Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 116(26): 13097-13106, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182610

ABSTRACT

Stress can either promote or impair learning and memory. Such opposing effects depend on whether synapses persist or decay after learning. Maintenance of new synapses formed at the time of learning upon neuronal network activation depends on the stress hormone-activated glucocorticoid receptor (GR) and neurotrophic factor release. Whether and how concurrent GR and neurotrophin signaling integrate to modulate synaptic plasticity and learning is not fully understood. Here, we show that deletion of the neurotrophin brain-derived neurotrophic factor (BDNF)-dependent GR-phosphorylation (PO4) sites impairs long-term memory retention and maintenance of newly formed postsynaptic dendritic spines in the mouse cortex after motor skills training. Chronic stress and the BDNF polymorphism Val66Met disrupt the BDNF-dependent GR-PO4 pathway necessary for preserving training-induced spines and previously acquired memories. Conversely, enrichment living promotes spine formation but fails to salvage training-related spines in mice lacking BDNF-dependent GR-PO4 sites, suggesting it is essential for spine consolidation and memory retention. Mechanistically, spine maturation and persistence in the motor cortex depend on synaptic mobilization of the glutamate receptor subunit A1 (GluA1) mediated by GR-PO4 Together, these findings indicate that regulation of GR-PO4 via activity-dependent BDNF signaling is important for the formation and maintenance of learning-dependent synapses. They also define a signaling mechanism underlying these effects.


Subject(s)
Memory Consolidation/physiology , Motor Cortex/physiopathology , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Stress, Psychological/physiopathology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Circadian Rhythm/physiology , Dendritic Spines/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Glucocorticoids/metabolism , Homeostasis/physiology , Humans , Intravital Microscopy , Male , Mice , Motor Cortex/diagnostic imaging , Phosphorylation/physiology , Polymorphism, Single Nucleotide , Receptors, AMPA/metabolism , Receptors, Glucocorticoid/genetics , Signal Transduction/physiology , Synapses/metabolism
2.
Stress ; 24(2): 130-153, 2021 03.
Article in English | MEDLINE | ID: mdl-32755268

ABSTRACT

The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.


Subject(s)
Receptors, Glucocorticoid , Stress, Psychological , Bayes Theorem , Brain/metabolism , Glucocorticoids , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
3.
J Neurosci ; 38(6): 1335-1350, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29295823

ABSTRACT

The energetic costs of behavioral chronic stress are unlikely to be sustainable without neuronal plasticity. Mitochondria have the capacity to handle synaptic activity up to a limit before energetic depletion occurs. Protective mechanisms driven by the induction of neuronal genes likely evolved to buffer the consequences of chronic stress on excitatory neurons in prefrontal cortex (PFC), as this circuitry is vulnerable to excitotoxic insults. Little is known about the genes involved in mitochondrial adaptation to the buildup of chronic stress. Using combinations of genetic manipulations and stress for analyzing structural, transcriptional, mitochondrial, and behavioral outcomes, we characterized NR4A1 as a stress-inducible modifier of mitochondrial energetic competence and dendritic spine number in PFC. NR4A1 acted as a transcription factor for changing the expression of target genes previously involved in mitochondrial uncoupling, AMP-activated protein kinase activation, and synaptic growth. Maintenance of NR4A1 activity by chronic stress played a critical role in the regressive synaptic organization in PFC of mouse models of stress (male only). Knockdown, dominant-negative approach, and knockout of Nr4a1 in mice and rats (male only) protected pyramidal neurons against the adverse effects of chronic stress. In human PFC tissues of men and women, high levels of the transcriptionally active NR4A1 correlated with measures of synaptic loss and cognitive impairment. In the context of chronic stress, prolonged expression and activity of NR4A1 may lead to responses of mitochondria and synaptic connectivity that do not match environmental demand, resulting in circuit malfunction between PFC and other brain regions, constituting a pathological feature across disorders.SIGNIFICANCE STATEMENT The bioenergetic cost of chronic stress is too high to be sustainable by pyramidal prefrontal neurons. Cellular checkpoints have evolved to adjust the responses of mitochondria and synapses to the buildup of chronic stress. NR4A1 plays such a role by controlling the energetic competence of mitochondria with respect to synapse number. As an immediate-early gene, Nr4a1 promotes neuronal plasticity, but sustained expression or activity can be detrimental. NR4A1 expression and activity is sustained by chronic stress in animal models and in human studies of neuropathologies sensitive to the buildup of chronic stress. Therefore, antagonism of NR4A1 is a promising avenue for preventing the regressive synaptic reorganization in cortical systems in the context of chronic stress.


Subject(s)
Mitochondria/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Prefrontal Cortex/physiopathology , Stress, Psychological/physiopathology , Synapses/metabolism , Aged , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Count , Chronic Disease , Cognition Disorders/etiology , Cognition Disorders/psychology , Dendritic Spines , Female , Gene Expression Regulation/genetics , Hindlimb Suspension , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Prefrontal Cortex/cytology , Pyramidal Cells/physiology , Rats , Stress, Psychological/psychology
4.
Proc Natl Acad Sci U S A ; 113(5): 1423-8, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787858

ABSTRACT

Complications of dopamine replacement for Parkinson's disease (PD) can limit therapeutic options, leading to interest in identifying novel pathways that can be exploited to improve treatment. p11 (S100A10) is a cellular scaffold protein that binds to and potentiates the activity of various ion channels and neurotransmitter receptors. We have previously reported that p11 can influence ventral striatal function in models of depression and drug addiction, and thus we hypothesized that dorsal striatal p11 might mediate motor function and drug responses in parkinsonian mice. To focally inhibit p11 expression in the dorsal striatum, we injected an adeno-associated virus (AAV) vector producing a short hairpin RNA (AAV.sh.p11). This intervention reduced the impairment in motor function on forced tasks, such as rotarod and treadmill tests, caused by substantia nigra lesioning in mice. Measures of spontaneous movement and gait in an open-field test declined as expected in control lesioned mice, whereas AAV.sh.p11 mice remained at or near normal baseline. Mice with unilateral lesions were then challenged with l-dopa (levodopa) and various dopamine receptor agonists, and resulting rotational behaviors were significantly reduced after ipsilateral inhibition of dorsal striatal p11 expression. Finally, p11 knockdown in the dorsal striatum dramatically reduced l-dopa-induced abnormal involuntary movements compared with control mice. These data indicate that focal inhibition of p11 action in the dorsal striatum could be a promising PD therapeutic target to improve motor function while reducing l-dopa-induced dyskinesias.


Subject(s)
Annexin A2/genetics , Corpus Striatum/physiology , Dyskinesias/physiopathology , Genetic Therapy , Motor Activity , Parkinsonian Disorders/physiopathology , S100 Proteins/genetics , Animals , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/therapy
5.
Proc Natl Acad Sci U S A ; 112(51): 15737-42, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26630005

ABSTRACT

Neurotrophins and glucocorticoids are robust synaptic modifiers, and deregulation of their activities is a risk factor for developing stress-related disorders. Low levels of brain-derived neurotrophic factor (BDNF) increase the desensitization of glucocorticoid receptors (GR) and vulnerability to stress, whereas higher levels of BDNF facilitate GR-mediated signaling and the response to antidepressants. However, the molecular mechanism underlying neurotrophic-priming of GR function is poorly understood. Here we provide evidence that activation of a TrkB-MAPK pathway, when paired with the deactivation of a GR-protein phosphatase 5 pathway, resulted in sustained GR phosphorylation at BDNF-sensitive sites that is essential for the transcription of neuronal plasticity genes. Genetic strategies that disrupted GR phosphorylation or TrkB signaling in vivo impaired the neuroplasticity to chronic stress and the effects of the antidepressant fluoxetine. Our findings reveal that the coordinated actions of BDNF and glucocorticoids promote neuronal plasticity and that disruption in either pathway could set the stage for the development of stress-induced psychiatric diseases.


Subject(s)
Antidepressive Agents/pharmacology , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/physiology , Signal Transduction/physiology , Stress, Psychological/physiopathology , Animals , Brain-Derived Neurotrophic Factor/physiology , Female , Fluoxetine/pharmacology , MAP Kinase Signaling System , Membrane Glycoproteins/physiology , Mice , Neuronal Plasticity/drug effects , Phosphorylation , Protein-Tyrosine Kinases/physiology , Rats , Rats, Sprague-Dawley , Receptor, trkB
6.
Neurobiol Dis ; 88: 107-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26774030

ABSTRACT

Clinical and experimental evidence point to a possible role of cerebrovascular dysfunction in Alzheimer's disease (AD). The 5xFAD mouse model of AD expresses human amyloid precursor protein and presenilin genes with mutations found in AD patients. It remains unknown whether amyloid deposition driven by these mutations is associated with cerebrovascular changes. 5xFAD and wild type mice (2 to 12months old; M2 to M12) were used. Thinned skull in vivo 2-photon microscopy was used to determine Aß accumulation on leptomeningeal or superficial cortical vessels over time. Parenchymal microvascular damage was assessed using FITC-microangiography. Collagen-IV and CD31 were used to stain basal lamina and endothelial cells. Methoxy-XO4, Thioflavin-S or 6E10 were used to visualize Aß accumulation in living mice or in fixed brain tissues. Positioning of reactive IBA1 microglia and GFAP astrocytes at the vasculature was rendered using confocal microscopy. Platelet-derived growth factor receptor beta (PDGFRß) staining was used to visualize perivascular pericytes. In vivo 2-photon microscopy revealed Methoxy-XO4(+) amyloid perivascular deposits on leptomeningeal and penetrating cortical vessels in 5xFAD mice, typical of cerebral amyloid angiopathy (CAA). Amyloid deposits were visible in vivo at M3 and aggravated over time. Progressive microvascular damage was concomitant to parenchymal Aß plaque accumulation in 5xFAD mice. Microvascular inflammation in 5xFAD mice presented with sporadic FITC-albumin leakages at M4 becoming more prevalent at M9 and M12. 3D colocalization showed inflammatory IBA1(+) microglia proximal to microvascular FITC-albumin leaks. The number of perivascular PDGFRß(+) pericytes was significantly decreased at M4 in the fronto-parietal cortices, with a trend decrease observed in the other structures. At M9-M12, PDGFRß(+) pericytes displayed hypertrophic perivascular ramifications contiguous to reactive microglia. Cerebral amyloid angiopathy and microvascular inflammation occur in 5xFAD mice concomitantly to parenchymal plaque deposition. The prospect of cerebrovascular pharmacology in AD is discussed.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Blood Vessels/pathology , Cerebrovascular Circulation/genetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Calcium-Binding Proteins/metabolism , Collagen Type IV/metabolism , Disease Models, Animal , Disease Progression , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Pericytes/metabolism , Pericytes/pathology , Plaque, Amyloid/metabolism , Platelet Endothelial Cell Adhesion Molecule-1 , Presenilin-1/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism
7.
Pharmacol Res ; 113(Pt A): 1-17, 2016 11.
Article in English | MEDLINE | ID: mdl-27498156

ABSTRACT

Glucocorticoid actions are tailored to the organs and cells responding thanks to complex integration with ongoing signaling mediated by cytokines, hormones, neurotransmitters, and growth factors. Disruption of: (1) the amount of signaling molecules available locally; (2) the timing with other signaling pathways; (3) the post-translational modifications on glucocorticoid receptors; and (4) the receptors-interacting proteins within cellular organelles and functional compartments, can modify the sensitivity and efficacy of glucocorticoid responses with implications in physiology, diseases and treatments. Tissue sensitivity to glucocorticoids is sustained by multiple systems that do not operate in isolation. We take the example of the interplay between the glucocorticoid and brain-derived neurotrophic factor signaling pathways to deconstruct context-dependent glucocorticoid responses that play key roles in physiology, diseases and therapies.


Subject(s)
Cytokines/metabolism , Glucocorticoids/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neurotransmitter Agents/metabolism , Signal Transduction/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Humans , Protein Processing, Post-Translational/physiology , Receptors, Glucocorticoid/metabolism
8.
Neural Plast ; 2016: 3985063, 2016.
Article in English | MEDLINE | ID: mdl-26885402

ABSTRACT

The brain evolved cellular mechanisms for adapting synaptic function to energy supply. This is particularly evident when homeostasis is challenged by stress. Signaling loops between the mitochondria and synapses scale neuronal connectivity with bioenergetics capacity. A biphasic "inverted U shape" response to the stress hormone glucocorticoids is demonstrated in mitochondria and at synapses, modulating neural plasticity and physiological responses. Low dose enhances neurotransmission, synaptic growth, mitochondrial functions, learning, and memory whereas chronic, higher doses produce inhibition of these functions. The range of physiological effects by stress and glucocorticoid depends on the dose, duration, and context at exposure. These criteria are met by neuronal activity and the circadian, stress-sensitive and ultradian, stress-insensitive modes of glucocorticoid secretion. A major hallmark of stress-related neuropsychiatric disorders is the disrupted glucocorticoid rhythms and tissue resistance to signaling with the glucocorticoid receptor (GR). GR resistance could result from the loss of context-dependent glucocorticoid signaling mediated by the downregulation of the activity-dependent neurotrophin BDNF. The coincidence of BDNF and GR signaling changes glucocorticoid signaling output with consequences on mitochondrial respiration efficiency, synaptic plasticity, and adaptive trajectories.


Subject(s)
Mental Disorders/metabolism , Stress, Psychological/metabolism , Brain/metabolism , Humans , Mitochondria , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/metabolism
9.
Adv Exp Med Biol ; 872: 33-57, 2015.
Article in English | MEDLINE | ID: mdl-26215989

ABSTRACT

Well-defined as signaling hormones for the programming of cell type-specific and context-dependent gene expression signatures, glucocorticoids control experience-driven allostasis. One unifying model is that glucocorticoids help maintaining the integrity and plasticity of cellular networks in changing environments through the mobilization of cellular energy stores, profiling of gene expression, and changes in the electrical and morphological properties of cells. The nucleus is their primary site of action, yet recent discoveries point to additional gene transcription-independent functions at the plasma membrane of neuronal synapses. Glucocorticoids are secreted factors that reflect intrinsically the changes coming from the external world, temporally and regionally, during development and adulthood. In this review, we will enumerate the properties and signaling attributes of glucocorticoids and their receptors that characterize them as allostatic modulators. The molecular mechanisms used to support their role at the synapse will be highlighted.


Subject(s)
Glucocorticoids/metabolism , Signal Transduction , Allosteric Regulation , Animals , Biological Availability , Humans , Phosphorylation , Receptors, Glucocorticoid/metabolism
10.
Proc Natl Acad Sci U S A ; 109(28): 11360-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733786

ABSTRACT

A large number of studies have demonstrated that the nucleus accumbens (NAC) is a critical site in the neuronal circuits controlling reward responses, motivation, and mood, but the neuronal cell type(s) underlying these processes are not yet known. Identification of the neuronal cell types that regulate depression-like states will guide us in understanding the biological basis of mood and its regulation by diseases like major depressive disorder. Taking advantage of recent findings demonstrating that the serotonin receptor chaperone, p11, is an important molecular regulator of depression-like states, here we identify cholinergic interneurons (CINs) as a primary site of action for p11 in the NAC. Depression-like behavior is observed in mice after decrease of p11 levels in NAC CINs. This phenotype is recapitulated by silencing neuronal transmission in these cells, demonstrating that accumbal cholinergic neuronal activity regulates depression-like behaviors and suggesting that accumbal CIN activity is crucial for the regulation of mood and motivation.


Subject(s)
Annexin A2/metabolism , Depression/physiopathology , Interneurons/metabolism , Nucleus Accumbens/metabolism , S100 Proteins/metabolism , Acetylcholine/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Depression/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Chaperones/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Receptors, Cholinergic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL