Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(30): e2408109121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39028694

ABSTRACT

The prevalence of "long COVID" is just one of the conundrums highlighting how little we know about the lung's response to viral infection, particularly to syndromecoronavirus-2 (SARS-CoV-2), for which the lung is the point of entry. We used an in vitro human lung system to enable a prospective, unbiased, sequential single-cell level analysis of pulmonary cell responses to infection by multiple SARS-CoV-2 strains. Starting with human induced pluripotent stem cells and emulating lung organogenesis, we generated and infected three-dimensional, multi-cell-type-containing lung organoids (LOs) and gained several unexpected insights. First, SARS-CoV-2 tropism is much broader than previously believed: Many lung cell types are infectable, if not through a canonical receptor-mediated route (e.g., via Angiotensin-converting encyme 2(ACE2)) then via a noncanonical "backdoor" route (via macropinocytosis, a form of endocytosis). Food and Drug Administration (FDA)-approved endocytosis blockers can abrogate such entry, suggesting adjunctive therapies. Regardless of the route of entry, the virus triggers a lung-autonomous, pulmonary epithelial cell-intrinsic, innate immune response involving interferons and cytokine/chemokine production in the absence of hematopoietic derivatives. The virus can spread rapidly throughout human LOs resulting in mitochondrial apoptosis mediated by the prosurvival protein Bcl-xL. This host cytopathic response to the virus may help explain persistent inflammatory signatures in a dysfunctional pulmonary environment of long COVID. The host response to the virus is, in significant part, dependent on pulmonary Surfactant Protein-B, which plays an unanticipated role in signal transduction, viral resistance, dampening of systemic inflammatory cytokine production, and minimizing apoptosis. Exogenous surfactant, in fact, can be broadly therapeutic.


Subject(s)
COVID-19 , Lung , Organoids , SARS-CoV-2 , Virus Internalization , Humans , SARS-CoV-2/physiology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , Lung/virology , Lung/immunology , Lung/pathology , Organoids/virology , COVID-19 Drug Treatment , Induced Pluripotent Stem Cells/virology , Angiotensin-Converting Enzyme 2/metabolism , Inflammation , Cytokines/metabolism , Apoptosis
2.
bioRxiv ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36747824

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) causes an acute respiratory distress syndrome (ARDS) that resembles surfactant deficient RDS. Using a novel multi-cell type, human induced pluripotent stem cell (hiPSC)-derived lung organoid (LO) system, validated against primary lung cells, we found that inflammatory cytokine/chemokine production and interferon (IFN) responses are dynamically regulated autonomously within the lung following SARS-CoV-2 infection, an intrinsic defense mechanism mediated by surfactant proteins (SP). Single cell RNA sequencing revealed broad infectability of most lung cell types through canonical (ACE2) and non-canonical (endocytotic) viral entry routes. SARS-CoV-2 triggers rapid apoptosis, impairing viral dissemination. In the absence of surfactant protein B (SP-B), resistance to infection was impaired and cytokine/chemokine production and IFN responses were modulated. Exogenous surfactant, recombinant SP-B, or genomic correction of the SP-B deletion restored resistance to SARS-CoV-2 and improved viability.

3.
Front Synaptic Neurosci ; 14: 1067420, 2022.
Article in English | MEDLINE | ID: mdl-36713088

ABSTRACT

Introduction: Two weeks of voluntary exercise in group-housed mice produces a reduction in anxiety-like behaviors across a number of different measures, including a reduction in the anxiety levels typically produced by the anxiogenic serotonergic drug m-chlorophenylpiperazine (mCPP), an agonist at 5-HT2C/2b receptors. We have previously demonstrated that 2-weeks of voluntary exercise blunted the anxiogenic effects of systemic mCPP, and we have also shown that mCPP infused into the bed nucleus of the stria terminalis (BNST) is anxiogenic. Here we follow up on these reports. Methods: In Experiment 1 we infused several doses of mCPP into the BNST with or without the 5-HT2C antagonist SB242084. In Experiment 2, we administered mCPP into amygdala subregions and the dorsal hippocampus to investigate site specificity. In Experiment 4 we lesioned the BNST and subsequently infused mCPP systemically, and in Experiment 4 we used RNAscope® to assess BNST 5-HT2C transcripts following wheel running. Results: BNST mCPP infusion increased acoustic startle responding, which was by 5-HT2C antagonism, while neither mCPP infused into the amygdala nor hippocampus was anxiogenic. Lesions of the BNST prevented the anxiogenic effect of systemically administered mCPP. Lastly, exercise reduced 5-HT2C transcripts in the BNST. Discussion: These results suggest that the BNST is a critical site of action for the effects of exercise on mCPP. Together these data suggest that exercise may reduce 5-HT2C receptor function in the BNST, which may, in part, explain some of the anxiolytic effects associated with wheel running.

4.
J Neuroinflammation ; 17(1): 157, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32410624

ABSTRACT

BACKGROUND: Fractalkine (CX3CL1; FKN) is a chemokine expressed by neurons that mediates communication between neurons and microglia. By regulating microglial activity, CX3CL1 can mitigate the damaging effects of chronic microglial inflammation within the brain, a state that plays a major role in aging and neurodegeneration. CX3CL1 is present in two forms, a full-length membrane-bound form and a soluble cleaved form (sFKN), generated by a disintegrin and metalloproteinase (ADAM) 10 or 17. Levels of sFKN decrease with aging, which could lead to enhanced inflammation, deficits in synaptic remodeling, and subsequent declines in cognition. Recently, the idea that these two forms of CX3CL1 may display differential activities within the CNS has garnered increased attention, but remains unresolved. METHODS: Here, we assessed the consequences of CX3CL1 knockout (CX3CL1-/-) on cognitive behavior as well as the functional rescue with the two different forms of CX3CL1 in mice. CX3CL1-/- mice were treated with adeno-associated virus (AAV) expressing either green fluorescent protein (GFP), sFKN, or an obligate membrane-bound form of CX3CL1 (mFKN) and then subjected to behavioral testing to assess cognition and motor function. Following behavioral analysis, brains were collected and analyzed for markers of neurogenesis, or prepared for electrophysiology to measure long-term potentiation (LTP) in hippocampal slices. RESULTS: CX3CL1-/- mice showed significant deficits in cognitive tasks for long-term memory and spatial learning and memory in addition to demonstrating enhanced basal motor performance. These alterations correlated with deficits in both hippocampal neurogenesis and LTP. Treatment of CX3CL1-/- mice with AAV-sFKN partially corrected changes in both cognitive and motor function and restored neurogenesis and LTP to levels similar to wild-type animals. Treatment with AAV-mFKN partially restored spatial learning and memory in CX3CL1-/- mice, but did not rescue long-term memory, or neurogenesis. CONCLUSIONS: These results are the first to demonstrate that CX3CL1 knockout causes significant cognitive deficits that can be rescued by treatment with sFKN and only partially rescued with mFKN. This suggests that treatments that restore signaling of soluble forms of CX3CL1 may be a viable therapeutic option for aging and disease.


Subject(s)
Brain/metabolism , Chemokine CX3CL1/metabolism , Cognitive Dysfunction/metabolism , Animals , Mice , Mice, Knockout , Neurogenesis/physiology , Protein Isoforms
5.
Geroscience ; 41(1): 77-87, 2019 02.
Article in English | MEDLINE | ID: mdl-30739297

ABSTRACT

The incidence of neurodegenerative disorders and cognitive impairment is increasing. Rising prevalence of age-related medical conditions is associated with a dramatic economic burden; therefore, developing strategies to manage these health concerns is of great public health interest. Nutritionally based interventions have shown promise in treatment of these age-associated conditions. Astaxanthin is a carotenoid with reputed neuroprotective properties in the context of disease and injury, while emerging evidence suggests that astaxanthin may also have additional biological activities relating to neurogenesis and synaptic plasticity. Here, we investigate the potential for astaxanthin to modulate cognitive function and neural plasticity in young and aged mice. We show that feeding astaxanthin to aged mice for 1 month improves performance on several hippocampal-dependent cognitive tasks and increases long-term potentiation. However, we did not observe an alteration in neurogenesis, nor did we observe a change in microglial-associated IBA1 immunostaining. This demonstrates the potential for astaxanthin to modulate neural plasticity and cognitive function in aging.


Subject(s)
Aging/drug effects , Cognition/drug effects , Dietary Supplements , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Aging/pathology , Animals , Behavior, Animal/drug effects , Cognitive Dysfunction/diet therapy , Hippocampus/drug effects , Hippocampus/physiology , Inflammation/diet therapy , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/physiology , Neurodegenerative Diseases/diet therapy , Neurogenesis/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Xanthophylls/administration & dosage , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
6.
Geroscience ; 39(1): 19-32, 2017 02.
Article in English | MEDLINE | ID: mdl-28299644

ABSTRACT

Astaxanthin (AXT) is a carotenoid with multiple health benefits. It is currently marketed as a health supplement and is well known for its antioxidant capacity. Recent evidence has emerged to suggest a broad range of biological activities. The interest in this compound has increased dramatically over the last few years and many studies are now applying this molecule across many disease models. Results from the current research are beginning to come together to suggest neuroprotective properties including anti-inflammatory, anti-apoptotic, and antioxidant effects, as well as the potential to promote or maintain neural plasticity. These emergent mechanisms of actions implicate AXT as a promising therapeutic agent for neurodegenerative disease. This review will examine and extrapolate from the recent literature to build support for the use of AXT in mitigating neuropathy in normal aging and neurodegenerative disease.


Subject(s)
Aging/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotection/drug effects , Neuroprotective Agents/administration & dosage , Aging/physiology , Animals , Dietary Supplements , Humans , Neurodegenerative Diseases/prevention & control , Prognosis , Risk Assessment , Treatment Outcome , Xanthophylls/therapeutic use
7.
Exp Gerontol ; 94: 4-8, 2017 08.
Article in English | MEDLINE | ID: mdl-28163132

ABSTRACT

Aging is the primary risk factor for many neurodegenerative diseases. Thus, understanding the basic biological changes that take place with aging that lead to the brain being less resilient to disease progression of neurodegenerative diseases such as Parkinson's disease or Alzheimer's disease or insults to the brain such as stroke or traumatic brain injuries. Clearly this will not cure the disease per se, yet increasing the ability of the brain to respond to injury could improve long term outcomes. The focus of this review is examining changes in microglia with age and possible therapeutic interventions involving the use of polyphenol rich dietary supplements.


Subject(s)
Aging/metabolism , Brain/metabolism , Cellular Senescence , Microglia/metabolism , Nerve Degeneration , Neurodegenerative Diseases/etiology , Age Factors , Aging/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cellular Senescence/drug effects , Dietary Supplements , Humans , Inflammation Mediators/metabolism , Microglia/drug effects , Microglia/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuronal Plasticity , Neuroprotective Agents/therapeutic use , Polyphenols/therapeutic use , Stem Cell Niche
8.
Brain Sci ; 6(4)2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27669315

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder that primarily afflicts the elderly. It is characterized by motor dysfunction due to extensive neuron loss in the substantia nigra pars compacta. There are multiple biological processes that are negatively impacted during the pathogenesis of PD, and are implicated in the cell death in this region. Neuroinflammation is evidently involved in PD pathology and mitigating the inflammatory cascade has been a therapeutic strategy. Age is the number one risk factor for PD and thus needs to be considered in the context of disease pathology. Here, we discuss the role of neuroinflammation within the context of aging as it applies to the development of PD, and the potential for two representative compounds, fractalkine and astaxanthin, to attenuate the pathophysiology that modulates neurodegeneration that occurs in Parkinson's disease.

10.
Cell Transplant ; 25(11): 1967-1977, 2016 11.
Article in English | MEDLINE | ID: mdl-28836850

ABSTRACT

The prevalence of HIV-associated neurocognitive disorders (HAND) remains high despite combination antiretroviral therapy (cART). There is evidence that neural stem cells (NSCs) can migrate to sites of brain injury such as those caused by inflammation and oxidative stress, which are pathological features of HAND. Thus, reductions in NSCs may contribute to HAND pathogenesis. Since the HIV non-nucleoside reverse transcriptase inhibitor efavirenz (EFV) has previously been associated with cognitive deficits and promotion of oxidative stress pathways, we examined its effect on NSCs in vitro as well as in C57BL/6J mice. Here we report that EFV induced a decrease in NSC proliferation in vitro as indicated by MTT assay, as well as BrdU and nestin immunocytochemistry. In addition, EFV decreased intracellular NSC adenosine triphosphate (ATP) stores and NSC mitochondrial membrane potential (MMP). Further, we found that EFV promoted increased lactate dehydrogenase (LDH) release, activation of p38 mitogen-activated protein kinase (MAPK), and increased Bax expression in cultured NSCs. Moreover, EFV reduced the quantity of proliferating NSCs in the subventricular zone (SVZ) of C57BL/6J mice as suggested by BrdU, and increased apoptosis as measured by active caspase-3 immunohistochemistry. If these in vitro and in vivo models translate to the clinical syndrome, then a pharmacological or cell-based therapy aimed at opposing EFV-mediated reductions in NSC proliferation may be beneficial to prevent or treat HAND in patients receiving EFV.


Subject(s)
Benzoxazines/toxicity , Cell Proliferation/drug effects , Reverse Transcriptase Inhibitors/toxicity , Adenosine Triphosphate/metabolism , Alkynes , Animals , Apoptosis/drug effects , Benzoxazines/administration & dosage , Caspase 3/metabolism , Cells, Cultured , Cyclopropanes , Female , Immunohistochemistry , Injections, Intraperitoneal , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Phosphorylation/drug effects , Rats , Reverse Transcriptase Inhibitors/administration & dosage , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Age (Dordr) ; 37(5): 103, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26410618

ABSTRACT

Aging is associated with a decline in function in many of the stem cell niches of the body. An emerging body of literature suggests that one of the reasons for this decline in function is due to cell non-autonomous influences on the niche from the body. For example, studies using the technique of parabiosis have demonstrated a negative influence of blood from aged mice on muscle satellite cells and neurogenesis in young mice. We examined if we could reverse this effect of aged serum on stem cell proliferation by treating aged rats with NT-020, a dietary supplement containing blueberry, green tea, vitamin D3, and carnosine that has been shown to increase neurogenesis in aged rats. Young and aged rats were administered either control NIH-31 diet or one supplemented with NT-020 for 28 days, and serum was collected upon euthanasia. The serum was used in cultures of both rat hippocampal neural progenitor cells (NPCs) and rat bone marrow-derived mesenchymal stem cells (MSCs). Serum from aged rats significantly reduced cell proliferation as measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-bromo-2'-deoxyuridine (BrdU) assays in both NPCs and MSCs. Serum from aged rats treated with NT-020 was not different from serum from young rats. Therefore, NT-020 rescued the effect of serum from aged rats to reduce stem cell proliferation.


Subject(s)
Aging/blood , Aging/drug effects , Dietary Supplements , Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis/drug effects , Animals , Cell Proliferation/drug effects , Male , Mesenchymal Stem Cells/drug effects , Mice , Neural Stem Cells/drug effects , Rats , Rats, Inbred F344
12.
PLoS One ; 10(2): e0116841, 2015.
Article in English | MEDLINE | ID: mdl-25658425

ABSTRACT

The protein α-synuclein (α-Syn) has a central role in the pathogenesis of Parkinson's disease (PD) and immunotherapeutic approaches targeting this molecule have shown promising results. In this study, novel antibodies were generated against specific peptides from full length human α-Syn and evaluated for effectiveness in ameliorating α-Syn-induced cell death and behavioral deficits in an AAV-α-Syn expressing rat model of PD. Fisher 344 rats were injected with rAAV vector into the right substantia nigra (SN), while control rats received an AAV vector expressing green fluorescent protein (GFP). Beginning one week after injection of the AAV-α-Syn vectors, rats were treated intraperitoneally with either control IgG or antibodies against the N-terminal (AB1), or central region (AB2) of α-Syn. An unbiased stereological estimation of TH+, NeuN+, and OX6 (MHC-II) immunostaining revealed that the α-Syn peptide antibodies (AB1 and AB2) significantly inhibited α-Syn-induced dopaminergic cell (DA) and NeuN+ cell loss (one-way ANOVA (F (3, 30) = 5.8, p = 0.002 and (F (3, 29) = 7.92, p = 0.002 respectively), as well as decreasing the number of activated microglia in the ipsilateral SN (one-way ANOVA F = 14.09; p = 0.0003). Antibody treated animals also had lower levels of α-Syn in the ipsilateral SN (one-way ANOVA F (7, 37) = 9.786; p = 0.0001) and demonstrated a partial intermediate improvement of the behavioral deficits. Our data suggest that, in particular, an α-Syn peptide antibody against the N-terminal region of the protein can protect against DA neuron loss and, to some extent behavioral deficits. As such, these results may be a potential therapeutic strategy for halting the progression of PD.


Subject(s)
Antibodies/immunology , Cell Death/physiology , Dopaminergic Neurons/physiology , Immunotherapy/methods , Parkinson Disease/drug therapy , Parkinson Disease/immunology , alpha-Synuclein/immunology , Analysis of Variance , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Behavior, Animal/physiology , Cell Death/drug effects , Dependovirus , Dopaminergic Neurons/drug effects , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins , Humans , Immunohistochemistry , Injections, Intraperitoneal , Rats , Substantia Nigra/metabolism , alpha-Synuclein/adverse effects
13.
PLoS One ; 8(1): e53376, 2013.
Article in English | MEDLINE | ID: mdl-23301065

ABSTRACT

The long-term consequences of traumatic brain injury (TBI), specifically the detrimental effects of inflammation on the neurogenic niches, are not very well understood. In the present in vivo study, we examined the prolonged pathological outcomes of experimental TBI in different parts of the rat brain with special emphasis on inflammation and neurogenesis. Sixty days after moderate controlled cortical impact injury, adult Sprague-Dawley male rats were euthanized and brain tissues harvested. Antibodies against the activated microglial marker, OX6, the cell cycle-regulating protein marker, Ki67, and the immature neuronal marker, doublecortin, DCX, were used to estimate microglial activation, cell proliferation, and neuronal differentiation, respectively, in the subventricular zone (SVZ), subgranular zone (SGZ), striatum, thalamus, and cerebral peduncle. Stereology-based analyses revealed significant exacerbation of OX6-positive activated microglial cells in the striatum, thalamus, and cerebral peduncle. In parallel, significant decrements in Ki67-positive proliferating cells in SVZ and SGZ, but only trends of reduced DCX-positive immature neuronal cells in SVZ and SGZ were detected relative to sham control group. These results indicate a progressive deterioration of the TBI brain over time characterized by elevated inflammation and suppressed neurogenesis. Therapeutic intervention at the chronic stage of TBI may confer abrogation of these deleterious cell death processes.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Gene Expression Regulation , Inflammation/metabolism , Up-Regulation , Animals , Brain Mapping/methods , Cell Death , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Doublecortin Protein , Genes, MHC Class II , Immunohistochemistry , Male , Microglia/metabolism , Neurogenesis , Rats , Rats, Sprague-Dawley
14.
J Neurosci ; 32(42): 14592-601, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23077045

ABSTRACT

The chemokine CX3CL1/fractalkine is expressed by neurons as a transmembrane-anchored protein that can be cleaved to yield a soluble isoform. However, the roles for these two types of endogenous CX3CL1 in neurodegenerative pathophysiology remain elusive. As such, it has been difficult to delineate the function of the two isoforms of CX3CL1, as both are natively present in the brain. In this study we examined each isoform's ability to regulate neuroinflammation in a mouse model of Parkinson's disease initiated by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We were able to delineate the function of both CX3CL1 isoforms by using adeno-associated virus-mediated gene therapy to selectively express synthetic variants of CX3CL1 that remain either permanently soluble or membrane bound. In the present study we injected each CX3CL1 variant or a GFP-expressing vector directly into the substantia nigra of CX3CL1(-/-) mice. Our results show that only the soluble isoform of CX3CL1 is sufficient for neuroprotection after exposure to MPTP. Specifically, we show that the soluble CX3CL1 isoform reduces impairment of motor coordination, decreases dopaminergic neuron loss, and ameliorates microglial activation and proinflammatory cytokine release resulting from MPTP exposure. Furthermore, we show that the membrane-bound isoform provides no neuroprotective capability to MPTP-induced pathologies, exhibiting similar motor coordination impairment, dopaminergic neuron loss, and inflammatory phenotypes as MPTP-treated CX3CL1(-/-) mice, which received the GFP-expressing control vector. Our results reveal that the neuroprotective capacity of CX3CL1 resides solely upon the soluble isoform in an MPTP-induced model of Parkinson's disease.


Subject(s)
Chemokine CX3CL1/physiology , Disease Models, Animal , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Chemokine CX3CL1/deficiency , Chemokine CX3CL1/therapeutic use , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/therapeutic use , Parkinson Disease/etiology , Protein Isoforms/biosynthesis , Protein Isoforms/deficiency , Protein Isoforms/therapeutic use , Random Allocation , Solubility
15.
J Neurosci ; 31(45): 16241-50, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22072675

ABSTRACT

The protective/neurotoxic role of fractalkine (CX3CL1) and its receptor CX3C chemokine receptor 1 (CX3CR1) signaling in neurodegenerative disease is an intricate and highly debated research topic and it is becoming even more complicated as new studies reveal discordant results. It appears that the CX3CL1/CX3CR1 axis plays a direct role in neurodegeneration and/or neuroprotection depending on the CNS insult. However, all the above studies focused on the role of CX3CL1/CX3CR1 signaling in pathological conditions, ignoring the relevance of CX3CL1/CX3CR1 signaling under physiological conditions. No approach to date has been taken to decipher the significance of defects in CX3CL1/CX3CR1 signaling in physiological condition. In the present study we used CX3CR1⁻/⁻, CX3CR1⁺/⁻, and wild-type mice to investigate the physiological role of CX3CR1 receptor in cognition and synaptic plasticity. Our results demonstrate for the first time that mice lacking the CX3CR1 receptor show contextual fear conditioning and Morris water maze deficits. CX3CR1 deficiency also affects motor learning. Importantly, mice lacking the receptor have a significant impairment in long-term potentiation (LTP). Infusion with IL-1ß receptor antagonist significantly reversed the deficit in cognitive function and impairment in LTP. Our results reveal that under physiological conditions, disruption in CX3CL1 signaling will lead to impairment in cognitive function and synaptic plasticity via increased action of IL-1ß.


Subject(s)
Cognition Disorders/pathology , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation/genetics , Receptors, Interleukin-8A/deficiency , Analysis of Variance , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Biophysics , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebellum/metabolism , Cognition Disorders/genetics , Conditioning, Psychological/physiology , Cytokines/metabolism , Disease Models, Animal , Electric Stimulation , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/physiology , Fear/physiology , Gene Expression Regulation/genetics , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein/pharmacology , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/pathology , Motor Activity/genetics , Neurogenesis/genetics , Patch-Clamp Techniques , Rotarod Performance Test
16.
J Mol Neurosci ; 42(3): 327-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20405238

ABSTRACT

Anxiety disorders are frequently long-lasting and debilitating for more than 40 million American adults. Although stressor exposure plays an important role in the etiology of some anxiety disorders, the mechanisms by which exposure to stressful stimuli alters central circuits that mediate anxiety-like emotional behavior are still unknown. Substantial evidence has implicated regions of the central extended amygdala, including the bed nucleus of the stria terminalis (BNST) and the central nucleus of the amygdala as critical structures mediating fear- and anxiety-like behavior in both humans and animals. These areas organize coordinated fear- and anxiety-like behavioral responses as well as peripheral stress responding to threats via direct and indirect projections to the paraventricular nucleus of the hypothalamus and brainstem regions (Walker et al. Eur J Pharmacol 463:199-216, 2003, Prog Neuropsychopharmacol Biol Psychiatry 33(8):1291-1308, 2009; Ulrich-Lai and Herman Nat Rev Neurosci 10:397-409, 2009). In particular, the BNST has been argued to mediate these central and peripheral responses when the perceived threat is of long duration (Waddell et al. Behav Neurosci 120:324-336, 2006) and/or when the anxiety-like response is sustained (Walker and Davis Brain Struct Funct 213:29-42, 2008); hence, the BNST may mediate pathological anxiety-like states that result from exposure to chronic stress. Indeed, chronic stress paradigms result in enhanced BNST neuroplasticity that has been associated with pathological anxiety-like states (Vyas et al. Brain Res 965:290-294, 2003; Pego et al. Eur J Neurosci 27:1503-1516, 2008). Here we review evidence that suggests that pituitary adenylate cyclase-activating polypeptide (PACAP) and corticotropin-releasing hormone (CRH) work together to modulate BNST function and increase anxiety-like behavior. Moreover, we have shown that BNST PACAP as well as its cognate PAC1 receptor is substantially upregulated following chronic stress, particularly in the BNST oval nucleus where PACAP-containing neurons closely interact with CRH-containing neurons (Kozicz et al. Brain Res 767:109-119, 1997; Hammack et al. Psychoneuroendocrinology 34:833-843, 2009). We describe how interactions between PACAP and CRH in the BNST may mediate stress-associated behaviors, including anorexia and anxiety-like behavior. These studies have the potential to define specific mechanisms underlying anxiety disorders, and may provide important therapeutic strategies for stress and anxiety management.


Subject(s)
Behavior, Animal/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Septal Nuclei/physiology , Signal Transduction/physiology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Animals , Anxiety/metabolism , Anxiety/physiopathology , Corticotropin-Releasing Hormone/metabolism , Humans , Neuronal Plasticity/physiology , Neuropeptides/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...