Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Psychiatry ; 27(11): 4485-4501, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36224260

ABSTRACT

Mood disorders (MD) are a major burden on society as their biology remains poorly understood, challenging both diagnosis and therapy. Among many observed biological dysfunctions, homeostatic dysregulation, such as metabolic syndrome (MeS), shows considerable comorbidity with MD. Recently, CREB-regulated transcription coactivator 1 (CRTC1), a regulator of brain metabolism, was proposed as a promising factor to understand this relationship. Searching for imaging biomarkers and associating them with pathophysiological mechanisms using preclinical models can provide significant insight into these complex psychiatric diseases and help the development of personalized healthcare. Here, we used neuroimaging technologies to show that deletion of Crtc1 in mice leads to an imaging fingerprint of hippocampal metabolic impairment related to depressive-like behavior. By identifying a deficiency in hippocampal glucose metabolism as the underlying molecular/physiological origin of the markers, we could assign an energy-boosting mood-stabilizing treatment, ebselen, which rescued behavior and neuroimaging markers. Finally, our results point toward the GABAergic system as a potential therapeutic target for behavioral dysfunctions related to metabolic disorders. This study provides new insights on Crtc1's and MeS's relationship to MD and establishes depression-related markers with clinical potential.


Subject(s)
Hippocampus , Transcription Factors , Mice , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Hippocampus/metabolism , Behavior, Animal/physiology , Depression/genetics , Depression/metabolism
2.
Front Mol Neurosci ; 15: 810641, 2022.
Article in English | MEDLINE | ID: mdl-35242012

ABSTRACT

Depression and obesity are major public health concerns, and there is mounting evidence that they share etiopathophysiological mechanisms. The neurobiological pathways involved in both mood and energy balance regulation are complex, multifactorial and still incompletely understood. As a coactivator of the pleiotropic transcription factor cAMP response element-binding protein (CREB), CREB-regulated transcription coactivator 1 (CRTC1) has recently emerged as a novel regulator of neuronal plasticity and brain functions, while CRTC1 dysfunction has been associated with neurodegenerative and psychiatric diseases. This review focuses on recent evidence emphasizing the critical role of CRTC1 in the neurobiology of depression and comorbid obesity. We discuss the role of CRTC1 downregulation in mediating chronic stress-induced depressive-like behaviors, and antidepressant response in the light of the previously characterized Crtc1 knockout mouse model of depression. The putative role of CRTC1 in the alteration of brain energy homeostasis observed in depression is also discussed. Finally, we highlight rodent and human studies supporting the critical involvement of CRTC1 in depression-associated obesity.

3.
J Mol Cell Cardiol ; 127: 31-43, 2019 02.
Article in English | MEDLINE | ID: mdl-30521840

ABSTRACT

The sympathetic nervous system is the main stimulator of cardiac function. While acute activation of the ß-adrenoceptors exerts positive inotropic and lusitropic effects by increasing cAMP and Ca2+, chronically enhanced sympathetic tone with changed ß-adrenergic signaling leads to alterations of gene expression and remodeling. The CREB-regulated transcription coactivator 1 (CRTC1) is activated by cAMP and Ca2+. In the present study, the regulation of CRTC1 in cardiomyocytes and its effect on cardiac function and growth was investigated. In cardiomyocytes, isoprenaline induced dephosphorylation, and thus activation of CRTC1, which was prevented by propranolol. Crtc1-deficient mice exhibited left ventricular dysfunction, hypertrophy and enlarged cardiomyocytes. However, isoprenaline-induced contractility of isolated trabeculae or phosphorylation of cardiac troponin I, cardiac myosin-binding protein C, phospholamban, and ryanodine receptor were not altered, suggesting that cardiac dysfunction was due to the global lack of Crtc1. The mRNA and protein levels of the Gαq GTPase activating protein regulator of G-protein signaling 2 (RGS2) were lower in hearts of Crtc1-deficient mice. Chromatin immunoprecipitation and reporter gene assays showed stimulation of the Rgs2 promoter by CRTC1. In Crtc1-deficient cardiomyocytes, phosphorylation of the Gαq-downstream kinase ERK was enhanced. CRTC1 content was higher in cardiac tissue from patients with aortic stenosis or hypertrophic cardiomyopathy and from two murine models mimicking these diseases. These data suggest that increased CRTC1 in maladaptive hypertrophy presents a compensatory mechanism to delay disease progression in part by enhancing Rgs2 gene transcription. Furthermore, the present study demonstrates an important role of CRTC1 in the regulation of cardiac function and growth.


Subject(s)
Cardiomegaly/metabolism , Transcription Factors/metabolism , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phosphorylation , Promoter Regions, Genetic , RGS Proteins/genetics , RGS Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Transcription Factors/deficiency
4.
Int J Obes (Lond) ; 43(6): 1295-1304, 2019 06.
Article in English | MEDLINE | ID: mdl-30301962

ABSTRACT

BACKGROUND/OBJECTIVES: High-fat diet consumption is known to trigger an inflammatory response in the hypothalamus, which has been characterized by an initial expression of pro-inflammatory genes followed by hypothalamic astrocytosis, microgliosis, and the appearance of neuronal injury markers. The specific effects of high-fat diet on hypothalamic energy metabolism and neurotransmission are however not yet known and have not been investigated before. SUBJECTS/METHODS: We used 1H and 13C magnetic resonance spectroscopy (MRS) and immunofluorescence techniques to evaluate in vivo the consequences of high-saturated fat diet administration to mice, and explored the effects on hypothalamic metabolism in three mouse cohorts at different time points for up to 4 months. RESULTS: We found that high-fat diet increases significantly the hypothalamic levels of glucose (P < 0.001), osmolytes (P < 0.001), and neurotransmitters (P < 0.05) from 2 months of diet, and alters the rates of metabolic (P < 0.05) and neurotransmission fluxes (P < 0.001), and the contribution of non-glycolytic substrates to hypothalamic metabolism (P < 0.05) after 10 weeks of high-fat feeding. CONCLUSIONS/INTERPRETATION: We report changes that reveal a high-fat diet-induced alteration of hypothalamic metabolism and neurotransmission that is quantifiable by 1H and 13C MRS in vivo, and present the first evidence of the extension of the inflammation pathology to a localized metabolic imbalance.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Energy Metabolism/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Animals , Dietary Fats/administration & dosage , Disease Models, Animal , Gene Expression Profiling , Hypothalamus/physiopathology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism
5.
Transl Psychiatry ; 7(12): 1269, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29217834

ABSTRACT

Obesity and depression are major public health concerns, and there is increasing evidence that they share etiological mechanisms. CREB-regulated transcription coactivator 1 (CRTC1) participates in neurobiological pathways involved in both mood and energy balance regulation. Crtc1 -/- mice rapidly develop a depressive-like and obese phenotype in early adulthood, and are therefore a relevant animal model to explore possible common mechanisms underlying mood disorders and obesity. Here, the obese phenotype of male and female Crtc1 -/- mice was further characterized by investigating CRTC1's role in the homeostatic and hedonic regulation of food intake, as well as its influence on daily locomotor activity. Crtc1 -/- mice showed a strong gender difference in the homeostatic regulation of energy balance. Mutant males were hyperphagic and rapidly developed obesity on normal chow diet, whereas Crtc1 -/- females exhibited mild late-onset obesity without hyperphagia. Overeating of mutant males was accompanied by alterations in the expression of several orexigenic and anorexigenic hypothalamic genes, thus confirming a key role of CRTC1 in the central regulation of food intake. No alteration in preference and conditioned response for saccharine was observed in Crtc1 -/- mice, suggesting that mutant males' hyperphagia was not due to an altered hedonic regulation of food intake. Intriguingly, mutant males exhibited a hyperphagic behavior only during the resting (diurnal) phase of the light cycle. This abnormal feeding behavior was associated with a higher diurnal locomotor activity indicating that the lack of CRTC1 may affect circadian rhythmicity. Collectively, these findings highlight the male-specific involvement of CRTC1 in the central control of energy balance and circadian locomotor activity.


Subject(s)
Circadian Rhythm/physiology , Depression/physiopathology , Energy Metabolism/physiology , Motor Activity/physiology , Transcription Factors/genetics , Animals , Behavior, Animal/physiology , Circadian Rhythm/genetics , Depression/genetics , Disease Models, Animal , Energy Metabolism/genetics , Female , Hyperphagia/genetics , Hyperphagia/physiopathology , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Motor Activity/genetics , Obesity/genetics , Obesity/physiopathology , Sex Factors
6.
Trends Neurosci ; 40(12): 720-733, 2017 12.
Article in English | MEDLINE | ID: mdl-29097017

ABSTRACT

The brain has the ability to sense, coordinate, and respond to environmental changes through biological processes involving activity-dependent gene expression. cAMP-response element binding protein (CREB)-regulated transcription coactivators (CRTCs) have recently emerged as novel transcriptional regulators of essential biological functions, while their deregulation is linked to age-related human diseases. In the brain, CRTCs are unique signaling factors that act as sensors and integrators of hormonal, metabolic, and neural signals contributing to brain plasticity and brain-body communication. In this review, we focus on the regulatory mechanisms and functions of CRTCs in brain metabolism, lifespan, circadian rhythm, and synaptic mechanisms underlying memory and emotion. We also discuss how CRTCs deregulation in cognitive and emotional disorders may provide the basis for potential clinical and therapeutic applications in neurodegenerative and psychiatric diseases.


Subject(s)
Brain/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Transcription Factors/metabolism , Animals , Brain/pathology , Humans
7.
Sci Rep ; 7: 43093, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266561

ABSTRACT

The homeodomain transcription factor Nkx2.1 (NK2 homeobox 1) controls cell differentiation of telencephalic GABAergic interneurons and oligodendrocytes. Here we show that Nkx2.1 also regulates astrogliogenesis of the telencephalon from embryonic day (E) 14.5 to E16.5. Moreover we identify the different mechanisms by which Nkx2.1 controls the telencephalic astrogliogenesis. In Nkx2.1 knockout (Nkx2.1-/-) mice a drastic loss of astrocytes is observed that is not related to cell death. Further, in vivo analysis using BrdU incorporation reveals that Nkx2.1 affects the proliferation of the ventral neural stem cells that generate early astrocytes. Also, in vitro neurosphere assays showed reduced generation of astroglia upon loss of Nkx2.1, which could be due to decreased precursor proliferation and possibly defects in glial specification/differentiation. Chromatin immunoprecipitation analysis and in vitro co-transfection studies with an Nkx2.1-expressing plasmid indicate that Nkx2.1 binds to the promoter of glial fibrillary acidic protein (GFAP), primarily expressed in astrocytes, to regulate its expression. Hence, Nkx2.1 controls astroglial production spatiotemporally in embryos by regulating proliferation of the contributing Nkx2.1-positive precursors.


Subject(s)
Astrocytes/metabolism , Cell Differentiation , Embryonic Development , Telencephalon/metabolism , Thyroid Nuclear Factor 1/physiology , Animals , Astrocytes/physiology , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Mice , Mice, Knockout , Telencephalon/physiology , Thyroid Nuclear Factor 1/metabolism
8.
Neuropharmacology ; 107: 111-121, 2016 08.
Article in English | MEDLINE | ID: mdl-26970016

ABSTRACT

Major depression is a highly complex disabling psychiatric disorder affecting millions of people worldwide. Despite the availability of several classes of antidepressants, a substantial percentage of patients are unresponsive to these medications. A better understanding of the neurobiology of depression and the mechanisms underlying antidepressant response is thus critically needed. We previously reported that mice lacking CREB-regulated transcription coactivator 1 (CRTC1) exhibit a depressive-like phenotype and a blunted antidepressant response to the selective serotonin reuptake inhibitor fluoxetine. In this study, we similarly show that Crtc1(-/-) mice are resistant to the antidepressant effect of chronic desipramine in a behavioral despair paradigm. Supporting the blunted response to this tricyclic antidepressant, we found that desipramine does not significantly increase the expression of Bdnf and Nr4a1-3 in the hippocampus and prefrontal cortex of Crtc1(-/-) mice. Epigenetic regulation of neuroplasticity gene expression has been associated with depression and antidepressant response, and histone deacetylase (HDAC) inhibitors have been shown to have antidepressant-like properties. Here, we show that unlike conventional antidepressants, chronic systemic administration of the HDAC inhibitor SAHA partially rescues the depressive-like behavior of Crtc1(-/-) mice. This behavioral effect is accompanied by an increased expression of Bdnf, but not Nr4a1-3, in the prefrontal cortex of these mice, suggesting that this epigenetic intervention restores the expression of a subset of genes by acting downstream of CRTC1. These findings suggest that CRTC1 alterations may be associated with treatment-resistant depression, and support the interesting possibility that targeting HDACs may be a useful therapeutic strategy in antidepressant development.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Transcription Factors/deficiency , Animals , Brain-Derived Neurotrophic Factor/metabolism , DNA-Binding Proteins/metabolism , Depressive Disorder, Major/metabolism , Depressive Disorder, Treatment-Resistant/metabolism , Desipramine/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Transcription Factors/genetics , Vorinostat
9.
J Affect Disord ; 198: 43-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27002284

ABSTRACT

BACKGROUND: Psychiatric disorders have been hypothesized to share common etiological pathways with obesity, suggesting related neurobiological bases. We aimed to examine whether CRTC1 polymorphisms were associated with major depressive disorder (MDD) and to test the association of these polymorphisms with obesity markers in several large case-control samples with MDD. METHODS: The association between CRTC1 polymorphisms and MDD was investigated in three case-control samples with MDD (PsyCoLaus n1=3,362, Radiant n2=3,148 and NESDA/NTR n3=4,663). The effect of CRTC1 polymorphisms on obesity markers was then explored. RESULTS: CRTC1 polymorphisms were not associated with MDD in the three samples. CRTC1 rs6510997C>T was significantly associated with fat mass in the PsyCoLaus study. In fact, a protective effect of this polymorphism was found in MDD cases (n=1,434, ß=-1.32%, 95% CI -2.07 to -0.57, p<0.001), but not in controls. In the Radiant study, CRTC1 polymorphisms were associated with BMI, exclusively in individuals with MDD (n=2,138, ß=-0.75kg/m(2), 95% CI -1.30 to -0.21, p=0.007), while no association with BMI was found in the NESDA/NTR study. LIMITATIONS: Estimated fat mass using bioimpedance that capture more accurately adiposity was only present in the PsyCoLaus sample. CONCLUSIONS: CRTC1 polymorphisms seem to play a role with obesity markers in individuals with MDD rather than non-depressive individuals. Therefore, the weak association previously reported in the population-based samples was driven by cases diagnosed with lifetime MDD. However, CRTC1 seems not to be implicated directly in the development of psychiatric diseases.


Subject(s)
Adipose Tissue , Body Mass Index , Depressive Disorder, Major/complications , Depressive Disorder, Major/genetics , Obesity/complications , Obesity/genetics , Transcription Factors/genetics , Adult , Biomarkers , Case-Control Studies , Female , Humans , Male , Middle Aged , Obesity/physiopathology , Polymorphism, Single Nucleotide/genetics
10.
Cell Signal ; 27(11): 2252-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26247811

ABSTRACT

CREB-binding protein (CBP) and p300 are transcriptional coactivators involved in numerous biological processes that affect cell growth, transformation, differentiation, and development. In this study, we provide evidence of the involvement of homeodomain-interacting protein kinase 2 (HIPK2) in the regulation of CBP activity. We show that HIPK2 interacts with and phosphorylates several regions of CBP. We demonstrate that serines 2361, 2363, 2371, 2376, and 2381 are responsible for the HIPK2-induced mobility shift of CBP C-terminal activation domain. Moreover, we show that HIPK2 strongly potentiates the transcriptional activity of CBP. However, our data suggest that HIPK2 activates CBP mainly by counteracting the repressive action of cell cycle regulatory domain 1 (CRD1), located between amino acids 977 and 1076, independently of CBP phosphorylation. Our findings thus highlight a complex regulation of CBP activity by HIPK2, which might be relevant for the control of specific sets of target genes involved in cellular proliferation, differentiation and apoptosis.


Subject(s)
CREB-Binding Protein/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Transcriptional Activation/genetics , Apoptosis/genetics , CREB-Binding Protein/genetics , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , E1A-Associated p300 Protein , HEK293 Cells , Humans , Phosphorylation , Protein Binding/genetics , Protein Structure, Tertiary , Transcription, Genetic
11.
JAMA Psychiatry ; 70(10): 1011-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23925723

ABSTRACT

IMPORTANCE: There is a high prevalence of obesity in psychiatric patients, possibly leading to metabolic complications and reducing life expectancy. The CREB-regulated transcription coactivator 1 (CRTC1) gene is involved in energy balance and obesity in animal models, but its role in human obesity is unknown. OBJECTIVE: To determine whether polymorphisms within the CRTC1 gene are associated with adiposity markers in psychiatric patients and the general population. DESIGN, SETTING, AND PARTICIPANTS: Retrospective and prospective data analysis and population-based samples at Lausanne and Geneva university hospitals in Switzerland and a private clinic in Lausanne, Switzerland. The effect of 3 CRTC1 polymorphisms on body mass index (BMI) and/or fat mass was investigated in a discovery cohort of psychiatric outpatients taking weight gain-inducing psychotropic drugs (sample 1, n = 152). The CRTC1 variant that was significantly associated with BMI and survived Bonferroni corrections for multiple comparison was then replicated in 2 independent psychiatric samples (sample 2, n = 174 and sample 3, n = 118) and 2 white population-based samples (sample 4, n = 5338 and sample 5, n = 123,865). INTERVENTION: Noninterventional studies. MAIN OUTCOME AND MEASURE: Difference in BMI and/or fat mass between CRTC1 genotype groups. RESULTS: Among the CRTC1 variants tested in the first psychiatric sample, only rs3746266A>G was associated with BMI (P(adjusted) = .003). In the 3 psychiatric samples, carriers of the rs3746266 G allele had a lower BMI than noncarriers (AA genotype) (sample 1, P = .001; sample 2, P = .05; and sample 3, P = .0003). In the combined analysis, excluding patients taking other weight gain-inducing drugs, G allele carriers (n = 98) had a 1.81-kg/m² lower BMI than noncarriers (n = 226; P < .0001). The strongest association was observed in women younger than 45 years, with a 3.87-kg/m² lower BMI in G allele carriers (n = 25) compared with noncarriers (n = 48; P < .0001), explaining 9% of BMI variance. In the population-based samples, the T allele of rs6510997C>T (a proxy of the rs3746266 G allele; r² = 0.7) was associated with lower BMI (sample 5, n = 123,865; P = .01) and fat mass (sample 4, n = 5338; P = .03). The strongest association with fat mass was observed in premenopausal women (n = 1192; P = .02). CONCLUSIONS AND RELEVANCE: These findings suggest that CRTC1 contributes to the genetics of human obesity in psychiatric patients and the general population. Identification of high-risk subjects could contribute to a better individualization of the pharmacological treatment in psychiatry.


Subject(s)
Adiposity/genetics , Body Mass Index , Genetic Predisposition to Disease/genetics , Mental Disorders/genetics , Obesity/genetics , Transcription Factors/genetics , Adult , Case-Control Studies , Female , Humans , Male , Mental Disorders/complications , Obesity/complications , Polymorphism, Single Nucleotide
12.
Biol Psychiatry ; 72(7): 528-36, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22592058

ABSTRACT

BACKGROUND: Mood disorders are polygenic disorders in which the alteration of several susceptibility genes results in dysfunctional mood regulation. However, the molecular mechanisms underlying their transcriptional dysregulation are still unclear. The transcription factor cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) and the neurotrophin brain-derived neurotrophic factor (BDNF) have been implicated in rodent models of depression. We previously provided evidence that Bdnf expression critically rely on a potent CREB coactivator called CREB-regulated transcription coactivator 1 (CRTC1). METHODS: To further evaluate the role of CRTC1 in the brain, we generated a knockout mouse line and analyzed its behavioral and molecular phenotype. RESULTS: We found that mice lacking CRTC1 associate neurobehavioral endophenotypes related to mood disorders. Crtc1(-/-) mice exhibit impulsive aggressiveness, social withdrawal, and decreased sexual motivation, together with increased behavioral despair, anhedonia, and anxiety-related behavior in the novelty-induced hypophagia test. They also present psychomotor retardation as well as increased emotional response to stressful events. Crtc1(-/-) mice have a blunted response to the antidepressant fluoxetine in behavioral despair paradigms, whereas fluoxetine normalizes their aggressiveness and their behavioral response in the novelty-induced hypophagia test. Crtc1(-/-) mice strikingly show, in addition to a reduced dopamine and serotonin turnover in the prefrontal cortex, a concomitant decreased expression of several susceptibility genes involved in neuroplasticity, including Bdnf, its receptor TrkB, the nuclear receptors Nr4a1-3, and several other CREB-regulated genes. CONCLUSIONS: Collectively, these findings support a role for the CRTC1-CREB pathway in mood disorders etiology and behavioral response to antidepressants and identify CRTC1 as an essential coactivator of genes involved in mood regulation.


Subject(s)
Aggression/physiology , Depressive Disorder/genetics , Gene Expression Regulation/genetics , Neuronal Plasticity/genetics , Signal Transduction/genetics , Transcription Factors/deficiency , Aggression/drug effects , Animals , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Second-Generation/therapeutic use , Arabidopsis Proteins/metabolism , Biogenic Monoamines/metabolism , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Chromatography, High Pressure Liquid , Corticosterone/blood , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Disease Models, Animal , Electroshock/adverse effects , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Fear/drug effects , Fear/psychology , Female , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Food Preferences/drug effects , Food Preferences/physiology , Hindlimb Suspension , Intramolecular Transferases/metabolism , Male , Maternal Behavior/drug effects , Maternal Behavior/physiology , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nesting Behavior/drug effects , Nesting Behavior/physiology , Neuronal Plasticity/drug effects , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Signal Transduction/drug effects , Social Behavior , Swimming/psychology , Transcription Factors/metabolism
13.
J Biol Chem ; 285(37): 28587-95, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20639200

ABSTRACT

Dendritic growth is essential for the establishment of a functional nervous system. Among extrinsic signals that control dendritic development, substantial evidence indicates that BDNF regulates dendritic morphology. However, little is known about the underlying mechanisms by which BDNF controls dendritic growth. In this study, we show that the MAPK signaling pathway and the transcription factor cAMP response element-binding protein (CREB) mediate the effects of BDNF on dendritic length and complexity. However, phosphorylation of CREB alone is not sufficient for the stimulation of dendritic growth by BDNF. Thus, using a mutant form of CREB unable to bind CREB-regulated transcription coactivator (CRTC1), we demonstrate that this effect also requires a functional interaction between CREB and CRTC1. Moreover, inhibition of CRTC1 expression by shRNA-mediated knockdown abolished BDNF-induced dendritic growth of cortical neurons. Interestingly, we found that nuclear translocation of CRTC1 results from activation of NMDA receptors by glutamate, a process that is essential for the effects of BDNF on dendritic development. Together, these data identify a previously unrecognized mechanism by which CREB and the coactivator CRTC1 mediate the effects of BDNF on dendritic growth.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dendrites/metabolism , Glutamic Acid/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Line , Cell Nucleus/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Gene Knockdown Techniques , Glutamic Acid/pharmacology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mutation , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics
15.
Proc Natl Acad Sci U S A ; 104(11): 4700-5, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360587

ABSTRACT

A key feature of memory processes is to link different input signals by association and to preserve this coupling at the level of synaptic connections. Late-phase long-term potentiation (L-LTP), a form of synaptic plasticity thought to encode long-term memory, requires gene transcription and protein synthesis. In this study, we report that a recently cloned coactivator of cAMP-response element-binding protein (CREB), called transducer of regulated CREB activity 1 (TORC1), contributes to this process by sensing the coincidence of calcium and cAMP signals in neurons and by converting it into a transcriptional response that leads to the synthesis of factors required for enhanced synaptic transmission. We provide evidence that TORC1 is involved in L-LTP maintenance at the Schaffer collateral-CA1 synapses in the hippocampus.


Subject(s)
Cyclic AMP/metabolism , Hippocampus/metabolism , Synapses , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , Calcineurin/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Long-Term Potentiation , Male , Mice , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Trans-Activators/physiology , Transcription Factors/physiology
16.
J Biol Chem ; 282(2): 956-67, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17110376

ABSTRACT

Transcription factor function can be modulated by post-translational modifications. Because the transcription factor CCAAT/enhancer-binding protein (C/EBP) beta associates with the nuclear coactivator p300, which contains acetyltransferase activity, acetylation of C/EBPbeta was examined to understand its regulation and function. C/EBPbeta is acetylated by acetyltransferases p300 and p300/CREB-binding protein associated factor. Endogenous C/EBPbeta in 3T3-F442A preadipocytes is also recognized by an acetyl-lysine-specific antibody. Analysis of truncations of C/EBPbeta and peptides based on C/EBPbeta sequences identified multiple lysines within C/EBPbeta that can be acetylated. Among these, a novel acetylation site at lysine 39 of C/EBPbeta was identified. Mutation of Lys-39 to arginine or alanine impairs its acetylation and the ability of C/EBPbeta to activate transcription at the promoters for C/EBPalpha and c-fos. Different C/EBPbeta-responsive promoters require different patterns of acetylated lysines in C/EBPbeta for transcription activation. Furthermore, C/EBPbeta acetylation was increased by growth hormone, and mutation of Lys-39 impaired growth hormone-stimulated c-fos promoter activation. These data suggest that acetylation of Lys-39 of C/EBPbeta, alone or in combination with acetylation at other lysines, may play a role in C/EBPbeta-mediated transcriptional activation.


Subject(s)
CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Transcriptional Activation/physiology , 3T3 Cells , Acetylation , Animals , CCAAT-Binding Factor/chemistry , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Cycle Proteins/metabolism , Growth Hormone/metabolism , Histone Acetyltransferases/metabolism , Humans , In Vitro Techniques , Lysine/metabolism , Mice , Mutagenesis, Site-Directed , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/genetics , Serine/metabolism , Threonine/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors
17.
J Neurochem ; 98(5): 1390-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16771829

ABSTRACT

Dopamine-induced changes in striatal gene expression are thought to play an important role in drug addiction and compulsive behaviour. In this study we report that dopamine induces the expression of the transcription factor CCAAT/Enhancer Binding Protein beta (C/EBP)-beta in primary cultures of striatal neurones. We identified the preprotachykinin-A (PPT-A) gene coding for substance P and neurokinin-A as a potential target gene of C/EBPbeta. We demonstrated that C/EBPbeta physically interacts with an element of the PPT-A promoter, thereby facilitating substance P precursor gene transcription. The regulation of PPT-A gene by C/EBPbeta could subserve many important physiological processes involving substance P, such as nociception, neurogenic inflammation and addiction. Given that substance P is known to increase dopamine signalling in the striatum and, in turn, dopamine increases substance P expression in medium spiny neurones, our results implicate C/EBPbeta in a positive feedback loop, changes of which might contribute to the development of drug addiction.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Corpus Striatum/cytology , Dopamine/pharmacology , Gene Expression/drug effects , Neurons/drug effects , Protein Precursors/metabolism , Signal Transduction/drug effects , Tachykinins/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/pharmacology , CCAAT-Enhancer-Binding Protein-beta/genetics , Cells, Cultured , DNA Footprinting/methods , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Drug Interactions , Electrophoretic Mobility Shift Assay/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/metabolism , Luciferases/metabolism , Mice , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Protein Precursors/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Tachykinins/genetics , Transfection/methods
18.
J Biol Chem ; 278(38): 36959-65, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12857754

ABSTRACT

CCAAT/enhancer-binding protein (C/EBP) family members are transcription factors involved in important physiological processes, such as cellular proliferation and differentiation, regulation of energy homeostasis, inflammation, and hematopoiesis. Transcriptional activation by C/EBPalpha and C/EBPbeta involves the coactivators CREB-binding protein (CBP) and p300, which promote transcription by acetylating histones and recruiting basal transcription factors. In this study, we show that C/EBPdelta is also using CBP as a coactivator. Based on sequence homology with C/EBPalpha and -beta, we identify in C/EBPdelta two conserved amino acid segments that are necessary for the physical interaction with CBP. Using reporter gene assays, we demonstrate that mutation of these residues prevents CBP recruitment and diminishes the transactivating potential of C/EBPdelta. In addition, our results indicate that C/EBP family members not only recruit CBP but specifically induce its phosphorylation. We provide evidence that CBP phosphorylation depends on its interaction with C/EBPdelta and define point mutations within one of the two conserved amino acid segments of C/EBPdelta that abolish CBP phosphorylation as well as transcriptional activation, suggesting that this new mechanism could be important for C/EBP-mediated transcription.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-delta , CREB-Binding Protein , Cell Division , Cell Line , Escherichia coli/metabolism , Genes, Reporter , Glutathione Transferase/metabolism , Humans , Immunoblotting , Luciferases/metabolism , Mutation , PC12 Cells , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Transcription, Genetic , Transcriptional Activation , Transfection
19.
Neuron ; 34(2): 235-44, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11970865

ABSTRACT

Activity-regulated transcription has been implicated in adaptive plasticity in the CNS. In many instances, this plasticity depends upon the transcription factor CREB. Precisely how neuronal activity regulates CREB remains unclear. To address this issue, we examined the phosphorylation state of components of the CREB transcriptional pathway. We show that NMDA activates transcription of CREB-responsive genes in hippocampal neurons, with ERK responsible for persistent CREB phosphorylation and CaM kinase IV (CaMKIV) responsible for phosphorylating the CREB coactivator, CBP. Ser301 of CBP was identified as a major target of CaMKIV phosphorylation in vitro and in vivo. CaM kinase inhibitors attenuated phosphorylation at Ser301 and blocked CBP-dependent transcription. Additionally, mutation of Ser301 impaired NMDA- and CaMKIV-stimulated transcription. These findings demonstrate that activity-induced CaMKIV signaling contributes to CREB/CBP-dependent transcription by phosphorylating CBP at Ser301.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Neurons/physiology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic/physiology , Amino Acid Sequence/genetics , Animals , COS Cells , CREB-Binding Protein , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Cyclic AMP Response Element-Binding Protein/physiology , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/physiology , N-Methylaspartate/pharmacology , Nuclear Proteins/genetics , Phosphorylation/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/physiology , Trans-Activators/genetics , Transcription, Genetic/genetics
20.
J Biol Chem ; 277(12): 10283-91, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11782455

ABSTRACT

MEKK1, a 196-kDa mitogen-activated protein kinase (MAPK) kinase kinase, generates anti-apoptotic signaling as a full-length protein but induces apoptosis when cleaved by caspases. Here, we show that caspase-dependent cleavage of MEKK1 relocalizes the protease-generated 91-kDa kinase fragment from a particulate fraction to a soluble cytoplasmic fraction. Relocalization of MEKK1 catalytic activity is necessary for the pro-apoptotic function of MEKK1. The addition of a membrane-targeting signal to the 91-kDa fragment inhibits caspase activation and the induction of apoptosis but does not change the activation of JNK, ERK, NFkappaB, or p300. These results identify the caspase cleavage of MEKK1 as a dynamic regulatory mechanism that alters the subcellular distribution of MEKK1, changing its function to pro-apoptotic signaling, which does not depend on the currently described MEKK1 effectors.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinase 1 , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Catalysis , Cell Line , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cytoplasm/enzymology , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , E1A-Associated p300 Protein , Humans , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Subcellular Fractions/metabolism , Time Factors , Trans-Activators/metabolism , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...