Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Mol Psychiatry ; 28(7): 3002-3012, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37131071

ABSTRACT

Chronic stress constitutes a major risk factor for depression that can disrupt various aspects of homeostasis, including the gut microbiome (GM). We have recently shown that GM imbalance affects adult hippocampal (HPC) neurogenesis and induces depression-like behaviors, with the exact mechanisms being under active investigation. Here we hypothesized that the vagus nerve (VN), a key bidirectional route of communication between the gut and the brain, could relay the effects of stress-induced GM changes on HPC plasticity and behavior. We used fecal samples derived from mice that sustained unpredictable chronic mild stress (UCMS) to inoculate healthy mice and assess standard behavioral readouts for anxiety- and depressive-like behavior, conduct histological and molecular analyses for adult HPC neurogenesis and evaluate neurotransmission pathways and neuroinflammation. To study the potential role of the VN in mediating the effects of GM changes on brain functions and behavior, we used mice that sustained subdiaphragmatic vagotomy (Vx) prior the GM transfer. We found that inoculation of healthy mice with GM from UCMS mice activates the VN and induces early and sustained changes in both serotonin and dopamine neurotransmission pathways in the brainstem and HPC. These changes are associated with prompt and persistent deficits in adult HPC neurogenesis and induce early and sustained neuroinflammatory responses in the HPC. Remarkably, Vx abrogates adult HPC neurogenesis deficits, neuroinflammation and depressive-like behavior, suggesting that vagal afferent pathways are necessary to drive GM-mediated effects on the brain.


Subject(s)
Gastrointestinal Microbiome , Mice , Animals , Gastrointestinal Microbiome/physiology , Neuroinflammatory Diseases , Brain/metabolism , Vagus Nerve/physiology , Depression/metabolism , Stress, Psychological
2.
Int J Mol Sci ; 24(6)2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36982451

ABSTRACT

Cajal-Retzius cells (CRs) are a class of transient neurons in the mammalian cortex that play a critical role in cortical development. Neocortical CRs undergo almost complete elimination in the first two postnatal weeks in rodents and the persistence of CRs during postnatal life has been detected in pathological conditions related to epilepsy. However, it is unclear whether their persistence is a cause or consequence of these diseases. To decipher the molecular mechanisms involved in CR death, we investigated the contribution of the PI3K/AKT/mTOR pathway as it plays a critical role in cell survival. We first showed that this pathway is less active in CRs after birth before massive cell death. We also explored the spatio-temporal activation of both AKT and mTOR pathways and reveal area-specific differences along both the rostro-caudal and medio-lateral axes. Next, using genetic approaches to maintain an active pathway in CRs, we found that the removal of either PTEN or TSC1, two negative regulators of the pathway, lead to differential CR survivals, with a stronger effect in the Pten model. Persistent cells in this latter mutant are still active. They express more Reelin and their persistence is associated with an increase in the duration of kainate-induced seizures in females. Altogether, we show that the decrease in PI3K/AKT/mTOR activity in CRs primes these cells to death by possibly repressing a survival pathway, with the mTORC1 branch contributing less to the phenotype.


Subject(s)
Kainic Acid , Proto-Oncogene Proteins c-akt , Animals , Female , Kainic Acid/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Seizures/chemically induced , Mammals/metabolism
3.
Cell Death Dis ; 15(1): 20, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195526

ABSTRACT

In recent years, primary familial brain calcification (PFBC), a rare neurological disease characterized by a wide spectrum of cognitive disorders, has been associated to mutations in the sodium (Na)-Phosphate (Pi) co-transporter SLC20A2. However, the functional roles of the Na-Pi co-transporters in the brain remain still largely elusive. Here we show that Slc20a1 (PiT-1) and Slc20a2 (PiT-2) are the most abundant Na-Pi co-transporters expressed in the brain and are involved in the control of hippocampal-dependent learning and memory. We reveal that Slc20a1 and Slc20a2 are differentially distributed in the hippocampus and associated with independent gene clusters, suggesting that they influence cognition by different mechanisms. Accordingly, using a combination of molecular, electrophysiological and behavioral analyses, we show that while PiT-2 favors hippocampal neuronal branching and survival, PiT-1 promotes synaptic plasticity. The latter relies on a likely Otoferlin-dependent regulation of synaptic vesicle trafficking, which impacts the GABAergic system. These results provide the first demonstration that Na-Pi co-transporters play key albeit distinct roles in the hippocampus pertaining to the control of neuronal plasticity and cognition. These findings could provide the foundation for the development of novel effective therapies for PFBC and cognitive disorders.


Subject(s)
Cognition , Symporters , Ion Transport , Neuronal Plasticity/genetics , Phosphates
4.
Nat Commun ; 14(1): 1531, 2023 03 18.
Article in English | MEDLINE | ID: mdl-36934089

ABSTRACT

Cajal-Retzius cells (CRs) are transient neurons, disappearing almost completely in the postnatal neocortex by programmed cell death (PCD), with a percentage surviving up to adulthood in the hippocampus. Here, we evaluate CR's role in the establishment of adult neuronal and cognitive function using a mouse model preventing Bax-dependent PCD. CRs abnormal survival resulted in impairment of hippocampus-dependent memory, associated in vivo with attenuated theta oscillations and enhanced gamma activity in the dorsal CA1. At the cellular level, we observed transient changes in the number of NPY+ cells and altered CA1 pyramidal cell spine density. At the synaptic level, these changes translated into enhanced inhibitory currents in hippocampal pyramidal cells. Finally, adult mutants displayed an increased susceptibility to lethal tonic-clonic seizures in a kainate model of epilepsy. Our data reveal that aberrant survival of a small proportion of postnatal hippocampal CRs results in cognitive deficits and epilepsy-prone phenotypes in adulthood.


Subject(s)
Hippocampus , Neurons , Hippocampus/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Neurons/metabolism , Pyramidal Cells/physiology , Seizures/genetics , Seizures/metabolism , Animals , Mice
5.
J Exp Med ; 219(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35254402

ABSTRACT

Crouzon syndrome with acanthosis nigricans (CAN, a rare type of craniosynostosis characterized by premature suture fusion and neurological impairments) has been linked to a gain-of-function mutation (p.Ala391Glu) in fibroblast growth factor receptor 3 (FGFR3). To characterize the CAN mutation's impact on the skull and on brain functions, we developed the first mouse model (Fgfr3A385E/+) of this syndrome. Surprisingly, Fgfr3A385E/+ mice did not exhibit craniosynostosis but did show severe memory impairments, a structurally abnormal hippocampus, low activity-dependent synaptic plasticity, and overactivation of MAPK/ERK and Akt signaling pathways in the hippocampus. Systemic or brain-specific pharmacological inhibition of FGFR3 overactivation by BGJ398 injections rescued the memory impairments observed in Fgfr3A385E/+ mice. The present study is the first to have demonstrated cognitive impairments associated with brain FGFR3 overactivation, independently of skull abnormalities. Our results provide a better understanding of FGFR3's functional role and the impact of its gain-of-function mutation on brain functions. The modulation of FGFR3 signaling might be of value for treating the neurological disorders associated with craniosynostosis.


Subject(s)
Acanthosis Nigricans , Craniofacial Dysostosis , Craniosynostoses , Acanthosis Nigricans/complications , Acanthosis Nigricans/genetics , Animals , Brain , Craniofacial Dysostosis/complications , Craniofacial Dysostosis/genetics , Craniosynostoses/genetics , Disease Models, Animal , Memory Disorders/genetics , Mice , Receptor, Fibroblast Growth Factor, Type 3/genetics
6.
Sci Rep ; 12(1): 6132, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35413967

ABSTRACT

Mutations in the Cystic Fibrosis Transmembrane Conductance Regulator gene (CFTR) are responsible for Cystic Fibrosis (CF). The most common CF-causing mutation is the deletion of the 508th amino-acid of CFTR (F508del), leading to dysregulation of the epithelial fluid transport in the airway's epithelium and the production of a thickened mucus favoring chronic bacterial colonization, sustained inflammation and ultimately respiratory failure. c407 is a bis-phosphinic acid derivative which corrects CFTR dysfunction in epithelial cells carrying the F508del mutation. This study aimed to investigate c407 in vivo activity in the F508del Cftrtm1Eur murine model of CF. Using nasal potential difference measurement, we showed that in vivo administration of c407 by topical, short-term intraperitoneal and long-term subcutaneous route significantly increased the CFTR dependent chloride (Cl-) conductance in F508del Cftrtm1Eur mice. This functional improvement was correlated with a relocalization of F508del-cftr to the apical membrane in nasal epithelial cells. Importantly, c407 long-term administration was well tolerated and in vitro ADME toxicologic studies did not evidence any obvious issue. Our data provide the first in vivo preclinical evidence of c407 efficacy and absence of toxicity after systemic administration for the treatment of Cystic Fibrosis.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Animals , Chlorides , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ion Transport , Mice , Mutation , Phosphinic Acids
7.
J Neurosci ; 29(50): 15745-55, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016090

ABSTRACT

Infant rats require maternal odor learning to guide pups' proximity-seeking of the mother and nursing. Maternal odor learning occurs using a simple learning circuit including robust olfactory bulb norepinephrine (NE), release from the locus ceruleus (LC), and amygdala suppression by low corticosterone (CORT). Early-life stress increases NE but also CORT, and we questioned whether early-life stress disrupted attachment learning and its neural correlates [2-deoxyglucose (2-DG) autoradiography]. Neonatal rats were normally reared or stressed-reared during the first 6 d of life by providing the mother with insufficient bedding for nest building and were odor-0.5 mA shock conditioned at 7 d old. Normally reared paired pups exhibited typical odor approach learning and associated olfactory bulb enhanced 2-DG uptake. However, stressed-reared pups showed odor avoidance learning and both olfactory bulb and amygdala 2-DG uptake enhancement. Furthermore, stressed-reared pups had elevated CORT levels, and systemic CORT antagonist injection reestablished the age-appropriate odor-preference learning, enhanced olfactory bulb, and attenuated amygdala 2-DG. We also assessed the neural mechanism for stressed-reared pups' abnormal behavior in a more controlled environment by injecting normally reared pups with CORT. This was sufficient to produce odor aversion, as well as dual amygdala and olfactory bulb enhanced 2-DG uptake. Moreover, we assessed a unique cascade of neural events for the aberrant effects of stress rearing: the amygdala-LC-olfactory bulb pathway. Intra-amygdala CORT or intra-LC corticotropin releasing hormone (CRH) infusion supported aversion learning with intra-LC CRH infusion associated with increased olfactory bulb NE (microdialysis). These results suggest that early-life stress disturbs attachment behavior via a unique cascade of events (amygdala-LC-olfactory bulb).


Subject(s)
Amygdala/physiology , Corticosterone/physiology , Corticotropin-Releasing Hormone/physiology , Locus Coeruleus/physiology , Norepinephrine/physiology , Olfactory Bulb/physiology , Stress, Psychological/metabolism , Age Factors , Animals , Animals, Newborn , Association Learning/physiology , Female , Male , Maternal Behavior/physiology , Maternal Behavior/psychology , Rats , Rats, Long-Evans , Stress, Psychological/psychology
8.
Dev Psychobiol ; 52(7): 651-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20730787

ABSTRACT

Here we review the neurobiology of infant odor learning in rats, and discuss the unique role of the stress hormone corticosterone (CORT) in the learning necessary for the developing rat. During the first 9 postnatal (PN) days, infants readily learn odor preferences, while aversion and fear learning are attenuated. Such restricted learning may ensure that pups only approach their mother. This sensitive period of preference learning overlaps with the stress hyporesponsive period (SHRP, PN4-14) when pups have a reduced CORT response to most stressors. Neural underpinnings responsible for sensitive-period learning include increased activity within the olfactory bulb and piriform "olfactory" cortex due to heightened release of norepinephrine from the locus coeruleus. After PN10 and with the decline of the SHRP, stress-induced CORT release permits amygdala activation and facilitates learned odor aversions and fear. Remarkably, odor preference and attenuated fear learning can be reestablished in PN10-15 pups if the mother is present, an effect due to her ability to suppress pups' CORT and amygdala activity. Together, these data indicate that functional changes in infant learning are modified by a unique interaction between the developing CORT system, the amygdala, and maternal presence, providing a learning system that becomes more flexible as pups mature.


Subject(s)
Aging/psychology , Amygdala/growth & development , Association Learning , Fear/psychology , Object Attachment , Odorants , Stress, Psychological/psychology , Aging/metabolism , Amygdala/metabolism , Animals , Animals, Newborn , Corticosterone/metabolism , Locus Coeruleus/growth & development , Locus Coeruleus/metabolism , Maternal Behavior/psychology , Motor Activity , Norepinephrine/metabolism , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Rats , Stress, Psychological/metabolism
9.
Nat Neurosci ; 9(8): 1004-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16829957

ABSTRACT

Odor-shock conditioning produces either olfactory preference or aversion in preweanling (12-15 days old) rats, depending on the context. In the mother's absence, odor-shock conditioning produces amygdala activation and learned odor avoidance. With maternal presence, this same conditioning yields an odor preference without amygdala activation. Maternal presence acts through modulation of pup corticosterone and corticosterone's regulation of amygdala activity. Over-riding maternal suppression of corticosterone through intra-amygdala corticosterone infusions permits fear conditioning and amygdala activation.


Subject(s)
Association Learning/physiology , Avoidance Learning/physiology , Fear , Learning , Mothers , Odorants , Amygdala/physiology , Animals , Conditioning, Classical , Corticosterone/metabolism , Rats
10.
Mol Autism ; 11(1): 22, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32228681

ABSTRACT

BACKGROUND: Formation and maintenance of appropriate neural networks require tight regulation of neural stem cell proliferation, differentiation, and neurogenesis. microRNAs (miRNAs) play an important role in brain development and plasticity, and dysregulated miRNA profiles have been linked to neurodevelopmental disorders including autism, schizophrenia, or intellectual disability. Yet, the functional role of miRNAs in neural development and postnatal brain functions remains unclear. METHODS: Using a combination of cell biology techniques as well as behavioral studies and brain imaging, we characterize mouse models with either constitutive inactivation or selectively hippocampal knockdown of the neurodevelopmental disease-associated gene Mir146a, the most commonly deregulated miRNA in developmental brain disorders (DBD). RESULTS: We first show that during development, loss of miR-146a impairs the differentiation of radial glial cells, neurogenesis process, and neurite extension. In the mouse adult brain, loss of miR-146a correlates with an increased hippocampal asymmetry coupled with defects in spatial learning and memory performances. Moreover, selective hippocampal downregulation of miR-146a in adult mice causes severe hippocampal-dependent memory impairments indicating for the first time a role for this miRNA in postnatal brain functions. CONCLUSION: Our results show that miR-146a expression is critical for correct differentiation of neural stem cell during brain development and provide for the first time a strong argument for a postnatal role of miR-146a in regulating hippocampal-dependent memory. Furthermore, the demonstration that the Mir146a-/- mouse recapitulates several aspects reported in DBD patients, including impaired neurogenesis, abnormal brain anatomy, and working and spatial memories deficits, provides convincing evidence that the dysregulation of miR146a contributes to the pathogenesis of DBDs.


Subject(s)
Learning , Memory Disorders/genetics , MicroRNAs , Neural Stem Cells/cytology , Neurodevelopmental Disorders/genetics , Animals , Cells, Cultured , Down-Regulation , Hippocampus/metabolism , Mice, Transgenic , Neurogenesis
11.
Aging Cell ; 19(9): e13189, 2020 09.
Article in English | MEDLINE | ID: mdl-32729663

ABSTRACT

Autophagy agonists have been proposed to slow down neurodegeneration. Spermidine, a polyamine that acts as an autophagy agonist, is currently under clinical trial for the treatment of age-related memory decline. How Spermidine and other autophagy agonists regulate memory and synaptic plasticity is under investigation. We set up a novel mouse model of mild cognitive impairment (MCI), in which middle-aged (12-month-old) mice exhibit impaired memory capacity, lysosomes engulfed with amyloid fibrils (ß-amyloid and α-synuclein) and impaired task-induced GluA1 hippocampal post-translation modifications. Subchronic treatment with Spermidine as well as the autophagy agonist TAT-Beclin 1 rescued memory capacity and GluA1 post-translational modifications by favouring the autophagy/lysosomal-mediated degradation of amyloid fibrils. These findings provide new mechanistic evidence on the therapeutic relevance of autophagy enhancers which, by improving the degradation of misfolded proteins, slow down age-related memory decline.


Subject(s)
Autophagy/genetics , Cognitive Dysfunction/genetics , Memory/drug effects , Aging , Animals , Disease Models, Animal , Mice
12.
Cell Death Dis ; 11(7): 502, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632162

ABSTRACT

Acyl coenzyme A binding protein (ACBP), also known as diazepam binding inhibitor (DBI) is a multifunctional protein with an intracellular action (as ACBP), as well as with an extracellular role (as DBI). The plasma levels of soluble ACBP/DBI are elevated in human obesity and reduced in anorexia nervosa. Accumulating evidence indicates that genetic or antibody-mediated neutralization of ACBP/DBI has anorexigenic effects, thus inhibiting food intake and inducing lipo-catabolic reactions in mice. A number of anorexiants have been withdrawn from clinical development because of their side effects including an increase in depression and suicide. For this reason, we investigated the psychiatric impact of ACBP/DBI in mouse models and patient cohorts. Intravenously (i.v.) injected ACBP/DBI protein conserved its orexigenic function when the protein was mutated to abolish acyl coenzyme A binding, but lost its appetite-stimulatory effect in mice bearing a mutation in the γ2 subunit of the γ-aminobutyric acid (GABA) A receptor (GABAAR). ACBP/DBI neutralization by intraperitoneal (i.p.) injection of a specific mAb blunted excessive food intake in starved and leptin-deficient mice, but not in ghrelin-treated animals. Neither i.v. nor i.p. injected anti-ACBP/DBI antibody affected the behavior of mice in the dark-light box and open-field test. In contrast, ACBP/DBI increased immobility in the forced swim test, while anti-ACBP/DBI antibody counteracted this sign of depression. In patients diagnosed with therapy-resistant bipolar disorder or schizophrenia, ACBP/DBI similarly correlated with body mass index (BMI), not with the psychiatric diagnosis. Patients with high levels of ACBP/DBI were at risk of dyslipidemia and this effect was independent from BMI, as indicated by multivariate analysis. In summary, it appears that ACBP/DBI neutralization has no negative impact on mood and that human depression is not associated with alterations in ACBP/DBI concentrations.


Subject(s)
Diazepam Binding Inhibitor/metabolism , Mental Disorders/metabolism , Animals , Appetite , Behavior, Animal , Body Mass Index , Darkness , Diazepam Binding Inhibitor/blood , Feeding Behavior , Immobilization , Male , Mental Disorders/blood , Mental Disorders/diagnosis , Metabolic Syndrome/blood , Mice, Inbred C57BL , Receptors, GABA-A/metabolism , Swimming/physiology
13.
Curr Biol ; 29(3): 435-448.e8, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30661803

ABSTRACT

Age-related declines in cognitive fitness are associated with a reduction in autophagy, an intracellular lysosomal catabolic process that regulates protein homeostasis and organelle turnover. However, the functional significance of autophagy in regulating cognitive function and its decline during aging remains largely elusive. Here, we show that stimulating memory upregulates autophagy in the hippocampus. Using hippocampal injections of genetic and pharmacological modulators of autophagy, we find that inducing autophagy in hippocampal neurons is required to form novel memory by promoting activity-dependent structural and functional synaptic plasticity, including dendritic spine formation, neuronal facilitation, and long-term potentiation. We show that hippocampal autophagy activity is reduced during aging and that restoring its levels is sufficient to reverse age-related memory deficits. Moreover, we demonstrate that systemic administration of young plasma into aged mice rejuvenates memory in an autophagy-dependent manner, suggesting a prominent role for autophagy to favor the communication between systemic factors and neurons in fostering cognition. Among these youthful factors, we identify osteocalcin, a bone-derived molecule, as a direct hormonal inducer of hippocampal autophagy. Our results reveal that inducing autophagy in hippocampal neurons is a necessary mechanism to enhance the integration of novel stimulations of memory and to promote the influence of systemic factors on cognitive fitness. We also demonstrate the potential therapeutic benefits of modulating autophagy in the aged brain to counteract age-related cognitive impairments.


Subject(s)
Aging/physiology , Autophagy/physiology , Hippocampus/physiology , Memory Disorders , Memory/physiology , Animals , Autophagy/drug effects , Autophagy/genetics , Disease Models, Animal , Male , Memory/drug effects , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL
14.
J Neurosci ; 26(25): 6737-48, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16793881

ABSTRACT

Rat pups must learn maternal odor to support attachment behaviors, including nursing and orientation toward the mother. Neonates have a sensitive period for rapid, robust odor learning characterized by increased ability to learn odor preferences and decreased ability to learn odor aversions. Specifically, odor-0.5 mA shock association paradoxically causes an odor preference and coincident failure of amygdala activation in pups until postnatal day 10 (P10). Because sensitive-period termination coincides with a declining "stress hyporesponsive period" when corticosterone release is attenuated, we explored the role of corticosterone in sensitive-period termination. Odor was paired with 0.5 mA shock in either sensitive-period (P8) or postsensitive-period (P12) pups while manipulating corticosterone. We then assessed preference/aversion learning and the olfactory neural circuitry underlying its acquisition. Although sensitive-period control paired odor-shock pups learned an odor preference without amygdala participation, systemic (3 mg/kg, i.p.; 24 h and 30 min before training) or intra-amygdala corticosterone (50 or 100 ng; during training) permitted precocious odor-aversion learning and evoked amygdala neural activity similar to that expressed by older pups. In postsensitive-period (P12) pups, control paired odor-shock pups showed an odor aversion and amygdala activation, whereas corticosterone-depleted (adrenalectomized) paired odor-shock pups showed odor-preference learning and activation of an odor learning circuit characteristic of the sensitive period. Intra-amygdala corticosterone receptor antagonist (0.3 ng; during training) infused into postsensitive-period (P12) paired odor-shock pups also showed odor-preference learning. These results suggest corticosterone is important in sensitive-period termination and developmental emergence of olfactory fear conditioning, acting via the amygdala as a switch between fear and attraction. Because maternal stimulation of pups modulates the pups' endogenous corticosterone, this suggests maternal care quality may alter sensitive-period duration.


Subject(s)
Amygdala/physiology , Conditioning, Classical/physiology , Corticosterone/metabolism , Nerve Net/physiology , Odorants , Shock , Adrenalectomy/methods , Age Factors , Amygdala/drug effects , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal , Conditioning, Classical/drug effects , Corticosterone/pharmacology , Deoxyglucose/pharmacokinetics , Emotions , Female , Hormone Antagonists/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mifepristone/pharmacology , Nerve Net/drug effects , Radioimmunoassay/methods , Rats , Rats, Long-Evans
15.
Biol Psychiatry ; 62(10): 1070-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17826749

ABSTRACT

BACKGROUND: Early life adverse experience alters adult emotional and cognitive development. Here we assess early life learning about adverse experience and its consequences on adult fear conditioning and amygdala activity. METHODS: Neonatal rats were conditioned daily from 8-12 days-old with paired odor (conditioned stimulus, CS) .5mA shock, unpaired, odor-only, or naive (no infant conditioning). In adulthood, each infant training group was divided into three adult training groups: paired, unpaired or odor-only, using either the same infant CS odor, or a novel adult CS odor without or with the infant CS present as context. Adults were cue tested for freezing (odor in novel environment), with amygdala (14)C 2-DG autoradiography and electrophysiology assessment. RESULTS: Infant paired odor-shock conditioning attenuated adult fear conditioning, but only if the same infant CS odor was used. The (14)C 2-DG activity correlated with infant paired odor-shock conditioning produced attenuated amygdala but heightened olfactory bulb activity. Electrophysiological amygdala assessment further suggests early experience causes changes in amygdala processing as revealed by increased paired-pulse facilitation in adulthood. CONCLUSIONS: This suggests some enduring effects of early life adversity (shock) are under CS control and dependent upon learning for their impact on later adult fear learning.


Subject(s)
Aging , Amygdala/physiology , Conditioning, Classical/physiology , Fear , Memory/physiology , Odorants , Amygdala/diagnostic imaging , Analysis of Variance , Animals , Animals, Newborn , Autoradiography/methods , Avoidance Learning , Behavior, Animal , Deoxyglucose/metabolism , Electric Stimulation/methods , Evoked Potentials/radiation effects , Freezing Reaction, Cataleptic/physiology , Male , Maze Learning , Olfactory Bulb/diagnostic imaging , Radiography , Rats , Rats, Long-Evans , Reaction Time/physiology , Reaction Time/radiation effects
16.
J Exp Med ; 214(10): 2859-2873, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28851741

ABSTRACT

That osteocalcin (OCN) is necessary for hippocampal-dependent memory and to prevent anxiety-like behaviors raises novel questions. One question is to determine whether OCN is also sufficient to improve these behaviors in wild-type mice, when circulating levels of OCN decline as they do with age. Here we show that the presence of OCN is necessary for the beneficial influence of plasma from young mice when injected into older mice on memory and that peripheral delivery of OCN is sufficient to improve memory and decrease anxiety-like behaviors in 16-mo-old mice. A second question is to identify a receptor transducing OCN signal in neurons. Genetic, electrophysiological, molecular, and behavioral assays identify Gpr158, an orphan G protein-coupled receptor expressed in neurons of the CA3 region of the hippocampus, as transducing OCN's regulation of hippocampal-dependent memory in part through inositol 1,4,5-trisphosphate and brain-derived neurotrophic factor. These results indicate that exogenous OCN can improve hippocampal-dependent memory in mice and identify molecular tools to harness this pathway for therapeutic purposes.


Subject(s)
Cognition/physiology , Osteocalcin/physiology , Receptors, G-Protein-Coupled/physiology , Aging/physiology , Animals , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Cognition/drug effects , Electrophysiology , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteocalcin/pharmacology
17.
Horm Mol Biol Clin Investig ; 28(2): 69-83, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27626767

ABSTRACT

Reciprocal relationships between organs are essential to maintain whole body homeostasis. An exciting interplay between two apparently unrelated organs, the bone and the brain, has emerged recently. Indeed, it is now well established that the brain is a powerful regulator of skeletal homeostasis via a complex network of numerous players and pathways. In turn, bone via a bone-derived molecule, osteocalcin, appears as an important factor influencing the central nervous system by regulating brain development and several cognitive functions. In this paper we will discuss this complex and intimate relationship, as well as several pathologic conditions that may reinforce their potential interdependence.


Subject(s)
Bone Diseases, Metabolic/epidemiology , Bone and Bones/physiology , Brain Diseases, Metabolic/epidemiology , Brain/physiology , Cell Communication , Animals , Bone Diseases, Metabolic/pathology , Brain Diseases, Metabolic/pathology , Humans
18.
J Neurosci ; 24(5): 1182-9, 2004 Feb 04.
Article in English | MEDLINE | ID: mdl-14762136

ABSTRACT

Imprinting ensures that the infant forms the caregiver attachment necessary for altricial species survival. In our mammalian model of imprinting, neonatal rats rapidly learn the odor-based maternal attachment. This rapid learning requires reward-evoked locus ceruleus (LC) release of copious amounts of norepinephrine (NE) into the olfactory bulb. This imprinting ends at postnatal day 10 (P10) and is associated with a dramatic reduction in reward-evoked LC NE release. Here we assess whether the functional emergence of LC alpha2 inhibitory autoreceptors and the downregulation of LC alpha1 excitatory autoreceptors underlie the dramatic reduction in NE release associated with termination of the sensitive period. Postsensitive period pups (P12) were implanted with either LC or olfactory bulb cannulas, classically conditioned with intracranial drug infusions (P14), and tested for an odor preference (P15). During conditioning, a novel odor was paired with either olfactory bulb infusion of abeta-receptor agonist (isoproterenol) to assess the target effects of NE or direct LC cholinergic stimulation combined with alpha2 antagonists and alpha1 agonists in a mixture to reinstate neonatal levels of LC autoreceptor activity to assess the source of NE. Pups learned an odor preference when the odor was paired with either olfactory bulb isoproterenol infusion or reinstatement of neonatal LC receptor activity. These results suggest that LC autoreceptor functional changes rather than olfactory bulb changes underlie sensitive period termination.


Subject(s)
Learning/physiology , Nerve Net/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Smell/physiology , Acetylcholine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Newborn , Choice Behavior/drug effects , Choice Behavior/physiology , Cholinergic Agents/pharmacology , Critical Period, Psychological , Drug Administration Routes , Female , Imprinting, Psychological/physiology , Infusion Pumps , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Male , Maze Learning/drug effects , Norepinephrine/pharmacology , Olfactory Bulb/drug effects , Phenylephrine/pharmacology , Rats , Rats, Long-Evans
19.
Behav Neurosci ; 118(2): 274-81, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15113251

ABSTRACT

Infant rats exhibit sensitive-period odor learning characterized by olfactory bulb neural changes and odor preference acquisitions critical for survival. This sensitive period is coincident with low endogenous corticosterone (CORT) levels and stress hyporesponsivity. The authors hypothesized that low corticosterone levels modulate sensitive-period learning. They assessed the effects of manipulating CORT levels by increasing and removing CORT during (Postnatal Day 8) and after (Postnatal Day 12) the sensitive period. Results show that (a) exogenous CORT prematurely ends sensitive-period odor-shock-induced preferences; (b) adrenalectomy developmentally extends the sensitive period as indicated by odor-shock-induced odor-preference learning in older pups, whereas CORT replacement can reinstate fear learning; and (c) CORT manipulation modulates olfactory bulb correlates of sensitive-period odor learning in a manner consistent with behavior.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Corticosterone/pharmacology , Critical Period, Psychological , Learning/drug effects , Adrenalectomy , Animals , Animals, Newborn , Behavior, Animal/drug effects , Female , Male , Maze Learning/drug effects , Object Attachment , Olfactory Bulb/physiology , Rats , Smell/drug effects
20.
Int J Dev Neurosci ; 22(5-6): 415-22, 2004.
Article in English | MEDLINE | ID: mdl-15380840

ABSTRACT

In many altricial species, fear responses such as freezing do not emerge until sometime later in development. In infant rats, fear to natural predator odors emerges around postnatal day (PN) 10 when infant rats begin walking. The behavioral emergence of fear is correlated with two physiological events: functional emergence of the amygdala and increasing corticosterone (CORT) levels. Here, we hypothesize that increasing corticosterone levels influence amygdala activity to permit the emergence of fear expression. We assessed the relationship between fear expression (immobility similar to freezing), amygdala function (c-fos) and the level of corticosterone in pups in response to presentation of novel male odor (predator), littermate odor and no odor. CORT levels were increased in PN8 pups (no fear, normally low CORT) by exogenous CORT (3 mg/kg) and decreased in PN12 pups (express fear, CORT levels higher) through adrenalectomy and CORT replacement. Results showed that PN8 expression of fear to a predator odor and basolateral/lateral amygdala activity could be prematurely evoked with exogenous CORT, while adrenalectomy in PN12 pups prevented both fear expression and amygdala activation. These results suggest that low neonatal CORT level serves to protect pups from responding to fear inducing stimuli and attenuate amygdala activation. This suggests that alteration of the neonatal CORT system by environmental insults such as alcohol, stress and illegal drugs, may also alter the neonatal fear system and its underlying neural control.


Subject(s)
Amygdala/physiology , Avoidance Learning/physiology , Behavior, Animal/physiology , Corticosterone/pharmacology , Fear/drug effects , Fear/physiology , Proto-Oncogene Proteins c-fos/metabolism , Amygdala/drug effects , Animals , Animals, Newborn , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Female , Male , Odorants , Predatory Behavior , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL