Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Biol Psychiatry Glob Open Sci ; 4(1): 203-212, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38298799

ABSTRACT

Background: There is a growing importance for environmental contributions to psychiatric disorders and understanding the impact of the exposome (i.e., pollutants and toxins). For example, increased biomonitoring and epidemiological studies suggest that daily phthalate chemical exposure contributes to neurological and behavioral abnormalities; however, these mechanisms remain poorly understood. Therefore, the current study was aimed at examining the effects of chronic phthalate exposure on rodent anxiety behaviors and cognition and the impact on hypothalamic-pituitary-adrenal axis function. Methods: Adult male mice (C57BL6/J) were administered MEHP via drinking water (1 mg/mL), and anxiety-like behavior and cognition combined with hypothalamic-pituitary-adrenal axis and inflammatory assays were assessed after 3 weeks of MEHP exposure. Results: MEHP-treated mice exhibited enhanced generalized anxiety-like behaviors, as demonstrated by reduced time spent in the open-arm of the elevated plus maze and center exploration in the open field. Tests of spatial memory and cognition were unchanged. Following MEHP administration, circulating levels of corticosterone and proinflammatory cytokines were significantly increased, while at the tissue level, there were MEHP-dependent reductions in glucocorticoid metabolism genes Hsd11b1 and Hsd11b2. Conclusions: These data suggest that chronic MEHP exposure leads to enhanced generalized anxiety behaviors independent of rodent measures of cognition and memory, which may be driven by MEHP-dependent effects on hypothalamic-pituitary-adrenal axis and peripheral glucocorticoid metabolism function.

2.
bioRxiv ; 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-38045293

ABSTRACT

Background: The renin-angiotensin system (RAS) has been identified as a potential therapeutic target for PTSD, though its mechanisms are not well understood. Brain angiotensin type 2 receptors (AT2Rs) are a subtype of angiotensin II receptors located in stress and anxiety-related regions, including the medial prefrontal cortex (mPFC), but their function and mechanism in the mPFC remain unexplored. We therefore used a combination of imaging, cre/lox, and behavioral methods to investigate mPFC-AT2R-expressing neuron involvement in fear learning. Methods: To characterize mPFC-AT2R-expressing neurons in the mPFC, AT2R-Cre/td-Tomato male and female mice were used for immunohistochemistry (IHC). mPFC brain sections were stained with glutamatergic or interneuron markers, and density of AT2R+ cells and colocalization with each marker was quantified. To assess fear-related behaviors in AT2R-flox mice, we selectively deleted AT2R from mPFC neurons using an AAV-Cre virus. Mice then underwent Pavlovian auditory fear conditioning, approach/avoidance, and locomotion testing. Results: IHC results revealed that AT2R is densely expressed in the mPFC. Furthermore, AT2R is primarily expressed in somatostatin interneurons in females but not males. Following fear conditioning, mPFC-AT2R deletion impaired extinction in female but not male mice. Locomotion was unaltered by mPFC-AT2R deletion in males or females, while AT2R-deleted females had increased exploratory behavior. Conclusion: These results lend support for mPFC-AT2R+ neurons as a novel subgroup of somatostatin interneurons that influence fear extinction in a sex-dependent manner. This furthers underscores the role of mPFC in top-down regulation and a unique role for peptidergic (ie., angiotensin) mPFC regulation of fear and sex differences.

3.
bioRxiv ; 2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37886449

ABSTRACT

There is a growing importance for environmental contributions to psychiatric disorders and understanding the impact of the exposome (i.e., pollutants and toxins). Increased biomonitoring and epidemiological studies, for example, suggest that daily phthalate chemical exposure contribute to neurological and behavioral abnormalities, however these mechanisms remain poorly understood. The current study therefore aimed to examine the effects of chronic phthalate exposure on rodent anxiety behaviors, cognition, and the impact on hypothalamic-pituitary- adrenal (HPA)-axis function. Adult male mice (C57BL6/J) were administered mono-2-ethylhexyl phthalate (MEHP) via drinking water (1 mg/ml), and anxiety-like behavior, cognition combined with HPA- axis and inflammatory assays were assessed after 3 weeks of MEHP exposure. MEHP-treated mice exhibited enhanced generalized anxiety-like behaviors, as demonstrated by reduced time spent in the open-arm of the elevated plus maze (EPM) and center exploration in the open field (OF). Tests of spatial, cognition and memory function were unchanged. Following MEHP administration, circulating levels of corticosterone and pro- inflammatory cytokines were significantly increased, while at the tissue level, MEHP-dependent reductions in glucocorticoid metabolism genes 11ß-hydroxysteroid dehydrogenase (11ß-HSD) 1 and 2. These data suggest that chronic MEHP exposure leads to enhanced generalized-anxiety behaviors independent of rodent measures of cognition and memory, which maybe driven by MEHP-dependent effects on HPA-axis and peripheral glucocorticoid metabolism function.

4.
Neuropharmacology ; 229: 109460, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36801399

ABSTRACT

The renin-angiotensin system (RAS) has been linked to the pathophysiology of posttraumatic stress disorder (PTSD) however, the underlying neurobiological mechanism(s) remain elusive. Here we utilized angiotensin II receptor type 1 (AT1R) transgenic mice combined with neuroanatomical, behavioral, and electrophysiological approaches, to examine the role of the central amygdala (CeA) expressing AT1R neurons in fear and anxiety-related behavior. Within the major amygdala subdivisions, AT1R+ neurons were localized to gamma-aminobutyric acid (GABA) expressing neurons in the lateral division of the central amygdala (CeL), and the majority of them were identified as protein kinase C-δ positive (PKCδ+) neurons. Following CeA-AT1R deletion using cre-expressing lentiviral delivery in AT1R-Flox mice, generalized anxiety and locomotor activity as well as the acquisition of conditioned fear were unaltered while the acquisition of extinction learning, as measured by percent freezing behavior, was significantly enhanced. During electrophysiological recordings of CeL-AT1R+ neurons, the application of angiotensin II (1 µm) increased the amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) and decreased the excitability of CeL-AT1R+ neurons. Overall, these findings demonstrate that CeL-AT1R-expressing neurons play a role in fear extinction, potentially through facilitated CeL-AT1R+ GABAergic inhibition. These results provide new evidence for mechanisms of angiotensinergic neuromodulation of the CeL and its role in fear extinction and may aid in further advancing targeted novel therapies for improving maladaptive fear learning processes associated with PTSD.


Subject(s)
Central Amygdaloid Nucleus , Fear , Mice , Animals , Fear/physiology , Central Amygdaloid Nucleus/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Extinction, Psychological , Mice, Transgenic , Neurons/metabolism
5.
Anal Chem ; 94(25): 9018-9025, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35696295

ABSTRACT

While the role of the renin-angiotensin system (RAS) in peripheral circulation is well characterized, we still lack an in-depth understanding of its role within the brain. This knowledge gap is sustained by lacking technologies for trace-level angiotensin detection throughout tissues, such as the brain. To provide a bridging solution, we enhanced capillary electrophoresis (CE) nanoflow electrospray ionization (ESI) with large-volume sample stacking and employed trapped ion mobility time-of-flight (timsTOF) tandem HRMS detection. A dynamic pH junction helped stack approximately 10 times more of the sample than optimal using the field-amplified reference. In conjunction, the efficiency of ion generation was maximized by a cone-jet nanospray on a low sheath-flow tapered-tip nano-electrospray emitter. The platform provided additional peptide-dependent information, the collision cross section, to filter chemical noise and improve sequence identification and detection limits. The lower limit of detection reached sub-picomolar or ∼30 zmol (∼18,000 copies) level. All nine targeted angiotensin peptides in mouse tissue samples were detectable and quantifiable from the paraventricular nucleus (PVN) of the hypothalamus even after removal of circulatory blood components (perfusion). We anticipate CE-ESI with timsTOF HRMS to be broadly applicable for the ultrasensitive detection of brain peptidomes in pursuit of a better understanding of the brain.


Subject(s)
Angiotensins , Spectrometry, Mass, Electrospray Ionization , Animals , Brain , Electrophoresis, Capillary/methods , Mice , Peptides/analysis , Spectrometry, Mass, Electrospray Ionization/methods
6.
Depress Anxiety ; 39(10-11): 663-674, 2022 10.
Article in English | MEDLINE | ID: mdl-35708302

ABSTRACT

Posttraumatic stress disorder (PTSD) has long been associated with a heightened risk of cardiovascular disease (CVD). A number of mechanisms have been implicated to underlie this brain-heart axis relationship, such as altered functioning of the autonomic nervous system and increased systemic inflammation. While neural alterations have repeatedly been observed in PTSD, they are rarely considered in the PTSD-CVD link. The brain-heart axis is a pathway connecting frontal and limbic brain regions to the brainstem and periphery via the autonomic nervous system and it may be a promising model for understanding CVD risk in PTSD given its overlap with PTSD neural deficits. We first provide a summary of the primary mechanisms implicated in the association between PTSD and CVD. We then review the brain-heart axis and its relevance to PTSD, as well as findings from PTSD trials demonstrating that a number of PTSD treatments have effects on areas of the brain-heart axis. Finally, we discuss sex considerations in the PTSD-CVD link. A critical next step in this study is to determine if PTSD treatments that affect the brain-heart axis (e.g., brain stimulation that improves autonomic function) also reduce the risk of CVD.


Subject(s)
Cardiovascular Diseases , Stress Disorders, Post-Traumatic , Autonomic Nervous System , Brain/diagnostic imaging , Brain/metabolism , Cardiovascular Diseases/epidemiology , Humans , Inflammation/complications , Stress Disorders, Post-Traumatic/complications , Stress Disorders, Post-Traumatic/epidemiology
7.
Biol Psychiatry ; 90(7): 473-481, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34275593

ABSTRACT

BACKGROUND: Evidence-based pharmacological treatments for posttraumatic stress disorder (PTSD) are few and of limited efficacy. Previous work suggests that angiotensin type 1 receptor inhibition facilitates fear inhibition and extinction, important for recovery from PTSD. This study tests the efficacy of the angiotensin type 1 receptor antagonist losartan, an antihypertensive drug, repurposed for the treatment of PTSD. METHODS: A randomized controlled trial was conducted for 10 weeks in 149 men and women meeting DSM-5 PTSD criteria. Losartan (vs. placebo) was flexibly titrated from 25 to 100 mg/day by week 6 and held at highest tolerated dose until week 10. Primary outcome was the Clinician-Administered PTSD Scale for DSM-5 (CAPS-5) change score at 10 weeks from baseline. A key secondary outcome was change in CAPS-5 associated with a single nucleotide polymorphism of the ACE gene. Additional secondary outcomes included changes in the PTSD Checklist for DSM-5 and the Patient Health Questionnaire-9, and proportion of responders with a Clinical Global Impressions-Improvement scale of "much improved" or "very much improved." RESULTS: Both groups had robust improvement in PTSD symptoms, but there was no significant difference on the primary end point, CAPS-5 measured as week 10 change from baseline, between losartan and placebo (mean change difference, 0.9, 95% confidence interval, -3.2 to 5.0). There was no significant difference in the proportion of Clinical Global Impressions-Improvement scale responders for losartan (58.6%) versus placebo (57.9%), no significant differences in changes in PTSD Checklist for DSM-5 or Patient Health Questionnaire-9, and no association between ACE genotype and CAPS-5 improvement on losartan. CONCLUSIONS: At these doses and durations, there was no significant benefit of losartan compared with placebo for the treatment of PTSD. We discuss implications for failure to determine the benefit of a repurposed drug with strong a priori expectations of success based on preclinical and epidemiological data.


Subject(s)
Stress Disorders, Post-Traumatic , Angiotensin Receptor Antagonists , Double-Blind Method , Female , Humans , Losartan/therapeutic use , Male , Stress Disorders, Post-Traumatic/drug therapy , Treatment Outcome
8.
Physiol Behav ; 236: 113414, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33819454

ABSTRACT

To further understand mechanisms of neuropsychiatric disease(s) and their impact on physiological systems, improved pre-clinical models and innovative methodology are needed to assess the internal physiological state of the animal in real-time. To address this challenge we developed a customizable software-based program for Ponemah™ that takes into account the animals diurnal and resting cardiovascular state in a home-cage environment. Using an integrated Pavlovian fear conditioning and cardiovascular telemetry approach in mice, we demonstrate for the first time a novel software add-on application that can remotely trigger a conditioned stimulus (CS) (i.e., audible tone) based on the animals instantaneous cardiovascular state while in its home-cage environment. This new software tool extends the ability to quantify integrated physiological correlates of learned threat and defensive behavior and may aid in further understanding mechanisms related to enhanced cardiovascular and autonomic arousal in anxiety-based disorders.


Subject(s)
Conditioning, Classical , Fear , Animals , Arousal , Conditioning, Operant , Mice , Telemetry
9.
J Neurosci ; 41(5): 901-910, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33472824

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by hypervigilance, increased reactivity to unpredictable versus predictable threat signals, deficits in fear extinction, and an inability to discriminate between threat and safety. First-line pharmacotherapies for psychiatric disorders have limited therapeutic efficacy in PTSD. However, recent studies have advanced our understanding of the roles of several limbic neuropeptides in the regulation of defensive behaviors and in the neural processes that are disrupted in PTSD. For example, preclinical studies have shown that blockers of tachykinin pathways, such as the Tac2 pathway, attenuate fear memory consolidation in mice and thus might have unique potential as early post-trauma interventions to prevent PTSD development. Targeting this pathway might also be beneficial in regulating other symptoms of PTSD, including trauma-induced aggressive behavior. In addition, preclinical and clinical studies have shown the important role of angiotensin receptors in fear extinction and the promise of using angiotensin II receptor blockade to reduce PTSD symptom severity. Additional preclinical studies have demonstrated that the oxytocin receptors foster accurate fear discrimination by facilitating fear responses to predictable versus unpredictable threats. Complementary human imaging studies demonstrate unique neural targets of intranasal oxytocin and compare its efficacy with well-established anxiolytic treatments. Finally, promising data from human subjects have demonstrated that a selective vasopressin 1A receptor antagonist reduces anxiety induced by unpredictable threats. This review highlights these novel promising targets for the treatment of unique core elements of PTSD pathophysiology.


Subject(s)
Anxiety/metabolism , Emotions/physiology , Limbic System/metabolism , Neuropeptides/metabolism , Stress Disorders, Post-Traumatic/metabolism , Animals , Anxiety/drug therapy , Anxiety/psychology , Emotions/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Humans , Limbic System/drug effects , Nerve Net/drug effects , Nerve Net/metabolism , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Receptors, Tachykinin/antagonists & inhibitors , Receptors, Tachykinin/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Tachykinins/antagonists & inhibitors , Tachykinins/metabolism
10.
Neuropsychopharmacology ; 46(4): 750-755, 2021 03.
Article in English | MEDLINE | ID: mdl-33318633

ABSTRACT

Prior observational studies have suggested that medications targeting the renin-angiotensin system, such as angiotensin-converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs), may be associated with decreased PTSD symptoms. Given known sex differences in PTSD prevalence and cardiovascular disease, here we tested whether the effects of ACE-I/ARB status on PTSD differ by sex. We also expanded these observations with replication analyses in a large biorepository database. Participants in the initial sample included 840 trauma-exposed individuals recruited as part of the Grady Trauma Project. The Modified PTSD Symptom Scale (M-PSS) was administered and ACE-I/ARB status was determined by self-report. Replication analyses were conducted using a large biorepository database (Partners Healthcare Biobank, N = 116,389) with diagnoses and medication status based on available electronic health records. Among individuals treated with ACE-Is/ARBs in the initial sample, women had significantly higher M-PSS total and Re-experiencing severity compared to men (p's < 0.05). Analyses with the large biorepository sample robustly replicated the overall effects of ACE-I/ARB medication associated with lower rate of PTSD diagnosis (p < 0.001). We also demonstrated that this effect may be specific to the renin-angiotensin system as it did not replicate for beta-blockers, calcium channel blockers, or diuretics. When we examined more specific drug classes, results indicated that the ACE-I/ARB effect on PTSD may be driven more by ARBs (e.g., Losartan) than by ACE-Is. Post-hoc analyses indicated that racial differences may exist in these effects. Overall, our results replicate and extend prior observations that the renin-angiotensin system is associated with PTSD. Medications targeting this system may be worthy of further investigation for PTSD treatment. Our findings suggest that sex and race effects should be considered in future treatment research.


Subject(s)
Cardiovascular Diseases , Stress Disorders, Post-Traumatic , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Renin-Angiotensin System , Stress Disorders, Post-Traumatic/drug therapy
11.
Transl Psychiatry ; 10(1): 363, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110066

ABSTRACT

Inhibition of the angiotensin type 1 receptor (AT1R) has been shown to decrease fear responses in both humans and rodents. These effects are attributed to modulation of extinction learning, however the contribution of AT1R to alternative memory processes remains unclear. Using classic Pavlovian conditioning combined with radiotelemetry and whole-genome RNA sequencing, we evaluated the effects of the AT1R antagonist losartan on fear memory reconsolidation. Following the retrieval of conditioned auditory fear memory, animals were given a single intraperitoneal injection of losartan or saline. In response to the conditioned stimulus (CS), losartan-treated animals exhibited significantly less freezing at 24 h and 1 week; an effect that was dependent upon memory reactivation and independent of conditioned cardiovascular reactivity. Using an unbiased whole-genome RNA sequencing approach, transcriptomic analysis of the basolateral amygdala (BLA) identified losartan-dependent differences in gene expression during the reconsolidation phase. These findings demonstrate that post-retrieval losartan modifies behavioral and transcriptomic markers of conditioned fear memory, supporting an important regulatory role for this receptor in reconsolidation and as a potential pharmacotherapeutic target for maladaptive fear disorders such as PTSD.


Subject(s)
Amygdala , Receptor, Angiotensin, Type 1 , Animals , Conditioning, Classical , Extinction, Psychological , Fear , Memory
12.
J Psychiatr Res ; 124: 85-90, 2020 05.
Article in English | MEDLINE | ID: mdl-32126364

ABSTRACT

BACKGROUND: Trauma and symptoms of posttraumatic stress disorder (PTSD) have repeatedly been linked to impaired cardiovascular functioning. Poor fear extinction is a well-established biomarker of PTSD that may provide insight into mechanisms underlying cardiovascular risk. The current study probed the cardiovascular response to extinction in a sample of trauma-exposed individuals. METHODS: Participants were 51 trauma-exposed women who underwent a fear conditioning paradigm. Heart rate (HR) during extinction was examined in response to a conditioned stimulus that was previously paired with an aversive unconditioned stimulus (CS+) and one that was never paired (CS-). Heart rate variability (HRV) was calculated at baseline and during the extinction session. RESULTS: Consistent with fear bradycardia, initial HR deceleration (.5-2s) after CS + onset occurred during early extinction and appeared to extinguish over time. Higher baseline HRV was significantly associated with greater fear bradycardia during early extinction. CONCLUSIONS: This is the first study to demonstrate a pattern of fear bradycardia in early extinction, which was associated with higher HRV levels and decreased over the course of the extinction phase. These results suggest that increased fear bradycardia may be indicative of greater vagal control (i.e., HRV), both of which are psychophysiological biomarkers that may influence cardiovascular and autonomic disease risk in trauma-exposed individuals.


Subject(s)
Extinction, Psychological , Stress Disorders, Post-Traumatic , Conditioning, Classical , Fear , Female , Heart Rate , Humans
13.
Brain Behav Immun ; 83: 260-269, 2020 01.
Article in English | MEDLINE | ID: mdl-31682970

ABSTRACT

Post-traumatic stress disorder (PTSD) is associated with a greater risk of incident hypertension and cardiovascular disease. Inflammation, impaired baroreflex sensitivity (BRS) decreased parasympathetic nervous system (PNS) and overactive sympathetic nervous system (SNS) activity are suggested as contributing mechanisms. Increasing severity of PTSD symptoms has been linked to greater cardiovascular risk; however, the impact of PTSD symptom severity on inflammation and autonomic control of blood pressure has not yet been explored. We hypothesized that increasing PTSD symptom severity is linked to higher inflammation, greater SNS activity, lower PNS reactivity and impaired BRS. Seventy Veterans participated in this study: 28 with severe PTSD ((Clinical Administered PTSD Scale (CAPS) > 60; S-PTSD), 16 with moderate PTSD (CAPS ≥ 45 ≤ 60; M-PTSD) and 26 Controls (CAPS < 45; NO-PTSD). We recorded continuous blood pressure (BP), heart rate (HR) via EKG, heart rate variability (HRV) markers reflecting PNS and muscle sympathetic nerve activity (MSNA) at rest, during arterial baroreflex sensitivity (BRS) testing via the modified Oxford technique, and during 3 min of mental stress via mental arithmetic. Blood samples were analyzed for 12 biomarkers of systemic and vascular inflammation. While BP was comparable between severity groups, HR tended to be higher (p = 0.055) in S-PTSD (76 ±â€¯2 beats/min) than in Controls (67 ±â€¯2 beats/min) but comparable to M-PTSD (70 ±â€¯3 beats/min). There were no differences in resting HRV and MSNA between groups; however, cardiovagal BRS was blunted (p = 0.021) in S-PTSD (10 ±â€¯1 ms/mmHg) compared to controls (16 ±â€¯3 ms/mmHg) but comparable to M-PTSD (12 ±â€¯2 ms/mmHg). Veterans in the S-PTSD group had a higher (p < 0.001) combined inflammatory score compared to both M-PTSD and NO-PTSD. Likewise, while mental stress induced similar SNS and cardiovascular responses between the groups, there was a greater reduction in HRV in S-PTSD compared to both M-PTSD and NO-PTSD. In summary, individuals with severe PTSD symptoms have higher inflammation, greater impairment of BRS, a trend towards higher resting HR and exaggerated PNS withdrawal at the onset of mental stress that may contribute to cardiovascular risk in severe PTSD.


Subject(s)
Inflammation/physiopathology , Stress Disorders, Post-Traumatic/physiopathology , Sympathetic Nervous System/physiopathology , Adult , Baroreflex , Blood Pressure , Female , Heart Rate , Humans , Male , Stress Disorders, Post-Traumatic/pathology
15.
Biol Psychiatry ; 86(12): 899-909, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31420088

ABSTRACT

BACKGROUND: The renin-angiotensin system has been implicated in posttraumatic stress disorder; however, the mechanisms responsible for this connection and the therapeutic potential of targeting the renin-angiotensin system in posttraumatic stress disorder remain unknown. Using an angiotensin receptor bacterial artificial chromosome (BAC) and enhanced green fluorescent protein (eGFP) reporter mouse, combined with neuroanatomical, pharmacological, and behavioral approaches, we examined the role of angiotensin II type 2 receptor (AT2R) in fear-related behavior. METHODS: Dual immunohistochemistry with retrograde labeling was used to characterize AT2R-eGFP+ cells in the amygdala of the AT2R-eGFP-BAC reporter mouse. Pavlovian fear conditioning and behavioral pharmacological analyses were used to demonstrate the effects of AT2R activation on fear memory in male C57BL/6 mice. RESULTS: AT2R-eGFP+ neurons in the amygdala were predominantly expressed in the medial amygdala and the medial division of the central amygdala (CeM), with little AT2R-eGFP expression in the basolateral amygdala or lateral division of the central amygdala. Characterization of AT2R-eGFP+ neurons in the CeM demonstrated distinct localization to gamma-aminobutyric acidergic projection neurons. Mice receiving acute intra-central amygdala injections of the selective AT2R agonist compound 21 prior to tests for cued or contextual fear expression displayed less freezing. Retrograde labeling of AT2R-eGFP+ neurons projecting to the periaqueductal gray revealed AT2R-eGFP+ neuronal projections from the CeM to the periaqueductal gray, a key brain structure mediating fear-related freezing. CONCLUSIONS: These findings suggest that CeM AT2R-expressing neurons can modulate central amygdala outputs that play a role in fear expression, providing new evidence for a novel angiotensinergic circuit in the regulation of fear.


Subject(s)
Central Amygdaloid Nucleus/physiology , Fear/physiology , Neurons/physiology , Receptor, Angiotensin, Type 2/physiology , Animals , Anxiety/physiopathology , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/metabolism , Conditioning, Classical , Corticosterone/blood , Locomotion , Male , Mice, Inbred C57BL , Neural Pathways/cytology , Neurons/metabolism , Periaqueductal Gray/cytology , Receptor, Angiotensin, Type 2/metabolism
16.
Physiol Rep ; 7(7): e14057, 2019 04.
Article in English | MEDLINE | ID: mdl-30968587

ABSTRACT

Elevated Resting Blood Pressure (ERBP) in the prehypertensive range is associated with increased risk of hypertension and cardiovascular disease, the mechanisms of which remain unclear. Prior studies have suggested that ERBP may be associated with overactivation and dysregulation of the sympathetic nervous system (SNS). We hypothesized that compared to normotensives (≤120/80 mmHg), ERBP (120/80-139/89 mmHg) has higher SNS activity, impaired arterial baroreflex sensitivity (BRS), and increased vascular inflammation. Twenty-nine participants were studied: 16 otherwise healthy individuals with ERBP (blood pressure (BP) 130 ± 2/85 ± 2 mmHg) and 13 matched normotensive controls (mean BP 114 ± 2/73 ± 2 mmHg). We measured muscle sympathetic nerve activity (MSNA), beat-to-beat BP, and continuous electrocardiogram at rest and during arterial BRS testing via the modified Oxford technique. Blood was analyzed for the following biomarkers of vascular inflammation: lipoprotein-associated phospholipase A2 (Lp-PLA2), E-selectin, and intercellular adhesion molecule 1 (ICAM-1). Resting MSNA burst frequency (22 ± 2 vs. 16 ± 2 bursts/min, P = 0.036) and burst incidence (36 ± 3 vs. 25 ± 3 bursts/100 heart beats, P = 0.025) were higher in ERBP compared to controls. Cardiovagal BRS was blunted in ERBP compared to controls (13 ± 2 vs. 20 ± 3 msec/mmHg, P = 0.032), while there was no difference in sympathetic BRS between groups. Lp-PLA2 (169 ± 8 vs. 142 ± 9 nmol/min/mL, P = 0.020) and E-selectin (6.89 ± 0.6 vs. 4.45 ± 0.51 ng/mL, P = 0.004) were higher in ERBP versus controls. E-selectin (r = 0.501, P = 0.011) and ICAM-1 (r = 0.481, P = 0.015) were positively correlated with MSNA, while E-selectin was negatively correlated with cardiovagal BRS (r = -0.427, P = 0.030). These findings demonstrate that individuals with ERBP have SNS overactivity and impaired arterial BRS that are linked to biomarkers of vascular inflammation.


Subject(s)
Baroreflex/physiology , Blood Pressure/physiology , Inflammation/physiopathology , Prehypertension/physiopathology , Sympathetic Nervous System/physiopathology , Vascular Diseases/physiopathology , Adult , Blood Pressure Determination , Female , Heart Rate/physiology , Hemodynamics/physiology , Humans , Male
17.
Anal Bioanal Chem ; 411(19): 4661-4671, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30953113

ABSTRACT

The renin-angiotensin system (RAS) of the brain produces a series of biologically active angiotensinogen-derived peptides involved in physiological homeostasis and pathophysiology of disease. Despite significant research efforts to date, a comprehensive understanding of brain RAS physiology is lacking. A significant challenge has been the limited set of bioanalytical assays capable of detecting angiotensin (Ang) peptides at physiologically low concentrations (2-15 fmol/g of wet tissue) and sufficient chemical specificity for unambiguous molecular identifications. Additionally, a complex brain anatomy calls for microanalysis of specific tissue regions, thus further taxing sensitivity requirements for identification and quantification in studies of the RAS. To fill this technology gap, we here developed a microanalytical assay by coupling a laboratory-built capillary electrophoresis (CE) nano-electrospray ionization (nano-ESI) platform to a high-resolution mass spectrometer (HRMS). Using parallel reaction monitoring, we demonstrated that this technology achieved confident identification and quantification of the Ang peptides at approx. 5 amol to 300 zmol sensitivity. This microanalytical assay revealed differential Ang peptide profiles between tissues that were micro-sampled from the subfornical organ and the paraventricular nucleus of the hypothalamus, important brain regions involved in thirst and water homeostasis and neuroendocrine regulation to stress. Microanalytical CE-nano-ESI-HRMS extends the analytical toolbox of neuroscience to help better understand the RAS.


Subject(s)
Angiotensins/metabolism , Brain/metabolism , Electrophoresis, Capillary/methods , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Male , Mice , Mice, Inbred C57BL , Proof of Concept Study
18.
Front Behav Neurosci ; 12: 276, 2018.
Article in English | MEDLINE | ID: mdl-30483079

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by a heightened emotional and physiological state and an impaired ability to suppress or extinguish traumatic fear memories. Exaggerated physiological responses may contribute to increased cardiovascular disease (CVD) risk in this population, but whether treatment for PTSD can offset CVD risk remains unknown. To further evaluate physiological correlates of fear learning, we used a novel pre-clinical conditioned cardiovascular testing paradigm and examined the effects of Pavlovian fear conditioning and extinction training on mean arterial pressure (MAP) and heart rate (HR) responses. We hypothesized that a fear conditioned cardiovascular response could be detected in a novel context and attenuated by extinction training. In a novel context, fear conditioned mice exhibited marginal increases in MAP (∼3 mmHg) and decreases in HR (∼20 bpm) during CS presentation. In a home cage context, the CS elicited significant increases in both HR (100 bpm) and MAP (20 mmHg). Following extinction training, the MAP response was suppressed while CS-dependent HR responses were variable. These pre-clinical data suggest that extinction learning attenuates the acute MAP responses to conditioned stimuli over time, and that MAP and HR responses may extinguish at different rates. These results suggest that in mouse models of fear learning, conditioned cardiovascular responses are modified by extinction training. Understanding these processes in pre-clinical disease models and in humans with PTSD may be important for identifying interventions that facilitate fear extinction and attenuate hyper-physiological responses, potentially leading to improvements in the efficacy of exposure therapy and PTSD-CVD comorbidity outcomes.

19.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1272-R1280, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30303706

ABSTRACT

Posttraumatic stress disorder (PTSD) is characterized by increased sympathetic nervous system (SNS) activity, blunted parasympathetic nervous system (PNS) activity, and impaired baroreflex sensitivity (BRS), which contribute to accelerated cardiovascular disease. Patients with PTSD also have chronic stress-related elevations in resting blood pressure (BP), often in the prehypertensive range; yet, it is unclear if elevated resting blood pressure (ERBP) augments these autonomic derangements in PTSD. We hypothesized that compared with normotensive PTSD (N-PTSD), those with ERBP (E-PTSD) have further increased SNS, decreased PNS activity, and impaired BRS at rest and exaggerated SNS reactivity, PNS withdrawal, and pressor responses during stress. In 16 E-PTSD and 17 matched N-PTSD, we measured continuous BP, ECG, muscle sympathetic nerve activity (MSNA), and heart rate variability (HRV) markers reflecting cardiac PNS activity [standard deviation of R-R intervals (SDNN), root mean square of differences in successive R-R intervals (RMSSD), and high frequency power (HF)] during 5 min of rest and 3 min of mental arithmetic. Resting MSNA ( P = 0.943), sympathetic BRS ( P = 0.189), and cardiovagal BRS ( P = 0.332) were similar between groups. However, baseline SDNN (56 ± 6 vs. 78 ± 8 ms, P = 0.019), RMSSD (39 ± 6 vs. 63 ± 9 ms, P = 0.018), and HF (378 ± 103 vs. 693 ± 92 ms2, P = 0.015) were lower in E-PTSD versus N-PTSD. During mental stress, the systolic blood pressure response ( P = 0.011) was augmented in E-PTSD. Although MSNA reactivity was not different ( P > 0.05), the E-PTSD group had an exaggerated reduction in HRV during mental stress ( P < 0.05). PTSD with ERBP have attenuated resting cardiac PNS activity, coupled with exaggerated BP reactivity and PNS withdrawal during stress.


Subject(s)
Autonomic Nervous System Diseases/physiopathology , Blood Pressure/physiology , Parasympathetic Nervous System/physiopathology , Sympathetic Nervous System/physiopathology , Adult , Autonomic Nervous System/physiopathology , Baroreflex/physiology , Cardiovascular Diseases/physiopathology , Female , Heart Rate/physiology , Humans , Male , Middle Aged , Stress, Psychological/physiopathology , Sympathetic Nervous System/injuries
20.
Psychoneuroendocrinology ; 94: 143-151, 2018 08.
Article in English | MEDLINE | ID: mdl-29783162

ABSTRACT

Alterations in peripheral immune markers are observed in individuals with post-traumatic stress disorder (PTSD). PTSD is characterized in part by impaired extinction of fear memory for a traumatic experience. We hypothesized that fear memory extinction is regulated by immune signaling stimulated when fear memory is retrieved. The relationship between fear memory and the peripheral immune response was tested using auditory Pavlovian fear conditioning in mice. Memory for the association was quantified by the amount of conditioned freezing exhibited in response to the conditioned stimulus (CS), extinction and time-dependent changes in circulating inflammatory cytokines. Brief extinction training with 12 CS rapidly and acutely increased circulating levels of the cytokine interleukin-6 (IL-6), downstream IL-6 signaling, other IL-6 related pro-inflammatory cytokines. Transgenic manipulations or neutralizing antibodies that inhibit IL-6 activity did not affect conditioned freezing during the acquisition of fear conditioning or extinction but significantly reduced conditioned freezing 24 h after extinction training with 12 CS. Conversely, conditioned freezing after extinction training was unchanged by IL-6 inhibition when 40 CS were used during the extinction training session. In addition to effectively diminishing conditioned freezing, extinction training with 40 CS also diminished the subsequent IL-6 response to the CS. These data demonstrate that IL-6 released following fear memory retrieval contributes to the maintenance of that fear memory and that this effect is extinction dependent. These findings extend the current understanding for the role of the immune system in PTSD and suggest that IL-6 and other IL-6 related pro-inflammatory cytokines may contribute to the persistence of fear memory in PTSD where fear memory extinction is impaired.


Subject(s)
Fear/physiology , Memory/physiology , Stress, Psychological/immunology , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Extinction, Psychological/physiology , Interleukin-6/analysis , Interleukin-6/metabolism , Male , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Prefrontal Cortex/physiology , Stress Disorders, Post-Traumatic/immunology , Stress Disorders, Post-Traumatic/psychology , Wounds and Injuries/immunology , Wounds and Injuries/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...