Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hippocampus ; 33(8): 970-992, 2023 08.
Article in English | MEDLINE | ID: mdl-37096324

ABSTRACT

How the development and function of neural circuits governing learning and memory are affected by insults in early life remains poorly understood. The goal of this study was to identify putative changes in cortico-hippocampal signaling mechanisms that could lead to learning and memory deficits in a clinically relevant developmental pathophysiological rodent model, Febrile status epilepticus (FSE). FSE in both pediatric cases and the experimental animal model, is associated with enduring physiological alterations of the hippocampal circuit and cognitive impairment. Here, we deconstruct hippocampal circuit throughput by inducing slow theta oscillations in rats under urethane anesthesia and isolating the dendritic compartments of CA1 and dentate gyrus subfields, their reception of medial and lateral entorhinal cortex inputs, and the efficacy of signal propagation to each somatic cell layer. We identify FSE-induced theta-gamma decoupling at cortical synaptic input pathways and altered signal phase coherence along the CA1 and dentate gyrus somatodendritic axes. Moreover, increased DG synaptic activity levels are predictive of poor cognitive outcomes. We propose that these alterations in cortico-hippocampal coordination interfere with the ability of hippocampal dendrites to receive, decode and propagate neocortical inputs. If this frequency-specific syntax is necessary for cortico-hippocampal coordination and spatial learning and memory, its loss could be a mechanism for FSE cognitive comorbidities.


Subject(s)
Seizures, Febrile , Status Epilepticus , Rats , Animals , Seizures, Febrile/chemically induced , Seizures, Febrile/complications , Seizures, Febrile/metabolism , Spatial Learning , Hippocampus/physiology , Entorhinal Cortex/physiology , Status Epilepticus/chemically induced , Dentate Gyrus/physiology
2.
J Neurosci ; 41(6): 1242-1250, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33328295

ABSTRACT

Across species, unpredictable patterns of maternal behavior are emerging as novel predictors of aberrant cognitive and emotional outcomes later in life. In animal models, exposure to unpredictable patterns of maternal behavior alters brain circuit maturation and cognitive and emotional outcomes. However, whether exposure to such signals in humans alters the development of brain pathways is unknown. In mother-child dyads, we tested the hypothesis that exposure to more unpredictable maternal signals in infancy is associated with aberrant maturation of corticolimbic pathways. We focused on the uncinate fasciculus, the primary fiber bundle connecting the amygdala to the orbitofrontal cortex and a key component of the medial temporal lobe-prefrontal cortex circuit. Infant exposure to unpredictable maternal sensory signals was assessed at 6 and 12 months. Using high angular resolution diffusion imaging, we quantified the integrity of the uncinate fasciculus using generalized fractional anisotropy (GFA). Higher maternal unpredictability during infancy presaged greater uncinate fasciculus GFA in children 9-11 years of age (n = 69, 29 female). In contrast to the uncinate, GFA of a second corticolimbic projection, the hippocampal cingulum, was not associated with maternal unpredictability. Addressing the overall functional significance of the uncinate and cingulum relationships, we found that the resulting imbalance of medial temporal lobe-prefrontal cortex connectivity partially mediated the association between unpredictable maternal sensory signals and impaired episodic memory function. These results suggest that unbalanced maturation of corticolimbic circuits is a mechanism by which early unpredictable sensory signals may impact cognition later in life.SIGNIFICANCE STATEMENT Our prior work across species demonstrated that unpredictable patterns of maternal care are associated with compromised memory function. However, the neurobiological mechanisms by which this occurs in humans remain unknown. Here, we identify an association of exposure to unpredictable patterns of maternal sensory signals with the integrity of corticolimbic circuits involved in emotion and cognition using state-of-the-art diffusion imaging techniques and analyses. We find that exposure to early unpredictability is associated with higher integrity of the uncinate fasciculus with no effect on a second corticolimbic pathway, the cingulum. The resulting imbalance of corticolimbic circuit development is a novel mediator of the association between unpredictable patterns of maternal care and poorer episodic memory.


Subject(s)
Maternal Behavior/physiology , Maternal Behavior/psychology , Mother-Child Relations/psychology , Perception/physiology , Uncinate Fasciculus/diagnostic imaging , Uncinate Fasciculus/growth & development , Adult , Child , Cohort Studies , Female , Humans , Infant , Longitudinal Studies , Male , Nerve Net/diagnostic imaging , Nerve Net/growth & development , Prospective Studies
3.
J Neurosci ; 41(4): 648-662, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33262247

ABSTRACT

Stress may promote emotional and cognitive disturbances, which differ by sex. Adverse outcomes, including memory disturbances, are typically observed following chronic stress, but are now being recognized also after short events, including mass shootings, assault, or natural disasters, events that consist of concurrent multiple acute stresses (MAS). Prior work has established profound and enduring effects of MAS on memory in males. Here we examined the effects of MAS on female mice and probed the role of hormonal fluctuations during the estrous cycle on MAS-induced memory problems and the underlying brain network and cellular mechanisms. Female mice were impacted by MAS in an estrous cycle-dependent manner: MAS impaired hippocampus-dependent spatial memory in early-proestrous mice, characterized by high levels of estradiol, whereas memory of mice stressed during estrus (low estradiol) was spared. As spatial memory requires an intact dorsal hippocampal CA1, we examined synaptic integrity in mice stressed at different cycle phases and found a congruence of dendritic spine density and spatial memory deficits, with reduced spine density only in mice stressed during high estradiol cycle phases. Assessing MAS-induced activation of brain networks interconnected with hippocampus, we identified differential estrous cycle-dependent activation of memory- and stress-related regions, including the amygdala. Network analyses of the cross-correlation of fos expression among these regions uncovered functional connectivity that differentiated impaired mice from those not impaired by MAS. In conclusion, the estrous cycle modulates the impact of MAS on spatial memory, and fluctuating physiological levels of sex hormones may contribute to this effect.SIGNIFICANCE STATEMENT: Effects of stress on brain functions, including memory, are profound and sex-dependent. Acute stressors occurring simultaneously result in spatial memory impairments in males, but effects on females are unknown. Here we identified estrous cycle-dependent effects of such stresses on memory in females. Surprisingly, females with higher physiological estradiol experienced stress-induced memory impairment and a loss of underlying synapses. Memory- and stress-responsive brain regions interconnected with hippocampus were differentially activated across high and low estradiol mice, and predicted memory impairment. Thus, at functional, network, and cellular levels, physiological estradiol influences the effects of stress on memory in females, providing insight into mechanisms of prominent sex differences in stress-related memory disorders, such as post-traumatic stress disorder.


Subject(s)
Estrogens , Memory Disorders/physiopathology , Memory Disorders/psychology , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Animals , Brain/physiopathology , CA1 Region, Hippocampal/physiopathology , Dendritic Spines , Estrous Cycle , Estrus , Female , Male , Maze Learning , Mice , Mice, Inbred C57BL , Nerve Net/physiopathology , Proto-Oncogene Proteins c-fos/genetics , Spatial Memory , Uterus/innervation , Uterus/physiopathology
4.
Eur J Neurosci ; 55(9-10): 2170-2195, 2022 05.
Article in English | MEDLINE | ID: mdl-33825217

ABSTRACT

Addiction is a chronic relapsing disorder with devastating personal, societal, and economic consequences. In humans, early-life adversity (ELA) such as trauma, neglect, and resource scarcity are linked with increased risk of later-life addiction, but the brain mechanisms underlying this link are still poorly understood. Here, we focus on data from rodent models of ELA and addiction, in which causal effects of ELA on later-life responses to drugs and the neurodevelopmental mechanisms by which ELA increases vulnerability to addiction can be determined. We first summarize evidence for a link between ELA and addiction in humans, then describe how ELA is commonly modeled in rodents. Since addiction is a heterogeneous disease with many individually varying behavioral aspects that may be impacted by ELA, we next discuss common rodent assays of addiction-like behaviors. We then summarize the specific addiction-relevant behavioral phenotypes caused by ELA in male and female rodents and discuss some of the underlying changes in brain reward and stress circuits that are likely responsible. By better understanding the behavioral and neural mechanisms by which ELA promotes addiction vulnerability, we hope to facilitate development of new approaches for preventing or treating addiction in those with a history of ELA.


Subject(s)
Behavior, Addictive , Stress, Psychological , Substance-Related Disorders , Animals , Female , Male , Models, Animal , Reward , Rodentia
5.
Mol Psychiatry ; 26(8): 4409-4416, 2021 08.
Article in English | MEDLINE | ID: mdl-31822817

ABSTRACT

The origins and neural bases of the current opioid addiction epidemic are unclear. Genetics plays a major role in addiction vulnerability, but cannot account for the recent exponential rise in opioid abuse, so environmental factors must contribute. Individuals with history of early life adversity (ELA) are disproportionately prone to opioid addiction, yet whether ELA interacts with factors such as increased access to opioids to directly influence brain development and function, and cause opioid addiction vulnerability, is unknown. We simulated ELA in female rats and this led to a striking opioid addiction-like phenotype. This was characterized by resistance to extinction, increased relapse-like behavior, and, as in addicted humans, major increases in opioid economic demand. By contrast, seeking of a less salient natural reward was unaffected by ELA, whereas demand for highly palatable treats was augmented. These discoveries provide novel insights into the origins and nature of reward circuit malfunction that may set the stage for addiction.


Subject(s)
Behavior, Addictive , Opioid-Related Disorders , Analgesics, Opioid , Animals , Female , Origin of Life , Rats , Reward
6.
Depress Anxiety ; 39(10-11): 706-717, 2022 10.
Article in English | MEDLINE | ID: mdl-35833573

ABSTRACT

BACKGROUND: Recent studies in both human and experimental animals have identified fragmented and unpredictable parental and environmental signals as a novel source of early-life adversity. Early-life unpredictability may be a fundamental developmental factor that impacts brain development, including reward and emotional memory circuits, affecting the risk for psychopathology later in life. Here, we tested the hypothesis that self-reported early-life unpredictability is associated with psychiatric symptoms in adult clinical populations. METHODS: Using the newly validated Questionnaire of Unpredictability in Childhood, we assessed early-life unpredictability in 156 trauma-exposed adults, of which 65% sought treatment for mood, anxiety, and/or posttraumatic stress disorder (PTSD) symptoms. All participants completed symptom measures of PTSD, depression and anhedonia, anxiety, alcohol use, and chronic pain. Relative contributions of early-life unpredictability versus childhood trauma and associations with longitudinal outcomes over a 6-month period were determined. RESULTS: Early-life unpredictability, independent of childhood trauma, was significantly associated with higher depression, anxiety symptoms, and anhedonia, and was related to higher overall symptom ratings across time. Early-life unpredictability was also associated with suicidal ideation, but not alcohol use or pain symptoms. CONCLUSIONS: Early-life unpredictability is an independent and consistent predictor of specific adult psychiatric symptoms, providing impetus for studying mechanisms of its effects on the developing brain that promote risk for psychopathology.


Subject(s)
Anhedonia , Stress Disorders, Post-Traumatic , Adult , Animals , Anxiety , Anxiety Disorders , Emotions , Humans , Stress Disorders, Post-Traumatic/psychology
7.
Epilepsia ; 62(12): 3117-3130, 2021 12.
Article in English | MEDLINE | ID: mdl-34562024

ABSTRACT

OBJECTIVE: Febrile seizures (FSs) are the most common form of seizures in children. Single short FSs are benign, but FSs lasting longer than 30 min, termed febrile status epilepticus, may result in neurological sequelae. However, there is little information about an intermediary condition, brief recurrent FSs (RFSs). The goal of this study was to determine the role of RFSs on spatial learning and memory and the properties of spontaneous hippocampal signals. METHODS: A hippocampus-dependent active avoidance task was used to assess spatial learning and memory in adult rats that underwent experimental RFSs (eRFSs) in early life compared with their littermate controls. Following completion of the task, we utilized high-density laminar probes to measure spontaneous hippocampal CA1 circuit activity under urethane anesthesia, which allowed for the simultaneous recording of input regions in CA1 associated with both CA3 and entorhinal cortex. RESULTS: RFSs did not result in deficits in the active avoidance spatial test, a hippocampus-dependent test of spatial learning and memory. However, in vivo high-density laminar electrode recordings from eRFS rats had significantly altered power and frequency expression of theta and gamma bandwidths as well as signaling efficacy along the CA1 somatodendritic axis. Thus, although eRFS modified CA1 neuronal input/output dynamics, these alterations were not sufficient to impair active avoidance spatial behavior. SIGNIFICANCE: These findings indicate that although eRFSs do not result in spatial cognitive deficits in the active avoidance task, recurrent seizures do alter the brain and result in longstanding changes in the temporal organization of the hippocampus.


Subject(s)
Seizures, Febrile , Status Epilepticus , Animals , Hippocampus/physiology , Rats , Seizures/chemically induced , Seizures/complications , Seizures, Febrile/chemically induced , Seizures, Febrile/complications , Spatial Learning/physiology , Status Epilepticus/chemically induced , Status Epilepticus/complications
8.
Epilepsia ; 62(3): 647-658, 2021 03.
Article in English | MEDLINE | ID: mdl-33475157

ABSTRACT

OBJECTIVE: Prolonged fever-induced seizures (febrile status epilepticus [FSE]) during early childhood increase the risk for later epilepsy, but the underlying mechanisms are incompletely understood. Experimental FSE (eFSE) in rats successfully models human FSE, recapitulating the resulting epileptogenesis in a subset of affected individuals. However, the powerful viral and genetic tools that may enhance mechanistic insights into epileptogenesis and associated comorbidities, are better-developed for mice. Therefore, we aimed to determine if eFSE could be generated in mice and if it provoked enduring changes in hippocampal-network excitability and the development of spontaneous seizures. METHODS: We employed C57BL/6J male mice, the strain used most commonly in transgenic manipulations, and examined if early life eFSE could be sustained and if it led to hyperexcitability of hippocampal networks and to epilepsy. Outcome measures included vulnerability to the subsequent administration of the limbic convulsant kainic acid (KA) and the development of spontaneous seizures. In the first mouse cohort, adult naive and eFSE-experiencing mice were exposed to KA. A second cohort of control and eFSE-experiencing young adult mice was implanted with bilateral hippocampal electrodes and recorded using continuous video-electroencephalography (EEG) for 2 to 3 months to examine for spontaneous seizures (epileptogenesis). RESULTS: Induction of eFSE was feasible and eFSE increased the susceptibility of adult C57BL/6J mice to KA, thereby reducing latency to seizure onset and increasing seizure severity. Of 24 chronically recorded eFSE mice, 4 (16.5%) developed hippocampal epilepsy with a latent period of ~3 months, significantly different from the expectation by chance (P = .04). The limbic epilepsy that followed eFSE was progressive. SIGNIFICANCE: eFSE promotes pro-epileptogenic network changes in a majority of C57BL/6J male mice and frank "temporal lobe-like" epilepsy in one sixth of the cohort. Mouse eFSE may thus provide a useful tool for investigating molecular, cellular, and circuit changes during the development of temporal lobe epilepsy and its comorbidities.


Subject(s)
Hippocampus/physiopathology , Seizures, Febrile/etiology , Status Epilepticus/etiology , Animals , Disease Models, Animal , Disease Susceptibility/physiopathology , Electrodes, Implanted , Electroencephalography , Excitatory Amino Acid Agonists/pharmacology , Female , Hot Temperature/adverse effects , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Seizures, Febrile/physiopathology , Status Epilepticus/physiopathology , Translational Research, Biomedical
9.
Front Neuroendocrinol ; 53: 100736, 2019 04.
Article in English | MEDLINE | ID: mdl-30711600

ABSTRACT

Mental illnesses originate early in life, governed by environmental and genetic factors. Because parents are a dominant source of signals to the developing child, parental signals - beginning with maternal signals in utero - are primary contributors to children's mental health. Existing literature on maternal signals has focused almost exclusively on their quality and valence (e.g. maternal depression, sensitivity). Here we identify a novel dimension of maternal signals: their patterns and especially their predictability/unpredictability, as an important determinant of children's neurodevelopment. We find that unpredictable maternal mood and behavior presage risk for child and adolescent psychopathology. In experimental models, fragmented/unpredictable maternal care patterns directly induce aberrant synaptic connectivity and disturbed maturation of cognitive and emotional brain circuits, with commensurate memory problems and anhedonia-like behaviors. Together, our findings across species demonstrate that patterns of maternal signals influence brain circuit maturation, promoting resilience or vulnerability to mental illness.


Subject(s)
Brain/growth & development , Emotions/physiology , Maternal Behavior/psychology , Mental Disorders/physiopathology , Parents/psychology , Animals , Humans , Stress, Psychological/psychology
10.
Proc Natl Acad Sci U S A ; 114(39): 10390-10395, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28893979

ABSTRACT

Maternal care is a critical determinant of child development. However, our understanding of processes and mechanisms by which maternal behavior influences the developing human brain remains limited. Animal research has illustrated that patterns of sensory information is important in shaping neural circuits during development. Here we examined the relation between degree of predictability of maternal sensory signals early in life and subsequent cognitive function in both humans (n = 128 mother/infant dyads) and rats (n = 12 dams; 28 adolescents). Behaviors of mothers interacting with their offspring were observed in both species, and an entropy rate was calculated as a quantitative measure of degree of predictability of transitions among maternal sensory signals (visual, auditory, and tactile). Human cognitive function was assessed at age 2 y with the Bayley Scales of Infant Development and at age 6.5 y with a hippocampus-dependent delayed-recall task. Rat hippocampus-dependent spatial memory was evaluated on postnatal days 49-60. Early life exposure to unpredictable sensory signals portended poor cognitive performance in both species. The present study provides evidence that predictability of maternal sensory signals early in life impacts cognitive function in both rats and humans. The parallel between experimental animal and observational human data lends support to the argument that predictability of maternal sensory signals causally influences cognitive development.


Subject(s)
Behavior, Animal/physiology , Child Development/physiology , Cognition/physiology , Hippocampus/growth & development , Maternal Behavior/psychology , Stress, Psychological/psychology , Animals , Child , Child, Preschool , Female , Hippocampus/physiology , Humans , Longitudinal Studies , Male , Maternal Behavior/physiology , Maternal-Fetal Relations/psychology , Rats , Rats, Sprague-Dawley , Spatial Memory/physiology
11.
Dev Psychobiol ; 62(6): 882-888, 2020 09.
Article in English | MEDLINE | ID: mdl-32115696

ABSTRACT

BACKGROUND: Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is associated with poor physical and mental health. Early-life adversity may dysregulate cortisol response to subsequent stress. This study examines the association between patterns of maternal behavior and infant stress response to a challenge. Specifically, we test whether infant exposure to unpredictable maternal sensory signals is related to the cortisol response to a painful stressor. METHOD: Participants were 102 mothers and their children enrolled in a longitudinal study. Patterns of maternal sensory signals were evaluated at 6 and 12 months during a 10-min mother-infant play episode. Entropy rate was calculated as a quantitative measure of the degree of unpredictability of maternal sensory signals (visual, auditory, and tactile) exhibited during the play episode. Infant saliva samples were collected for cortisol analysis before and after inoculation at 12 months. RESULTS: Unpredictable patterns of maternal sensory signals were associated with a blunted infant cortisol response to a painful stressor. This relation persisted after evaluation of covariates including maternal sensitivity and maternal psychological distress. CONCLUSIONS: This study provides evidence that unpredictable patterns of maternal sensory signals are one process through which caregiving affects the function of infant stress response systems.


Subject(s)
Child Development/physiology , Hydrocortisone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Maternal Behavior/physiology , Stress, Psychological/metabolism , Adult , Female , Humans , Infant , Longitudinal Studies , Male , Psychological Distress
12.
Learn Mem ; 26(7): 206-218, 2019 07.
Article in English | MEDLINE | ID: mdl-31209115

ABSTRACT

Spatial memory, the aspect of memory involving encoding and retrieval of information regarding one's environment and spatial orientation, is a complex biological function incorporating multiple neuronal networks. Hippocampus-dependent spatial memory is not innate and emerges during development in both humans and rodents. In children, nonhippocampal dependent egocentric (self-to-object) memory develops before hippocampal-dependent allocentric (object-to-object) memory. The onset of allocentric spatial memory abilities in children around 22 mo of age occurs at an age-equivalent time in rodents when spatially tuned grid and place cells arise from patterned activity propagating through the entorhinal-hippocampal circuit. Neuronal activity, often driven by specific sensory signals, is critical for the normal maturation of brain circuits This patterned activity fine-tunes synaptic connectivity of the network and drives the emergence of specific firing necessary for spatial memory. Whereas normal activity patterns are required for circuit maturation, aberrant neuronal activity during development can have major adverse consequences, disrupting the development of spatial memory. Seizures during infancy, involving massive bursts of synchronized network activity, result in impaired spatial memory when animals are tested as adolescents or adults. This impaired spatial memory is accompanied by alterations in spatial and temporal coding of place cells. The molecular mechanisms by which early-life seizures lead to disruptions at the cellular and network levels are now becoming better understood, and provide a target for intervention, potentially leading to improved cognitive outcome in individuals experiencing early-life seizures.


Subject(s)
Nerve Net/growth & development , Spatial Memory/physiology , Animals , Child , Child Development , Child, Preschool , Connectome , Entorhinal Cortex/cytology , Entorhinal Cortex/growth & development , Entorhinal Cortex/physiology , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/physiology , Humans , Infant , Memory Disorders/etiology , Memory Disorders/physiopathology , Models, Neurological , Nerve Net/physiology , Neurons/classification , Neurons/physiology , Seizures/complications
13.
J Neurosci ; 37(14): 3799-3812, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28275159

ABSTRACT

In a subset of children experiencing prolonged febrile seizures (FSs), the most common type of childhood seizures, cognitive outcomes are compromised. However, the underlying mechanisms are unknown. Here we identified significant, enduring spatial memory problems in male rats following experimental prolonged FS (febrile status epilepticus; eFSE). Remarkably, these deficits were abolished by transient, post hoc interference with the chromatin binding of the transcriptional repressor neuron restrictive silencing factor (NRSF or REST). This transcriptional regulator is known to contribute to neuronal differentiation during development and to programmed gene expression in mature neurons. The mechanisms of the eFSE-provoked memory problems involved complex disruption of memory-related hippocampal oscillations recorded from CA1, likely resulting in part from impairments of dendritic filtering of cortical inputs as well as abnormal synaptic function. Accordingly, eFSE provoked region-specific dendritic loss in the hippocampus, and aberrant generation of excitatory synapses in dentate gyrus granule cells. Blocking NRSF transiently after eFSE prevented granule cell dysmaturation, restored a functional balance of γ-band network oscillations, and allowed treated eFSE rats to encode and retrieve spatial memories. Together, these studies provide novel insights into developing networks that underlie memory, the mechanisms by which early-life seizures influence them, and the means to abrogate the ensuing cognitive problems.SIGNIFICANCE STATEMENT Whereas seizures have been the central focus of epilepsy research, they are commonly accompanied by cognitive problems, including memory impairments that contribute to poor quality of life. These deficits often arise before the onset of spontaneous seizures, or independent from them, yet the mechanisms involved are unclear. Here, using a rodent model of common developmental seizures that provoke epilepsy in a subset of individuals, we identify serious consequent memory problems. We uncover molecular, cellular, and circuit-level mechanisms that underlie these deficits and successfully abolish them by targeted therapeutic interventions. These findings may be important for understanding and preventing cognitive problems in individuals suffering long febrile seizures.


Subject(s)
Memory Disorders/metabolism , Memory Disorders/physiopathology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology , Animals , Animals, Newborn , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Memory Disorders/etiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Seizures, Febrile/complications
14.
Neuroimage ; 175: 402-412, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29649560

ABSTRACT

Adolescence is a complex period of concurrent mental and physical development that facilitates adult functioning at multiple levels. Despite the growing number of neuroimaging studies of cognitive development in adolescence focusing on regional activation patterns, there remains a paucity of information about the functional interactions across these participating regions that are critical for cognitive functioning, including memory. The current study used structural equation modeling (SEM) to determine how interactions among brain regions critical for memory change over the course of adolescence. We obtained functional MRI in 77 individuals aged 8-16 years old, divided into younger (ages 8-10) and older (ages > 11) cohorts, using an incidental encoding memory task to activate hippocampus formation and associated brain networks, as well as behavioral data on memory function. SEM was performed on the imaging data for four groups (younger girls, younger boys, older girls, and older boys) that were subsequently compared using a stacked model approach. Significant differences were seen between the models for these groups. Younger boys had a predominantly posterior distribution of connections originating in primary visual regions and terminating on multi-modal processing regions. In older boys, there was a relatively greater anterior connection distribution, with increased effective connectivity within association and multi-modal processing regions. Connection patterns in younger girls were similar to those of older boys, with a generally anterior-posterior distributed network among sensory, multi-modal, and limbic regions. In contrast, connections in older girls were widely distributed but relatively weaker. Memory performance increased with age, without a significant difference between the sexes. These findings suggest a progressive reorganization among brain regions, with a commensurate increase in efficiency of cognitive functioning, from younger to older individuals in both girls and boys, providing insight into the age- and gender-specific processes at play during this critical transition period.


Subject(s)
Adolescent Development/physiology , Child Development/physiology , Connectome/methods , Hippocampus/physiology , Nerve Net/physiology , Adolescent , Child , Female , Humans , Magnetic Resonance Imaging , Male , Memory/physiology , Sex Factors
15.
Epilepsia ; 59(11): 2005-2018, 2018 11.
Article in English | MEDLINE | ID: mdl-30256385

ABSTRACT

OBJECTIVE: A subset of children with febrile status epilepticus (FSE) are at risk for development of temporal lobe epilepsy later in life. We sought a noninvasive predictive marker of those at risk that can be identified soon after FSE, within a clinically realistic timeframe. METHODS: Longitudinal T2 -weighted magnetic resonance imaging (T2 WI MRI) of rat pups at several time points after experimental FSE (eFSE) was performed on a high-field scanner followed by long-term continuous electroencephalography. In parallel, T2 WI MRI scans were performed on a 3.0-T clinical scanner. Finally, chronic T2 WI MRI signal changes were examined in rats that experienced eFSE and were imaged months later in adulthood. RESULTS: Epilepsy-predicting T2 changes, previously observed at 2 hours after eFSE, persisted for at least 6 hours, enabling translation to the clinic. Repeated scans, creating MRI trajectories of T2 relaxation times following eFSE, provided improved prediction of epileptogenesis compared with a single MRI scan. Predictive signal changes centered on limbic structures, such as the basolateral and medial amygdala. T2 WI MRI changes, originally described on high-field scanners, can also be measured on clinical MRI scanners. Chronically elevated T2 relaxation times in hippocampus were observed months after eFSE in rats, as noted for post-FSE changes in children. SIGNIFICANCE: Early T2 WI MRI changes after eFSE provide a strong predictive measure of epileptogenesis following eFSE, on both high-field and clinical MRI scanners. Importantly, the extension of the acute signal changes to at least 6 hours after the FSE enables its inclusion in clinical studies. Chronic elevations of T2 relaxation times within the hippocampal formation and related structures are common to human and rodent FSE, suggesting that similar processes are involved across species.


Subject(s)
Brain/diagnostic imaging , Brain/growth & development , Disease Progression , Image Processing, Computer-Assisted , Magnetic Resonance Imaging/methods , Status Epilepticus/diagnostic imaging , Animals , Animals, Newborn , Disease Models, Animal , Electroencephalography , Female , Fever/complications , Male , ROC Curve , Rats , Rats, Sprague-Dawley , Status Epilepticus/etiology , Time Factors
16.
J Neurosci ; 36(44): 11295-11307, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807170

ABSTRACT

Stress influences memory, an adaptive process crucial for survival. During stress, hippocampal synapses are bathed in a mixture of stress-released molecules, yet it is unknown whether or how these interact to mediate the effects of stress on memory. Here, we demonstrate novel synergistic actions of corticosterone and corticotropin-releasing hormone (CRH) on synaptic physiology and dendritic spine structure that mediate the profound effects of acute concurrent stresses on memory. Spatial memory in mice was impaired enduringly after acute concurrent stresses resulting from loss of synaptic potentiation associated with disrupted structure of synapse-bearing dendritic spines. Combined application of the stress hormones corticosterone and CRH recapitulated the physiological and structural defects provoked by acute stresses. Mechanistically, corticosterone and CRH, via their cognate receptors, acted synergistically on the spine-actin regulator RhoA, promoting its deactivation and degradation, respectively, and destabilizing spines. Accordingly, blocking the receptors of both hormones, but not each alone, rescued memory. Therefore, the synergistic actions of corticosterone and CRH at hippocampal synapses underlie memory impairments after concurrent and perhaps also single, severe acute stresses, with potential implications to spatial memory dysfunction in, for example, posttraumatic stress disorder. SIGNIFICANCE STATEMENT: Stress influences memory, an adaptive process crucial for survival. During stress, adrenal corticosterone and hippocampal corticotropin-releasing hormone (CRH) permeate memory-forming hippocampal synapses, yet it is unknown whether (and how) these hormones interact to mediate effects of stress. Here, we demonstrate novel synergistic actions of corticosterone and CRH on hippocampal synaptic plasticity and spine structure that mediate the memory-disrupting effects of stress. Combined application of both hormones provoked synaptic function collapse and spine disruption. Mechanistically, corticosterone and CRH synergized at the spine-actin regulator RhoA, promoting its deactivation and degradation, respectively, and destabilizing spines. Notably, blocking both hormones, but not each alone, prevented the enduring memory problems after acute concurrent stresses. Therefore, synergistic actions of corticosterone and CRH underlie enduring memory impairments after concurrent acute stresses, which might be relevant to spatial memory deficits described in posttraumatic stress disorder.


Subject(s)
Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Hippocampus/physiopathology , Memory Disorders/physiopathology , Spatial Memory , Stress, Psychological/physiopathology , Acute Disease , Animals , Corticosterone/administration & dosage , Corticotropin-Releasing Hormone/administration & dosage , Drug Synergism , Hippocampus/drug effects , Male , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Stress, Psychological/complications
17.
Stress ; 20(5): 421-448, 2017 09.
Article in English | MEDLINE | ID: mdl-28617197

ABSTRACT

The immediate and long-term effects of exposure to early life stress (ELS) have been documented in humans and animal models. Even relatively brief periods of stress during the first 10 days of life in rodents can impact later behavioral regulation and the vulnerability to develop adult pathologies, in particular an impairment of cognitive functions and neurogenesis, but also modified social, emotional, and conditioned fear responses. The development of preclinical models of ELS exposure allows the examination of mechanisms and testing of therapeutic approaches that are not possible in humans. Here, we describe limited bedding and nesting (LBN) procedures, with models that produce altered maternal behavior ranging from fragmentation of care to maltreatment of infants. The purpose of this paper is to discuss important issues related to the implementation of this chronic ELS procedure and to describe some of the most prominent endpoints and consequences, focusing on areas of convergence between laboratories. Effects on the hypothalamic-pituitary adrenal (HPA) axis, gut axis and metabolism are presented in addition to changes in cognitive and emotional functions. Interestingly, recent data have suggested a strong sex difference in some of the reported consequences of the LBN paradigm, with females being more resilient in general than males. As both the chronic and intermittent variants of the LBN procedure have profound consequences on the offspring with minimal external intervention from the investigator, this model is advantageous ecologically and has a large translational potential. In addition to the direct effect of ELS on neurodevelopmental outcomes, exposure to adverse early environments can also have intergenerational impacts on mental health and function in subsequent generation offspring. Thus, advancing our understanding of the effect of ELS on brain and behavioral development is of critical concern for the health and wellbeing of both the current population, and for generations to come.


Subject(s)
Child Abuse , Cognition , Emotions , Maternal Behavior , Nesting Behavior , Stress, Psychological/psychology , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Bedding and Linens , Behavior, Animal , Epigenesis, Genetic , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Infant, Newborn , Male , Models, Animal , Neurogenesis , Pituitary-Adrenal System/metabolism , Reproducibility of Results , Resilience, Psychological , Rodentia , Sex Factors , Stress, Psychological/metabolism
18.
Epilepsia ; 58 Suppl 3: 11-19, 2017 07.
Article in English | MEDLINE | ID: mdl-28675560

ABSTRACT

Epilepsy can be a devastating disorder. In addition to debilitating seizures, epilepsy can cause cognitive and emotional problems with reduced quality of life. Therefore, the major aim is to prevent the disorder in the first place: identify, detect, and reverse the processes responsible for its onset, and monitor and treat its progression. Epilepsy often occurs following a latent period of months to years (epileptogenesis) as a consequence of a brain insult, such as head trauma, stroke, or status epilepticus. Although this latent period clearly represents a therapeutic window, we are not able to stratify patients at risk for long-term epilepsy, which is prerequisite for preventative clinical trials. Moreover, because of the length of the latent period, an early biomarker for treatment response would be of high value. Finally, mechanistic biomarkers of epileptogenesis may provide more profound insight in the process of disease development.


Subject(s)
Biomarkers/analysis , Epilepsy/immunology , Neurogenic Inflammation/immunology , Animals , Astrocytes/physiology , Brain/diagnostic imaging , Brain/immunology , Brain/physiopathology , Brain Injuries/complications , Brain Injuries/diagnostic imaging , Brain Injuries/immunology , Brain Injuries/physiopathology , Disease Models, Animal , Disease Progression , Electroencephalography , Epilepsy/diagnostic imaging , Epilepsy/physiopathology , Epilepsy/prevention & control , Humans , Image Enhancement , Magnetic Resonance Imaging , Neurogenic Inflammation/diagnostic imaging , Neurogenic Inflammation/physiopathology , Neurogenic Inflammation/prevention & control , Proton Magnetic Resonance Spectroscopy , Rats , Risk Factors , Seizures, Febrile/diagnostic imaging , Seizures, Febrile/physiopathology , Status Epilepticus/complications , Status Epilepticus/diagnostic imaging , Status Epilepticus/immunology , Status Epilepticus/physiopathology , Stroke/complications , Stroke/diagnostic imaging , Stroke/immunology , Stroke/physiopathology , Vascular Cell Adhesion Molecule-1/analysis , Video Recording
19.
Hippocampus ; 26(12): 1618-1632, 2016 12.
Article in English | MEDLINE | ID: mdl-27657911

ABSTRACT

Memory and related cognitive functions are progressively impaired in a subgroup of individuals experiencing childhood adversity and stress. However, it is not possible to identify vulnerable individuals early, a crucial step for intervention. In this study, high-resolution magnetic resonance imaging (MRI) and intra-hippocampal diffusion tensor imaging (DTI) were employed to examine for structural signatures of cognitive adolescent vulnerabilities in a rodent model of early-life adversity. These methods were complemented by neuroanatomical and functional assessments of hippocampal network integrity during adolescence, adulthood and middle-age. The high-resolution MRI identified selective loss of dorsal hippocampal volume, and intra-hippocampal DTI uncovered disruption of dendritic structure, consistent with disrupted local connectivity, already during late adolescence in adversity-experiencing rats. Memory deteriorated over time, and stunting of hippocampal dendritic trees was apparent on neuroanatomical analyses. Thus, disrupted hippocampal neuronal structure and connectivity, associated with cognitive impairments, are detectable via non-invasive imaging modalities in rats experiencing early-life adversity. These high-resolution imaging approaches may constitute promising tools for prediction and assessment of at-risk individuals in the clinic. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/diagnostic imaging , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Stress, Psychological/complications , Stress, Psychological/diagnostic imaging , Animals , Cohort Studies , Corticosterone/blood , Crowding , Diffusion Tensor Imaging , Environment , Female , Hippocampus/growth & development , Hippocampus/pathology , Housing, Animal , Light , Magnetic Resonance Imaging , Male , Memory Disorders/blood , Memory Disorders/pathology , Models, Animal , Noise , Organ Size , Pyramidal Cells/pathology , Radioimmunoassay , Random Allocation , Rats, Sprague-Dawley , Stress, Psychological/blood , Stress, Psychological/pathology
20.
J Neurosci ; 34(26): 8672-84, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24966369

ABSTRACT

A significant proportion of temporal lobe epilepsy (TLE), a common, intractable brain disorder, arises in children with febrile status epilepticus (FSE). Preventative therapy development is hampered by our inability to identify early the FSE individuals who will develop TLE. In a naturalistic rat model of FSE, we used high-magnetic-field MRI and long-term video EEG to seek clinically relevant noninvasive markers of epileptogenesis and found that reduced amygdala T2 relaxation times in high-magnetic-field MRI hours after FSE predicted experimental TLE. Reduced T2 values likely represented paramagnetic susceptibility effects derived from increased unsaturated venous hemoglobin, suggesting augmented oxygen utilization after FSE termination. Indeed, T2 correlated with energy-demanding intracellular translocation of the injury-sensor high-mobility group box 1 (HMGB1), a trigger of inflammatory cascades implicated in epileptogenesis. Use of deoxyhemoglobin-sensitive MRI sequences enabled visualization of the predictive changes on lower-field, clinically relevant scanners. This novel MRI signature delineates the onset and suggests mechanisms of epileptogenesis that follow experimental FSE.


Subject(s)
Brain/physiopathology , Electroencephalography/methods , Epilepsy/diagnosis , Magnetic Resonance Imaging/methods , Seizures, Febrile/complications , Status Epilepticus/complications , Animals , Biomarkers , Brain/pathology , Disease Models, Animal , Epilepsy/etiology , Epilepsy/pathology , Epilepsy/physiopathology , Rats , Rats, Sprague-Dawley , Seizures, Febrile/pathology , Seizures, Febrile/physiopathology , Status Epilepticus/pathology , Status Epilepticus/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL