Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Sci Rep ; 12(1): 14476, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-36008431

RESUMO

Drug resistance caused by mutations is a public health threat for existing and emerging viral diseases. A wealth of evidence about these mutations and their clinically associated phenotypes is scattered across the literature, but a comprehensive perspective is usually lacking. This work aimed to produce a clinically relevant view for the case of Hepatitis B virus (HBV) mutations by combining a chronic HBV clinical study with a compendium of genetic mutations systematically gathered from the scientific literature. We enriched clinical mutation data by systematically mining 2,472,725 scientific articles from PubMed Central in order to gather information about the HBV mutational landscape. By performing this analysis, we were able to identify mutational hotspots for each HBV genotype (A-E) and gene (C, X, P, S), as well as the location of disulfide bonds associated with these mutations. Through a modelling study, we also identified a mutation position common in both the clinical data and the literature that is located at the binding pocket for a known anti-HBV drug, namely entecavir. The results of this novel approach show the potential of integrated analyses to assist in the development of new drugs for viral diseases that are more robust to resistance. Such analyses should be of particular interest due to the increasing importance of viral resistance in established and emerging viruses, such as for newly developed drugs against SARS-CoV-2.


Assuntos
Tratamento Farmacológico da COVID-19 , Hepatite B Crônica , Antivirais/farmacologia , Antivirais/uso terapêutico , DNA Viral/genética , Farmacorresistência Viral/genética , Genótipo , Vírus da Hepatite B/genética , Humanos , Mutação , SARS-CoV-2/genética
2.
Cancer Discov ; 8(4): 395-402, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29545369

RESUMO

Checkpoint inhibitor therapy has been a breakthrough in cancer research, but only some patients with cancer derive substantial benefit. Although mechanisms underlying sensitivity and resistance to checkpoint inhibitors are being elucidated, the importance of organ-specific regulation of immunity is currently underappreciated. Here, we call for a greater understanding of tissue-specific immunoregulation, namely, "tissue-specific immunostats," to make advances in treatments for cancer. A better understanding of how individual organs at baseline regulate the immune system could enable an improved precision medicine approach to cancer immunotherapy. Cancer Discov; 8(4); 395-402. ©2018 AACR.


Assuntos
Sistema Imunitário , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos , Imunoterapia , Camundongos , Especificidade de Órgãos , Medicina de Precisão
3.
Nat Rev Drug Discov ; 17(4): 280-299, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29217836

RESUMO

Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.


Assuntos
Síndrome do Cromossomo X Frágil/tratamento farmacológico , Transtornos do Neurodesenvolvimento/tratamento farmacológico , Neurotransmissores/farmacologia , Neurotransmissores/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Desenvolvimento de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
4.
PLoS Biol ; 15(12): e2002690, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29283992

RESUMO

Response to antidepressant treatment in major depressive disorder (MDD) cannot be predicted currently, leading to uncertainty in medication selection, increasing costs, and prolonged suffering for many patients. Despite tremendous efforts in identifying response-associated genes in large genome-wide association studies, the results have been fairly modest, underlining the need to establish conceptually novel strategies. For the identification of transcriptome signatures that can distinguish between treatment responders and nonresponders, we herein submit a novel animal experimental approach focusing on extreme phenotypes. We utilized the large variance in response to antidepressant treatment occurring in DBA/2J mice, enabling sample stratification into subpopulations of good and poor treatment responders to delineate response-associated signature transcript profiles in peripheral blood samples. As a proof of concept, we translated our murine data to the transcriptome data of a clinically relevant human cohort. A cluster of 259 differentially regulated genes was identified when peripheral transcriptome profiles of good and poor treatment responders were compared in the murine model. Differences in expression profiles from baseline to week 12 of the human orthologues selected on the basis of the murine transcript signature allowed prediction of response status with an accuracy of 76% in the patient population. Finally, we show that glucocorticoid receptor (GR)-regulated genes are significantly enriched in this cluster of antidepressant-response genes. Our findings point to the involvement of GR sensitivity as a potential key mechanism shaping response to antidepressant treatment and support the hypothesis that antidepressants could stimulate resilience-promoting molecular mechanisms. Our data highlight the suitability of an appropriate animal experimental approach for the discovery of treatment response-associated pathways across species.


Assuntos
Antidepressivos/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Paroxetina/farmacologia , Receptores de Glucocorticoides/fisiologia , Animais , Antidepressivos/uso terapêutico , Biomarcadores Farmacológicos , Encéfalo/metabolismo , Corticosterona/sangue , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos DBA , Família Multigênica , Paroxetina/metabolismo , Paroxetina/uso terapêutico , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
5.
J Pharmacol Exp Ther ; 353(1): 213-33, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25665805

RESUMO

Major depressive disorder (MDD) is a serious public health burden and a leading cause of disability. Its pharmacotherapy is currently limited to modulators of monoamine neurotransmitters and second-generation antipsychotics. Recently, glutamatergic approaches for the treatment of MDD have increasingly received attention, and preclinical research suggests that metabotropic glutamate receptor 5 (mGlu5) inhibitors have antidepressant-like properties. Basimglurant (2-chloro-4-[1-(4-fluoro-phenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]-pyridine) is a novel mGlu5 negative allosteric modulator currently in phase 2 clinical development for MDD and fragile X syndrome. Here, the comprehensive preclinical pharmacological profile of basimglurant is presented with a focus on its therapeutic potential for MDD and drug-like properties. Basimglurant is a potent, selective, and safe mGlu5 inhibitor with good oral bioavailability and long half-life supportive of once-daily administration, good brain penetration, and high in vivo potency. It has antidepressant properties that are corroborated by its functional magnetic imaging profile as well as anxiolytic-like and antinociceptive features. In electroencephalography recordings, basimglurant shows wake-promoting effects followed by increased delta power during subsequent non-rapid eye movement sleep. In microdialysis studies, basimglurant had no effect on monoamine transmitter levels in the frontal cortex or nucleus accumbens except for a moderate increase of accumbal dopamine, which is in line with its lack of pharmacological activity on monoamine reuptake transporters. These data taken together, basimglurant has favorable drug-like properties, a differentiated molecular mechanism of action, and antidepressant-like features that suggest the possibility of also addressing important comorbidities of MDD including anxiety and pain as well as daytime sleepiness and apathy or lethargy.


Assuntos
Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Imidazóis/farmacologia , Piridinas/farmacologia , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Regulação Alostérica , Animais , Ansiolíticos/farmacocinética , Ansiolíticos/uso terapêutico , Antidepressivos/farmacocinética , Antidepressivos/uso terapêutico , Monoaminas Biogênicas/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Cricetulus , Depressão/metabolismo , Depressão/psicologia , Agonismo Inverso de Drogas , Eletroencefalografia , Feminino , Imidazóis/farmacocinética , Imidazóis/uso terapêutico , Macaca fascicularis , Masculino , Camundongos , Dor/tratamento farmacológico , Dor/fisiopatologia , Piridinas/farmacocinética , Piridinas/uso terapêutico , Ensaio Radioligante , Ratos Sprague-Dawley , Ratos Wistar , Receptor de Glutamato Metabotrópico 5/metabolismo , Bexiga Urinária Hiperativa/tratamento farmacológico , Bexiga Urinária Hiperativa/fisiopatologia
6.
Curr Opin Pharmacol ; 20: 124-34, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25488569

RESUMO

Fragile X syndrome (FXS) is the most common monogenic form of inherited mental retardation caused by a trinucleotid repeat expansion and transcriptional shutdown of the FMR1 gene. FXS patients present a complex and often severe neuropsychiatric phenotype yet have mild somatic symptoms, normal life expectancies, and no indications of neurodegeneration. The therapeutic potential of mGlu5 inhibitors was proposed in the 'mGluR theory of FXS' based on early insights into the molecular pathophysiology of FXS. Studies in Fragile X mental retardation 1 (Fmr1) knock-out mice, a widely used disease model, demonstrated that mGlu5 inhibitors can correct a broad range of disease-related phenotypes. Recent clinical trials, however, with two different mGlu5 inhibitors (basimglurant and mavoglurant) showed no therapeutic benefit in FXS patients for reasons as yet unclear.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Síndrome do Cromossomo X Frágil/fisiopatologia , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Indóis/farmacologia , Indóis/uso terapêutico , Camundongos , Camundongos Knockout , Terapia de Alvo Molecular , Fenótipo , Piridinas/farmacologia , Piridinas/uso terapêutico
7.
Psychoneuroendocrinology ; 48: 98-110, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24998413

RESUMO

Understanding the molecular mechanisms by which stress is translated into changes in complex behavior may help to identify novel treatment strategies for stress-associated psychiatric disorders. The tumor suppressor gene down-regulated in renal cell carcinoma 1 (DRR1) was recently characterized as a new molecular link between stress, synaptic efficacy and behavioral performance, most likely through its ability to modulate actin dynamics. The lateral septum is one of the brain regions prominently involved in the stress response. This brain region features high DRR1 expression in adult mice, even under basal conditions. We therefore aimed to characterize and dissect the functional role of septal DRR1 in modulating complex behavior. DRR1 protein expression was shown to be expressed in both neurons and astrocytes of the lateral septum of adult mice. Septal DRR1 mRNA expression increased after acute defeat stress and glucocorticoid receptor activation. To mimic the stress-induced DRR1 increase in the lateral septum of mice, we performed adenovirus-mediated region-specific overexpression of DRR1 and characterized the behavior of these mice. Overexpression of DRR1 in the septal region increased sociability, but did not change cognitive, anxiety-like or anhedonic behavior. The observed changes in social behavior did not involve alterations of the expression of vasopressin or oxytocin receptors, the canonical social neuropeptidergic circuits of the lateral septum. In summary, our data suggest that the stress-induced increase of DRR1 expression in the lateral septum could be a protective mechanism to buffer or counterbalance negative consequences of stress exposure on social behavior.


Assuntos
Comportamento Animal , Transtornos Mentais/genética , Comportamento Social , Proteínas Supressoras de Tumor/fisiologia , Actinas/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ligação Proteica , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia
8.
Endocrinology ; 155(7): 2500-10, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24773341

RESUMO

Chronic stress is a risk factor for psychiatric disorders but does not necessarily lead to uniform long-term effects on mental health, suggesting modulating factors such as genetic predispositions. Here we address the question whether natural genetic variations in the mouse CRH receptor 1 (Crhr1) locus modulate the effects of adolescent chronic social stress (ACSS) on long-term stress hormone dysregulation in outbred CD1 mice, which allows a better understanding of the currently reported genes × environment interactions of early trauma and CRHR1 in humans. We identified 2 main haplotype variants in the mouse Crhr1 locus that modulate the long-term effects of ACSS on basal hypothalamic-pituitary-adrenal axis activity. This effect is likely mediated by higher levels of CRHR1, because Crhr1 mRNA expression and CRHR1 binding were enhanced in risk haplotype carriers. Furthermore, a CRHR1 receptor antagonist normalized these long-term effects. Deep sequencing of the Crhr1 locus in CD1 mice revealed a large number of linked single-nucleotide polymorphisms with some located in important regulatory regions, similar to the location of human CRHR1 variants implicated in modulating gene × stress exposure interactions. Our data support that the described gene × stress exposure interaction in this animal model is based on naturally occurring genetic variations in the Crhr1 gene associated with enhanced CRHR1-mediated signaling. Our results suggest that patients with a specific genetic predisposition in the CRHR1 gene together with an exposure to chronic stress may benefit from a treatment selectively antagonizing CRHR1 hyperactivity.


Assuntos
Predisposição Genética para Doença/genética , Polimorfismo de Nucleotídeo Único , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Psicológico/genética , Animais , Comportamento Animal/efeitos dos fármacos , Ligação Competitiva , Corticosterona/sangue , Feminino , Expressão Gênica , Frequência do Gene , Interação Gene-Ambiente , Genótipo , Haplótipos , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Hibridização In Situ , Masculino , Camundongos , Hipófise/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Pirazóis/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Transdução de Sinais/genética , Triazinas/farmacologia
9.
Eur Neuropsychopharmacol ; 24(6): 907-18, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24589292

RESUMO

Chronic stress is one of the predominant environmental risk factors for a number of psychiatric disorders, particularly for major depression. Different hypotheses have been formulated to address the interaction between early and adult chronic stress in psychiatric disease vulnerability. The match/mismatch hypothesis of psychiatric disease states that the early life environment shapes coping strategies in a manner that enables individuals to optimally face similar environments later in life. We tested this hypothesis in female Balb/c mice that underwent either stress or enrichment early in life and were in adulthood further subdivided in single or group housed, in order to provide aversive or positive adult environments, respectively. We studied the effects of the environmental manipulation on anxiety-like, depressive-like and sociability behaviors and gene expression profiles. We show that continuous exposure to adverse environments (matched condition) is not necessarily resulting in an opposite phenotype compared to a continuous supportive environment (matched condition). Rather, animals with mismatched environmental conditions behaved differently from animals with matched environments on anxious, social and depressive like phenotypes. These results further support the match/mismatch hypothesis and illustrate how mild or moderate aversive conditions during development can shape an individual to be optimally adapted to similar conditions later in life.


Assuntos
Transtornos de Ansiedade/etiologia , Transtorno Depressivo/etiologia , Meio Ambiente , Modelos Psicológicos , Comportamento Social , Estresse Psicológico/complicações , Adaptação Psicológica , Glândulas Suprarrenais/fisiopatologia , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Transtornos de Ansiedade/fisiopatologia , Transtornos de Ansiedade/psicologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corticosterona/sangue , Transtorno Depressivo/fisiopatologia , Transtorno Depressivo/psicologia , Modelos Animais de Doenças , Ciclo Estral/fisiologia , Feminino , Hipocampo/fisiopatologia , Abrigo para Animais , Camundongos Endogâmicos BALB C , Testes Neuropsicológicos , Fenótipo , Isolamento Social/psicologia , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Timo/fisiopatologia
10.
Nat Neurosci ; 16(6): 706-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23644483

RESUMO

Stress impairs cognition via corticotropin-releasing hormone receptor 1 (CRHR1), but the molecular link between abnormal CRHR1 signaling and stress-induced cognitive impairments remains unclear. We investigated whether the cell adhesion molecule nectin-3 is required for the effects of CRHR1 on cognition and structural remodeling after early-life stress exposure. Postnatally stressed adult mice had decreased hippocampal nectin-3 levels, which could be attenuated by CRHR1 inactivation and mimicked by corticotropin-releasing hormone (CRH) overexpression in forebrain neurons. Acute stress dynamically reduced hippocampal nectin-3 levels, which involved CRH-CRHR1, but not glucocorticoid receptor, signaling. Suppression of hippocampal nectin-3 caused spatial memory deficits and dendritic spine loss, whereas enhancing hippocampal nectin-3 expression rescued the detrimental effects of early-life stress on memory and spine density in adulthood. Our findings suggest that hippocampal nectin-3 is necessary for the effects of stress on memory and structural plasticity and indicate that the CRH-CRHR1 system interacts with the nectin-afadin complex to mediate such effects.


Assuntos
Moléculas de Adesão Celular/fisiologia , Espinhas Dendríticas/metabolismo , Hipocampo/fisiopatologia , Memória/fisiologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Transdução de Sinais/fisiologia , Estresse Psicológico , Animais , Comportamento Animal/fisiologia , Moléculas de Adesão Celular/antagonistas & inibidores , Hormônio Liberador da Corticotropina/fisiologia , Espinhas Dendríticas/patologia , Regulação para Baixo/genética , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Nectinas , Prosencéfalo/patologia , Prosencéfalo/fisiologia , Transdução de Sinais/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Regulação para Cima/genética
11.
Neuropharmacology ; 72: 38-46, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23602985

RESUMO

Exposure to chronic stress during developmental periods is a risk factor for a number of psychiatric disorders. While the direct effects of stress exposure have been studied extensively, little is known about the long-lasting effects and the interaction with ageing. The same holds true for the treatment with selective serotonin reuptake inhibitors (SSRIs), which have been shown to prevent or reverse some stress-induced effects. Here, we studied the direct and long-lasting impact of chronic social stress during adolescence and the impact of chronic treatment with the SSRI paroxetine in adulthood and aged animals. Therefore, male CD1 mice at the age of 28 days were subjected to 7 weeks of chronic social stress. Treatment with paroxetine was performed per os with a dosage of 20 mg/g BW. We were able to reverse most of the effects of chronic social stress in adult mice (4 months old) and to some extend in aged animals (15 months old) with the SSRI treatment. Especially the regulation of the HPA axis seems to be affected in aged mice with a shift to the use of vasopressin. Our results demonstrate that chronic stress exposure and antidepressant treatment at the end of the developmental period can have a significant and long-lasting impact, highly relevant for healthy ageing.


Assuntos
Envelhecimento , Paroxetina/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Isolamento Social/psicologia , Estresse Psicológico/tratamento farmacológico , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Peso Corporal/efeitos dos fármacos , Doença Crônica , Corticosterona/sangue , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/genética , Dexametasona , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Estudos Longitudinais , Masculino , Camundongos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/sangue , Timo/efeitos dos fármacos
12.
Neuropsychopharmacology ; 37(13): 2797-808, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22871917

RESUMO

Various clinical studies have identified FK506-binding protein 51 (FKBP51) as a target gene involved in the development of psychiatric disorders such as depression. Furthermore, FKBP51 has been shown to affect glucocorticoid receptor signaling by sensitivity modulation and it is implicated in stress reactivity as well as in molecular mechanisms of stress vulnerability and resilience. We investigated the physiological, behavioral, and neuroendocrine parameters in an established chronic stress model both directly after stress and after a recovery period of 3 weeks and also studied the efficacy of paroxetine in this model. We then examined FKBP51 mRNA levels in the dorsal and ventral part of the hippocampus and correlated the expression to behavioral and endocrine parameters. We show robust chronic stress effects in physiological, behavioral, and neuroendocrine parameters, which were only slightly affected by paroxetine treatment. On the contrary, paroxetine led to a disruption of the neuroendocrine system. FKBP51 expression was significantly increased directly after the stress period and correlated with behavioral and neuroendocrine parameters. Taken together, we were able to further elucidate the role of FKBP51 in the mechanisms of stress resilience and vulnerability, especially with respect to behavioral and neuroendocrine parameters. These findings strongly support the concept of FKBP51 as a marker for glucocorticoid receptor sensitivity and its involvement in the development of psychiatric disorders.


Assuntos
Paroxetina/uso terapêutico , Comportamento Social , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Antidepressivos de Segunda Geração/uso terapêutico , Depressão/tratamento farmacológico , Depressão/metabolismo , Depressão/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/psicologia , Resultado do Tratamento
13.
Psychoneuroendocrinology ; 37(12): 2009-21, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22641006

RESUMO

Aversive life events represent one of the main risk factors for the development of many psychiatric diseases, but the interplay between environmental factors and genetic predispositions is still poorly understood. One major finding in many depressed patients is an impaired regulation of the hypothalamic-pituitary-adrenal (HPA) axis. The negative feedback loop of the HPA axis is mediated via the glucocorticoid receptor (GR) and the mineralocorticoid receptor. The co-chaperones FK506-binding protein 51 (FKBP51) and FK506-binding protein 52 (FKBP52) are components of the heat shock protein 90-receptor-heterocomplex and are functionally divergent regulators of both receptors. Here, we characterized heterozygous Fkbp52 knockout (Fkbp52(+/-)) mice under basal or chronic social defeat stress (CSDS) conditions with regard to physiological, neuroendocrine, behavioral and mRNA expression alterations. Fkbp52(+/-) mice displayed symptoms of increased stress sensitivity in a subset of behavioral and neuroendocrine parameters. These included increased anxiety-related behavior in the elevated plus-maze and an enhanced neuroendocrine response to a forced swim test (FST), possibly mediated by reduced GR sensitivity. At the same time, Fkbp52(+/-) mice also demonstrated signs of stress resilience in other behavioral and neuroendocrine aspects, such as reduced basal corticosterone levels and more active stress-coping behavior in the FST following CSDS. These contrasting results are in line with previous reports showing that FKBP52 is not involved in all branches of GR signaling, but rather acts in a gene-specific manner to regulate GR transcriptional activation.


Assuntos
Comportamento Animal/fisiologia , Corticosterona/metabolismo , Heterozigoto , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/fisiologia , Animais , Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Estresse Psicológico/sangue , Proteínas de Ligação a Tacrolimo/biossíntese , Proteínas de Ligação a Tacrolimo/metabolismo , Vasopressinas/metabolismo
14.
Curr Psychiatry Rep ; 14(2): 159-65, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278810

RESUMO

Stress has been identified as a key risk factor for a multitude of human pathologies. However, stress by itself is often not sufficient to induce a disease, as a large contribution comes from an individual's genetic background. Therefore, many stress models have been created to investigate this so-called gene-environment interaction for different diseases. Recently, evidence has been accumulating to indicate that not only the exposure to stress, but also the vulnerability to such an exposure can have a significant impact on the development of disease. Herein we review recent animal models of stress vulnerability and resilience, with special attention devoted to the readout parameters and the potential for translatability of the results.


Assuntos
Resiliência Psicológica , Estresse Psicológico/psicologia , Animais , Suscetibilidade a Doenças/psicologia , Interação Gene-Ambiente , Camundongos , Camundongos Transgênicos , Modelos Animais , Fatores de Risco , Estresse Psicológico/etiologia
15.
Neuropharmacology ; 62(1): 332-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21839098

RESUMO

Chronic stress is increasingly considered to be a main risk factor for the development of a variety of psychiatric diseases such as depression. This is further supported by an impaired negative feedback of the hypothalamic-pituitary-adrenal (HPA) axis, which has been observed in the majority of depressed patients. The effects of glucocorticoids, the main hormonal endpoint of the HPA axis, are mediated via the glucocorticoid receptor (GR) and the mineralocorticoid receptor. The FK506-binding protein 51 (FKBP5), a co-chaperone of the Hsp90 and component of the chaperone-receptor heterocomplex, has been shown to reduce ligand sensitivity of the GR. This study aimed to investigate the function of FKBP5 as a possible mediator of the stress response system and its potential role in the development of stress-related diseases. Therefore, we assessed whether mice lacking the gene encoding FKBP5 (51KO mice) were less vulnerable to the adverse effects of three weeks of chronic social defeat stress. Mice were subsequently analyzed with regards to physiological, neuroendocrine, behavioral and mRNA expression alterations. Our results show a less vulnerable phenotype of 51KO mice with respect to physiological and neuroendocrine parameters compared to wild-type animals. 51KO mice demonstrated lower adrenal weights and basal corticosterone levels, a diminished response to a novel acute stimulus and an enhanced recovery, as well as more active stress-coping behavior. These results suggest an enhanced negative glucocorticoid feedback within the HPA axis of 51KO mice, possibly modulated by an increased sensitivity of the GR. This article is part of a Special Issue entitled 'Anxiety and Depression'.


Assuntos
Sistemas Neurossecretores/metabolismo , Sistemas Neurossecretores/fisiopatologia , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Estresse Psicológico , Proteínas de Ligação a Tacrolimo/metabolismo , Análise de Variância , Animais , Corticosterona/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP90/metabolismo , Locomoção/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia , Natação/psicologia , Proteínas de Ligação a Tacrolimo/deficiência
16.
Proc Natl Acad Sci U S A ; 108(41): 17213-8, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21969592

RESUMO

Stress has been identified as a major causal factor for many mental disorders. However, our knowledge about the chain of molecular and cellular events translating stress experience into altered behavior is still rather scant. Here, we have characterized a murine ortholog of the putative tumor suppressor gene DRR1 as a unique stress-induced protein in brain. It binds to actin, promotes bundling and stabilization of actin filaments, and impacts on actin-dependent neurite outgrowth. Endogenous DRR1 localizes to some, but not all, synapses, with preference for the presynaptic region. Hippocampal virus-mediated enhancement of DRR1 expression reduced spine density, diminished the probability of synaptic glutamate release, and altered cognitive performance. DRR1 emerges as a protein to link stress with actin dynamics, which in addition is able to act on synaptic function and cognition.


Assuntos
Cognição/fisiologia , Sinapses/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Actinas/metabolismo , Animais , Comportamento Animal/fisiologia , Encéfalo/citologia , Encéfalo/fisiologia , Genes Supressores de Tumor , Células HEK293 , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuritos/metabolismo , Neuritos/ultraestrutura , Ligação Proteica , Estresse Fisiológico , Proteínas Supressoras de Tumor/genética
17.
J Neurosci ; 31(38): 13625-34, 2011 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-21940453

RESUMO

Childhood traumatic events hamper the development of the hippocampus and impair declarative memory in susceptible individuals. Persistent elevations of hippocampal corticotropin-releasing factor (CRF), acting through CRF receptor 1 (CRF1), in experimental models of early-life stress have suggested a role for this endogenous stress hormone in the resulting structural modifications and cognitive dysfunction. However, direct testing of this possibility has been difficult. In the current study, we subjected conditional forebrain CRF1 knock-out (CRF1-CKO) mice to an impoverished postnatal environment and examined the role of forebrain CRF1 in the long-lasting effects of early-life stress on learning and memory. Early-life stress impaired spatial learning and memory in wild-type mice, and postnatal forebrain CRF overexpression reproduced these deleterious effects. Cognitive deficits in stressed wild-type mice were associated with disrupted long-term potentiation (LTP) and a reduced number of dendritic spines in area CA3 but not in CA1. Forebrain CRF1 deficiency restored cognitive function, LTP and spine density in area CA3, and augmented CA1 LTP and spine density in stressed mice. In addition, early-life stress differentially regulated the amount of hippocampal excitatory and inhibitory synapses in wild-type and CRF1-CKO mice, accompanied by alterations in the neurexin-neuroligin complex. These data suggest that the functional, structural and molecular changes evoked by early-life stress are at least partly dependent on persistent forebrain CRF1 signaling, providing a molecular target for the prevention of cognitive deficits in adults with a history of early-life adversity.


Assuntos
Transtornos Cognitivos/fisiopatologia , Hormônio Liberador da Corticotropina/fisiologia , Prosencéfalo/metabolismo , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular Neuronais/metabolismo , Transtornos Cognitivos/complicações , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Transtornos Cognitivos/psicologia , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Hipocampo/citologia , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Comportamento Espacial/fisiologia , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Estresse Psicológico/psicologia
18.
Neurobiol Dis ; 42(3): 300-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21296667

RESUMO

Chronic stress evokes profound structural and molecular changes in the hippocampus, which may underlie spatial memory deficits. Corticotropin-releasing hormone (CRH) and CRH receptor 1 (CRHR1) mediate some of the rapid effects of stress on dendritic spine morphology and modulate learning and memory, thus providing a potential molecular basis for impaired synaptic plasticity and spatial memory by repeated stress exposure. Using adult male mice with CRHR1 conditionally inactivated in the forebrain regions, we investigated the role of CRH-CRHR1 signaling in the effects of chronic social defeat stress on spatial memory, the dendritic morphology of hippocampal CA3 pyramidal neurons, and the hippocampal expression of nectin-3, a synaptic cell adhesion molecule important in synaptic remodeling. In chronically stressed wild-type mice, spatial memory was disrupted, and the complexity of apical dendrites of CA3 neurons reduced. In contrast, stressed mice with forebrain CRHR1 deficiency exhibited normal dendritic morphology of CA3 neurons and mild impairments in spatial memory. Additionally, we showed that the expression of nectin-3 in the CA3 area was regulated by chronic stress in a CRHR1-dependent fashion and associated with spatial memory and dendritic complexity. Moreover, forebrain CRHR1 deficiency prevented the down-regulation of hippocampal glucocorticoid receptor expression by chronic stress but induced increased body weight gain during persistent stress exposure. These findings underscore the important role of forebrain CRH-CRHR1 signaling in modulating chronic stress-induced cognitive, structural and molecular adaptations, with implications for stress-related psychiatric disorders.


Assuntos
Dendritos/metabolismo , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Prosencéfalo/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Psicológico/metabolismo , Análise de Variância , Animais , Western Blotting , Peso Corporal/genética , Moléculas de Adesão Celular/metabolismo , Dominação-Subordinação , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Nectinas , Neurônios/metabolismo , Estresse Psicológico/genética
19.
PLoS One ; 6(2): e16883, 2011 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21347384

RESUMO

BACKGROUND: Chronic stress has been found to be a major risk factor for various human pathologies. Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, which is tightly regulated via, among others, the glucocorticoid receptor (GR). The activity of the GR is modulated by a variety of proteins, including the co-chaperone FK506 binding protein 51 (FKBP5). Although FKBP5 has been associated with risk for affective disorders and has been implicated in GR sensitivity, previous studies focused mainly on peripheral blood, while information about basal distribution and induction in the central nervous system are sparse. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we describe the basal expression pattern of Fkbp5 mRNA in the brain of adult male mice and show the induction of Fkbp5 mRNA via dexamethasone treatment or different stress paradigms. We could show that Fkbp5 is often, but not exclusively, expressed in regions also known for GR expression, for example the hippocampus. Furthermore, we were able to induce Fkbp5 expression via dexamethasone in the CA1 and DG subregions of the hippocampus, the paraventricular nucleus (PVN) and the central amygdala (CeA). Increase of Fkbp5 mRNA was also found after restrained stress and 24 hours of food deprivation in the PVN and the CeA, while in the hippocampus only food deprivation caused an increase in Fkbp5 mRNA. CONCLUSIONS/SIGNIFICANCE: Interestingly, regions with a low basal expression showed higher increase in Fkbp5 mRNA following induction than regions with high basal expression, supporting the hypothesis that GR sensitivity is, at least partly, mediated via Fkbp5. In addition, this also supports the use of Fkbp5 gene expression as a marker for GR sensitivity. In summary, we were able to give an overview of the basal expression of fkbp5 mRNA as well as to extend the findings of induction of Fkbp5 and its regulatory influence on GR sensitivity from peripheral blood to the brain.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica , Proteínas de Ligação a Tacrolimo/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Dexametasona/farmacologia , Privação de Alimentos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética
20.
Psychoneuroendocrinology ; 36(4): 579-87, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20940090

RESUMO

The incidence of chronic stress is frequently related to the development of psychiatric disorders like depression. The hypothalamic-pituitary-adrenal (HPA) axis is a major physiological system that mediates the stress response. Tight HPA axis regulation through negative feedback mechanisms is essential for health and environmental adaptation. This feedback regulation acts in part through the glucocorticoid receptor (GR) on several organizational levels, including the pituitary, the hypothalamus and the hippocampus. However, the precise role of the different anatomical structures, specifically the pituitary, in HPA axis regulation is yet largely unknown. Here, we show that a conditional pituitary GR knockout is not necessarily detrimental for the animal's ability to cope with chronic stress situations. Mice with a deletion of the GR at the pituitary (GR(POMCCre)) were subjected to 3 weeks of chronic social defeat stress. We analyzed both the behavioral and neuroendocrine phenotype as well as the central nervous system expression of genes involved in HPA axis function in these animals. Our results show a more resilient phenotype of GR(POMCCre) mice with respect to anxiety-related behavior and neuroendocrine parameters compared to stressed wild type animals. In light of the previously reported high corticosterone levels during postnatal development in GR(POMCCre) mice, our findings suggest that adverse early life events may have beneficial developmental effects on the organism to improve stress coping later in life.


Assuntos
Hipófise/metabolismo , Receptores de Glucocorticoides/genética , Estresse Psicológico/genética , Adaptação Psicológica/fisiologia , Animais , Comportamento Animal , Doença Crônica , Deleção de Genes , Predisposição Genética para Doença , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Sistemas Neurossecretores/metabolismo , Sistemas Neurossecretores/fisiologia , Especificidade de Órgãos/genética , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...