Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Compr Psychoneuroendocrinol ; 6: 100045, 2021 May.
Article in English | MEDLINE | ID: mdl-35757356

ABSTRACT

17ß-estradiol (E2) levels in women correlate with multiple neuropsychiatric symptoms, including those that are stress-related. Furthermore, prior work from our group has demonstrated that E2 status influences DNA methylation (DNAm) across the genome. We developed and validated a DNAm-based predictor of E2 (one of four naturally occurring estrogens) using a training set of 183 females and a test set of 79 females from the same traumatized cohort. We showed that predicted E2 levels were highly correlated with measured E2 concentrations in our testing set (r â€‹= â€‹0.75, p â€‹= â€‹1.8e-15). We further demonstrated that predicted E2 concentrations, in combination with measured values, negatively correlated with current post-traumatic stress disorder (PTSD) (ߠ​= â€‹-0.38, p â€‹= â€‹0.01) and major depressive disorder (MDD) diagnoses (ߠ​= â€‹-0.45, p â€‹= â€‹0.02), as well as a continuous measure of PTSD symptom severity (ߠ​= â€‹-2.3, p â€‹= â€‹0.007) in females. Finally, we tested our predictor in an independent data set (n â€‹= â€‹85) also comprised of recently traumatized female subjects to determine if the predictor would generalize to a different population than the one on which it was developed. We found that the correlation between predicted and actual E2 concentrations in the external validation data set was also high (r â€‹= â€‹0.48, p â€‹= â€‹3.0e-6). While further validation is warranted, a DNAm predictor of E2 concentrations will advance our understanding of hormone-epigenetic interactions. Furthermore, such a DNAm predictor may serve as an epigenetic proxy for E2 concentrations and thus provide an important biomarker to better evaluate the contribution of E2 to current and potentially future psychiatric symptoms in samples for which E2 is not measured.

2.
Psychol Med ; 50(16): 2790-2798, 2020 12.
Article in English | MEDLINE | ID: mdl-31615590

ABSTRACT

BACKGROUND: Preclinical and human studies suggest an association between chronic inflammation and the development of depressive behaviors. This is proposed to occur through downstream effects of inflammatory cytokines on neuroplasticity, neurogenesis and neurotransmitter function, although the neural correlates remain poorly understood in humans. METHODS: In Study 1, structural magnetic resonance imaging and serum inflammatory cytokine data were analyzed from 53 psychiatrically healthy female participants. Correlational analyses were conducted between interleukin-6 (IL-6) and volume in a priori regions implicated in the pathophysiology of major depressive disorder (MDD). In Study 2, medical data [including serum inflammatory acute phase reactants (C-reactive protein)] were analyzed for 12 589 participants. Participants were classified as having (n = 2541) v. not having (n = 10 048) probable lifetime MDD using phenotypes derived using machine-learning approaches. Non-parametric analyses compared inflammation between groups, whereas regression analyses probed whether inflammation predicted probable MDD classification while accounting for other variables. RESULTS: In Study 1, significant negative correlations emerged between IL-6 and hippocampal, caudate, putamen and amygdalar volume. In Study 2, the MDD group showed a higher probability of elevated inflammation than the non-MDD group. Moreover, elevated inflammation was a significant predictor of probable MDD classification. CONCLUSIONS: Findings indicate that inflammation is cross-sectionally related to reduced volume in brain regions implicated in MDD phenotypes among a sample of psychiatrically healthy women, and is associated with the presence of probable MDD in a large clinical dataset. Future investigations may identify specific inflammatory markers predicting first MDD onset.


Subject(s)
Brain/physiopathology , Depressive Disorder, Major/blood , Depressive Disorder, Major/physiopathology , Inflammation/psychology , Adult , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Chronic Disease , Comorbidity , Female , Humans , Inflammation/blood , Interleukin-6/blood , Logistic Models , Magnetic Resonance Imaging , Male , Medical Records , Phenotype , Young Adult
3.
Neuron ; 102(1): 60-74, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30946827

ABSTRACT

Threat processing is central to understanding debilitating fear- and trauma-related disorders such as posttraumatic stress disorder (PTSD). Progress has been made in understanding the neural circuits underlying the "engram" of threat or fear memory formation that complements a decades-old appreciation of the neurobiology of fear and threat involving hub structures such as the amygdala. In this review, we examine key recent findings, as well as integrate the importance of hormonal and physiological approaches, to provide a broader perspective of how bodily systems engaged in threat responses may interact with amygdala-based circuits in the encoding and updating of threat-related memory. Understanding how trauma-related memories are encoded and updated throughout the brain and the body will ultimately lead to novel biologically-driven approaches for treatment and prevention.


Subject(s)
Brain/physiopathology , Fear/physiology , Memory/physiology , Psychological Trauma/physiopathology , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology , Amygdala/metabolism , Amygdala/physiology , Amygdala/physiopathology , Brain/metabolism , Brain/physiology , Central Amygdaloid Nucleus/physiology , Central Amygdaloid Nucleus/physiopathology , Corticotropin-Releasing Hormone/metabolism , Fear/psychology , Glucocorticoids/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Hippocampus/physiopathology , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Hypothalamus/physiopathology , Interneurons/metabolism , Interneurons/physiology , Psychological Trauma/metabolism , Psychological Trauma/psychology , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Thalamus/metabolism , Thalamus/physiology , Thalamus/physiopathology
4.
Addict Biol ; 23(2): 653-664, 2018 03.
Article in English | MEDLINE | ID: mdl-28635037

ABSTRACT

Epigenetic mechanisms have been proposed to contribute to persistent aspects of addiction-related behaviors. One family of epigenetic molecules that may regulate maladaptive behavioral changes produced by cocaine use are the histone deacetylases (HDACs)-key regulators of chromatin and gene expression. In particular, the class IIa HDACs (HDAC4, HDAC5, HDAC7 and HDAC9) respond to changes in neuronal activity by modulating their distribution between the nucleus and cytoplasm-a process controlled in large part by changes in phosphorylation of conserved residues. Cocaine triggers a transient nuclear accumulation of HDAC5 that functions to limit the development of cocaine reward behavior. However, the role and regulation of the close family member, HDAC4, in cocaine behaviors remain largely unknown. In this study, we report that cocaine and cAMP signaling in striatum produced differential phosphorylation and subcellular localization of HDAC4 and HDAC5. Unlike HDAC5, cocaine exposure induced a modest hyperphosphorylation and nuclear export of HDAC4. Genetic deletion of HDAC4 in the nucleus accumbens reduced acute cocaine-produced locomotion, maximum locomotor sensitization and cocaine reward-related behavior. Interestingly, overexpression of an HDAC4 cytoplasm-concentrated mutant (S266E) increased cocaine reward behavior in the cocaine conditioned place preference assay, suggesting that cocaine-induced nuclear export of HDAC4 might function to facilitate the development of cocaine reward behaviors through a role in the cell cytoplasm. Together, our findings suggest that, despite high sequence homology, HDAC4 and HDAC5 are oppositely regulated by cocaine-induced signaling in vivo and have distinct roles in regulating cocaine behaviors.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Histone Deacetylases/drug effects , Neurons/drug effects , Animals , Behavior, Animal , Cell Nucleus , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Cytoplasm , Epigenesis, Genetic , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mice , Mutation , Neurons/metabolism , Phosphorylation , Protein Transport/drug effects , Rats
5.
Front Psychiatry ; 9: 778, 2018.
Article in English | MEDLINE | ID: mdl-30705647

ABSTRACT

Stress and trauma profoundly influence psychiatric biobehavioral outcomes. The identification of treatment and biomarker targets would be accelerated by a broad understanding of the biological responses to these events. The goal of this study was to determine genes responsive to auditory fear conditioning (FC), a well-characterized amygdala-dependent rodent model of threat-exposure, in the presence or absence of prior stress history, providing insight into the physiological processes underlying response to trauma. RNA-sequencing was performed in blood and amygdala from mice that underwent fear conditioning with (Immo+FC) and without (FC) prior immobilization stress, a paradigm that induces HPA axis, and behavioral stress sensitization. In the amygdala, 607 genes were regulated by FC vs. home-cage (HC) controls, and 516 genes differed in stress-sensitized mice (Immo+FC vs. FC). In the former, we observed an enhancement of specific biological processes involved in learning and synaptic transmission, and in the latter processes associated with cell proliferation and the cellular response to drugs. In the blood of stress-sensitized animals, 468 genes were dynamically regulated when compared to FC, and were enriched for the biological pathways of inflammation and cytokine signaling. This study identified genes and pathways that respond to threat in the amygdala and blood of mice with and without a prior stress history and reveals the impact of stress history on subsequent inflammation. Future studies will be needed to examine the role of these dynamically regulated genes may play in human clinical stress and trauma-related disorders.

6.
PLoS One ; 12(7): e0181892, 2017.
Article in English | MEDLINE | ID: mdl-28715509

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0054463.].

7.
Neurobiol Learn Mem ; 107: 93-100, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24291571

ABSTRACT

We have previously shown that auditory Pavlovian fear conditioning is associated with an increase in DNA methyltransferase (DNMT) expression in the lateral amygdala (LA) and that intra-LA infusion or bath application of an inhibitor of DNMT activity impairs the consolidation of an auditory fear memory and long-term potentiation (LTP) at thalamic and cortical inputs to the LA, in vitro. In the present study, we use awake behaving neurophysiological techniques to examine the role of DNMT activity in memory-related neurophysiological changes accompanying fear memory consolidation and reconsolidation in the LA, in vivo. We show that auditory fear conditioning results in a training-related enhancement in the amplitude of short-latency auditory-evoked field potentials (AEFPs) in the LA. Intra-LA infusion of a DNMT inhibitor impairs both fear memory consolidation and, in parallel, the consolidation of training-related neural plasticity in the LA; that is, short-term memory (STM) and short-term training-related increases in AEFP amplitude in the LA are intact, while long-term memory (LTM) and long-term retention of training-related increases in AEFP amplitudes are impaired. In separate experiments, we show that intra-LA infusion of a DNMT inhibitor following retrieval of an auditory fear memory has no effect on post-retrieval STM or short-term retention of training-related changes in AEFP amplitude in the LA, but significantly impairs both post-retrieval LTM and long-term retention of AEFP amplitude changes in the LA. These findings are the first to demonstrate the necessity of DNMT activity in the consolidation and reconsolidation of memory-associated neural plasticity, in vivo.


Subject(s)
Amygdala/enzymology , Amygdala/physiology , DNA Modification Methylases/physiology , Memory/physiology , Neuronal Plasticity/physiology , Acoustic Stimulation , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Fear/physiology , Male , Memory/drug effects , Mental Recall/drug effects , Mental Recall/physiology , Neuronal Plasticity/drug effects , Phthalimides/pharmacology , Rats , Rats, Sprague-Dawley , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
8.
Front Psychiatry ; 4: 62, 2013.
Article in English | MEDLINE | ID: mdl-23847551

ABSTRACT

This review examines recent work on epigenetic mechanisms underlying animal models of fear learning as well as its translational implications in disorders of fear regulation, such as Post-traumatic Stress Disorder (PTSD). Specifically, we will examine work outlining roles of differential histone acetylation and DNA-methylation associated with consolidation, reconsolidation, and extinction in Pavlovian fear paradigms. We then focus on the numerous studies examining the epigenetic modifications of the Brain-derived neurotrophin factor (BDNF) pathway and the extension of these findings from animal models to recent work in human clinical populations. We will also review recently published data on FKBP5 regulation of glucocorticoid receptor function, and how this is modulated in animal models of PTSD and in human clinical populations via epigenetic mechanisms. As glucocorticoid regulation of memory consolidation is well established in fear models, we examine how these recent data contribute to our broader understanding of fear memory formation. The combined recent progress in epigenetic modulation of memory with the advances in fear neurobiology suggest that this area may be critical to progress in our understanding of fear-related disorders with implications for new approaches to treatment and prevention.

9.
PLoS One ; 8(1): e54463, 2013.
Article in English | MEDLINE | ID: mdl-23349897

ABSTRACT

The study of the cellular and molecular mechanisms underlying the consolidation and reconsolidation of traumatic fear memories has progressed rapidly in recent years, yet few compounds have emerged that are readily useful in a clinical setting for the treatment of anxiety disorders such as post-traumatic stress disorder (PTSD). Here, we use a combination of biochemical, behavioral, and neurophysiological methods to systematically investigate the ability of garcinol, a naturally-occurring histone acetyltransferase (HAT) inhibitor derived from the rind of the fruit of the Kokum tree (Garcina indica), to disrupt the consolidation and reconsolidation of Pavlovian fear conditioning, a widely studied rodent model of PTSD. We show that local infusion of garcinol into the rat lateral amygdala (LA) impairs the training and retrieval-related acetylation of histone H3 in the LA. Further, we show that either intra-LA or systemic administration of garcinol within a narrow window after either fear conditioning or fear memory retrieval significantly impairs the consolidation and reconsolidation of a Pavlovian fear memory and associated neural plasticity in the LA. Our findings suggest that a naturally-occurring compound derived from the diet that regulates chromatin function may be useful in the treatment of newly acquired or recently reactivated traumatic memories.


Subject(s)
Anxiety Disorders/drug therapy , Fear , Memory/drug effects , Stress Disorders, Post-Traumatic/drug therapy , Terpenes/administration & dosage , Animals , Anxiety Disorders/complications , Anxiety Disorders/physiopathology , Brain/drug effects , Brain/physiopathology , Chromatin/drug effects , Fear/drug effects , Fear/psychology , Garcinia/chemistry , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Neuronal Plasticity/drug effects , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/complications , Stress Disorders, Post-Traumatic/physiopathology
10.
Learn Mem ; 20(2): 109-19, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23328899

ABSTRACT

Modifications in chromatin structure have been widely implicated in memory and cognition, most notably using hippocampal-dependent memory paradigms including object recognition, spatial memory, and contextual fear memory. Relatively little is known, however, about the role of chromatin-modifying enzymes in amygdala-dependent memory formation. Here, we use a combination of biochemical, behavioral, and neurophysiological methods to systematically examine the role of p300/CBP histone acetyltransferase (HAT) activity in the consolidation and reconsolidation of auditory Pavlovian fear memories. We show that local infusions of c646, a selective pharmacological inhibitor of p300/CBP activity, shortly following either fear conditioning or fear memory retrieval impair training and retrieval-related regulation of histone acetylation in the lateral nucleus of the amygdala (LA). Furthermore, we show that intra-LA infusion of c646 significantly impairs fear memory consolidation, reconsolidation, and associated neural plasticity in the LA. Our findings collectively suggest that p300/CBP HAT activity is critical for the consolidation and reconsolidation of amygdala-dependent Pavlovian fear memories.


Subject(s)
Amygdala/physiology , Conditioning, Classical/physiology , Fear , Memory/physiology , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Acoustic Stimulation , Amygdala/drug effects , Analysis of Variance , Animals , Conditioning, Classical/drug effects , Electrodes, Implanted , Electroshock , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Male , Memory/drug effects , Memory Disorders/chemically induced , Rats , Rats, Sprague-Dawley , Time Factors
11.
Learn Mem ; 18(9): 579-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21868438

ABSTRACT

Epigenetic mechanisms have been widely implicated in synaptic plasticity and in memory consolidation, yet little is known about the role of epigenetic mechanisms in memory reconsolidation processes. In the present study, we systematically examine the role of histone acetylation and DNA methylation in the reconsolidation of an amygdala-dependent Pavlovian fear memory. We first show that the acetylation of histone 3 (H3), but not histone 4 (H4), is regulated following auditory fear memory retrieval in the lateral nucleus of the amygdala (LA). We next show that histone deacetylase (HDAC) inhibition in the LA enhances both retrieval-induced histone acetylation and reconsolidation of an auditory fear memory. Conversely, inhibition of DNA methytransferase (DNMT) activity in the LA significantly impairs both retrieval-related H3 acetylation and fear memory reconsolidation. The effects of HDAC and DNMT inhibitors on fear memory reconsolidation were observed to be time-limited and were not evident in the absence of memory reactivation. Further, memories lost following DNMT inhibition were not observed to be vulnerable to spontaneous recovery, reinstatement, or to a shift in testing context, suggesting that memory impairment was not the result of facilitated extinction. Finally, pretreatment with the HDAC inhibitor was observed to rescue the reconsolidation deficit induced by the DNMT inhibitor. These findings collectively suggest that histone acetylation and DNA methylation are critical for reconsolidation of fear memories in the LA.


Subject(s)
Amygdala/metabolism , Conditioning, Classical/physiology , Epigenesis, Genetic/genetics , Epigenomics , Fear/physiology , Histones/metabolism , Acetylation/drug effects , Acoustic Stimulation/methods , Amygdala/drug effects , Animals , Conditioning, Classical/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Electroshock/adverse effects , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Fear/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Histones/genetics , Male , Rats , Rats, Sprague-Dawley , Time Factors
12.
J Neurosci ; 31(19): 7073-82, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21562269

ABSTRACT

The activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) is an immediate-early gene that has been widely implicated in synaptic plasticity and in the consolidation of a variety of hippocampal- and amygdala-dependent memory tasks. The functional role of Arc/Arg3.1 in memory reconsolidation processes, however, has not been systematically studied. In the present study, we examined the role of Arc/Arg3.1 in the reconsolidation of an amygdala-dependent auditory pavlovian fear memory. We show that Arc/Arg3.1 protein is regulated in the lateral nucleus of the amygdala (LA) by retrieval of an auditory fear memory. Next, we show that antisense knockdown of Arc/Arg3.1 in the LA impairs fear memory reconsolidation of both a recent (1-d-old) as well as a well-consolidated (2-week-old) fear memory; that is, post-retrieval short-term memory, tested at 3 h after retrieval, is intact, whereas post-retrieval long-term memory, tested approximately 24 h after retrieval, is significantly impaired. The effect of Arc/Arg3.1 knockdown was observed to be time limited and specific to an actively reactivated fear memory. Moreover, the reconsolidation deficit induced by Arc/Arg3.1 knockdown was not found to be sensitive to spontaneous recovery, reinstatement, or a shift in the testing context, suggesting that our behavioral effects are not attributable to facilitated extinction. Collectively, our findings provide the first comprehensive look at the functional role of Arc/Arg3.1 in memory reconsolidation processes in the mammalian brain.


Subject(s)
Amygdala/metabolism , Conditioning, Classical/physiology , Cytoskeletal Proteins/metabolism , Fear/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Analysis of Variance , Animals , Behavior, Animal/drug effects , Blotting, Western , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
13.
Learn Mem ; 18(1): 24-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21177377

ABSTRACT

The immediate-early gene early growth response gene-1 (EGR-1, zif-268) has been extensively studied in synaptic plasticity and memory formation in a variety of memory systems. However, a convincing role for EGR-1 in amygdala-dependent memory consolidation processes has yet to emerge. In the present study, we have examined the role of EGR-1 in the consolidation and reconsolidation of amygdala-dependent auditory Pavlovian fear conditioning. In our first series of experiments, we show that EGR-1 is regulated following auditory fear conditioning in the lateral nucleus of the amygdala (LA). Next, we use antisense oligodeoxynucleotide (ODN) knockdown of EGR-1 in the LA to show that training-induced expression of EGR-1 is required for memory consolidation of auditory fear conditioning; that is, long-term memory (LTM) is significantly impaired while acquisition and short-term memory (STM) are intact. In a second set of experiments, we show that EGR-1 is regulated in the LA by retrieval of an auditory fear memory. We then show that retrieval-induced expression of EGR-1 in the LA is required for memory reconsolidation of auditory fear conditioning; that is, post-retrieval (PR)-LTM is significantly impaired while memory retrieval and PR-STM are intact. Additional experiments show these effects to be restricted to the LA, to be temporally graded, and unlikely to be due to nonspecific toxicity within the LA. Collectively, our findings strongly implicate a role for EGR-1 in both the initial consolidation and in the reconsolidation of auditory fear memories in the LA.


Subject(s)
Amygdala/metabolism , Conditioning, Classical/physiology , Early Growth Response Protein 1/metabolism , Fear , Memory/physiology , Acoustic Stimulation/adverse effects , Amygdala/drug effects , Animals , Association Learning/drug effects , Behavior, Animal , Conditioning, Classical/drug effects , Early Growth Response Protein 1/genetics , Electroshock/adverse effects , Fear/drug effects , Male , Memory/drug effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Time Factors
14.
Neurobiol Learn Mem ; 88(3): 369-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17613252

ABSTRACT

Previous work in our laboratory demonstrated that galantamine, a cholinesterase inhibitor and weak cholinergic agonist, facilitated classical trace eyeblink conditioning in healthy, young rabbits [Simon, B. B., Knuckley, B., & Powell, D. A. (2004). Galantamine facilitates acquisition of a trace-conditioned eyeblink response in healthy, young rabbits. Learning & Memory, 11(1), 116-122.]. The current study investigated the effects of galantamine (0.0 or 3.0mg/kg) in rabbits sustaining knife-cut lesions to the fimbria-fornix, a major projection pathway connecting the hippocampus to cortical and subcortical brain structures involved in the formation of long-term memories. Two experiments were conducted. Experiment one assessed the effects of knife-cut lesions to the fornix or sham surgeries on trace eyeblink (EB) conditioning. Results indicate that fornix lesions significantly retarded EB conditioning when trace parameters were employed. Experiment 2 assessed whether treatment with galantamine would reverse the deficits caused by fornix damage. Results indicate that 3.0mg/kg GAL reversed trace EB conditioning deficits in animals with fornix knife-cut lesions. These findings suggest that galantamine may provide benefit in the reversal of cognitive dysfunction following certain types of brain damage, especially damage involving hippocampal structures.


Subject(s)
Cholinergic Agents/pharmacology , Conditioning, Classical/physiology , Conditioning, Eyelid/physiology , Fornix, Brain/physiology , Galantamine/pharmacology , Analysis of Variance , Animals , Conditioning, Classical/drug effects , Conditioning, Eyelid/drug effects , Female , Fornix, Brain/drug effects , Hippocampus/physiology , Male , Rabbits , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...