Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Eur J Neurosci ; 60(1): 3544-3556, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38695253

RESUMO

Empathetic relationships and the social transference of behaviours have been shown to occur in humans, and more recently through the development of rodent models, where both fear and pain phenotypes develop in observer animals. Clinically, observing traumatic events can induce 'trauma and stressor-related disorders' as defined in the DSM 5. These disorders are often comorbid with pain and gastrointestinal disturbances; however, our understanding of how gastrointestinal - or visceral - pain can be vicariously transmitted is lacking. Visceral pain originates from the internal organs, and despite its widespread prevalence, remains poorly understood. We established an observation paradigm to assess the impact of witnessing visceral pain. We utilised colorectal distension (CRD) to induce visceral pain behaviours in a stimulus rodent while the observer rodent observed. Twenty four hours post-observation, the observer rodent's visceral sensitivity was assessed using CRD. The observer rodents were found to have significant hyperalgesia as determined by lower visceral pain threshold and higher number of total pain behaviours compared with controls. The behaviours of the observer animals during the observation were found to be correlated with the behaviours of the stimulus animal employed. We found that observer animals had hypoactivity of the hypothalamic-pituitary-adrenal (HPA) axis, highlighted by reduced corticosterone at 90 minutes post-CRD. Using c-Fos immunohistochemistry we showed that observer animals also had increased activation of the anterior cingulate cortex, and decreased activation of the paraventricular nucleus, compared with controls. These results suggest that witnessing another animal in pain produces a behavioural phenotype and impacts the brain-gut axis.


Assuntos
Modelos Animais de Doenças , Estresse Psicológico , Dor Visceral , Animais , Masculino , Dor Visceral/fisiopatologia , Dor Visceral/psicologia , Ratos , Estresse Psicológico/fisiopatologia , Ratos Sprague-Dawley , Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipotálamo-Hipofisário/metabolismo , Hiperalgesia/fisiopatologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Limiar da Dor/fisiologia
2.
Brain Behav Immun ; 120: 315-326, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38852762

RESUMO

Social anxiety disorder is a common psychiatric condition that severely affects quality of life of individuals and is a significant societal burden. Although many risk factors for social anxiety exist, it is currently unknown how social fear sensitivity manifests biologically. Furthermore, since some individuals are resilient and others are susceptible to social fear, it is important to interrogate the mechanisms underpinning individual response to social fear situations. The microbiota-gut-brain axis has been associated with social behaviour, has recently been linked with social anxiety disorder, and may serve as a therapeutic target for modulation. Here, we assess the potential of this axis to be linked with social fear extinction processes in a murine model of social anxiety disorder. To this end, we correlated differential social fear responses with microbiota composition, central gene expression, and immune responses. Our data provide evidence that microbiota variability is strongly correlated with alterations in social fear behaviour. Moreover, we identified altered gene candidates by amygdalar transcriptomics that are linked with social fear sensitivity. These include genes associated with social behaviour (Armcx1, Fam69b, Kcnj9, Maoa, Serinc5, Slc6a17, Spata2, and Syngr1), inflammation and immunity (Cars, Ckmt1, Klf5, Maoa, Map3k12, Pex5, Serinc5, Sidt1, Spata2), and microbe-host interaction (Klf5, Map3k12, Serinc5, Sidt1). Together, these data provide further evidence for a role of the microbiota-gut-brain axis in social fear responses.


Assuntos
Eixo Encéfalo-Intestino , Extinção Psicológica , Medo , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Animais , Medo/fisiologia , Camundongos , Microbioma Gastrointestinal/fisiologia , Extinção Psicológica/fisiologia , Masculino , Eixo Encéfalo-Intestino/fisiologia , Encéfalo/metabolismo , Comportamento Social , Fobia Social/metabolismo , Fobia Social/psicologia , Tonsila do Cerebelo/metabolismo , Modelos Animais de Doenças , Ansiedade/metabolismo
3.
Mol Psychiatry ; 28(11): 4729-4741, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37644175

RESUMO

Psychological loss is a common experience that erodes well-being and negatively impacts quality of life. The molecular underpinnings of loss are poorly understood. Here, we investigate the mechanisms of loss using an environmental enrichment removal (ER) paradigm in male rats. The basolateral amygdala (BLA) was identified as a region of interest, demonstrating differential Fos responsivity to ER and having an established role in stress processing and adaptation. A comprehensive multi-omics investigation of the BLA, spanning multiple cohorts, platforms, and analyses, revealed alterations in microglia and the extracellular matrix (ECM). Follow-up studies indicated that ER decreased microglia size, complexity, and phagocytosis, suggesting reduced immune surveillance. Loss also substantially increased ECM coverage, specifically targeting perineuronal nets surrounding parvalbumin interneurons, suggesting decreased plasticity and increased inhibition within the BLA following loss. Behavioral analyses suggest that these molecular effects are linked to impaired BLA salience evaluation, leading to a mismatch between stimulus and reaction intensity. These loss-like behaviors could be rescued by depleting BLA ECM during the removal period, helping us understand the mechanisms underlying loss and revealing novel molecular targets to ameliorate its impact.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Ratos , Animais , Masculino , Complexo Nuclear Basolateral da Amígdala/fisiologia , Neurobiologia , Qualidade de Vida , Interneurônios , Matriz Extracelular
4.
Brain Behav Immun ; 107: 385-396, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36400332

RESUMO

Mounting evidence suggests a link between gut microbiota abnormalities and post-traumatic stress disorder (PTSD). However, whether and how the gut microbiota influences PTSD susceptibility is poorly understood. Here using the arousal-based individual screening model, we provide evidence for pre-trauma and post-trauma gut microbiota alterations in susceptible mice exhibiting persistent PTSD-related phenotypes. A more in-depth analysis revealed an increased abundance of bacteria affecting brain processes including myelination, and brain systems like the dopaminergic neurotransmission. Because dopaminergic dysfunctions play a key role in the pathophysiological mechanisms subserving PTSD, we assessed whether these alterations in gut microbiota composition could be associated with abnormal levels of metabolites inducing dopaminergic dysfunctions. We found high levels of the l-tyrosine-derived metabolite p-cresol exclusively in the prefrontal cortex of susceptible mice. We further uncovered abnormal levels of dopamine and DOPAC, together with a detrimental increase of dopamine D3 receptor expression, exclusively in the prefrontal cortex of susceptible mice. Conversely, we observed either resilience mechanisms aimed at counteracting these p-cresol-induced dopaminergic dysfunctions or myelination-related resilience mechanisms only in the prefrontal cortex of resilient mice. These findings reveal that gut microbiota abnormalities foster trauma susceptibility and thus it may represent a promising target for therapeutic interventions.


Assuntos
Dopamina , Camundongos , Animais
5.
J Pharmacol Exp Ther ; 357(1): 84-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26787772

RESUMO

Chronic pain is a multifaceted and complex condition. Broadly classified into somatic, visceral, or neuropathic pain, it is poorly managed despite its prevalence. Current drugs used for the treatment of chronic pain are limited by tolerance with long-term use, abuse potential, and multiple adverse side effects. The persistent nature of pain suggests that epigenetic machinery may be a critical factor driving chronic pain. In this review, we discuss the latest insights into epigenetic processes, including DNA methylation, histone modifications, and microRNAs, and we describe their involvement in the pathophysiology of chronic pain and whether epigenetic modifications could be applied as future therapeutic targets for chronic pain. We provide evidence from experimental models and translational research in human tissue that have enhanced our understanding of epigenetic processes mediating nociception, and we then speculate on the potential future use of more specific and selective agents that target epigenetic mechanisms to attenuate pain.


Assuntos
Analgésicos/farmacologia , Analgésicos/uso terapêutico , Dor Crônica/tratamento farmacológico , Dor Crônica/genética , Epigênese Genética/efeitos dos fármacos , Manejo da Dor/métodos , Animais , Humanos , Neuralgia
6.
Brain Behav Immun ; 48: 165-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25866195

RESUMO

BACKGROUND: There is growing appreciation for the importance of bacteria in shaping brain development and behaviour. Adolescence and early adulthood are crucial developmental periods during which exposure to harmful environmental factors can have a permanent impact on brain function. Such environmental factors include perturbations of the gut bacteria that may affect gut-brain communication, altering the trajectory of brain development, and increasing vulnerability to psychiatric disorders. Here we assess the effects of gut bacterial depletion from weaning onwards on adult cognitive, social and emotional behaviours and markers of gut-brain axis dysfunction in mice. METHODS: Mice were treated with a combination of antibiotics from weaning onwards and effects on behaviours and potential gut-brain axis neuromodulators (tryptophan, monoamines, and neuropeptides) and BDNF expression were assessed in adulthood. RESULTS: Antibiotic-treatment depleted and restructured gut microbiota composition of caecal contents and decreased spleen weights in adulthood. Depletion of the gut microbiota from weaning onwards reduced anxiety, induced cognitive deficits, altered dynamics of the tryptophan metabolic pathway, and significantly reduced BDNF, oxytocin and vasopressin expression in the adult brain. CONCLUSIONS: Microbiota depletion from weaning onwards by means of chronic treatment with antibiotics in mice impacts on anxiety and cognitive behaviours as well as key neuromodulators of gut-brain communication in a manner that is similar to that reported in germ-free mice. This model may represent a more amenable alternative for germ-free mice in the assessment of microbiota modulation of behaviour. Finally, these data suggest that despite the presence of a normal gut microbiome in early postnatal life, reduced abundance and diversity of the gut microbiota from weaning influences adult behaviours and key neuromodulators of the microbiota-gut-brain axis suggesting that dysregulation of this axis in the post-weaning period may contribute to the pathogenesis of disorders associated with altered anxiety and cognition.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Animais , Antibacterianos/farmacologia , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Triptofano/metabolismo
7.
Adv Appl Microbiol ; 91: 1-62, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25911232

RESUMO

The realization that the microbiota-gut-brain axis plays a critical role in health and disease has emerged over the past decade. The brain-gut axis is a bidirectional communication system between the central nervous system (CNS) and the gastrointestinal tract. Regulation of the microbiota-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. The routes of this communication are not fully elucidated but include neural, humoral, immune, and metabolic pathways. A number of approaches have been used to interrogate this axis including the use of germ-free animals, probiotic agents, antibiotics, or animals exposed to pathogenic bacterial infections. Together, it is clear that the gut microbiota can be a key regulator of mood, cognition, pain, and obesity. Understanding microbiota-brain interactions is an exciting area of research which may contribute new insights into individual variations in cognition, personality, mood, sleep, and eating behavior, and how they contribute to a range of neuropsychiatric diseases ranging from affective disorders to autism and schizophrenia. Finally, the concept of psychobiotics, bacterial-based interventions with mental health benefit, is also emerging.


Assuntos
Encéfalo/microbiologia , Trato Gastrointestinal/microbiologia , Microbiota/fisiologia , Animais , Antibacterianos , Sistema Nervoso Central , Cognição , Mamíferos
8.
Mamm Genome ; 25(1-2): 49-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24281320

RESUMO

Bacterial colonisation of the gut plays a major role in postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Individually, these systems have been implicated in the neuropathology of many CNS disorders and collectively they form an important bidirectional pathway of communication between the microbiota and the brain in health and disease. Regulation of the microbiome-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. Moreover, there is now expanding evidence for the view that commensal organisms within the gut play a role in early programming and later responsivity of the stress system. Research has focused on how the microbiota communicates with the CNS and thereby influences brain function. The routes of this communication are not fully elucidated but include neural, humoral, immune and metabolic pathways. This view is underpinned by studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics which indicate a role for the gut microbiota in the regulation of mood, cognition, pain and obesity. Thus, the concept of a microbiome-brain-gut axis is emerging which suggests that modulation of the gut microflora may be a tractable strategy for developing novel therapeutics for complex stress-related CNS disorders where there is a huge unmet medical need.


Assuntos
Encéfalo/fisiologia , Trato Gastrointestinal/microbiologia , Microbiota , Animais , Comportamento , Humanos , Transtornos Mentais/etiologia , Transdução de Sinais , Estresse Fisiológico , Estresse Psicológico
9.
Adv Exp Med Biol ; 817: 373-403, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24997043

RESUMO

There is increasing evidence that host-microbe interactions play a key role in maintaining homeostasis. Alterations in gut microbial composition is associated with marked changes in behaviors relevant to mood, pain and cognition, establishing the critical importance of the bi-directional pathway of communication between the microbiota and the brain in health and disease. Dysfunction of the microbiome-brain-gut axis has been implicated in stress-related disorders such as depression, anxiety and irritable bowel syndrome and neurodevelopmental disorders such as autism. Bacterial colonization of the gut is central to postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Moreover, there is now expanding evidence for the view that enteric microbiota plays a role in early programming and later response to acute and chronic stress. This view is supported by studies in germ-free mice and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics. Although communication between gut microbiota and the CNS are not fully elucidated, neural, hormonal, immune and metabolic pathways have been suggested. Thus, the concept of a microbiome-brain-gut axis is emerging, suggesting microbiota-modulating strategies may be a tractable therapeutic approach for developing novel treatments for CNS disorders.


Assuntos
Comportamento , Encéfalo/fisiologia , Microbiota/fisiologia , Animais , Antibacterianos/farmacologia , Encefalopatias/terapia , Cognição , Humanos , Intestinos/microbiologia , Probióticos/farmacologia , Transdução de Sinais , Triptofano/metabolismo
10.
Biol Psychiatry Glob Open Sci ; 3(2): 274-282, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37124346

RESUMO

Background: Adolescent brains are sensitive to stressors. However, under certain circumstances, developmental stress can promote an adaptive phenotype, allowing individuals to cope better with adverse situations in adulthood, thereby contributing to resilience. Methods: Sprague Dawley rats (50 males, 48 females) were subjected to adolescent chronic variable stress (adol CVS) for 2 weeks at postnatal day 45. At postnatal day 85, a group was subjected to single prolonged stress (SPS). After a week, animals were evaluated in an auditory-cued fear conditioning paradigm, and neuronal recruitment during reinstatement was assessed by Fos expression. Patch clamp electrophysiology (17-35 cells/group) was performed in male rats to examine physiological changes associated with resilience. Results: Adol CVS blocked fear potentiation evoked by SPS. We observed that SPS impaired extinction (males) and enhanced reinstatement (both sexes) of the conditioned freezing response. Prior adol CVS prevented both effects. SPS effects were associated with a reduction of infralimbic (IL) cortex neuronal recruitment after reinstatement in males and increased engagement of the central amygdala in females, both also prevented by adol CVS, suggesting different neurocircuits involved in generating resilience between sexes. We explored the mechanism behind reduced IL recruitment in males by studying the intrinsic excitability of IL pyramidal neurons. SPS reduced excitability of IL neurons, and prior adol CVS prevented this effect. Conclusions: Our data indicate that adolescent stress can impart resilience to the effects of traumatic stress on neuroplasticity and behavior. Our data provide a mechanistic link behind developmental stress-induced behavioral resilience and prefrontal (IL) cortical excitability in males.

11.
J Psychopharmacol ; 37(11): 1051-1057, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37522187

RESUMO

Animal models are important in preclinical psychopharmacology to study mechanisms and potential treatments for psychiatric disorders. A working group of 14 volunteers, comprising an international team of researchers from academia and industry, convened in 2021 to discuss how to improve the translational relevance and interpretation of findings from animal models that are used in preclinical psychopharmacology. The following paper distils the outcomes of the working group's discussions into 10 key considerations for the planning and reporting of behavioural studies in animal models relevant to psychiatric disorders. These form the iTRIPP guidelines (Improving Translational Relevance In Preclinical Psychopharmacology). These guidelines reflect the key considerations that the group thinks will likely have substantial impact in terms of improving the translational relevance of behavioural studies in animal models that are used to study psychiatric disorders and their treatment. They are relevant to the research community when drafting and reviewing manuscripts, presentations and grant applications. The iTRIPP guidelines are intended to complement general recommendations for planning and reporting animal studies that have been published elsewhere, by enabling researchers to fully consider the most appropriate animal model for the research purpose and to interpret their findings appropriately. This in turn will increase the clinical benefit of such research and is therefore important not only for the scientific community but also for patients and the lay public.


Assuntos
Transtornos Mentais , Psicofarmacologia , Animais , Humanos , Transtornos Mentais/tratamento farmacológico , Modelos Animais de Doenças
12.
eNeuro ; 7(5)2020.
Artigo em Inglês | MEDLINE | ID: mdl-33055196

RESUMO

Hypofunction of the prefrontal cortex (PFC) contributes to stress-related neuropsychiatric illnesses. Mechanisms leading to prefrontal hypoactivity remain to be determined. Prior evidence suggests that chronic stress leads to an increase in activity of parvalbumin (PV) expressing GABAergic interneurons (INs) in the PFC. The purpose of the study was to determine whether reducing PV IN activity in the Infralimbic (IL) PFC would prevent stress-related phenotypes. We used a chemogenetic approach to inhibit IL PFC PV INs during stress. Mice were first tested in the tail suspension test (TST) to determine the impact of PV IN inhibition on behavioral responses to acute stress. The long-term impact of PV IN inhibition during a modified chronic variable stress (CVS) was tested in the forced swim test (FST). Acute PV IN inhibition reduced active (struggling) and increased passive coping behaviors (immobility) in the TST. In contrast, inhibition of PV INs during CVS increased active and reduced passive coping behaviors in the FST. Moreover, chronic inhibition of PV INs attenuated CVS-induced changes in Fos expression in the prelimbic cortex (PrL), basolateral amygdala (BLA), and ventrolateral periaqueductal gray (vlPAG) and also attenuated adrenal hypertrophy and body weight loss associated with chronic stress. Our results suggest differential roles of PV INs in acute versus chronic stress, indicative of distinct biological mechanisms underlying acute versus chronic stress responses. Our results also indicate a role for PV INs in driving chronic stress adaptation and support literature evidence suggesting cortical GABAergic INs as a therapeutic target in stress-related illnesses.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Interneurônios , Parvalbuminas , Estresse Fisiológico , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Córtex Cerebral/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo
13.
Psychoneuroendocrinology ; 112: 104490, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31786480

RESUMO

Adolescent animals are vulnerable to the effects of stress on brain development. We hypothesized that long-term effects of adolescent chronic stress are mediated by glucocorticoid receptor (GR) signaling. We used a specific GR modulator (CORT108297) to pharmacologically disrupt GR signaling in adolescent rats during exposure to chronic variable stress (CVS). Male and female rats received 30 mg/kg of drug during a 2-week CVS protocol starting at PND46. Emotional reactivity (open field) and coping behaviors (forced swim test (FST)) were then tested in adulthood, 5 weeks after the end of the CVS protocol. Blood samples were collected two days before FST and serial samples after the onset of the swim test to determine baseline and stress response levels of HPA hormones respectively. Our results support differential behavioral, physiological and stress circuit reactivity to adolescent chronic stress exposure in males and females, with variable involvement of GR signaling. In response to adolescent stress, males had heightened reactivity to novelty and exhibited marked reduction in neuronal excitation following swim stress in adulthood, whereas females developed a passive coping strategy in the FST and enhanced HPA axis stress reactivity. Only the latter effect was attenuated by treatment with the GR modulator C108297. In summary, our data suggest that adolescent stress differentially affects emotional behavior and circuit development in males and females, and that GR manipulation during stress can reverse at least some of these effects.


Assuntos
Adaptação Psicológica/fisiologia , Compostos Aza/farmacologia , Comportamento Animal/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Receptores de Glucocorticoides/fisiologia , Transdução de Sinais/fisiologia , Estresse Psicológico/fisiopatologia , Adaptação Psicológica/efeitos dos fármacos , Fatores Etários , Animais , Compostos Aza/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/administração & dosagem , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/efeitos dos fármacos , Fatores Sexuais , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/metabolismo
14.
Curr Biol ; 30(19): 3761-3774.e6, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32822606

RESUMO

Birth by Caesarean (C)-section impacts early gut microbiota colonization and is associated with an increased risk of developing immune and metabolic disorders. Moreover, alterations of the microbiome have been shown to affect neurodevelopmental trajectories. However, the long-term effects of C-section on neurobehavioral processes remain unknown. Here, we demonstrated that birth by C-section results in marked but transient changes in microbiome composition in the mouse, in particular, the abundance of Bifidobacterium spp. was depleted in early life. Mice born by C-section had enduring social, cognitive, and anxiety deficits in early life and adulthood. Interestingly, we found that these specific behavioral alterations induced by the mode of birth were also partially corrected by co-housing with vaginally born mice. Finally, we showed that supplementation from birth with a Bifidobacterium breve strain, or with a dietary prebiotic mixture that stimulates the growth of bifidobacteria, reverses selective behavioral alterations in C-section mice. Taken together, our data link the gut microbiota to behavioral alterations in C-section-born mice and suggest the possibility of developing adjunctive microbiota-targeted therapies that may help to avert long-term negative consequences on behavior associated with C-section birth mode.


Assuntos
Cesárea/efeitos adversos , Microbioma Gastrointestinal/fisiologia , Doenças do Sistema Nervoso/microbiologia , Animais , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Cesárea/psicologia , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Camundongos , Gravidez
15.
Biol Psychiatry ; 86(9): 669-681, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31326084

RESUMO

The medial prefrontal cortex (mPFC) receives information regarding stimuli and appropriately orchestrates neurophysiological, autonomic, and behavioral responses to stress. The cellular and neurochemical heterogeneity of the mPFC and its projections are key to fine-tuning of stress responses and adaptation. Output of the mPFC is mediated by glutamatergic pyramidal neurons whose activity is coordinated by an intricate network of interneurons. Excitatory/inhibitory (E/I) balance in the mPFC is critical for appropriate responsiveness to stress, and E/I imbalance occurs in numerous neuropsychiatric disorders that co-occur with chronic stress. Moreover, there is mounting data suggesting that chronic stress may precipitate E/I imbalance. This review will provide information regarding the cellular and anatomical makeup of the mPFC and discuss the impact of acute and chronic stress in adulthood and early life on interneuron function, with implications for E/I balance affecting functional connectivity. Specifically, the review will highlight the importance of interneuron type, connectivity, and location (both layer- and subregion-specific). The discussion of local mPFC networks will focus on stress context, including stressor duration (acute vs. chronic) and timing (early life vs. adulthood), as these factors have significant implications for the interpretation of experiments and mPFC E/I balance. Indeed, interneurons appear to play a prominent role in prefrontal adaptation, and a better understanding of the interactions between stress and interneuron function may yield insight to the transition from adaptation to pathology. Ultimately, determining the mechanisms mediating adaptive versus pathologic plasticity will promote the development of novel treatments for neuropsychiatric disorders related to prefrontal E/I imbalance.


Assuntos
Comportamento , Glucocorticoides/fisiologia , Interneurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Estresse Psicológico , Adaptação Psicológica , Animais , Sistema Nervoso Autônomo/fisiologia , Humanos , Sistemas Neurossecretores/fisiologia
16.
Elife ; 82019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31329100

RESUMO

Glucocorticoid receptors (GR) have diverse functions relevant to maintenance of homeostasis and adaptation to environmental challenges. Understanding the importance of tissue-specific GR function in physiology and behavior has been hampered by near-ubiquitous localization in brain and body. Here we use CRISPR/Cas9 gene editing to create a conditional GR knockdown in Sprague Dawley rats. To test the impact of cell- and region-specific GR knockdown on physiology and behavior, we targeted GR knockdown to output neurons of the prelimbic cortex. Prelimbic knockdown of GR in females caused deficits in acquisition and extinction of fear memory during auditory fear conditioning, whereas males exhibited enhanced active-coping behavior during forced swim. Our data support the utility of this conditional knockdown rat to afford high-precision knockdown of GR across a variety of contexts, ranging from neuronal depletion to circuit-wide manipulations, leveraging the behavioral tractability and enhanced brain size of the rat as a model organism.


Assuntos
Adaptação Psicológica , Comportamento Animal , Encéfalo/enzimologia , Encéfalo/fisiologia , Medo , Técnicas de Silenciamento de Genes , Receptores de Glucocorticoides/metabolismo , Animais , Edição de Genes , Ratos Sprague-Dawley , Fatores Sexuais
17.
Microbiome ; 5(1): 102, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28838324

RESUMO

BACKGROUND: There is growing evidence for a role of the gut microbiome in shaping behaviour relevant to many psychiatric and neurological disorders. Preclinical studies using germ-free (GF) animals have been essential in contributing to our current understanding of the potential importance of the host microbiome for neurodevelopment and behaviour. In particular, it has been repeatedly demonstrated that manipulation of the gut microbiome modulates anxiety-like behaviours. The neural circuits that underlie anxiety- and fear-related behaviours are complex and heavily depend on functional communication between the amygdala and prefrontal cortex (PFC). Previously, we have shown that the transcriptional networks within the amygdala and PFC of GF mice are altered. MicroRNAs (miRNAs) act through translational repression to control gene translation and have also been implicated in anxiety-like behaviours. However, it is unknown whether these features of host post-transcriptional machinery are also recruited by the gut microbiome to exert control over CNS transcriptional networks. RESULTS: We conducted Illumina® next-generation sequencing (NGS) in the amygdala and PFC of conventional, GF and germ-free colonized mice (exGF). We found a large proportion of miRNAs to be dysregulated in GF animals in both brain regions (103 in the amygdala and 31 in the PFC). Additionally, colonization of GF mice normalized some of the noted alterations. Next, we used a complementary approach to GF by manipulating the adult rat microbiome with an antibiotic cocktail to deplete the gut microbiota and found that this strategy also impacted the expression of relevant miRNAs. CONCLUSION: These results suggest that the microbiome is necessary for appropriate regulation of miRNA expression in brain regions implicated in anxiety-like behaviours.


Assuntos
Tonsila do Cerebelo/metabolismo , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Vida Livre de Germes , MicroRNAs/genética , Córtex Pré-Frontal/metabolismo , Animais , Ansiedade/genética , Encéfalo , Redes Reguladoras de Genes/genética , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Ratos
18.
Biol Psychiatry ; 82(7): 472-487, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28242013

RESUMO

BACKGROUND: The realization that the microbiota-gut-brain axis plays a critical role in health and disease, including neuropsychiatric disorders, is rapidly advancing. Nurturing a beneficial gut microbiome with prebiotics, such as fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS), is an appealing but underinvestigated microbiota manipulation. Here we tested whether chronic prebiotic treatment modifies behavior across domains relevant to anxiety, depression, cognition, stress response, and social behavior. METHODS: C57BL/6J male mice were administered FOS, GOS, or a combination of FOS+GOS for 3 weeks prior to testing. Plasma corticosterone, microbiota composition, and cecal short-chain fatty acids were measured. In addition, FOS+GOS- or water-treated mice were also exposed to chronic psychosocial stress, and behavior, immune, and microbiota parameters were assessed. RESULTS: Chronic prebiotic FOS+GOS treatment exhibited both antidepressant and anxiolytic effects. Moreover, the administration of GOS and the FOS+GOS combination reduced stress-induced corticosterone release. Prebiotics modified specific gene expression in the hippocampus and hypothalamus. Regarding short-chain fatty acid concentrations, prebiotic administration increased cecal acetate and propionate and reduced isobutyrate concentrations, changes that correlated significantly with the positive effects seen on behavior. Moreover, FOS+GOS reduced chronic stress-induced elevations in corticosterone and proinflammatory cytokine levels and depression-like and anxiety-like behavior in addition to normalizing the effects of stress on the microbiota. CONCLUSIONS: Taken together, these data strongly suggest a beneficial role of prebiotic treatment for stress-related behaviors. These findings strengthen the evidence base supporting therapeutic targeting of the gut microbiota for brain-gut axis disorders, opening new avenues in the field of nutritional neuropsychopharmacology.


Assuntos
Ansiolíticos/uso terapêutico , Antidepressivos/uso terapêutico , Ansiedade/terapia , Depressão/terapia , Oligossacarídeos/uso terapêutico , Prebióticos , Animais , Ansiedade/sangue , Ansiedade/etiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Corticosterona/sangue , Depressão/sangue , Depressão/etiologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Expressão Gênica/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurotransmissores/sangue , Nociceptividade/efeitos dos fármacos , Estresse Psicológico/complicações , Triptofano/sangue
19.
Curr Drug Targets ; 17(5): 538-616, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25777273

RESUMO

Extensive research over the past thirty years has demonstrated a vital role for metabotropic glutamate (mGlu) receptors in the major functions of the central nervous system (CNS). A wealth of preclinical studies provide evidence that pharmacological targeting of mGlu receptors can effectively attenuate the development of symptoms and progression of many CNS disorders in animal models. In this review we summarize the current knowledge on the involvement of mGlu receptors in the pathophysiology of neuropsychiatric disorders (schizophrenia, depression, anxiety and cognitive disorders, pain perception and addiction), as well as neurodegenerative (Alzheimer's, Huntington's and Parkinson's diseases) and neurodevelopmental (fragile X syndrome and autism spectrum disorders) diseases. We further emphasize the therapeutic potential of mGlu receptors' pharmacological modulators in these diseases, describe the results of clinical trials with these compounds and discuss the potential sources of translational difficulties.


Assuntos
Doenças do Sistema Nervoso Central/tratamento farmacológico , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Doenças do Sistema Nervoso Central/metabolismo , Ensaios Clínicos como Assunto , Humanos , Receptores de Glutamato Metabotrópico/agonistas , Transdução de Sinais/efeitos dos fármacos
20.
Biol Sex Differ ; 7: 33, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27429736

RESUMO

BACKGROUND: Early-life stress (ELS) is a recognized risk factor for chronic pain disorders, and females appear to be more sensitive to the negative effects of stress. Moreover, estrous cycle-related fluctuations in estrogen levels have been linked with alternating pain sensitivity. Aberrant central circuitry involving both the anterior cingulate cortex (ACC) and the lumbosacral spinal cord has also been implicated in the modulation of visceral pain in clinical and preclinical studies. Here we further investigate changes in visceral pain sensitivity and central glutamatergic systems in rats with respect to estrous cycle and ELS. METHODS: We investigated visceral sensitivity in adult female Sprague-Dawley rats, which had undergone maternal separation (MS) in early life or remained non-separated (NS), by performing colorectal distension (CRD). We also assessed excitatory amino acid uptake through excitatory amino acid transporters (EAATs) in the lumbosacral spinal cord and ACC. RESULTS: NS animals in proestrus and estrus exhibited reduced EAAT uptake and decreased threshold to CRD. Moreover, total pain behaviors were increased in these stages. MS rats exhibited lower pain thresholds and higher total pain behaviors to CRD across all stages of the estrous cycle. Interestingly, cortical EAAT function in MS rats was inhibited in the low estrogen state-an effect completely opposite to that seen in NS rats. CONCLUSIONS: This data confirms that estrous cycle and ELS are significant factors in visceral sensitivity and fluctuations in EAAT function may be a perpetuating factor mediating central sensitization.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA