Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Scand J Med Sci Sports ; 34(2): e14575, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38339809

ABSTRACT

INTRODUCTION: The number of randomized controlled trials (RCTs) investigating the effects of exercise among cancer survivors has increased in recent years; however, participants dropping out of the trials are rarely described. The objective of the present study was to assess which combinations of participant and exercise program characteristics were associated with dropout from the exercise arms of RCTs among cancer survivors. METHODS: This study used data collected in the Predicting OptimaL cAncer RehabIlitation and Supportive care (POLARIS) study, an international database of RCTs investigating the effects of exercise among cancer survivors. Thirty-four exercise trials, with a total of 2467 patients without metastatic disease randomized to an exercise arm were included. Harmonized studies included a pre and a posttest, and participants were classified as dropouts when missing all assessments at the post-intervention test. Subgroups were identified with a conditional inference tree. RESULTS: Overall, 9.6% of the participants dropped out. Five subgroups were identified in the conditional inference tree based on four significant associations with dropout. Most dropout was observed for participants with BMI >28.4 kg/m2 , performing supervised resistance or unsupervised mixed exercise (19.8% dropout) or had low-medium education and performed aerobic or supervised mixed exercise (13.5%). The lowest dropout was found for participants with BMI >28.4 kg/m2 and high education performing aerobic or supervised mixed exercise (5.1%), and participants with BMI ≤28.4 kg/m2 exercising during (5.2%) or post (9.5%) treatment. CONCLUSIONS: There are several systematic differences between cancer survivors completing and dropping out from exercise trials, possibly affecting the external validity of exercise effects.


Subject(s)
Cancer Survivors , Neoplasms , Humans , Quality of Life , Exercise , Exercise Therapy , Neoplasms/rehabilitation , Randomized Controlled Trials as Topic
2.
Neurobiol Stress ; 13: 100282, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33344733

ABSTRACT

Exposure to adversity during early life can have profound influences on brain function and behavior later in life. The peripubertal period is emerging as an important time-window of susceptibility to stress, with substantial evidence documenting long-term consequences in the emotional and social domains. However, little is known about how stress during this period impacts subsequent cognitive functioning. Here, we assessed potential long-term effects of peripubertal stress on spatial learning and memory using the water maze task. In addition, we interrogated whether individual differences in stress-induced behavioral and endocrine changes are related to the degree of adaptation of the corticosterone response to repeated stressor exposure during the peripubertal period. We found that, when tested at adulthood, peripubertally stressed animals displayed a slower learning rate. Strikingly, the level of spatial orientation in the water maze completed on the last training day was predicted by the degree of adaptation of the recovery -and not the peak-of the corticosterone response to stressor exposure (i.e., plasma levels at 60 min post-stressor) across the peripubertal stress period. In addition, peripubertal stress led to changes in emotional and glucocorticoid reactivity to novelty exposure, as well as in the expression levels of the plasticity molecule PSA-NCAM in the hippocampus. Importantly, by assessing the same endpoints in another peripubertally stressed cohort tested during adolescence, we show that the observed effects at adulthood are the result of a delayed programming manifested at adulthood and not protracted effects of stress. Altogether, our results support the view that the degree of stress-induced adaptation of the hypothalamus-pituitary-adrenal axis responsiveness at the important transitional period of puberty relates to the long-term programming of cognition, behavior and endocrine reactivity.

3.
Neuropharmacology ; 164: 107896, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31811875

ABSTRACT

The LPA1 receptor, one of the six characterized G protein-coupled receptors (LPA1-6) through which lysophosphatidic acid acts, is likely involved in promoting normal emotional behaviours. Current data suggest that the LPA-LPA1-receptor pathway may be involved in mediating the negative consequences of stress on hippocampal function. However, to date, there is no available information regarding the mechanisms whereby the LPA1 receptor mediates this adaptation. To gain further insight into how the LPA-LPA1 pathway may prevent the negative consequences of chronic stress, we assessed the effects of the continuous delivery of LPA on depressive-like behaviours induced by a chronic restraint stress protocol. Because a proper excitatory/inhibitory balance seems to be key for controlling the stress response system, the gene expression of molecular markers of excitatory and inhibitory neurotransmission was also determined. In addition, the hippocampal expression of mineralocorticoid receptor genes and glucocorticoid receptor genes and proteins as well as plasma corticosterone levels were determined. Contrary to our expectations, the continuous delivery of LPA in chronically stressed animals potentiated rather than inhibited some (e.g., anhedonia, reduced latency to the first immobility period), though not all, behavioural effects of stress. Furthermore, this treatment led to an alteration in the genes coding for proteins involved in the excitatory/inhibitory balance in the ventral hippocampus and to changes in corticosterone levels. In conclusion, the results of this study reinforce the assumption that LPA is involved in emotional regulation, mainly through the LPA1 receptor, and regulates the effects of stress on hippocampal gene expression and hippocampus-dependent behaviour.


Subject(s)
Behavior, Animal , Hippocampus/physiopathology , Receptors, Lysophosphatidic Acid/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology , Anhedonia , Animals , Chronic Disease , Corticosterone/blood , Depression/psychology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Neural Inhibition , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/genetics , Stress, Psychological/physiopathology , Swimming/psychology , Synaptic Transmission
4.
Neuropharmacology ; 159: 107498, 2019 11 15.
Article in English | MEDLINE | ID: mdl-30660627

ABSTRACT

Most socially living species are organized hierarchically, primarily based on individual differences in social dominance. Dominant individuals typically gain privileged access to important resources, such as food, mating partners and territories, whereas submissive conspecifics are often devoid of such benefits. The benefits associated with a high social status provide a strong incentive to become dominant. Importantly, motivational- and reward-related processes are regulated, to a large extent, by the mesolimbic system. Consequently, several studies point to a key role for the mesolimbic system in social hierarchy formation. This review summarizes the growing body of literature that implicates the mesolimbic system, and associated neural circuits, on social hierarchies. In particular, we discuss the neurochemical and pharmacological studies that have highlighted the contributions of the mesolimbic system and associated circuits including dopamine signaling through the D1 or D2 receptors, GABAergic neurotransmission, the androgen receptor system, and mitochondria and bioenergetics. Given that low social status has been linked to the emergence of anxiety- and depressive-like disorders, a greater understanding of the neurochemistry underlying social dominance could be of tremendous benefit for the development of pharmacological treatments to dysfunctions in social behaviors. This article is part of the Special Issue entitled 'The neuropharmacology of social behavior: from bench to bedside'.


Subject(s)
Dopamine Agents/pharmacology , Hierarchy, Social , Limbic System/physiology , Nerve Net/physiology , Ventral Tegmental Area/physiology , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Humans , Limbic System/drug effects , Nerve Net/drug effects , Neuropharmacology , Ventral Tegmental Area/drug effects
5.
Mol Psychiatry ; 23(3): 569-578, 2018 03.
Article in English | MEDLINE | ID: mdl-28727688

ABSTRACT

Benzodiazepines can ameliorate social disturbances and increase social competition, particularly in high-anxious individuals. However, the neural circuits and mechanisms underlying benzodiazepines' effects in social competition are not understood. Converging evidence points to the mesolimbic system as a potential site of action for at least some benzodiazepine-mediated effects. Furthermore, mitochondrial function in the nucleus accumbens (NAc) has been causally implicated in the link between anxiety and social competitiveness. Here, we show that diazepam facilitates social dominance, ameliorating both the competitive disadvantage and low NAc mitochondrial function displayed by high-anxious rats, and identify the ventral tegmental area (VTA) as a key site of action for direct diazepam effects. We also show that intra-VTA diazepam infusion increases accumbal dopamine and DOPAC, as well as activity of dopamine D1- but not D2-containing cells. In addition, intra-NAc infusion of a D1-, but not D2, receptor agonist facilitates social dominance and mitochondrial respiration. Conversely, intra-VTA diazepam actions on social dominance and NAc mitochondrial respiration are blocked by pharmacological NAc micro-infusion of a mitochondrial complex I inhibitor or an antagonist of D1 receptors. Our data support the view that diazepam disinhibits VTA dopaminergic neurons, leading to the release of dopamine into the NAc where activation of D1-signaling transiently facilitates mitochondrial function, that is, increased respiration and enhanced ATP levels, which ultimately enhances social competitive behavior. Therefore, our findings critically involve the mesolimbic system in the facilitating effects of diazepam on social competition and highlight mitochondrial function as a potential therapeutic target for anxiety-related social dysfunctions.


Subject(s)
Anxiety/drug therapy , Diazepam/pharmacology , Ventral Tegmental Area/drug effects , Animals , Benzazepines/pharmacology , Brain/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Mitochondria/metabolism , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Receptors, Dopamine D1/metabolism , Social Dominance
6.
Eur J Neurosci ; 45(7): 877-885, 2017 04.
Article in English | MEDLINE | ID: mdl-27606489

ABSTRACT

Decision-making processes can be modulated by stress, and the time elapsed from stress induction seems to be a crucial factor in determining the direction of the effects. Although current approaches consider the first post-stress hour a uniform period, the dynamic pattern of activation of the physiological stress systems (i.e., the sympathetic nervous system and hypothalamic-pituitary-adrenal axis) suggests that its neurobehavioural impact might be heterogeneous. Here, we evaluate economic risk preferences on the gain domain (i.e., risk aversion) at three time points following exposure to psychosocial stress (immediately after, and 20 and 45 min from onset). Using lottery games, we examine decisions at both the individual and social levels. We find that risk aversion shows a time-dependent change across the first post-stress hour, evolving from less risk aversion shortly after stress to more risk averse behaviour at the last testing time. When risk implied an antisocial outcome to a third party, stressed individuals showed less regard for this person in their decisions. Participants' cortisol levels explained their behaviour in the risk, but not the antisocial, game. Our findings reveal differential stress effects in self- and other-regarding decision-making and highlight the multidimensional nature of the immediate aftermath of stress for cognition.


Subject(s)
Reaction Time , Risk-Taking , Social Behavior , Stress, Psychological/physiopathology , Decision Making , Female , Games, Experimental , Humans , Hydrocortisone/blood , Male , Reward , Stress, Psychological/blood , Young Adult
7.
Cell Death Dis ; 7: e2237, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27228352

ABSTRACT

Friedreich's ataxia (FRDA) is an inherited neurodegenerative disease. The mutation consists of a GAA repeat expansion within the FXN gene, which downregulates frataxin, leading to abnormal mitochondrial iron accumulation, which may in turn cause changes in mitochondrial function. Although, many studies of FRDA patients and mouse models have been conducted in the past two decades, the role of frataxin in mitochondrial pathophysiology remains elusive. Are the mitochondrial abnormalities only a side effect of the increased accumulation of reactive iron, generating oxidative stress? Or does the progressive lack of iron-sulphur clusters (ISCs), induced by reduced frataxin, cause an inhibition of the electron transport chain complexes (CI, II and III) leading to reactive oxygen species escaping from oxidative phosphorylation reactions? To answer these crucial questions, we have characterised the mitochondrial pathophysiology of a group of disease-relevant and readily accessible neurons, cerebellar granule cells, from a validated FRDA mouse model. By using live cell imaging and biochemical techniques we were able to demonstrate that mitochondria are deregulated in neurons from the YG8R FRDA mouse model, causing a decrease in mitochondrial membrane potential (▵Ψm) due to an inhibition of Complex I, which is partially compensated by an overactivation of Complex II. This complex activity imbalance leads to ROS generation in both mitochondrial matrix and cytosol, which results in glutathione depletion and increased lipid peroxidation. Preventing this increase in lipid peroxidation, in neurons, protects against in cell death. This work describes the pathophysiological properties of the mitochondria in neurons from a FRDA mouse model and shows that lipid peroxidation could be an important target for novel therapeutic strategies in FRDA, which still lacks a cure.


Subject(s)
Iron-Binding Proteins/genetics , Lipid Peroxidation/genetics , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Neurons/metabolism , Animals , Cerebellum/metabolism , Cerebellum/pathology , Disease Models, Animal , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Gene Expression Regulation , Glutathione/metabolism , Humans , Iron/metabolism , Iron-Binding Proteins/metabolism , Mice , Mitochondria/pathology , Mutation , Neurons/pathology , Oxidative Stress , Primary Cell Culture , Reactive Oxygen Species/metabolism , Signal Transduction , Frataxin
8.
Eur Neuropsychopharmacol ; 26(2): 357-367, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26776368

ABSTRACT

Early-life stress and biological predispositions are linked to mood and personality disorders related to aggressive behavior. We previously showed that exposure to peripubertal stress leads to increased anxiety-like behaviors and aggression against males and females, as well as increased aggression against females in their male offspring. Here, we investigated whether paternal (pS) and individual (iS) exposure to peripubertal stress may exert additive effects on the long-term programming of anxiety-like and aggressive behaviors in rats. Given the key role of the lateral septum (LS) in the regulation of anxiety and aggressive behaviors and the hypothesized alterations in balance between neural excitation and inhibition in aggression-related disorders, markers for these processes were examined in the LS. Peripubertal stress was applied both in naïve male rats and in the offspring of peripubertally stressed males, and anxiety-like and aggressive behaviors were assessed at adulthood. Proton magnetic resonance spectroscopy at 6-months, and post-mortem analysis of glutamic acid decarboxylase 67 (GAD67) at 12-months were conducted in LS. We confirmed that aggressive behavior was increased by pS and iS, while only iS increased anxiety-like behavior. Individual stress led to reduced GABA, confirmed by reduced GAD67 immunolabelling, and increased glutamate, N-acetyl-aspartate, phosphocholine and creatine; while pS specifically led to reduced phosphocreatine. pS and iS do not interact and exert a differential impact on the analyzed aspects of brain function and anxiety-like behaviors. These data support the view that early-life stress can affect the behavioral and neurodevelopmental trajectories of individuals and their offspring, which may involve different neurobiological mechanisms.


Subject(s)
Aggression/physiology , Anxiety Disorders/etiology , Glutamate Decarboxylase/metabolism , Septal Nuclei/metabolism , Stress, Psychological , Age Factors , Animals , Animals, Newborn , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Creatine/metabolism , Disease Models, Animal , Female , Glutamic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Maze Learning/physiology , Phosphorylcholine/metabolism , Rats , Rats, Wistar , Septal Nuclei/diagnostic imaging , Stress, Psychological/complications , Stress, Psychological/diagnostic imaging , Stress, Psychological/psychology , Tritium/pharmacokinetics
9.
Neuroscience ; 311: 508-18, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26548415

ABSTRACT

Infancy is a critical period for brain development. Emerging evidence indicates that stress experienced during that period can have long-term programming effects on the brain and behavior. However, whether different time periods represent different vulnerabilities to the programming of different neurobehavioral domains is not yet known. Disrupted maternal care is known to interfere with neurodevelopmental processes and may lead to the manifestation of behavioral abnormalities in adulthood. Mouse dams confronted with insufficient bedding/nesting material have been shown to provide fragmented maternal care to their offspring. Here, we compared the impact of this model of early-life stress (ELS) during different developmental periods comprising either postnatal days (PNDs) 2-9 (ELS-early) or PND 10-17 (ELS-late) on behavior and hippocampal cell adhesion molecules in male mice in adulthood. ELS-early treatment caused a permanent reduction in bodyweight, whereas this reduction only occurred transiently during juvenility in ELS-late mice. Anxiety was only affected in ELS-late mice, while cognition and sociability were equally impaired in both ELS-treated groups. We analyzed hippocampal gene expression of the γ2 subunit of the GABAa receptor (Gabrg2) and of genes encoding cell adhesion molecules. Gabrg2 expression was increased in the ventral hippocampus in ELS-late-treated animals and was correlated with anxiety-like behavior in the open-field (OF) test. ELS-early-treated animals exhibited an increase in nectin-1 expression in the dorsal hippocampus, and this increase was associated with the social deficits seen in these animals. Our findings highlight the relevance of developmental age on stress-induced long-term behavioral alterations. They also suggest potential links between early stress-induced alterations in hippocampal Gabrg2 expression and the developmental programming of anxiety and between changes in hippocampal nectin-1 expression and stress-induced social impairments.


Subject(s)
Hippocampus/growth & development , Hippocampus/physiopathology , Neuronal Plasticity/physiology , Stress, Psychological/physiopathology , Animals , Animals, Newborn , Anxiety Disorders/etiology , Anxiety Disorders/physiopathology , Cell Adhesion Molecules/metabolism , Cognition/physiology , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Critical Period, Psychological , Disease Models, Animal , Housing, Animal , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Nectins , Receptors, GABA-A/metabolism , Social Behavior , Stress, Psychological/complications
10.
Gynecol Oncol ; 136(1): 87-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448454

ABSTRACT

OBJECTIVES: Few studies have assessed the risk and impact of lymphedema among women treated for endometrial cancer. We aimed to quantify cumulative incidence of, and risk factors for developing lymphedema following treatment for endometrial cancer and estimate absolute risk for individuals. Further, we report unmet needs for help with lymphedema-specific issues. METHODS: Women treated for endometrial cancer (n = 1243) were followed-up 3-5 years after diagnosis; a subset of 643 completed a follow-up survey that asked about lymphedema and lymphedema-related support needs. We identified a diagnosis of secondary lymphedema from medical records or self-report. Multivariable logistic regression was used to evaluate risk factors and estimates. RESULTS: Overall, 13% of women developed lymphedema. Risk varied markedly with the number of lymph nodes removed and, to a lesser extent, receipt of adjuvant radiation or chemotherapy treatment, and use of nonsteroidal anti-inflammatory drugs (pre-diagnosis). The absolute risk of developing lymphedema was >50% for women with 15+ nodes removed and 2-3 additional risk factors, 30-41% for those with 15+ nodes removed plus 0-1 risk factors or 6-14 nodes removed plus 3 risk factors, but ≤ 8% for women with no nodes removed or 1-5 nodes but no additional risk factors. Over half (55%) of those who developed lymphedema reported unmet need(s), particularly with lymphedema-related costs and pain. CONCLUSION: Lymphedema is common; experienced by one in eight women following endometrial cancer. Women who have undergone lymphadenectomy have very high risks of lymphedema and should be informed how to self-monitor for symptoms. Affected women need greater levels of support.


Subject(s)
Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/therapy , Lymphedema/epidemiology , Lymphedema/therapy , Palliative Care/methods , Aged , Australia/epidemiology , Endometrial Neoplasms/pathology , Female , Humans , Incidence , Leg , Logistic Models , Middle Aged , Needs Assessment , Risk Factors
11.
Transl Psychiatry ; 4: e410, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25004390

ABSTRACT

Early-life stress is a critical risk factor for developing psychopathological alterations later in life. This early adverse environment has been modeled in rats by exposure to stress during the peripubertal period-that is, corresponding to childhood and puberty-and has been shown to lead to increased emotionality, decreased sociability and pathological aggression. The amygdala, particularly its central nucleus (CeA), is hyperactivated in this model, consistent with evidence implicating this nucleus in the regulation of social and aggressive behaviors. Here, we investigated potential changes in the gene expression of molecular markers of excitatory and inhibitory neurotransmission in the CeA. We found that peripubertal stress led to an increase in the expression of mRNA encoding NR1 (the obligatory subunit of the N-methyl D-aspartate (NMDA) receptor) but to a reduction in the level of mRNA encoding glutamic acid decarboxylase 67 (GAD67), an enzyme that is critically involved in the activity-dependent synthesis of GABA, and to an increase in the vesicular glutamate transporter 1 (VGLUT1)/vesicular GABA transporter (VGAT) ratio in the CeA. These molecular alterations were present in addition to increased novelty reactivity, sociability deficits and increased aggression. Our results also showed that the full extent of the peripubertal protocol was required for the observed behavioral and neurobiological effects because exposure during only the childhood/prepubertal period (Juvenile Stress) or the male pubertal period (Puberty Stress) was insufficient to elicit the same effects. These findings highlight peripuberty as a period in which stress can lead to long-term programming of the genes involved in excitatory and inhibitory neurotransmission in the CeA.


Subject(s)
Behavior, Animal/physiology , Central Amygdaloid Nucleus/metabolism , Gene Expression/genetics , Sexual Maturation , Stress, Psychological/metabolism , Age Factors , Animals , Male , Random Allocation , Rats , Rats, Wistar , Stress, Psychological/physiopathology
12.
J Neuroendocrinol ; 26(10): 724-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25059307

ABSTRACT

We review the impact of early adversities on the development of violence and antisocial behaviour in humans, and present three aetiological animal models of escalated rodent aggression, each disentangling the consequences of one particular adverse early-life factor. A review of the human data, as well as those obtained with the animal models of repeated maternal separation, post-weaning social isolation and peripubertal stress, clearly shows that adverse developmental conditions strongly affect aggressive behaviour displayed in adulthood, the emotional responses to social challenges and the neuronal mechanisms activated by conflict. Although similarities between models are evident, important differences were also noted, demonstrating that the behavioural, emotional and neuronal consequences of early adversities are to a large extent dependent on aetiological factors. These findings support recent theories on human aggression, which suggest that particular developmental trajectories lead to specific forms of aggressive behaviour and brain dysfunctions. However, dissecting the roles of particular aetiological factors in humans is difficult because these occur in various combinations; in addition, the neuroscientific tools employed in humans still lack the depth of analysis of those used in animal research. We suggest that the analytical approach of the rodent models presented here may be successfully used to complement human findings and to develop integrative models of the complex relationship between early adversity, brain development and aggressive behaviour.


Subject(s)
Aggression , Behavior, Animal , Social Behavior , Animals , Female , Humans , Male , Models, Animal , Neurons/cytology , Social Isolation
13.
J Pediatr ; 165(1): 117-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24679609

ABSTRACT

OBJECTIVE: To compare symptoms of obstructive sleep apnea (OSA) and polysomnography (PSG) results in children with Down syndrome and typically developing children. STUDY DESIGN: A total of 49 children with Down syndrome referred for PSG between 2008 and 2012 were matched with typically developing children of the same sex, age, and OSA severity who had undergone PSG in the same year. A parent completed a sleep symptom questionnaire for each child. Sleep quality and measures of gas exchange were compared between the matched groups. RESULTS: The 98 children (46 females, 52 males) had mean age of 6.2 years (range, 0.3-16.9 years). Fourteen children had primary snoring, and 34 had OSA (9 mild, 7 moderate, and 19 severe). Children with Down syndrome had more severe OSA compared with 278 typically developing children referred in 2012. Symptom scores were not different between the matched groups. Those with Down syndrome had a higher average pCO2 during sleep (P = .03) and worse McGill oximetry scores. CONCLUSION: Compared with closely matched typically developing children with OSA of comparable severity, children with Down syndrome had a similar symptom profile and slightly worse gas exchange. Referred children with Down syndrome had more severe OSA than referred typically developing children, suggesting a relative reluctance by parents or doctors to investigate symptoms of OSA in children with Down syndrome. These findings highlight the need for formal screening tools for OSA in children with Down syndrome to improve detection of the condition in this high-risk group.


Subject(s)
Down Syndrome/complications , Mass Screening/methods , Sleep Apnea, Obstructive/diagnosis , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Polysomnography , Sleep Apnea, Obstructive/epidemiology , Surveys and Questionnaires
14.
Front Behav Neurosci ; 7: 35, 2013.
Article in English | MEDLINE | ID: mdl-23641204

ABSTRACT

Exposure to violence is traumatic and an important source of mental health disturbance, yet the factors associated with victimization remain incompletely understood. The aim of the present study was to investigate factors related to vulnerability to depression-like behaviors in females. An animal model of intimate partner violence, which was previously shown to produce long-lasting behavioral effects in females as a result of male partner aggression, was used. The associations among the degree of partner aggression, the long-term consequences on depressive-like behavior, and the impact of the anxious temperament of the female were examined. In a separate group, pre-selected neural markers were evaluated in the amygdala and the lateral septum of females. Expression was examined by analyses of targeted candidate genes, serotonin transporter (slc6a4), vasopressin receptor 1a, (avpr1a), and oxytocin receptor (oxtr). Structural equation modeling revealed that the female's temperament moderated depressive-like behavior that was induced by cohabitation aggression from the male partner. More specifically, increased floating in the forced swim test following male aggression was most apparent in females exhibiting more anxiety-like behavior (i.e., less open arm exploration in an elevated plus-maze) prior to the cohabitation. Aggression reduced slc6a4 levels in the lateral septum. However, the interaction between partner aggression and the anxious temperament of the female affected the expression of avpr1a in the amygdala. Although, aggression reduced levels of this marker in females with high anxiety, no such pattern was observed in females with low anxiety. These results identify important characteristics in females that moderate the impact of male aggression. Furthermore, these results provide potential therapeutic targets of interest in the amygdala and the lateral septum to help improve post-stress behavioral pathology and increase resilience to social adversity.

15.
Transl Psychiatry ; 3: e216, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23321813

ABSTRACT

Although adverse early life experiences have been found to increase lifetime risk to develop violent behaviors, the neurobiological mechanisms underlying these long-term effects remain unclear. We present a novel animal model for pathological aggression induced by peripubertal exposure to stress with face, construct and predictive validity. We show that male rats submitted to fear-induction experiences during the peripubertal period exhibit high and sustained rates of increased aggression at adulthood, even against unthreatening individuals, and increased testosterone/corticosterone ratio. They also exhibit hyperactivity in the amygdala under both basal conditions (evaluated by 2-deoxy-glucose autoradiography) and after a resident-intruder (RI) test (evaluated by c-Fos immunohistochemistry), and hypoactivation of the medial orbitofrontal (MO) cortex after the social challenge. Alterations in the connectivity between the orbitofrontal cortex and the amygdala were linked to the aggressive phenotype. Increased and sustained expression levels of the monoamine oxidase A (MAOA) gene were found in the prefrontal cortex but not in the amygdala of peripubertally stressed animals. They were accompanied by increased activatory acetylation of histone H3, but not H4, at the promoter of the MAOA gene. Treatment with an MAOA inhibitor during adulthood reversed the peripuberty stress-induced antisocial behaviors. Beyond the characterization and validation of the model, we present novel data highlighting changes in the serotonergic system in the prefrontal cortex-and pointing at epigenetic control of the MAOA gene-in the establishment of the link between peripubertal stress and later pathological aggression. Our data emphasize the impact of biological factors triggered by peripubertal adverse experiences on the emergence of violent behaviors.


Subject(s)
Aggression/psychology , Amygdala/physiopathology , Fear/psychology , Monoamine Oxidase/genetics , Prefrontal Cortex/physiopathology , Stress, Psychological/genetics , Aggression/physiology , Analysis of Variance , Animals , Clorgyline/therapeutic use , Conditioning, Psychological/physiology , Disease Models, Animal , Fear/physiology , Gene Expression , Immunohistochemistry , Male , Monoamine Oxidase/drug effects , Monoamine Oxidase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-fos/analysis , Rats , Sexual Maturation/physiology , Stress, Psychological/complications , Stress, Psychological/physiopathology
16.
Transl Psychiatry ; 2: e106, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22832906

ABSTRACT

Intimate partner violence is a ubiquitous and devastating phenomenon for which effective interventions and a clear etiological understanding are still lacking. A major risk factor for violence perpetration is childhood exposure to violence, prompting the proposal that social learning is a major contributor to the transgenerational transmission of violence. Using an animal model devoid of human cultural factors, we showed that male rats became highly aggressive against their female partners as adults after exposure to non-social stressful experiences in their youth. Their offspring also showed increased aggression toward females in the absence of postnatal father-offspring interaction or any other exposure to violence. Both the females that cohabited with the stressed males and those that cohabited with their male offspring showed behavioral (including anxiety- and depression-like behaviors), physiological (decreased body weight and basal corticosterone levels) and neurobiological symptoms (increased activity in dorsal raphe serotonergic neurons in response to an unfamiliar male) resembling the alterations described in abused and depressed women. With the caution required when translating animal work to humans, our findings extend current psychosocial explanations of the transgenerational transmission of intimate partner violence by strongly suggesting an important role for biological factors.


Subject(s)
Body Weight/physiology , Child Abuse/psychology , Corticosterone/blood , Disease Models, Animal , Intergenerational Relations , Raphe Nuclei/physiopathology , Serotonin/physiology , Spouse Abuse/psychology , Violence/psychology , Adult , Age Factors , Aggression/physiology , Aggression/psychology , Agonistic Behavior/physiology , Animals , Anxiety/physiopathology , Anxiety/psychology , Child , Depression/physiopathology , Depression/psychology , Epigenesis, Genetic/physiology , Fear/physiology , Female , Gender Identity , Gene-Environment Interaction , Humans , Life Change Events , Male , Neurons/physiology , Rats , Rats, Wistar , Risk Factors , Stress, Psychological/complications , Stress, Psychological/psychology
17.
BMC Cancer ; 11: 389, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21899778

ABSTRACT

BACKGROUND: Exercise interventions during adjuvant cancer therapy have been shown to increase functional capacity, relieve fatigue and distress and may assist rates of chemotherapy completion. These studies have been limited to breast, gastric and mixed cancer groups and it is not yet known if a similar intervention is even feasible among women with ovarian cancer. We aimed to assess safety, feasibility and potential effect of a walking intervention in women undergoing chemotherapy for ovarian cancer. METHODS: Women newly diagnosed with ovarian cancer were recruited to participate in an individualised walking intervention throughout chemotherapy and were assessed pre- and post-intervention. Feasibility measures included session adherence, compliance with exercise physiologist prescribed walking targets and self-reported program acceptability. Changes in objective physical functioning (6-minute walk test), self-reported distress (Hospital Anxiety and Depression Scale), symptoms (Memorial Symptom Assessment Scale - Physical) and quality of life (Functional Assessment of Cancer Therapy - Ovarian) were calculated, and chemotherapy completion and adverse intervention effects recorded. RESULTS: Seventeen women were enrolled (63% recruitment rate). Mean age was 60 years (SD = 8 years), 88% were diagnosed with FIGO stage III or IV disease, 14 women underwent adjuvant and three neo-adjuvant chemotherapy. On average, women adhered to > 80% of their intervention sessions and complied with 76% of their walking targets, with the majority walking four days a week at moderate intensity for 30 minutes per session. Meaningful improvements were found in physical functioning, physical symptoms, physical well-being and ovarian cancer-specific quality of life. Most women (76%) completed ≥85% of their planned chemotherapy dose. There were no withdrawals or serious adverse events and all women reported the program as being helpful. CONCLUSIONS: These positive preliminary results suggest that this walking intervention for women receiving chemotherapy for ovarian cancer is safe, feasible and acceptable and could be used in development of future work. TRIAL REGISTRATION: ACTRN12609000252213.


Subject(s)
Exercise Therapy , Ovarian Neoplasms/rehabilitation , Walking , Adult , Aged , Antineoplastic Agents/therapeutic use , Exercise Therapy/adverse effects , Female , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Pilot Projects , Treatment Outcome
18.
Mol Psychiatry ; 16(5): 533-47, 2011 May.
Article in English | MEDLINE | ID: mdl-20177408

ABSTRACT

Intensive research is devoted to unravel the neurobiological mechanisms mediating adult hippocampal neurogenesis, its regulation by antidepressants, and its behavioral consequences. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that is expressed in the CNS, where its function is unknown. Here, we show, for the first time, the relevance of MIF expression for adult hippocampal neurogenesis. We identify MIF expression in neurogenic cells (in stem cells, cells undergoing proliferation, and in newly proliferated cells undergoing maturation) in the subgranular zone of the rodent dentate gyrus. A causal function for MIF in cell proliferation was shown using genetic (MIF gene deletion) and pharmacological (treatment with the MIF antagonist Iso-1) approaches. Behaviorally, genetic deletion of MIF resulted in increased anxiety- and depression-like behaviors, as well as of impaired hippocampus-dependent memory. Together, our studies provide evidence supporting a pivotal function for MIF in both basal and antidepressant-stimulated adult hippocampal cell proliferation. Moreover, loss of MIF results in a behavioral phenotype that, to a large extent, corresponds with alterations predicted to arise from reduced hippocampal neurogenesis. These findings underscore MIF as a potentially relevant molecular target for the development of treatments linked to deficits in neurogenesis, as well as to problems related to anxiety, depression, and cognition.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Anxiety/pathology , Cell Proliferation/drug effects , Depression/pathology , Fluoxetine/pharmacology , Hippocampus/drug effects , Macrophage Migration-Inhibitory Factors/metabolism , Memory Disorders/pathology , Acoustic Stimulation/adverse effects , Animals , Antidepressive Agents, Second-Generation/therapeutic use , Anxiety/drug therapy , Anxiety/genetics , Bromodeoxyuridine/metabolism , C-Reactive Protein/drug effects , C-Reactive Protein/metabolism , Conditioning, Psychological/drug effects , Corticosterone/blood , Corticosterone/therapeutic use , Depression/drug therapy , Depression/genetics , Disease Models, Animal , Fear , Fluoxetine/therapeutic use , Hippocampus/pathology , Macrophage Migration-Inhibitory Factors/deficiency , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/genetics , Mice , Mice, Knockout , Microscopy, Confocal/methods , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Rats , Rats, Wistar , Receptors, Steroid/metabolism , Spatial Behavior/drug effects
19.
Genes Brain Behav ; 9(8): 958-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20659171

ABSTRACT

Previous studies using neuronal cell adhesion molecule (NCAM) -/- knockout (KO) mice provided evidence for a role of NCAMs in social behaviors. However, polysialic acid (PSA), the most important post-translational modification of NCAM, was also absent in these mice, which makes it difficult to distinguish between the specific involvement of either PSA or NCAM in social interactions. To address this issue, we assessed two lines of mice deficient for one of the two sialyltransferase enzymes required for the polysialylation of NCAM, sialyltransferase-X (St8SiaII or STX) and polysialyltransferase (ST8SiaIV or PST), in a series of tests for social behaviors. Results showed that PST KO mice display a decreased motivation in social interaction. This deficit can be partly explained by olfactory deficits and was associated with a clear decrease in PSA-NCAM expression in all brain regions analyzed (amygdala, septum, bed nucleus of the stria terminalis and frontal cortices). STX KO mice displayed both a decreased social motivation and an increased aggressive behavior that cannot be explained by olfactory deficits. This finding might be related to the reduced anxiety-like behavior, increased locomotion and stress-induced corticosterone secretion observed in these mice. Moreover, STX KO mice showed mild increase of PSA-NCAM expression in the lateral septum and the orbitofrontal cortex. Altogether, these findings support a role for PSA-NCAM in the regulation of social behaviors ranging from a lack of social motivation to aggression. They also underscore STX KO mice as an interesting animal model that combines a behavioral profile of violence and hyperactivity with reduced anxiety-like behavior.


Subject(s)
Aggression/physiology , Anxiety/genetics , Brain/enzymology , Sialyltransferases/genetics , Social Behavior , Animals , Anxiety/metabolism , Behavior, Animal/physiology , Corticosterone/blood , Exploratory Behavior/physiology , Gene Expression Regulation , Male , Matched-Pair Analysis , Mice , Mice, Knockout , Motivation/genetics , Motivation/physiology , Sialyltransferases/metabolism , Smell/genetics , Smell/physiology
20.
Genes Brain Behav ; 9(4): 353-64, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20059553

ABSTRACT

Chronic stress in rodents was shown to induce structural shrinkage and functional alterations in the hippocampus that were linked to spatial memory impairments. Effects of chronic stress on the amygdala have been linked to a facilitation of fear conditioning. Although the underlying molecular mechanisms are still poorly understood, increasing evidence highlights the neural cell adhesion molecule (NCAM) as an important molecular mediator of stress-induced structural and functional alterations. In this study, we investigated whether altered NCAM expression levels in the amygdala might be related to stress-induced enhancement of auditory fear conditioning and anxiety-like behavior. In adult C57BL/6J wild-type mice, chronic unpredictable stress resulted in an isoform-specific increase of NCAM expression (NCAM-140 and NCAM-180) in the amygdala, as well as enhanced auditory fear conditioning and anxiety-like behavior. Strikingly, forebrain-specific conditional NCAM-deficient mice (NCAM-floxed mice that express the cre-recombinase under the control of the promoter of the alpha-subunit of the calcium-calmodulin-dependent protein kinase II), whose amygdala NCAM expression levels are reduced, displayed impaired auditory fear conditioning which was not altered following chronic stress exposure. Likewise, chronic stress in these conditional NCAM-deficient mice did not modify NCAM expression levels in the amygdala or hippocampus, while they showed enhanced anxiety-like behavior, questioning the involvement of NCAM in this type of behavior. Together, our results strongly support the involvement of NCAM in the amygdala in the consolidation of auditory fear conditioning and highlight increased NCAM expression in the amygdala among the mechanisms whereby stress facilitates fear conditioning processes.


Subject(s)
Amygdala/metabolism , Conditioning, Psychological/physiology , Fear/psychology , Neural Cell Adhesion Molecules/physiology , Stress, Psychological/genetics , Stress, Psychological/metabolism , Acoustic Stimulation , Amygdala/physiopathology , Animals , Anxiety/genetics , Anxiety/metabolism , Auditory Perception/genetics , Brain Chemistry/genetics , Chronic Disease , Down-Regulation/genetics , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics
SELECTION OF CITATIONS
SEARCH DETAIL