocytes/pathology , Blotting, Western , Disease Models, Animal , Disease Progression , Female , HMGB1 Protein/metabolism , Hippocampus/pathology , Immunohistochemistry , Interleukin-1beta/metabolism , Magnetic Resonance Imaging , Male , Microglia/immunology , Microglia/pathology , Neurons/immunology , Neurons/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Seizures, Febrile/pathology , Status Epilepticus/pathology
18.
Neurobiol Dis ; 74: 347-58, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25511803

ABSTRACT

Mood disturbances are frequent in patients with multiple sclerosis (MS), even in non-disabled patients and in the remitting stages of the disease. It is still largely unknown how the pathophysiological process on MS causes anxiety and depression, but the dopaminergic system is likely involved. Aim of the present study was to investigate depressive-like behavior in mice with experimental autoimmune encephalomyelitis (EAE), a model of MS, and its possible link to dopaminergic neurotransmission. Behavioral, amperometric and biochemical experiments were performed to determine the role of inflammation in mood control in EAE. First, we assessed the independence of mood alterations from motor disability during the acute phase of the disease, by showing a depressive-like behavior in EAE mice with mild clinical score and preserved motor skills (mild-EAE). Second, we linked such behavioral changes to the selective increased striatal expression of interleukin-1beta (IL-1ß) in a context of mild inflammation and to dopaminergic system alterations. Indeed, in the striatum of EAE mice, we observed an impairment of dopamine (DA) neurotransmission, since DA release was reduced and signaling through DA D1- and D2-like receptors was unbalanced. In conclusion, the present study provides first evidence of the link between the depressive-like behavior and the alteration of dopaminergic system in EAE mice, raising the possibility that IL-1ß driven dysfunction of dopaminergic signaling might play a role in mood disturbances also in MS patients.


Subject(s)
Corpus Striatum/immunology , Depression/metabolism , Dopamine/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/psychology , Interleukin-1beta/metabolism , Acute Disease , Amygdala/drug effects , Amygdala/immunology , Amygdala/pathology , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Depression/drug therapy , Depression/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/pathology , Interleukin-1beta/antagonists & inhibitors , Mice, Inbred C57BL , Motor Skills , RNA, Messenger/metabolism , Random Allocation , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Severity of Illness Index , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
19.
Epilepsia ; 54(9): 1679-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23889589

ABSTRACT

PURPOSE: Limbic encephalitis is an autoimmune-mediated disease leading to temporal lobe epilepsy, mnestic deficits, and affective disturbances. Magnetic resonance imaging (MRI) usually shows signal and volume changes of the temporomesial structures. However, these abnormalities may be subtle, thereby hampering the diagnosis by conventional visual assessment. In the present study we evaluated the diagnostic value of a fully automated MRI postprocessing technique in limbic encephalitis and hippocampal sclerosis. METHODS: The MRI postprocessing was based largely on a recently described method allowing for an observer-independent quantification of the fluid-attenuated inversion recovery (FLAIR) signal intensities of amygdala and hippocampus. A 95% confidence region was calculated from the FLAIR intensities of 100 healthy controls. We applied this analysis to the MRI data of 39 patients with antibody-associated limbic encephalitis and 63 patients with hippocampal sclerosis. Moreover, the results were compared to those of visual assessment by an experienced neuroradiologist. KEY FINDINGS: The method detected limbic encephalitis and hippocampal sclerosis with a high sensitivity of 85% and 95%, respectively. The detection rate of the automated approach in limbic encephalitis was significantly superior to visual analysis (85% vs. 51%; p = 0.001), whereas no statistically significant difference for the detection rate in hippocampal sclerosis was found. Patients with limbic encephalitis had significantly higher absolute intensity values of the amygdala and a significantly higher percentage fell outside of the amygdalar confidence region compared to those with hippocampal sclerosis (79% vs. 27%; p < 0.001), whereas we found opposite results in the hippocampal analysis (38% vs. 95%; p < 0.001). SIGNIFICANCE: The FLAIR analysis applied in this study is a powerful tool to quantify signal changes of the amygdala and hippocampus in limbic encephalitis and hippocampal sclerosis. It significantly increases the diagnostic sensitivity in limbic encephalitis in comparison to conventional visual analysis. Furthermore, the method provides an interesting insight into the distinct properties of these two disease entities on MRI, indicating a predominant affection of the amygdala in limbic encephalitis, whereas the affection of the hippocampus is far less pronounced when compared to hippocampal sclerosis.


Subject(s)
Amygdala/pathology , Limbic Encephalitis/pathology , Magnetic Resonance Imaging/methods , Adolescent , Adult , Aged , Amygdala/immunology , Electroencephalography/methods , Epilepsy, Temporal Lobe/immunology , Epilepsy, Temporal Lobe/pathology , Female , Hippocampus/immunology , Hippocampus/pathology , Humans , Male , Middle Aged , Sclerosis/immunology , Sclerosis/pathology , Young Adult
20.
Brain Behav Immun ; 33: 102-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770090

ABSTRACT

Anxiety-like responses to stress are accompanied by elevation of brain cytokine-mRNAs. Because cytokines microinjected into central-amygdala (CeA) substitute for stress in a behavioral paradigm, the possibility was raised that cytokines increased by stress influence behavior by affecting CeA-neural activity. Previously, cytokines increased firing-rate of CeA-neurons comparable to that induced by corticotropin-releasing factor (CRF). In this investigation, tumor-necrosis-factor-α (TNFα) increased amplitude, but not frequency of mEPSCs from CeA-neurons. Additionally, TNFα decreased the threshold for triggering action potentials from CeA-neurons without altering membrane-properties during current-clamp recording. Glutamate-receptor-antagonist blockade of mEPSCs and the TNFα-induced reduction in firing threshold implicated glutamate in these changes. A phosphatidyl-inositol-3-kinase-antagonist prevented the TNFα-induced increased in amplitude of mEPSCs, documenting a TNFα intracellular influence. Additionally, TNFα increased frequency, but not amplitude of mIPSCs. CRF-receptor-antagonists were found to prevent the TNFα-induced increase in mIPSC-frequency, without altering the TNFα-induced amplitude increase in mEPSCs or the reduced threshold for action-potentials by TNFα. To clarify how TNFα was increasing CRF-release in the presence of tetrodotoxin, the possibility tested was whether preventing glial-activation would prevent this elevated mIPSC-frequency blocked by CRF-receptor antagonists. Minocycline, which blocks glial activation, prevented the TNFα-induced increase in mIPSC-frequency - a finding consistent with glia contributing to the CRF-involvement in this TNFα action. To fully understand the means by which a CRF1-receptor-antagonist and minocycline prevent TNFα from increasing mIPSC-frequency will require further clarification. Nonetheless, these data provide convincing evidence that release of TNFα by stress could alter neural activity of CeA-neurons by influencing GABA-and glutamate function.


Subject(s)
Amygdala/immunology , Excitatory Postsynaptic Potentials/immunology , Induced Pluripotent Stem Cells/immunology , Neural Inhibition/immunology , Neurons/immunology , Stress, Physiological/immunology , Synaptic Transmission/immunology , Tumor Necrosis Factor-alpha/physiology , Amygdala/cytology , Amygdala/metabolism , Animals , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...