Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Gut ; 72(10): 1942-1958, 2023 10.
Article in English | MEDLINE | ID: mdl-36593103

ABSTRACT

OBJECTIVE: The current treatment for hepatocellular carcinoma (HCC) to block angiogenesis and immunosuppression provides some benefits only for a subset of patients with HCC, thus optimised therapeutic regimens are unmet needs, which require a thorough understanding of the underlying mechanisms by which tumour cells orchestrate an inflamed tumour microenvironment with significant myeloid cell infiltration. MicroRNA-223 (miR-223) is highly expressed in myeloid cells but its role in regulating tumour microenvironment remains unknown. DESIGN: Wild-type and miR-223 knockout mice were subjected to two mouse models of inflammation-associated HCC induced by injection of diethylnitrosamine (DEN) or orthotopic HCC cell implantation in chronic carbon tetrachloride (CCl4)-treated mice. RESULTS: Genetic deletion of miR-223 markedly exacerbated tumourigenesis in inflammation-associated HCC. Compared with wild-type mice, miR-223 knockout mice had more infiltrated programmed cell death 1 (PD-1+) T cells and programmed cell death ligand 1 (PD-L1+) macrophages after DEN+CCl4 administration. Bioinformatic analyses of RNA sequencing data revealed a strong correlation between miR-223 levels and tumour hypoxia, a condition that is well-documented to regulate PD-1/PD-L1. In vivo and in vitro mechanistic studies demonstrated that miR-223 did not directly target PD-1 and PD-L1 in immune cells rather than indirectly downregulated them by modulating tumour microenvironment via the suppression of hypoxia-inducible factor 1α-driven CD39/CD73-adenosine pathway in HCC. Moreover, gene delivery of miR-223 via adenovirus inhibited angiogenesis and hypoxia-mediated PD-1/PD-L1 activation in both HCC models, thereby hindering HCC progression. CONCLUSION: The miR-223 plays a critical role in modulating hypoxia-induced tumour immunosuppression and angiogenesis, which may serve as a novel therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Mice , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Immunosuppression Therapy , Carcinogenesis , Mice, Knockout , MicroRNAs/genetics , Inflammation , Hypoxia , Tumor Microenvironment
2.
Neuroimage ; 188: 419-426, 2019 03.
Article in English | MEDLINE | ID: mdl-30576849

ABSTRACT

Traumatic Brain Injury (TBI) affects approximately 2.5 million people in the United States, of which 80% are considered to be mild (mTBI). Previous studies have shown that cerebral glucose uptake and metabolism are altered after brain trauma and functional metabolic deficits observed following mTBI are associated with changes in cognitive performance. Imaging of glucose uptake using [18F] Fluorodeoxyglucose (FDG) based Positron Emission Tomography (PET) with anesthesia during the uptake period demonstrated limited variability in results, but may have depressed uptake. Anesthesia has been found to interfere with blood glucose levels, and hence, FDG uptake. Conversely, forced cognitive testing during uptake may increase glucose demand in targeted regions, such as hippocampus, allowing for better differentiation of outcomes. Therefore, the objective of this study was to investigate the influence of a directed cognitive function task during the FDG uptake period on uptake measurements both in naïve rats and at 2 days after mild lateral fluid percussion (mLFP) TBI. Adult male Sprague Dawley rats underwent FDG uptake with either cognitive testing with the Novel Object Recognition (NOR) test or No Novel Object (NNO), followed by PET scans at baseline (prior to injury) and at 2days post mLFP. At baseline, FDG uptake in the right hippocampus was elevated in rats completing the NOR in comparison to the NNO (control group). Further, the NNO group rats demonstrated a greater fold change in the FDG uptake between baseline and post injury scans than the NOR group. Overall, these data suggest that cognitive activity during FDG uptake affects the regional uptake pattern in the brain, increasing uptake at baseline and suppressing the effects of injury.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Functional Laterality/physiology , Hippocampus/physiopathology , Recognition, Psychology/physiology , Animals , Behavior, Animal/physiology , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Fluorodeoxyglucose F18 , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Male , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley
3.
Molecules ; 20(11): 20355-80, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26569216

ABSTRACT

α-Linolenic acid (ALA) is a nutraceutical found in vegetable products such as flax and walnuts. The pleiotropic properties of ALA target endogenous neuroprotective and neurorestorative pathways in brain and involve the transcription factor nuclear factor kappa B (NF-κB), brain-derived neurotrophic factor (BDNF), a major neuroprotective protein in brain, and downstream signaling pathways likely mediated via activation of TrkB, the cognate receptor of BDNF. In this review, we discuss possible mechanisms of ALA efficacy against the highly toxic OP nerve agent soman. Organophosphate (OP) nerve agents are highly toxic chemical warfare agents and a threat to military and civilian populations. Once considered only for battlefield use, these agents are now used by terrorists to inflict mass casualties. OP nerve agents inhibit the critical enzyme acetylcholinesterase (AChE) that rapidly leads to a cholinergic crisis involving multiple organs. Status epilepticus results from the excessive accumulation of synaptic acetylcholine which in turn leads to the overactivation of muscarinic receptors; prolonged seizures cause the neuropathology and long-term consequences in survivors. Current countermeasures mitigate symptoms and signs as well as reduce brain damage, but must be given within minutes after exposure to OP nerve agents supporting interest in newer and more effective therapies. The pleiotropic properties of ALA result in a coordinated molecular and cellular program to restore neuronal networks and improve cognitive function in soman-exposed animals. Collectively, ALA should be brought to the clinic to treat the long-term consequences of nerve agents in survivors. ALA may be an effective therapy for other acute and chronic neurodegenerative disorders.


Subject(s)
Dietary Supplements , Nerve Agents/adverse effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Organophosphates/adverse effects , Signal Transduction/drug effects , alpha-Linolenic Acid/pharmacology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Cognition/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/metabolism , Humans , Models, Animal , Nervous System Diseases/chemically induced , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neuropathology , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Linolenic Acid/therapeutic use
4.
Nat Metab ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902331

ABSTRACT

Alcohol use disorder (AUD) affects millions of people worldwide, causing extensive morbidity and mortality with limited pharmacological treatments. The liver is considered as the principal site for the detoxification of ethanol metabolite, acetaldehyde (AcH), by aldehyde dehydrogenase 2 (ALDH2) and as a target for AUD treatment, however, our recent data indicate that the liver only plays a partial role in clearing systemic AcH. Here we show that a liver-gut axis, rather than liver alone, synergistically drives systemic AcH clearance and voluntary alcohol drinking. Mechanistically, we find that after ethanol intake, a substantial proportion of AcH generated in the liver is excreted via the bile into the gastrointestinal tract where AcH is further metabolized by gut ALDH2. Modulating bile flow significantly affects serum AcH level and drinking behaviour. Thus, combined targeting of liver and gut ALDH2, and manipulation of bile flow and secretion are potential therapeutic strategies to treat AUD.

5.
Exp Biol Med (Maywood) ; 248(7): 596-604, 2023 04.
Article in English | MEDLINE | ID: mdl-37208920

ABSTRACT

Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.


Subject(s)
Basolateral Nuclear Complex , Rats , Humans , Animals , Basolateral Nuclear Complex/metabolism , alpha-Linolenic Acid/pharmacology , Brain-Derived Neurotrophic Factor , Rats, Sprague-Dawley , Activities of Daily Living , Synaptic Transmission/physiology , Receptors, GABA-A/metabolism
6.
J Clin Invest ; 133(15)2023 08 01.
Article in English | MEDLINE | ID: mdl-37338984

ABSTRACT

The liver can fully regenerate after partial resection, and its underlying mechanisms have been extensively studied. The liver can also rapidly regenerate after injury, with most studies focusing on hepatocyte proliferation; however, how hepatic necrotic lesions during acute or chronic liver diseases are eliminated and repaired remains obscure. Here, we demonstrate that monocyte-derived macrophages (MoMFs) were rapidly recruited to and encapsulated necrotic areas during immune-mediated liver injury and that this feature was essential in repairing necrotic lesions. At the early stage of injury, infiltrating MoMFs activated the Jagged1/notch homolog protein 2 (JAG1/NOTCH2) axis to induce cell death-resistant SRY-box transcription factor 9+ (SOX9+) hepatocytes near the necrotic lesions, which acted as a barrier from further injury. Subsequently, necrotic environment (hypoxia and dead cells) induced a cluster of complement 1q-positive (C1q+) MoMFs that promoted necrotic removal and liver repair, while Pdgfb+ MoMFs activated hepatic stellate cells (HSCs) to express α-smooth muscle actin and induce a strong contraction signal (YAP, pMLC) to squeeze and finally eliminate the necrotic lesions. In conclusion, MoMFs play a key role in repairing the necrotic lesions, not only by removing necrotic tissues, but also by inducing cell death-resistant hepatocytes to form a perinecrotic capsule and by activating α-smooth muscle actin-expressing HSCs to facilitate necrotic lesion resolution.


Subject(s)
Actins , Liver Neoplasms , Humans , Actins/metabolism , Liver/metabolism , Hepatocytes/metabolism , Macrophages/metabolism , Hepatic Stellate Cells/metabolism , Necrosis/metabolism , Necrosis/pathology , Liver Neoplasms/metabolism
7.
iScience ; 25(6): 104354, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35601919

ABSTRACT

The NDE1 gene encodes a scaffold protein essential for brain development. Although biallelic NDE1 loss of function (LOF) causes microcephaly with profound mental retardation, NDE1 missense mutations and copy number variations are associated with multiple neuropsychiatric disorders. However, the etiology of the diverse phenotypes resulting from NDE1 aberrations remains elusive. Here we demonstrate Nde1 controls neurogenesis through facilitating H4K20 trimethylation-mediated heterochromatin compaction. This mechanism patterns diverse chromatin landscapes and stabilizes constitutive heterochromatin of neocortical neurons. We demonstrate that NDE1 can undergo dynamic liquid-liquid phase separation, partitioning to the nucleus and interacting with pericentromeric and centromeric satellite repeats. Nde1 LOF results in nuclear architecture aberrations and DNA double-strand breaks, as well as instability and derepression of pericentromeric satellite repeats in neocortical neurons. These findings uncover a pivotal role of NDE1/Nde1 in establishing and protecting neuronal heterochromatin. They suggest that heterochromatin instability predisposes a wide range of brain dysfunction.

8.
iScience ; 25(7): 104519, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35754718

ABSTRACT

Aging is an intricate process characterized by multiple hallmarks including stem cell exhaustion, genome instability, epigenome alteration, impaired proteostasis, and cellular senescence. Whereas each of these traits is detrimental at the cellular level, it remains unclear how they are interconnected to cause systemic organ deterioration. Here we show that abrogating Brap, a BRCA1-associated protein essential for neurogenesis, results in persistent DNA double-strand breaks and elevation of histone H2A mono- and poly-ubiquitination (H2Aub). These defects extend to cellular senescence and proteasome-mediated histone H2A proteolysis with alterations in cells' proteomic and epigenetic states. Brap deletion in the mouse brain causes neuroinflammation, impaired proteostasis, accelerated neurodegeneration, and substantially shortened the lifespan. We further show the elevation of H2Aub also occurs in human brain tissues with Alzheimer's disease. These data together suggest that chromatin aberrations mediated by H2Aub may act as a nexus of multiple aging hallmarks and promote tissue-wide degeneration.

9.
Sci Rep ; 10(1): 17446, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060648

ABSTRACT

Traumatic brain injury generated by blast may induce long-term neurological and psychiatric sequelae. We aimed to identify molecular, histopathological, and behavioral changes in rats 2 weeks after explosive-driven double-blast exposure. Rats received two 30-psi (~ 207-kPa) blasts 24 h apart or were handled identically without blast. All rats were behaviorally assessed over 2 weeks. At Day 15, rats were euthanized, and brains removed. Brains were dissected into frontal cortex, hippocampus, cerebellum, and brainstem. Western blotting was performed to measure levels of total-Tau, phosphorylated-Tau (pTau), amyloid precursor protein (APP), GFAP, Iba1, αII-spectrin, and spectrin breakdown products (SBDP). Kinases and phosphatases, correlated with tau phosphorylation were also measured. Immunohistochemistry for pTau, APP, GFAP, and Iba1 was performed. pTau protein level was greater in the hippocampus, cerebellum, and brainstem and APP protein level was greater in cerebellum of blast vs control rats (p < 0.05). GFAP, Iba1, αII-spectrin, and SBDP remained unchanged. No immunohistochemical or neurobehavioral changes were observed. The dissociation between increased pTau and APP in different regions in the absence of neurobehavioral changes 2 weeks after double blast exposure is a relevant finding, consistent with human data showing that battlefield blasts might be associated with molecular changes before signs of neurological and psychiatric disorders manifest.


Subject(s)
Blast Injuries/pathology , Brain Injuries/pathology , Explosions , Mental Disorders/etiology , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Male , Morris Water Maze Test , Phosphorylation , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
10.
J Neurosci ; 28(5): 1118-30, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18234890

ABSTRACT

Brain-derived neurotrophic factor (BDNF), via activation of TrkB receptors, mediates vital physiological functions in the brain, ranging from neuronal survival to synaptic plasticity, and has been implicated in the pathophysiology of neurodegenerative disorders. Although transcriptional regulation of the BDNF gene (Bdnf) has been extensively studied, much remains to be understood. We discovered a sequence within Bdnf promoter 4 that binds the basic helix-loop-helix protein BHLHB2 and is a target for BHLHB2-mediated transcriptional repression. NMDA receptor activation de-repressed promoter 4-mediated transcription and correlated with reduced occupancy of the promoter by BHLHB2 in cultured hippocampal neurons. Bhlhb2 gene -/- mice showed increased hippocampal exon 4-specific Bdnf mRNA levels compared with +/+ littermates under basal and activity-dependent conditions. Bhlhb2 knock-out mice also showed increased status epilepticus susceptibility, suggesting that BHLHB2 alters neuronal excitability. Together, these results support a role for BHLHB2 as a new modulator of Bdnf transcription and neuronal excitability.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Brain-Derived Neurotrophic Factor/metabolism , Homeodomain Proteins/physiology , Neurons/physiology , Promoter Regions, Genetic/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Female , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Neurons/metabolism , Promoter Regions, Genetic/genetics , Rats
11.
J Neurochem ; 109(5): 1375-88, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19476549

ABSTRACT

To determine the epigenetic events associated with NMDA receptor-mediated activation of brain-derived neurotrophic factor gene (Bdnf) promoter 1 by hippocampal neurons in culture, we screened 12 loci across 4.5 kb of genomic DNA 5' of the transcription start site (TSS) of rat Bdnf for specific changes in histone modification and transcription factor binding following NMDA receptor stimulation. Chromatin immunoprecipitation (ChIP) assays showed that NMDA receptor stimulation produced a durable, time-dependent decrease in histone H3 at lysine 9 dimethylation (H3K9me2), within 3 h after NMDA treatment across multiple loci. Concomitant increases in H3K4me2 and H3K9/14 acetylation (H3AcK9/14) were associated with transcriptional activation, but occurred at fewer sites within the promoter. The decrease in H3K9me2 was associated with release of HDAC1, MBD1, MeCP2, and REST from specific locations within promoter 1, although with different kinetics. In addition, occupancy of sites proximal to and distal to the TSS by the transcription factors NF-kappaB, CREB-binding protein (CBP), and cAMP-response element-binding protein were correlated with increased occupancy of RNA polymerase II at two loci proximal to the TSS following NMDA receptor stimulation. These temporal changes in promoter occupancy could occur thousands of base pairs 5' of the TSS, suggesting a mechanism that produces waves of Bdnf transcription.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Chromatin Assembly and Disassembly/physiology , Hippocampus/cytology , Neurons/metabolism , Promoter Regions, Genetic/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Acetylation/drug effects , Analysis of Variance , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Chromatin Assembly and Disassembly/drug effects , Chromatin Immunoprecipitation/methods , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Exons/physiology , Histone Deacetylase 1 , Histone Deacetylases/metabolism , Histones/metabolism , Hydroxamic Acids/pharmacology , In Vitro Techniques , Lysine/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Methylation/drug effects , N-Methylaspartate/pharmacology , Neurons/drug effects , Promoter Regions, Genetic/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Repressor Proteins/metabolism , Time Factors , Transcription Factors/metabolism
12.
Ageing Res Rev ; 7(1): 21-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17889623

ABSTRACT

The brain developed adaptive mechanisms in the face of changing environments and stresses imposed on the nervous system. The addition of glutamate as the major excitatory amino acid neurotransmitter to the brain's complement of amino acids and peptides dictated a coordinated transcriptional and translational program to meet the demands of excitatory neurotransmission. One such program is the ability of neurons to sustain and maintain their survival given the nature of glutamate-mediated receptor activation. The unique development of endogenous neuronal pathways activated by glutamate receptors transformed neurons and allowed them to survive under conditions of high energy demands. These same endogenous survival pathways also mediate plastic responses to meet another demand of the brain, adaptation. An endogenous protein that plays a central role in glutamate receptor-mediated survival pathways is brain-derived neurotrophic factor (BDNF). Intermittent but frequent synaptic ionotropic glutamate receptor activation ensures neuronal survival through a BDNF autocrine loop. In sharp contrast, overactivation of ionotropic glutamate receptors leads to neuronal cell death. Thus, innovative strategies that induce endogenous neuronal survival pathways through low-level activation of ionotropic glutamate receptors or those that bypass receptor activation but upregulate endogenous survival pathways may not only prevent neurodegenerative disorders that involve glutamate as a final common pathway that kills neurons, but may also provide treatment alternatives critical for neurons to survive stressful conditions such as stroke, status epilepticus and hypoglycemic-induced neuronal cell death.


Subject(s)
Neurodegenerative Diseases/drug therapy , Signal Transduction/physiology , Animals , Brain-Derived Neurotrophic Factor/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Glutamic Acid/physiology , Humans , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/drug effects
13.
Mol Neurobiol ; 55(1): 187-200, 2018 01.
Article in English | MEDLINE | ID: mdl-28844093

ABSTRACT

Approximately, 1.7 million Americans suffer a TBI annually and TBI is a major cause of death and disability. The majority of the TBI cases are of the mild type and while most patients recover completely from mild TBI (mTBI) about 10% result in persistent symptoms and some result in lifelong disability. Anxiety disorders are the second most common diagnosis post-TBI. Of note, TBI-induced anxiety disorders are difficult to treat and remain a chronic condition suggesting that new therapies are needed. Previous work from our laboratory demonstrated that a mild TBI induced an anxiety-like phenotype, a key feature of the human condition, associated with loss of GABAergic interneurons and hyperexcitability in the basolateral amygdala (BLA) in rodents 7 and 30 days after a controlled cortical impact (CCI) injury. We now confirm that animals display significantly increased anxiety-like behavior 30 days after CCI. The anxiety-like behavior was associated with a significant loss of GABAergic interneurons and significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor-mediated inhibitory postsynaptic currents (IPSCs) in the BLA. Significantly, subchronic treatment with alpha-linolenic acid (ALA) after CCI prevents the development of anxiety-like behavior, the loss of GABAergic interneurons, hyperexcitability in the BLA and reduces the impact injury. Taken together, administration of ALA after CCI is a potent therapy against the neuropathology and pathophysiological effects of mTBI in the BLA.


Subject(s)
Anxiety/prevention & control , Brain Injuries, Traumatic/drug therapy , Contusions/drug therapy , alpha-Linolenic Acid/therapeutic use , Animals , Anxiety/etiology , Anxiety/physiopathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/physiopathology , Contusions/etiology , Contusions/physiopathology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome , alpha-Linolenic Acid/pharmacology
14.
Transl Psychiatry ; 8(1): 263, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504810

ABSTRACT

Ketamine is a multimodal dissociative anesthetic, which provides powerful analgesia for victims with traumatic injury. However, the impact of ketamine administration in the peri-trauma period on the development of post-traumatic stress disorder (PTSD) remains controversial. Moreover, there is a major gap between preclinical and clinical studies because they utilize different doses and routes of ketamine administration. Here, we investigated the effects of sub-anesthetic doses of intravenous (IV) ketamine infusion on fear memory and brain glucose metabolism (BGluM) in rats. Male Sprague-Dawley rats received an IV ketamine infusion (0, 2, 10, and 20 mg/kg, 2 h) or an intraperitoneal (IP) injection (0 and 10 mg/kg) following an auditory fear conditioning (3 pairings of tone and foot shock [0.6 mA, 1 s]) on day 0. Fear memory retrieval, fear extinction, and fear recall were tested on days 2, 3, and 4, respectively. The effects of IV ketamine infusion (0 and 10 mg/kg) on BGluM were measured using 18F-fluoro-deoxyglucose positron emission tomography (FDG-PET) and computed tomography (CT). The IV ketamine infusion dose-dependently enhanced fear memory retrieval, delayed fear extinction, and increased fear recall in rats. The IV ketamine (10 mg/kg) increased BGluM in the hippocampus, amygdala, and hypothalamus, while decreasing it in the cerebellum. On the contrary, a single ketamine injection (10 mg/kg, IP) after fear conditioning facilitated fear memory extinction in rats. The current findings suggest that ketamine may produce differential effects on fear memory depending on the route and duration of ketamine administration.


Subject(s)
Brain/drug effects , Brain/metabolism , Fear , Glucose/metabolism , Ketamine/administration & dosage , Memory/drug effects , Administration, Intravenous , Animals , Conditioning, Classical , Extinction, Psychological/drug effects , Fluorodeoxyglucose F18 , Locomotion/drug effects , Male , Mental Recall/drug effects , Positron-Emission Tomography , Rats, Sprague-Dawley
15.
Neuromolecular Med ; 17(3): 251-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25920465

ABSTRACT

Exposure to organophosphorous (OP) nerve agents such as soman inhibits the critical enzyme acetylcholinesterase (AChE) leading to excessive acetylcholine accumulation in synapses, resulting in cholinergic crisis, status epilepticus and brain damage in survivors. The hippocampus is profoundly damaged after soman exposure leading to long-term memory deficits. We have previously shown that treatment with three sequential doses of alpha-linolenic acid, an essential omega-3 polyunsaturated fatty acid, increases brain plasticity in naïve animals. However, the effects of this dosing schedule administered after a brain insult and the underlying molecular mechanisms in the hippocampus are unknown. We now show that injection of three sequential doses of alpha-linolenic acid after soman exposure increases the endogenous expression of mature BDNF, activates Akt and the mammalian target of rapamycin complex 1 (mTORC1), increases neurogenesis in the subgranular zone of the dentate gyrus, increases retention latency in the passive avoidance task and increases animal survival. In sharp contrast, while soman exposure also increases mature BDNF, this increase did not activate downstream signaling pathways or neurogenesis. Administration of the inhibitor of mTORC1, rapamycin, blocked the alpha-linolenic acid-induced neurogenesis and the enhanced retention latency but did not affect animal survival. Our results suggest that alpha-linolenic acid induces a long-lasting neurorestorative effect that involves activation of mTORC1 possibly via a BDNF-TrkB-mediated mechanism.


Subject(s)
Avoidance Learning/drug effects , Hippocampus/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Soman/toxicity , alpha-Linolenic Acid/pharmacology , Animals , Antigens, Nuclear/biosynthesis , Antigens, Nuclear/genetics , Atropine Derivatives/therapeutic use , Avoidance Learning/physiology , Brain Damage, Chronic/etiology , Brain Damage, Chronic/physiopathology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , DNA Replication/drug effects , Diazepam/therapeutic use , Doublecortin Domain Proteins , Electroshock , Exploratory Behavior/drug effects , Hippocampus/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuropeptides/biosynthesis , Neuropeptides/genetics , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Neurotoxins/metabolism , Oximes/therapeutic use , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Pyridinium Compounds/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, trkB/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics , alpha-Linolenic Acid/antagonists & inhibitors , alpha-Linolenic Acid/therapeutic use
16.
Neurotoxicology ; 51: 38-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386148

ABSTRACT

Exposure to nerve agents results in severe seizures or status epilepticus caused by the inhibition of acetylcholinesterase, a critical enzyme that breaks down acetylcholine to terminate neurotransmission. Prolonged seizures cause brain damage and can lead to long-term consequences. Current countermeasures are only modestly effective against the brain damage supporting interest in the evaluation of new and efficacious therapies. The nutraceutical alpha-linolenic acid (LIN) is an essential omega-3 polyunsaturated fatty acid that has a wide safety margin. Previous work showed that a single intravenous injection of alpha-linolenic acid (500 nmol/kg) administered before or after soman significantly protected against soman-induced brain damage when analyzed 24h after exposure. Here, we show that administration of three intravenous injections of alpha-linolenic acid over a 7 day period after soman significantly improved motor performance on the rotarod, enhanced memory retention, exerted an anti-depressant-like activity and increased animal survival. This dosing schedule significantly reduced soman-induced neuronal degeneration in four major vulnerable brain regions up to 21 days. Taken together, alpha-linolenic acid reduces the profound behavioral deficits induced by soman possibly by decreasing neuronal cell death, and increases animal survival.


Subject(s)
Antidepressive Agents/administration & dosage , Cognition/drug effects , Neuroprotective Agents/administration & dosage , Soman/toxicity , alpha-Linolenic Acid/administration & dosage , Animals , Avoidance Learning/drug effects , Body Weight/drug effects , Brain/drug effects , Brain/pathology , Dietary Supplements , Male , Motor Activity/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Rotarod Performance Test
17.
Cell Mol Immunol ; 1(1): 43-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-16212920

ABSTRACT

Interleukin-22 (IL-22) is a recently identified T cell-derived cytokine whose biological significance remains obscure. Previously, we have shown that IL-22 plays a protective role in T cell-mediated hepatitis induced by Concanavalin A (Con A), acting as a survival factor for hepatocytes. In the present paper, we demonstrate that hydrodynamic gene delivery of IL-22 cDNA driven either by a liver-specific albumin promoter or a human cytomegalovirus (CMV) promoter results in IL-22 protein expression, STAT3 activation, and expression of several anti-apoptotic proteins, including Bcl-xL, Bcl-2, and Mcl-1 in the liver. Immunohistochemical analysis reveals that IL-22 protein expression is mainly detected in the cytoplasm of hepatocytes. Overexpression of IL-22 by hydrodynamic gene delivery significantly protects against liver injury, necrosis, and apoptosis induced by administration of Con A, carbon tetrachloride (CCl4), or the Fas agonist Jo-2 mAb. Western blot analyses show that overexpression of IL-22 significantly enhances activation of STAT3 and expression of Bcl-xL, Bcl-2, and Mcl-1 proteins in liver injury induced by Con A. In conclusion, hydrodynamic gene delivery of IL-22 protects against liver injury induced by a variety of toxins, suggesting the therapeutic potential of IL-22 in treating human liver disease.


Subject(s)
Carbon Tetrachloride/toxicity , Concanavalin A/metabolism , Gene Transfer Techniques , Interleukins/metabolism , Liver/physiology , Membrane Glycoproteins/metabolism , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factors/metabolism , Animals , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Fas Ligand Protein , Humans , Interleukins/genetics , Liver/pathology , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism , Interleukin-22
18.
PLoS One ; 9(6): e99818, 2014.
Article in English | MEDLINE | ID: mdl-24955574

ABSTRACT

Organophosphorus (OP) nerve agents are deadly chemical weapons that pose an alarming threat to military and civilian populations. The irreversible inhibition of the critical cholinergic degradative enzyme acetylcholinesterase (AChE) by OP nerve agents leads to cholinergic crisis. Resulting excessive synaptic acetylcholine levels leads to status epilepticus that, in turn, results in brain damage. Current countermeasures are only modestly effective in protecting against OP-induced brain damage, supporting interest for evaluation of new ones. (-)-Phenserine is a reversible AChE inhibitor possessing neuroprotective and amyloid precursor protein lowering actions that reached Phase III clinical trials for Alzheimer's Disease where it exhibited a wide safety margin. This compound preferentially enters the CNS and has potential to impede soman binding to the active site of AChE to, thereby, serve in a protective capacity. Herein, we demonstrate that (-)-phenserine protects neurons against soman-induced neuronal cell death in rats when administered either as a pretreatment or post-treatment paradigm, improves motoric movement in soman-exposed animals and reduces mortality when given as a pretreatment. Gene expression analysis, undertaken to elucidate mechanism, showed that (-)-phenserine pretreatment increased select neuroprotective genes and reversed a Homer1 expression elevation induced by soman exposure. These studies suggest that (-)-phenserine warrants further evaluation as an OP nerve agent protective strategy.


Subject(s)
Chemical Warfare Agents/toxicity , Cholinesterase Inhibitors/pharmacology , Physostigmine/analogs & derivatives , Soman/toxicity , Status Epilepticus , Animals , Carrier Proteins/biosynthesis , Gene Expression Regulation/drug effects , Homer Scaffolding Proteins , Male , Physostigmine/pharmacology , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism
19.
Neurotoxicology ; 33(5): 1219-29, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22884490

ABSTRACT

Nerve agents are deadly threats to military and civilian populations around the world. Nerve agents cause toxicity to peripheral and central sites through the irreversible inhibition of acetylcholinesterase, the enzyme that metabolizes acetylcholine. Excessive acetylcholine accumulation in synapses results in status epilepticus in the central nervous system. Prolonged status epilepticus leads to brain damage, neurological dysfunction and poor outcome. Anticonvulsants are effective but must be given rapidly following exposure. Because these agents cause mass casualties, effective neuroprotective agents are needed to reduce brain damage and improve cognitive outcome. α-Linolenic acid is an omega-3 fatty acid that is found in vegetable products and has no known side effects. α-Linolenic acid is neuroprotective against kainic acid-induced brain damage in vivo, but its neuroprotective efficacy against nerve agents is unknown. α-Linolenic acid also exerts anti-depressant and anti-inflammatory activities and enhances synaptic plasticity in vivo. These properties make this polyunsaturated fatty acid (PUFA) a potential candidate against nerve agent-induced neuropathology. Here we show that α-linolenic acid is neuroprotective against soman-induced neuropathology in either a pretreatment or post-treatment paradigm. We also show that subcutaneous injection of α-linolenic acid shows greater neuroprotective efficacy compared with intravenous injection in a brain region-specific manner.


Subject(s)
Cholinesterase Inhibitors/toxicity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , alpha-Linolenic Acid/therapeutic use , Analysis of Variance , Animals , Brain Injuries/etiology , Brain Injuries/prevention & control , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Electrophoretic Mobility Shift Assay , Fluoresceins , Male , NF-kappa B/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurotoxicity Syndromes/complications , Organic Chemicals , Rats , Rats, Sprague-Dawley , Soman/toxicity
20.
Dose Response ; 9(3): 416-33, 2011.
Article in English | MEDLINE | ID: mdl-22013403

ABSTRACT

Synaptic function is critical for the brain to process experiences dictated by the environment requiring change over the lifetime of the organism. Experience-driven adaptation requires that receptors, signal transduction pathways, transcription and translational mechanisms within neurons respond rapidly over its lifetime. Adaptive responses communicated through the rapid firing of neurons are dependent upon the integrity and function of synapses. These rapid responses via adaptation underlie the organism's ability to perceive, learn, remember, calculate and plan. Glutamate, the endogenous neurotransmitter required for physiological excitation in the brain, is critically involved in neuronal adaptive responses and in the pathophysiology of neurodegenerative disorders. Using neuronal experimental systems, we will discuss how compounds with low dose effects mediated via glutamate receptors can result either in a neuroprotective or neurotoxic response. Because the brain has evolved to respond rapidly to environmental cues, exposure of neurons to stressful stimuli can result in a pivotal response toward either synaptic adaptation or dysfunction and neuronal cell death. Understanding how neurons adapt to stressful stimuli will provide important clues toward the development of strategies to protect the brain against neurodegeneration.

SELECTION OF CITATIONS
SEARCH DETAIL