Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 364
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Psychiatry ; 27(5): 2563-2579, 2022 05.
Article in English | MEDLINE | ID: mdl-33931727

ABSTRACT

Heightened aggressive behavior is considered as one of the central symptoms of many neuropsychiatric disorders including autism, schizophrenia, and dementia. The consequences of aggression pose a heavy burden on patients and their families and clinicians. Unfortunately, we have limited treatment options for aggression and lack mechanistic insight into the causes of aggression needed to inform new efforts in drug discovery and development. Levels of proinflammatory cytokines in the periphery or cerebrospinal fluid were previously reported to correlate with aggressive traits in humans. However, it is still unknown whether cytokines affect brain circuits to modulate aggression. Here, we examined the functional role of interleukin 1ß (IL-1ß) in mediating individual differences in aggression using a resident-intruder mouse model. We found that nonaggressive mice exhibit higher levels of IL-1ß in the dorsal raphe nucleus (DRN), the major source of forebrain serotonin (5-HT), compared to aggressive mice. We then examined the effect of pharmacological antagonism and viral-mediated gene knockdown of the receptors for IL-1 within the DRN and found that both treatments consistently increased aggressive behavior of male mice. Aggressive mice also exhibited higher c-Fos expression in 5-HT neurons in the DRN compared to nonaggressive mice. In line with these findings, deletion of IL-1 receptor in the DRN enhanced c-Fos expression in 5-HT neurons during aggressive encounters, suggesting that modulation of 5-HT neuronal activity by IL-1ß signaling in the DRN controls expression of aggressive behavior.


Subject(s)
Aggression , Dorsal Raphe Nucleus , Interleukin-1beta , Serotonin , Aggression/physiology , Animals , Dorsal Raphe Nucleus/metabolism , Humans , Individuality , Interleukin-1beta/metabolism , Male , Mice , Serotonin/metabolism
2.
Proc Natl Acad Sci U S A ; 117(1): 650-655, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31843894

ABSTRACT

Even a brief exposure to severe stress strengthens synaptic connectivity days later in the amygdala, a brain area implicated in the affective symptoms of stress-related psychiatric disorders. However, little is known about the synaptic signaling mechanisms during stress that eventually culminate in its delayed impact on the amygdala. Hence, we investigated early stress-induced changes in amygdalar synaptic signaling in order to prevent its delayed effects. Whole-cell recordings in basolateral amygdala (BLA) slices from rats revealed higher frequency of miniature excitatory postsynaptic currents (mEPSCs) immediately after 2-h immobilization stress. This was replicated by inhibition of cannabinoid receptors (CB1R), suggesting a role for endocannabinoid (eCB) signaling. Stress also reduced N-arachidonoylethanolamine (AEA), an endogenous ligand of CB1R. Since stress-induced activation of fatty acid amide hydrolase (FAAH) reduces AEA, we confirmed that oral administration of an FAAH inhibitor during stress prevents the increase in synaptic excitation in the BLA soon after stress. Although stress also caused an immediate reduction in synaptic inhibition, this was not prevented by FAAH inhibition. Strikingly, FAAH inhibition during the traumatic stressor was also effective 10 d later on the delayed manifestation of synaptic strengthening in BLA neurons, preventing both enhanced mEPSC frequency and increased dendritic spine-density. Thus, oral administration of an FAAH inhibitor during a brief stress prevents the early synaptic changes that eventually build up to hyperexcitability in the amygdala. This framework is of therapeutic relevance because of growing interest in targeting eCB signaling to prevent the gradual development of emotional symptoms and underlying amygdalar dysfunction triggered by traumatic stress.


Subject(s)
Basolateral Nuclear Complex/physiology , Emotions/drug effects , Endocannabinoids/metabolism , Signal Transduction/physiology , Stress, Psychological/physiopathology , Administration, Oral , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Arachidonic Acids , Cannabinoid Receptor Antagonists/administration & dosage , Disease Models, Animal , Emotions/physiology , Enzyme Inhibitors/administration & dosage , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Humans , Male , Polyunsaturated Alkamides , Rats , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/psychology
3.
J Neurosci ; 41(24): 5190-5205, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33941651

ABSTRACT

Hypertension susceptibility in women increases at the transition to menopause, termed perimenopause, a state characterized by erratic estrogen fluctuation and extended hormone cycles. Elucidating the role of estrogen signaling in the emergence of hypertension during perimenopause has been hindered by animal models that are confounded by abrupt estrogen cessation or effects of aging. In the present study, accelerated ovarian failure (AOF) in estrogen receptor ß (ERß) reporter mice was induced by 4-vinylcyclohexene diepoxide in young mice to model early-stage ovarian failure (peri-AOF) characteristic of peri-menopause. It was found that administering ERß agonists suppressed elevated blood pressure in a model of neurogenic hypertension induced by angiotensin II (AngII) in peri-AOF, but not in age-matched male mice. It was also found that ERß agonist administration in peri-AOF females, but not males, suppressed the heightened NMDAR signaling and reactive oxygen production in ERß neurons in the hypothalamic paraventricular nucleus (PVN), a critical neural regulator of blood pressure. It was further shown that deleting ERß in the PVN of gonadally intact females produced a phenotype marked by a sensitivity to AngII hypertension. These results suggest that ERß signaling in the PVN plays an important role in blood pressure regulation in female mice and contributes to hypertension susceptibility in females at an early stage of ovarian failure comparable to human perimenopause.


Subject(s)
Estrogen Receptor beta/metabolism , Hypertension/metabolism , Neuronal Plasticity/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Perimenopause/metabolism , Animals , Disease Models, Animal , Female , Hypertension/etiology , Mice , Mice, Inbred C57BL
4.
Physiol Rev ; 95(3): 785-807, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26109339

ABSTRACT

Estrogen facilitates higher cognitive functions by exerting effects on brain regions such as the prefrontal cortex and hippocampus. Estrogen induces spinogenesis and synaptogenesis in these two brain regions and also initiates a complex set of signal transduction pathways via estrogen receptors (ERs). Along with the classical genomic effects mediated by activation of ER α and ER ß, there are membrane-bound ER α, ER ß, and G protein-coupled estrogen receptor 1 (GPER1) that can mediate rapid nongenomic effects. All key ERs present throughout the body are also present in synapses of the hippocampus and prefrontal cortex. This review summarizes estrogen actions in the brain from the standpoint of their effects on synapse structure and function, noting also the synergistic role of progesterone. We first begin with a review of ER subtypes in the brain and how their abundance and distributions are altered with aging and estrogen loss (e.g., ovariectomy or menopause) in the rodent, monkey, and human brain. As there is much evidence that estrogen loss induced by menopause can exacerbate the effects of aging on cognitive functions, we then review the clinical trials of hormone replacement therapies and their effectiveness on cognitive symptoms experienced by women. Finally, we summarize studies carried out in nonhuman primate models of age- and menopause-related cognitive decline that are highly relevant for developing effective interventions for menopausal women. Together, we highlight a new understanding of how estrogen affects higher cognitive functions and synaptic health that go well beyond its effects on reproduction.


Subject(s)
Aging/metabolism , Aging/psychology , Brain/metabolism , Cognition , Estrogens/metabolism , Synapses/metabolism , Synaptic Transmission , Age Factors , Animals , Cognition Disorders/metabolism , Cognition Disorders/psychology , Female , Humans , Male , Menopause/metabolism , Menopause/psychology , Progesterone/metabolism , Receptors, Estrogen/metabolism
5.
Mol Psychiatry ; 26(9): 5140-5149, 2021 09.
Article in English | MEDLINE | ID: mdl-32536688

ABSTRACT

Insulin signaling is critical for neuroplasticity, cerebral metabolism as well as for systemic energy metabolism. In rodent studies, impaired brain insulin signaling with resultant insulin resistance (IR) modulates synaptic plasticity and the corresponding behavioral functions. Despite discoveries of central actions of insulin, in vivo molecular mechanisms of brain IR until recently have proven difficult to study in the human brain. In the current study, we leveraged recent technological advances in molecular biology and herein report an increased number of exosomes enriched for L1CAM, a marker predominantly expressed in the brain, in subjects with major depressive disorder (MDD) as compared with age- and sex-matched healthy controls (HC). We also report increased concentration of the insulin receptor substrate-1 (IRS-1) in L1CAM+ exosomes in subjects with MDD as compared with age- and sex-matched HC. We found a relationship between expression of IRS-1 in L1CAM+ exosomes and systemic IR as assessed by homeostatic model assessment of IR in HC, but not in subjects with MDD. The increased IRS-1 levels in L1CAM+ exosomes were greater in subjects with MDD and were associated with suicidality and anhedonia. Finally, our data suggested sex differences in serine-312 phosphorylation of IRS-1 in L1CAM+ exosomes in subjects with MDD. These findings provide a starting point for creating mechanistic framework of brain IR in further development of personalized medicine strategies to effectively treat MDD.


Subject(s)
Depressive Disorder, Major , Exosomes , Insulin Resistance , Brain/metabolism , Depression , Depressive Disorder, Major/metabolism , Exosomes/metabolism , Female , Humans , Insulin/metabolism , Male , Phosphoproteins/metabolism , Phosphorylation , Receptor, Insulin/metabolism
6.
Brain ; 144(12): 3742-3755, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34145880

ABSTRACT

Dysregulation of glutamatergic neural circuits has been implicated in a cycle of toxicity, believed among the neurobiological underpinning of Alzheimer's disease. Previously, we reported preclinical evidence that the glutamate modulator riluzole, which is FDA approved for the treatment of amyotrophic lateral sclerosis, has potential benefits on cognition, structural and molecular markers of ageing and Alzheimer's disease. The objective of this study was to evaluate in a pilot clinical trial, using neuroimaging biomarkers, the potential efficacy and safety of riluzole in patients with Alzheimer's disease as compared to placebo. A 6-month phase 2 double-blind, randomized, placebo-controlled study was conducted at two sites. Participants consisted of males and females, 50 to 95 years of age, with a clinical diagnosis of probable Alzheimer's disease, and Mini-Mental State Examination between 19 and 27. Ninety-four participants were screened, 50 participants who met inclusion criteria were randomly assigned to receive 50 mg riluzole (n = 26) or placebo (n = 24) twice a day. Twenty-two riluzole-treated and 20 placebo participants completed the study. Primary end points were baseline to 6 months changes in (i) cerebral glucose metabolism as measured with fluorodeoxyglucose-PET in prespecified regions of interest (hippocampus, posterior cingulate, precuneus, lateral temporal, inferior parietal, frontal); and (ii) changes in posterior cingulate levels of the neuronal viability marker N-acetylaspartate as measured with in vivo proton magnetic resonance spectroscopy. Secondary outcome measures were neuropsychological testing for correlation with neuroimaging biomarkers and in vivo measures of glutamate in posterior cingulate measured with magnetic resonance spectroscopy as a potential marker of target engagement. Measures of cerebral glucose metabolism, a well-established Alzheimer's disease biomarker and predictor of disease progression, declined significantly less in several prespecified regions of interest with the most robust effect in posterior cingulate, and effects in precuneus, lateral temporal, right hippocampus and frontal cortex in riluzole-treated participants in comparison to the placebo group. No group effect was found in measures of N-acetylaspartate levels. A positive correlation was observed between cognitive measures and regional cerebral glucose metabolism. A group × visit interaction was observed in glutamate levels in posterior cingulate, potentially suggesting engagement of glutamatergic system by riluzole. In vivo glutamate levels positively correlated with cognitive performance. These findings support our main primary hypothesis that cerebral glucose metabolism would be better preserved in the riluzole-treated group than in the placebo group and provide a rationale for more powered, longer duration studies of riluzole as a potential intervention for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Glucose/metabolism , Neuroprotective Agents/therapeutic use , Riluzole/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Brain/metabolism , Double-Blind Method , Female , Humans , Male , Middle Aged
7.
Proc Natl Acad Sci U S A ; 116(45): 22821-22832, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31636210

ABSTRACT

Infant maltreatment increases vulnerability to physical and mental disorders, yet specific mechanisms embedded within this complex infant experience that induce this vulnerability remain elusive. To define critical features of maltreatment-induced vulnerability, rat pups were reared from postnatal day 8 (PN8) with a maltreating mother, which produced amygdala and hippocampal deficits and decreased social behavior at PN13. Next, we deconstructed the maltreatment experience to reveal sufficient and necessary conditions to induce this phenotype. Social behavior and amygdala deficits (volume, neurogenesis, c-Fos, local field potential) required combined chronic high corticosterone and maternal presence (not maternal behavior). Hippocampal deficits were induced by chronic high corticosterone regardless of social context. Causation was shown by blocking corticosterone during maltreatment and suppressing amygdala activity during social behavior testing. These results highlight (1) that early life maltreatment initiates multiple pathways to pathology, each with distinct causal mechanisms and outcomes, and (2) the importance of social presence on brain development.


Subject(s)
Amygdala/physiopathology , Hippocampus/physiopathology , Mothers/psychology , Social Behavior , Stress, Physiological , Animals , Corticosterone/administration & dosage , Corticosterone/blood , Female , Rats
8.
Proc Natl Acad Sci U S A ; 116(43): 21800-21811, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591195

ABSTRACT

The excitatory amino acid transporter 2 (EAAT2) is the major glutamate transporter in the brain expressed predominantly in astrocytes and at low levels in neurons and axonal terminals. EAAT2 expression is reduced in aging and sporadic Alzheimer's disease (AD) patients' brains. The role EAAT2 plays in cognitive aging and its associated mechanisms remains largely unknown. Here, we show that conditional deletion of astrocytic and neuronal EAAT2 results in age-related cognitive deficits. Astrocytic, but not neuronal EAAT2, deletion leads to early deficits in short-term memory and in spatial reference learning and long-term memory. Neuronal EAAT2 loss results in late-onset spatial reference long-term memory deficit. Neuronal EAAT2 deletion leads to dysregulation of the kynurenine pathway, and astrocytic EAAT2 deficiency results in dysfunction of innate and adaptive immune pathways, which correlate with cognitive decline. Astrocytic EAAT2 deficiency also shows transcriptomic overlaps with human aging and AD. Overall, the present study shows that in addition to the widely recognized astrocytic EAAT2, neuronal EAAT2 plays a role in hippocampus-dependent memory. Furthermore, the gene expression profiles associated with astrocytic and neuronal EAAT2 deletion are substantially different, with the former associated with inflammation and synaptic function similar to changes observed in human AD and gene expression changes associated with inflammation similar to the aging human brain.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Cognitive Dysfunction/pathology , Excitatory Amino Acid Transporter 2/deficiency , Memory Disorders/pathology , Neurons/metabolism , Adult , Aged, 80 and over , Aging/physiology , Animals , Cognition/physiology , Cognitive Dysfunction/genetics , Excitatory Amino Acid Transporter 2/genetics , Hippocampus/physiology , Humans , Kynurenine/metabolism , Male , Memory Disorders/genetics , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Mice , Mice, Knockout , Middle Aged , Young Adult
9.
Proc Natl Acad Sci U S A ; 116(22): 10988-10993, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31085646

ABSTRACT

Regular exercise and dietary supplements with antioxidants each have the potential to improve cognitive function and attenuate cognitive decline, and, in some cases, they enhance each other. Our current results reveal that low-intensity exercise (mild exercise, ME) and the natural antioxidant carotenoid astaxanthin (AX) each have equivalent beneficial effects on hippocampal neurogenesis and memory function. We found that the enhancement by ME combined with AX in potentiating hippocampus-based plasticity and cognition is mediated by leptin (LEP) made and acting in the hippocampus. In assessing the combined effects upon wild-type (WT) mice undergoing ME with or without an AX diet for four weeks, we found that, when administrated alone, ME and AX separately enhanced neurogenesis and spatial memory, and when combined they were at least additive in their effects. DNA microarray and bioinformatics analyses revealed not only the up-regulation of an antioxidant gene, ABHD3, but also that the up-regulation of LEP gene expression in the hippocampus of WT mice with ME alone is further enhanced by AX. Together, they also increased hippocampal LEP (h-LEP) protein levels and enhanced spatial memory mediated through AKT/STAT3 signaling. AX treatment also has direct action on human neuroblastoma cell lines to increase cell viability associated with increased LEP expression. In LEP-deficient mice (ob/ob), chronic infusion of LEP into the lateral ventricles restored the synergy. Collectively, our findings suggest that not only h-LEP but also exogenous LEP mediates effects of ME on neural functions underlying memory, which is further enhanced by the antioxidant AX.


Subject(s)
Antioxidants/pharmacology , Hippocampus , Leptin/metabolism , Neurogenesis/drug effects , Physical Conditioning, Animal , Spatial Memory/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice , Xanthophylls/pharmacology
10.
J Neurosci ; 40(1): 12-21, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31896560

ABSTRACT

Over the last 50 years, the concept of stress has evolved significantly, and our understanding of the underlying neurobiology has expanded dramatically. Rather than consider stress biology to be relevant only under unusual and threatening conditions, we conceive of it as an ongoing, adaptive process of assessing the environment, coping with it, and enabling the individual to anticipate and deal with future challenges. Though much remains to be discovered, the fundamental neurocircuitry that underlies these processes has been broadly delineated, key molecular players have been identified, and the impact of this system on neuroplasticity has been well established. More recently, we have come to appreciate the critical interaction between the brain and the rest of the body as it pertains to stress responsiveness. Importantly, this system can become overloaded due to ongoing environmental demands on the individual, be they physical, physiological, or psychosocial. The impact of this overload is deleterious to brain health, and it results in vulnerability to a range of brain disorders, including major depression and cognitive deficits. Thus, stress biology is one of the best understood systems in affective neuroscience and is an ideal target for addressing the pathophysiology of many brain-related diseases. The story we present began with the discovery of glucocorticoid receptors in hippocampus and has extended to other brain regions in both animal models and the human brain with the further discovery of structural and functional adaptive plasticity in response to stressful and other experiences.


Subject(s)
Brain/physiology , Glucocorticoids/physiology , Mood Disorders/physiopathology , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Adaptation, Physiological/physiology , Animals , Endocannabinoids/physiology , Epigenesis, Genetic , Feedback, Physiological , Fibroblast Growth Factor 2/physiology , Fibroblast Growth Factor 2/therapeutic use , Gene Expression Regulation/physiology , Hormones/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Intercellular Signaling Peptides and Proteins/physiology , Life Change Events , Models, Neurological , Models, Psychological , Mood Disorders/etiology , Mood Disorders/psychology , Nerve Tissue Proteins/physiology , Neuronal Plasticity , Pituitary-Adrenal System/physiology , Psychophysiology , Receptors, Cell Surface/physiology , Social Determinants of Health
11.
Synapse ; 75(10): e22218, 2021 10.
Article in English | MEDLINE | ID: mdl-34255372

ABSTRACT

Our prior studies demonstrated that the rat hippocampal opioid system can undergo sex-specific adaptations to external stimuli that can influence opioid-associated learning processes. This opioid system extensively overlaps with the cannabinoid system. Moreover, acute administration of Δ9 Tetrahydrocannabinoid (THC), the primary psychoactive constituent of cannabis, can alter cognitive behaviors that involve the hippocampus. Here, we use light and electron microscopic immunocytochemical methods to examine the effects of acute THC (5 mg/kg, i.p., 1 h) on mossy fiber Leu-Enkephalin (LEnk) levels and the distribution and phosphorylation levels of delta and mu opioid receptors (DORs and MORs, respectively) in CA3 pyramidal cells and parvalbumin dentate hilar interneurons of adult female and male Sprague-Dawley rats. In females with elevated estrogen states (proestrus/estrus stage), acute THC altered the opioid system so that it resembled that seen in vehicle-injected females with low estrogen states (diestrus) and males: (1) mossy fiber LEnk levels in CA2/3a decreased; (2) phosphorylated-DOR levels in CA2/3a pyramidal cells increased; and (3) phosphorylated-MOR levels increased in most CA3b laminae. In males, acute THC resulted in the internalization of MORs in parvalbumin-containing interneuron dendrites which would decrease disinhibition of granule cells. In both sexes, acute THC redistributed DORs to the near plasma membrane of CA3 pyramidal cell dendrites, however, the dendritic region varied with sex. Additionally, acute THC also resulted in a sex-specific redistribution of DORs within CA3 pyramidal cell dendrites which could differentially promote synaptic plasticity and/or opioid-associated learning processes in both females and males.


Subject(s)
Analgesics, Opioid , Dronabinol , Analgesics, Opioid/pharmacology , Animals , Dronabinol/pharmacology , Female , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
12.
Synapse ; 75(1): e22182, 2021 01.
Article in English | MEDLINE | ID: mdl-32654187

ABSTRACT

Oxycodone (Oxy) conditioned place preference (CPP) in Sprague Dawley rats results in sex-specific alterations in hippocampal opioid circuits in a manner that facilitates opioid-associative learning processes, particularly in females. Here, we examined if Oxy (3 mg/kg, I.P.) or saline (Sal) injections not paired with behavioral testing similarly affect the hippocampal opioid system. Sal-injected females compared to Sal-injected males had: (1) higher densities of cytoplasmic delta opioid receptors (DOR) in GABAergic hilar dendrites suggesting higher baseline reserve DOR pools and (2) elevated phosphorylated DOR levels, but lower phosphorylated mu opioid receptor (MOR) levels in CA3a suggesting that the baseline pools of activated opioid receptors vary in females and males. In contrast to CPP studies, Oxy-injections in the absence of behavioral tests resulted in few changes in the hippocampal opioid system in either females or males. Specifically, Oxy-injected males compared to Sal-injected males had fewer DORs near the plasma membrane of CA3 pyramidal cell dendrites and in CA3 dendritic spines contacted by mossy fibers, and lower pMOR levels in CA3a. Oxy-injected females compared to Sal-injected females had higher total DORs in GABAergic dendrites and lower total MORs in parvalbumin-containing dendrites. Thus, unlike Oxy CPP, Oxy-injections redistributed opioid receptors in hippocampal neurons in a manner that would either decrease (males) or not alter (females) excitability and plasticity processes. These results indicate that the majority of changes within hippocampal opioid circuits that would promote opioid-associative learning processes in both females and males do not occur with Oxy administration alone, and instead must be paired with CPP.


Subject(s)
Conditioning, Classical/physiology , Hippocampus/metabolism , Oxycodone/administration & dosage , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Sex Characteristics , Analgesics, Opioid/administration & dosage , Animals , Conditioning, Classical/drug effects , Female , Hippocampus/drug effects , Hippocampus/ultrastructure , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists
13.
Mol Psychiatry ; 25(3): 572-583, 2020 03.
Article in English | MEDLINE | ID: mdl-30356121

ABSTRACT

Premenstrual dysphoric disorder (PMDD) affects over 5% of women, with symptoms similar to anxiety and major depression, and is associated with differential sensitivity to circulating ovarian hormones. Little is known about the genetic and epigenetic factors that increase the risk to develop PMDD. We report that 17ß-estradiol (E2) affects the behavior and the epigenome in a mouse model carrying a single-nucleotide polymorphism of the brain-derived neurotrophic factor gene (BDNF Val66Met), in a way that recapitulates the hallmarks of PMDD. Ovariectomized mice heterozygous for the BDNF Met allele (Het-Met) and their matched wild-type (WT) mice were administered estradiol or vehicle in drinking water for 6 weeks. Using the open field and the splash test, we show that E2 add-back induces anxiety-like and depression-like behavior in Het-Met mice, but not in WT mice. RNA-seq of the ventral hippocampus (vHpc) highlights that E2-dependent gene expression is markedly different between WT mice and Het-Met mice. Through a comparative whole-genome RNA-seq analysis between mouse vHpc and lymphoblastoid cell line cultures from control women and women with PMDD, we discovered common epigenetic biomarkers that transcend species and cell types. Those genes include epigenetic modifiers of the ESC/E(Z) complex, an effector of response to ovarian steroids. Although the BDNF Met genotype intersects the behavioral and transcriptional traits of women with PMDD, we suggest that these similarities speak to the epigenetic factors by which ovarian steroids produce negative behavioral effects.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Premenstrual Dysphoric Disorder/drug therapy , Premenstrual Dysphoric Disorder/genetics , Adult , Animals , Anxiety/genetics , Brain-Derived Neurotrophic Factor/metabolism , Epigenesis, Genetic/genetics , Epigenomics/methods , Estradiol/pharmacology , Estrogens , Female , Gene Expression Profiling/methods , Gene Knock-In Techniques , Genotype , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Ovary/metabolism , Polymorphism, Single Nucleotide/genetics , Premenstrual Dysphoric Disorder/physiopathology , Transcriptome/genetics
14.
Proc Natl Acad Sci U S A ; 115(34): 8627-8632, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30061399

ABSTRACT

The lack of biomarkers to identify target populations greatly limits the promise of precision medicine for major depressive disorder (MDD), a primary cause of ill health and disability. The endogenously produced molecule acetyl-l-carnitine (LAC) is critical for hippocampal function and several behavioral domains. In rodents with depressive-like traits, LAC levels are markedly decreased and signal abnormal hippocampal glutamatergic function and dendritic plasticity. LAC supplementation induces rapid and lasting antidepressant-like effects via epigenetic mechanisms of histone acetylation. This mechanistic model led us to evaluate LAC levels in humans. We found that LAC levels, and not those of free carnitine, were decreased in patients with MDD compared with age- and sex-matched healthy controls in two independent study centers. Secondary exploratory analyses showed that the degree of LAC deficiency reflected both the severity and age of onset of MDD. Moreover, these analyses showed that the decrease in LAC was larger in patients with a history of treatment-resistant depression (TRD), among whom childhood trauma and, specifically, a history of emotional neglect and being female, predicted the decreased LAC. These findings suggest that LAC may serve as a candidate biomarker to help diagnose a clinical endophenotype of MDD characterized by decreased LAC, greater severity, and earlier onset as well as a history of childhood trauma in patients with TRD. Together with studies in rodents, these translational findings support further exploration of LAC as a therapeutic target that may help to define individualized treatments in biologically based depression subtype consistent with the spirit of precision medicine.


Subject(s)
Acetylcarnitine/blood , Acetylcarnitine/deficiency , Depressive Disorder, Major/blood , Adult , Age Factors , Aged , Carnitine/blood , Female , Humans , Male , Middle Aged , Sex Factors
15.
Proc Natl Acad Sci U S A ; 115(29): 7605-7610, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967158

ABSTRACT

Endocannabinoid signaling regulates feeding and metabolic processes and has been linked to obesity development. Several hormonal signals, such as glucocorticoids and ghrelin, regulate feeding and metabolism by engaging the endocannabinoid system. Similarly, studies have suggested that leptin interacts with the endocannabinoid system, yet the mechanism and functional relevance of this interaction remain elusive. Therefore, we explored the interaction between leptin and endocannabinoid signaling with a focus on fatty acid amide hydrolase (FAAH), the primary degradative enzyme for the endocannabinoid N-arachidonoylethanolamine (anandamide; AEA). Mice deficient in leptin exhibited elevated hypothalamic AEA levels and reductions in FAAH activity while leptin administration to WT mice reduced AEA content and increased FAAH activity. Following high fat diet exposure, mice developed resistance to the effects of leptin administration on hypothalamic AEA content and FAAH activity. At a functional level, pharmacological inhibition of FAAH was sufficient to prevent leptin-mediated effects on body weight and food intake. Using a novel knock-in mouse model recapitulating a common human polymorphism (FAAH C385A; rs324420), which reduces FAAH activity, we investigated whether human genetic variance in FAAH affects leptin sensitivity. While WT (CC) mice were sensitive to leptin-induced reductions in food intake and body weight gain, low-expressing FAAH (AA) mice were unresponsive. These data demonstrate that FAAH activity is required for leptin's hypophagic effects and, at a translational level, suggest that a genetic variant in the FAAH gene contributes to differences in leptin sensitivity in human populations.


Subject(s)
Amidohydrolases/metabolism , Arachidonic Acids/metabolism , Eating , Endocannabinoids/metabolism , Energy Metabolism/drug effects , Hypothalamus/metabolism , Leptin/pharmacology , Polyunsaturated Alkamides/metabolism , Amidohydrolases/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Dietary Fats/pharmacology , Eating/drug effects , Eating/genetics , Gene Knock-In Techniques , Leptin/deficiency , Male , Mice , Mice, Knockout , Polymorphism, Genetic
16.
Horm Behav ; 119: 104619, 2020 03.
Article in English | MEDLINE | ID: mdl-31790663

ABSTRACT

The investigation of hormones, brain function and behavior over the past 50 years has played a major role in elucidating how the brain and body communicate reciprocally via hormones and other mediators and how this impacts brain and body health both positively and negatively. This is illustrated here for the hippocampus, a uniquely sensitive and vulnerable brain region, study of which as a hormone target has provided a gateway into the rest of the brain. Hormone actions on the brain and hormones generated within the brain are now recognized to include not only steroid hormones but also metabolic hormones and chemical signals from bone and muscle. Moreover, steroid hormones, and some metabolic hormones, and their receptors, are generated by the brain for specific functions that synergize with effects of those circulating hormones. Hormone actions in hippocampus have revealed its capacity, and that of other brain regions, for adaptive plasticity, loss of which needs external intervention in, for example, mood disorders. Early life experiences as well as in utero and transgenerational effects are now appreciated for their lasting effects at the level of gene expression affecting the capacity for adaptive plasticity. Moreover sex differences are recognized as affecting the whole brain via both genetic and epigenetic mechanisms. The demonstrated plasticity of a healthy brain gives hope that interventions throughout the life course can ameliorate negative effects by reactivating that plasticity and the underlying epigenetic activity to produce compensatory changes in the brain with more positive consequences for the body.


Subject(s)
Behavior/physiology , Brain/physiology , Hormones/metabolism , Psychophysiology , Adaptation, Physiological/physiology , Animals , Brain/metabolism , Female , Gonadal Steroid Hormones/metabolism , Hippocampus/physiology , Hormones/physiology , Humans , Male , Mood Disorders/etiology , Mood Disorders/metabolism , Mood Disorders/physiopathology , Sex Characteristics , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
17.
Neuroendocrinology ; 110(3-4): 282-291, 2020.
Article in English | MEDLINE | ID: mdl-31220843

ABSTRACT

BACKGROUND: Use of local corticosteroids, especially the inhaled types, has increasingly been associated with systemic uptake and consequent adverse effects. In this study, we assessed the associations between the use of different corticosteroid types with cognitive and neuropsychiatric adverse effects related to high glucocorticoid exposure. METHODS: In 83,592 adults (mean age 44 years, 59% women) of the general population (Lifelines Cohort Study), we analyzed the relationship between corticosteroid use with executive cognitive functioning (Ruff Figural Fluency Test), and presence of mood and anxiety disorders (Mini-International Neuropsychiatric Interview survey). We performed additional exploration for effects of physical quality of life (QoL; RAND-36), and inflammation (high-sensitive C-reactive protein [CRP]). RESULTS: Cognitive scores were lower among corticosteroid users, in particular of systemic and inhaled types, when compared to nonusers. Users of inhaled types showed lower cognitive scores irrespective of physical QoL, psychiatric disorders, and high-sensitive CRP. Overall corticosteroid use was also associated with higher likelihood for mood and anxiety disorders. Users of inhaled corticosteroids were more likely to have mood disorders (OR 1.40 [95% CI 1.19-1.65], p < 0.001) and anxiety disorders (OR 1.19 [95% CI 1.06-1.33], p = 0.002). These findings were independent of physical QoL. A higher likelihood for mood disorders was also found for systemic users whereas nasal and dermal corticosteroid users were more likely to have anxiety disorders. CONCLUSIONS: Commonly used local corticosteroids, in particular inhaled types, and systemic corticosteroids are associated with reduced executive cognitive functioning and a higher likelihood of mood and anxiety disorders in the general adult population.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/adverse effects , Anxiety Disorders/chemically induced , Cognitive Dysfunction/chemically induced , Executive Function/drug effects , Mood Disorders/chemically induced , Administration, Inhalation , Adult , Cohort Studies , Female , Humans , Male , Middle Aged
18.
Proc Natl Acad Sci U S A ; 114(24): 6358-6363, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28515312

ABSTRACT

Brain glycogen stored in astrocytes provides lactate as an energy source to neurons through monocarboxylate transporters (MCTs) to maintain neuronal functions such as hippocampus-regulated memory formation. Although prolonged exhaustive exercise decreases brain glycogen, the role of this decrease and lactate transport in the exercising brain remains less clear. Because muscle glycogen fuels exercising muscles, we hypothesized that astrocytic glycogen plays an energetic role in the prolonged-exercising brain to maintain endurance capacity through lactate transport. To test this hypothesis, we used a rat model of exhaustive exercise and capillary electrophoresis-mass spectrometry-based metabolomics to observe comprehensive energetics of the brain (cortex and hippocampus) and muscle (plantaris). At exhaustion, muscle glycogen was depleted but brain glycogen was only decreased. The levels of MCT2, which takes up lactate in neurons, increased in the brain, as did muscle MCTs. Metabolomics revealed that brain, but not muscle, ATP was maintained with lactate and other glycogenolytic/glycolytic sources. Intracerebroventricular injection of the glycogen phosphorylase inhibitor 1,4-dideoxy-1,4-imino-d-arabinitol did not affect peripheral glycemic conditions but suppressed brain lactate production and decreased hippocampal ATP levels at exhaustion. An MCT2 inhibitor, α-cyano-4-hydroxy-cinnamate, triggered a similar response that resulted in lower endurance capacity. These findings provide direct evidence for the energetic role of astrocytic glycogen-derived lactate in the exhaustive-exercising brain, implicating the significance of brain glycogen level in endurance capacity. Glycogen-maintained ATP in the brain is a possible defense mechanism for neurons in the exhausted brain.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glycogen/metabolism , Lactic Acid/metabolism , Physical Exertion/physiology , Adenosine Triphosphate/metabolism , Animals , Astrocytes/drug effects , Brain/drug effects , Coumaric Acids/pharmacology , Energy Metabolism/drug effects , Fatigue/metabolism , Glycogenolysis/drug effects , Male , Metabolomics , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Wistar
19.
Proc Natl Acad Sci U S A ; 114(38): 10250-10255, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874569

ABSTRACT

Sleep-disordered breathing (SDB) is a common disorder in aging that is associated with cognitive decline, including significant executive dysfunction, for which the neurobiological underpinnings remain poorly understood. Using proton magnetic resonance spectroscopy (1H MRS), this study assessed whether dysregulation of the homeostatic balance of the major inhibitory and excitatory amino acid neurotransmitter systems of γ-aminobutyric acid (GABA) and glutamate, respectively, play a role in SDB. Levels of GABA and those of the combined resonances of glutamate and glutamine (Glx), were measured by 1H MRS in the left dorsolateral prefrontal cortex (l-DLPFC) and bilateral hippocampal regions of 19 older adults (age ± SD: 66.1 ± 1.9 years) with moderate to severe SDB, defined as having an Apnea-Hypopnea Index (AHI) greater than 15 as assessed by polysomnography, and in 14 older adults (age ± SD: 62.3 ± 1.3 years) without SDB (AHI < 5). In subjects with SDB, levels of l-DLPFC GABA, but not Glx, were significantly lower than in control subjects (P < 0.0002). Additionally, there was a negative correlation between l-DLPFC GABA levels, but not Glx, and SDB severity by AHI (r = -0.68, P < 0.0001), and a positive correlation between l-DLPFC GABA levels, but not Glx, and minimal oxygen saturation during sleep (r = 0.62, P = 0.0005). By contrast, no group differences or oxygenation associations were found for levels of GABA or Glx in right or left hippocampal region. These findings are interpreted in terms of a pathophysiological model of SDB in which hypoxia-mediated inhibitory neurotransmission deficit in DLPFC could lead to hyperexcitability and, potentially neuronal dysfunction and cognitive decline.


Subject(s)
Glutamates/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Sleep Apnea Syndromes/metabolism , gamma-Aminobutyric Acid/deficiency , Aged , Case-Control Studies , Female , Glutamine/metabolism , Humans , Male , Middle Aged
20.
Proc Natl Acad Sci U S A ; 114(50): 13296-13301, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29167373

ABSTRACT

Epitranscriptomics refers to posttranscriptional alterations on an mRNA sequence that are dynamic and reproducible, and affect gene expression in a similar way to epigenetic modifications. However, the functional relevance of those modifications for the transcript, the cell, and the organism remain poorly understood. Here, we focus on RNA editing and show that Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-1 (APOBEC1), together with its cofactor RBM47, mediates robust editing in different tissues. The majority of editing events alter the sequence of the 3'UTR of targeted transcripts, and we focus on one cell type (monocytes) and on a small set of highly edited transcripts within it to show that editing alters gene expression by modulating translation (but not RNA stability or localization). We further show that specific cellular processes (phagocytosis and transendothelial migration) are enriched for transcripts that are targets of editing and that editing alters their function. Finally, we survey bone marrow progenitors and demonstrate that common monocyte progenitor cells express high levels of APOBEC1 and are susceptible to loss of the editing enzyme. Overall, APOBEC1-mediated transcriptome diversification is required for the fine-tuning of protein expression in monocytes, suggesting an epitranscriptomic mechanism for the proper maintenance of homeostasis in innate immune cells.


Subject(s)
APOBEC-1 Deaminase/metabolism , Epigenesis, Genetic , RNA Editing , Transcriptome , APOBEC-1 Deaminase/genetics , Animals , Cell Movement , Cells, Cultured , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/cytology , Monocyte-Macrophage Precursor Cells/metabolism , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL