1
|
Şahin M, Yüzügüllü D. Outcomes of metabolic syndrome and anxiety levels in light and heavy smokers. PeerJ 2025; 13:e19069. [PMID: 40061234 PMCID: PMC11890032 DOI: 10.7717/peerj.19069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/10/2025] [Indexed: 05/13/2025] Open
Abstract
Background This study aimed to assess the impact of smoking status, as measured by pack-years (PY), on components of metabolic syndrome while considering the influence of anxiety. Design This cross-sectional study was conducted at a smoking cessation clinic in Turkey, enrolling individuals who visited the clinic in 2022. The Fagerstrom Test for Nicotine Dependence and the State-Trait Anxiety Inventory were utilized as assessment tools, while metabolic syndrome parameters (body mass index, hypertension, hyperglycemia, dyslipidemia) were evaluated. Smoking status was classified based on pack-years. Results The study revealed a dose-dependent relationship between smoking status and essential metabolic factors such as systolic blood pressure (SBP), diastolic blood pressure (DBP), hemoglobin A1c (HbA1c), and low-density lipoprotein (LDL). Notably, triglyceride (TG) levels exhibited a significant increase, particularly at 25 pack years. While anxiety levels did not exhibit a significant correlation with smoking status, they demonstrated an upward trend with increasing SBP and DBP values. Anxiety levels did not exhibit a significant correlation with smoking status. Conclusions A significant association was identified between nicotine addiction, as indicated by PY, and both metabolic syndrome parameters and anxiety levels. Early smoking cessation is strongly recommended for current smokers, and former smokers are advised to abstain from smoking to mitigate its adverse effects on metabolic syndrome components. These findings underscore the interconnectedness of cigarette smoking's effects on both physical and mental health, emphasizing the necessity of comprehensive approaches encompassing both metabolic disorder management and mental health support within cessation programs.
Collapse
Affiliation(s)
- Musa Şahin
- Provincial Health Directorate of Adana, Ministry of Health Türkiye, Adana, Turkey
| | - Didem Yüzügüllü
- Provincial Health Directorate of Adana, Ministry of Health Türkiye, Adana, Turkey
| |
Collapse
|
2
|
O'Leary KB, Khan JS. Pharmacotherapy for Anxiety Disorders. Psychiatr Clin North Am 2024; 47:689-709. [PMID: 39505448 DOI: 10.1016/j.psc.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Anxiety disorders are the most common psychiatric illness and include disorders such as generalized anxiety disorder (GAD), panic disorder (PD), and social anxiety disorder (SAD). Psychotherapy and pharmacotherapy are both effective treatments for anxiety disorders, with efficacy between 60% and 85%. Selective serotonin reuptake inhibitors and serotonin-norepinephrine reuptake inhibitors are first-line pharmacologic treatment for GAD, PD, and SAD. Recommendations for treating pediatric and geriatric populations vary slightly, but first-line treatments remain the same. Recent advancements in the treatment of anxiety disorders are limited although research has discovered novel pathways, which may lead to additional treatment options in the future.
Collapse
Affiliation(s)
- Kerry B O'Leary
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, 1 Baylor Plaza - BCM350, Houston, TX 77030, USA. Kerry.O'
| | - Jeffrey S Khan
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, 1977 Butler Boulevard, E4.203. Houston, TX 77030, USA
| |
Collapse
|
3
|
Kang S, Kim J, Yang JS, Jeon YJ, Lee HH, Suglia SF, Tsai AC, Kang JI, Jung SJ. Use of renin-angiotensin system blockers and posttraumatic stress disorder risk in the UK Biobank: a retrospective cohort study. BMC Med 2024; 22:489. [PMID: 39443947 PMCID: PMC11515478 DOI: 10.1186/s12916-024-03704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Previous research has shown that the use of renin-angiotensin system (RAS) blockers is linked to a lower prevalence of posttraumatic stress disorder (PTSD), but longitudinal studies are scarce. We aimed to estimate the association between the use of RAS blockers and the risk of PTSD among individuals taking antihypertensive medications. METHODS This longitudinal study included participants aged 40-69 from the UK Biobank. Exposure data were obtained from the initial assessment (2006-10), while outcome data were obtained from the online mental health questionnaire administered 6-11 years later (2016-17). We included participants who were under antihypertensive treatment and did not have a prior diagnosis of PTSD before the initial assessment. Use of RAS blockers was defined as self-reported regular use, at the initial assessment, of angiotensin-converting enzyme inhibitor (ACEi) or angiotensin receptor blocker (ARB). Among participants who experienced adverse life experiences, cases of probable PTSD were defined with the six-item PTSD Checklist-Civilian version score ≥ 14. Logistic regression with inverse probability of treatment weighting was used to estimate the odds ratios (ORs) and 95% confidence interval (CI) for the association between RAS blocker use and the risk of probable PTSD. RESULTS Of the 15,954 participants (mean age = 59.9 years; 42.6% women) under antihypertensive treatment with no prior history of PTSD at the initial assessment, 64.5% were taking RAS blockers. After a mean follow-up of 7.5 years, 1,249 (7.8%) were newly identified with probable PTSD. RAS blocker users had a lower risk of probable PTSD than RAS blocker non-users (OR = 0.84 [95% CI: 0.75-0.94]), whereas the use of other antihypertensive medications showed no such association (users vs. non-users; calcium channel blockers, OR = 0.99 [95% CI: 0.88-1.11]; beta-blockers, 1.20 [1.08-1.34]; and thiazide-related diuretics, 1.15 [1.03-1.29]). The association between probable PTSD risk and the use of ACEi vs. ARB showed no significant difference (p = 0.96). CONCLUSIONS Among individuals under antihypertensive treatment, the use of RAS blockers was associated with a decreased risk of probable PTSD. This added benefit of RAS blockers should be considered in the selection of antihypertensive medications.
Collapse
Affiliation(s)
- Sunghyuk Kang
- Department of Preventive Medicine, Yonsei University College of Medicine, Yonsei-Ro 50-1, Seodaemun-Gu, Seoul, 03722, South Korea
- Department of Psychiatry and Institute of Behavioural Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jimin Kim
- Department of Public Health, Graduate School, Yonsei University, Seoul, Korea
| | - Ji Su Yang
- Department of Public Health, Graduate School, Yonsei University, Seoul, Korea
| | - Ye Jin Jeon
- Department of Public Health, Graduate School, Yonsei University, Seoul, Korea
| | - Hyeok-Hee Lee
- Department of Preventive Medicine, Yonsei University College of Medicine, Yonsei-Ro 50-1, Seodaemun-Gu, Seoul, 03722, South Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Shakira F Suglia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Alexander C Tsai
- Center for Global Health and Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jee In Kang
- Department of Psychiatry and Institute of Behavioural Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Jae Jung
- Department of Preventive Medicine, Yonsei University College of Medicine, Yonsei-Ro 50-1, Seodaemun-Gu, Seoul, 03722, South Korea.
- Department of Public Health, Graduate School, Yonsei University, Seoul, Korea.
- Center for Global Health and Mongan Institute, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
4
|
Smith HC, Yu Z, Iyer L, Marvar PJ. Sex-Dependent Effects of Angiotensin Type 2 Receptor-Expressing Medial Prefrontal Cortex Interneurons in Fear Extinction Learning. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100340. [PMID: 39140003 PMCID: PMC11321323 DOI: 10.1016/j.bpsgos.2024.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 08/15/2024] Open
Abstract
Background The renin-angiotensin system has been identified as a potential therapeutic target for posttraumatic stress disorder, although 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. Therefore, we used a combination of imaging, cre/lox, and behavioral methods to investigate mPFC-AT2R-expressing neurons in fear and stess related behavior. Methods To characterize mPFC-AT2R-expressing neurons in the mPFC, AT2R-Cre/tdTomato male and female mice were used for immunohistochemistry. mPFC brain sections were stained with glutamatergic or interneuron markers, and density of AT2R+ cells and colocalization with each marker were quantified. To assess fear-related behaviors in AT2R-flox mice, we selectively deleted AT2R from mPFC neurons using a Cre-expressing adeno-associated virus. Mice then underwent Pavlovian auditory fear conditioning, elevated plus maze, and open field testing. Results Immunohistochemistry results revealed that AT2R was densely expressed throughout the mPFC and primarily expressed in somatostatin interneurons in a sex-dependent manner. Following fear conditioning, mPFC-AT2R Cre-lox deletion impaired extinction and increased exploratory behavior in female but not male mice, while locomotion was unaltered by mPFC-AT2R deletion in both sexes. Conclusions These results identify mPFC-AT2R+ neurons as a novel subgroup of somatostatin interneurons and reveal their role in regulating fear learning in a sex-dependent manner, potentially offering insights into novel therapeutic targets for posttraumatic stress disorder.
Collapse
Affiliation(s)
- Hannah C. Smith
- Department of Neuroscience, George Washington University, Washington, DC
| | - Zhe Yu
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| | - Laxmi Iyer
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| | - Paul J. Marvar
- Department of Neuroscience, George Washington University, Washington, DC
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC
| |
Collapse
|
5
|
Zika O, Appel J, Klinge C, Shkreli L, Browning M, Wiech K, Reinecke A. Reduction of Aversive Learning Rates in Pavlovian Conditioning by Angiotensin II Antagonist Losartan: A Randomized Controlled Trial. Biol Psychiatry 2024; 96:247-255. [PMID: 38309320 DOI: 10.1016/j.biopsych.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Angiotensin receptor blockade has been linked to aspects of aversive learning and memory formation and to the prevention of posttraumatic stress disorder symptom development. METHODS We investigated the influence of the angiotensin receptor blocker losartan on aversive Pavlovian conditioning using a probabilistic learning paradigm. In a double-blind, randomized, placebo-controlled design, we tested 45 (18 female) healthy volunteers during a baseline session, after application of losartan or placebo (drug session), and during a follow-up session. During each session, participants engaged in a task in which they had to predict the probability of an electrical stimulation on every trial while the true shock contingencies switched repeatedly between phases of high and low shock threat. Computational reinforcement learning models were used to investigate learning dynamics. RESULTS Acute administration of losartan significantly reduced participants' adjustment during both low-to-high and high-to-low threat changes. This was driven by reduced aversive learning rates in the losartan group during the drug session compared with baseline. The 50-mg drug dose did not induce reduction of blood pressure or change in reaction times, ruling out a general reduction in attention and engagement. Decreased adjustment of aversive expectations was maintained at a follow-up session 24 hours later. CONCLUSIONS This study shows that losartan acutely reduces Pavlovian learning in aversive environments, thereby highlighting a potential role of the renin-angiotensin system in anxiety development.
Collapse
Affiliation(s)
- Ondrej Zika
- Max Planck Institute for Human Development, Berlin, Germany
| | - Judith Appel
- Behavioural Science Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Corinna Klinge
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Lorika Shkreli
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Trust, Warneford Hospital, Oxford, United Kingdom
| | - Katja Wiech
- Wellcome Centre for Integrative Functional Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Andrea Reinecke
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Trust, Warneford Hospital, Oxford, United Kingdom.
| |
Collapse
|
6
|
Shkreli L, Thoroddsen T, Kobelt M, Martens MA, Browning M, Harmer CJ, Cowen P, Reinecke A. Acute Angiotensin II Receptor Blockade Facilitates Parahippocampal Processing During Memory Encoding in High-Trait-Anxious Individuals. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100286. [PMID: 38323154 PMCID: PMC10844816 DOI: 10.1016/j.bpsgos.2023.100286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024] Open
Abstract
Background Angiotensin II receptor blockers (ARBs) have been associated with preventing posttraumatic stress disorder symptom development and improving memory. However, the underlying neural mechanisms are poorly understood. This study investigated ARB effects on memory encoding and hippocampal functioning that have previously been implicated in posttraumatic stress disorder development. Methods In a double-blind randomized design, 40 high-trait-anxious participants (33 women) received the ARB losartan (50 mg) or placebo. At drug peak level, participants encoded images of animals and landscapes before undergoing functional magnetic resonance imaging, where they viewed the encoded familiar images and unseen novel images to be memorized and classified as animals/landscapes. Memory recognition was assessed 1 hour after functional magnetic resonance imaging. To analyze neural effects, whole-brain analysis, hippocampus region-of-interest analysis, and exploratory multivariate pattern similarity analysis were employed. Results ARBs facilitated parahippocampal processing. In the whole-brain analysis, losartan enhanced brain activity for familiar images in the parahippocampal gyrus (PHC), anterior cingulate cortex, and caudate. For novel images, losartan enhanced brain activity in the PHC only. Pattern similarity analysis showed that losartan increased neural stability in the PHC when processing novel and familiar images. However, there were no drug effects on memory recognition or hippocampal activation. Conclusions Given that the hippocampus receives major input from the PHC, our findings suggest that ARBs may modulate higher-order visual processing through parahippocampal involvement, potentially preserving intact memory input. Future research needs to directly investigate whether this effect may underlie the preventive effects of ARBs in the development of posttraumatic stress disorder.
Collapse
Affiliation(s)
- Lorika Shkreli
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | | | - Malte Kobelt
- Institute of Cognitive Neuroscience, Ruhr-Universität Bochum, Bochum, Germany
| | | | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Catherine J. Harmer
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Phil Cowen
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Andrea Reinecke
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
7
|
Xu T, Chen Z, Zhou X, Wang L, Zhou F, Yao D, Zhou B, Becker B. The central renin-angiotensin system: A genetic pathway, functional decoding, and selective target engagement characterization in humans. Proc Natl Acad Sci U S A 2024; 121:e2306936121. [PMID: 38349873 PMCID: PMC10895353 DOI: 10.1073/pnas.2306936121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024] Open
Abstract
Accumulating evidence suggests that the brain renin angiotensin system (RAS) plays a pivotal role in the regulation of cognition and behavior as well as in the neuropathology of neurological and mental disorders. The angiotensin II type 1 receptor (AT1R) mediates most functional and neuropathology-relevant actions associated with the central RAS. However, an overarching comprehension to guide translation and utilize the therapeutic potential of the central RAS in humans is currently lacking. We conducted a comprehensive characterization of the RAS using an innovative combination of transcriptomic gene expression mapping, image-based behavioral decoding, and pre-registered randomized controlled discovery-replication pharmacological resting-state functional magnetic resonance imaging (fMRI) trials (N = 132) with a selective AT1R antagonist. The AT1R exhibited a particular dense expression in a subcortical network encompassing the thalamus, striatum, and amygdalo-hippocampal formation. Behavioral decoding of the AT1R gene expression brain map showed an association with memory, stress, reward, and motivational processes. Transient pharmacological blockade of the AT1R further decreased neural activity in subcortical systems characterized by a high AT1R expression, while increasing functional connectivity in the cortico-basal ganglia-thalamo-cortical circuitry. Effects of AT1R blockade on the network level were specifically associated with the transcriptomic signatures of the dopaminergic, opioid, acetylcholine, and corticotropin-releasing hormone signaling systems. The robustness of the results was supported in an independent pharmacological fMRI trial. These findings present a biologically informed comprehensive characterization of the central AT1R pathways and their functional relevance on the neural and behavioral level in humans.
Collapse
Affiliation(s)
- Ting Xu
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Zhiyi Chen
- Experimental Research Center for Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing400037, People’s Republic of China
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Xinqi Zhou
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu 610066, People’s Republic of China
| | - Lan Wang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Feng Zhou
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Dezhong Yao
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Bo Zhou
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
| | - Benjamin Becker
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong999077, People’s Republic of China
- Department of Psychology, The University of Hong Kong, Hong Kong999077, People’s Republic of China
| |
Collapse
|
8
|
de Miranda AS, Macedo DS, Rocha NP, Teixeira AL. Targeting the Renin-Angiotensin System (RAS) for Neuropsychiatric Disorders. Curr Neuropharmacol 2024; 22:107-122. [PMID: 36173067 PMCID: PMC10716884 DOI: 10.2174/1570159x20666220927093815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/03/2022] [Accepted: 08/14/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Neuropsychiatric disorders, such as mood disorders, schizophrenia, and Alzheimer's disease (AD) and related dementias, are associated to significant morbidity and mortality worldwide. The pathophysiological mechanisms of neuropsychiatric disorders remain to be fully elucidated, which has hampered the development of effective therapies. The Renin Angiotensin System (RAS) is classically viewed as a key regulator of cardiovascular and renal homeostasis. The discovery that RAS components are expressed in the brain pointed out a potential role for this system in central nervous system (CNS) pathologies. The understanding of RAS involvement in the pathogenesis of neuropsychiatric disorders may contribute to identifying novel therapeutic targets. AIMS We aim to report current experimental and clinical evidence on the role of RAS in physiology and pathophysiology of mood disorders, schizophrenia, AD and related dementias. We also aim to discuss bottlenecks and future perspectives that can foster the development of new related therapeutic strategies. CONCLUSION The available evidence supports positive therapeutic effects for neuropsychiatric disorders with the inhibition/antagonism of the ACE/Ang II/AT1 receptor axis or the activation of the ACE2/Ang-(1-7)/Mas receptor axis. Most of this evidence comes from pre-clinical studies and clinical studies lag much behind, hampering a potential translation into clinical practice.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- Interdisciplinary Laboratory of Medical Investigation (LIIM), Faculty of Medicine, UFMG, Belo Horizonte, MG, Brazil
- Department of Morphology, Laboratory of Neurobiology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle S Macedo
- Department of Physiology and Pharmacology, Neuropharmacology Laboratory, Drug Research, and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Natalia P Rocha
- Department of Neurology, The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, McGovern Medical School, University of Texas Health Science Center at Houston, TX, USA
| | - Antonio L Teixeira
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, University of Texas Health Science Center at Houston, TX, USA
- Faculdade Santa Casa BH, Belo Horizonte, Brasil
| |
Collapse
|
9
|
Smith HC, Yu Z, Iyer L, Marvar PJ. Sex-dependent effects of angiotensin type 2 receptor expressing medial prefrontal cortex (mPFC) interneurons in fear extinction learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568156. [PMID: 38045293 PMCID: PMC10690250 DOI: 10.1101/2023.11.21.568156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
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.
Collapse
Affiliation(s)
- Hannah C. Smith
- Department of Neuroscience, George Washington University, Washington, DC
| | - Zhe Yu
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
| | - Laxmi Iyer
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
| | - Paul J. Marvar
- Department of Neuroscience, George Washington University, Washington, DC
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington DC
| |
Collapse
|
10
|
Ortiz-Nazario E, Denton-Ortiz CM, Soto-Escobar LDM, Mateo-Mayol Z, Colon-Romero M, Hernandez-Lopez A, Porter JT. Sex-dependent effects of angiotensin II type 1 receptor blocker on molecular and behavioral changes induced by single prolonged stress. Behav Brain Res 2023; 454:114639. [PMID: 37652238 PMCID: PMC10530531 DOI: 10.1016/j.bbr.2023.114639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that not only entails alterations in fear behavior and anxiety but also includes neuroendocrine dysfunctions involving the hypothalamic pituitary adrenal (HPA) axis and the renin-angiotensin system. Recent preclinical studies demonstrate that activation of the angiotensin type 1 receptor (AT1R) in the paraventricular region of the hypothalamus (PVR) promotes anxiety-like behaviors and enables microglia proliferation. An increase in microglia and anxiety-like behavior also occurs in the PTSD animal model single-prolonged stress (SPS). In the present study, we tested whether AT1Rs contribute to the effects of SPS on behavior and microglia in brain structures important for HPA axis regulation and fear behavior. To test this, male and female animals were exposed to SPS and then given the oral AT1R antagonist candesartan beginning one week later. Candesartan did not alter auditory fear conditioning or extinction in SPS-exposed male or female animals. However, we found that the male animals exposed to SPS showed increased anxiety-like behavior, which was reversed by candesartan. In contrast, neither SPS nor candesartan altered anxiety-like behavior in the female animals. At the molecular level, SPS increased the cellular expression of AT1Rs in the PVR of male animals and candesartan reversed this effect, whereas AT1Rs in the PVR of females were unaltered by either SPS or candesartan. Iba1-expressing microglia increased in the PVR after SPS exposure and was reversed by candesartan in both sexes suggesting that SPS stimulates AT1Rs to increase microglia in the PVR. Collectively, these results suggest that the contribution of AT1Rs to the molecular and behavioral effects of SPS is sex-dependent.
Collapse
Affiliation(s)
- Emily Ortiz-Nazario
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Carla M Denton-Ortiz
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Lawry D M Soto-Escobar
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Zaira Mateo-Mayol
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Maria Colon-Romero
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Anixa Hernandez-Lopez
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - James T Porter
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico.
| |
Collapse
|
11
|
Gradus JL, Smith ML, Szentkúti P, Rosellini AJ, Horváth-Puhó E, Lash TL, Galea S, Schnurr PP, Sumner JA, Sørensen HT. Antihypertensive Medications and PTSD Incidence in a Trauma Cohort. J Clin Psychiatry 2023; 84:22m14767. [PMID: 37530605 PMCID: PMC10545136 DOI: 10.4088/jcp.22m14767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Objective: Antihypertensive medications have been examined as agents for posttraumatic stress disorder (PTSD) prevention in trauma-exposed individuals, given well-documented associations between PTSD and increased risk of cardiovascular disease and purported trauma-relevant mechanisms of action for these medications. Evidence regarding the effectiveness of such drugs for this purpose remains mixed. Methods: We conducted a national population-based cohort study using data from Danish national registries to assess whether 4 classes of antihypertensive drugs (beta-adrenoceptor blockers [beta blockers], angiotensin II receptor blockers [ARBs], angiotensin-converting enzyme [ACE] inhibitors, and calcium channel blockers) were associated with a decreased incidence of PTSD (diagnosed according to ICD-10) over a 22-year study period. Data for this study originated from a population-based cohort of over 1.4 million persons who experienced a traumatic event between 1994 and 2016 in Denmark. We calculated the incidence rate of PTSD per 100,000 person-years among persons who filled a prescription for each class of drug in the 60 days prior to a traumatic event and for corresponding unexposed comparison groups. We then used Cox proportional hazards regression to compare the rate of PTSD among persons who filled an antihypertensive medication prescription within 60 days before their trauma to the rate among persons who did not. Results: We found evidence that calcium channel blockers were associated with a decreased incidence of PTSD (adjusted hazard ratio = 0.63, 95% confidence interval [CI] = 0.34, 1.2); all other antihypertensive medication classes had null or near null associations. Conclusions: These findings lay a foundation for additional research focusing on antihypertensive medications that appear most effective in reducing PTSD incidence following trauma and for additional replication work aimed at continuing to clarify the disparate findings reported in the literature to date.
Collapse
Affiliation(s)
- Jaimie L Gradus
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
- Department of Psychiatry, Boston University School of Medicine, Massachusetts
- Corresponding Author: Jaimie L. Gradus, DMSc, DSc, Boston University School of Public Health, 715 Albany St T317E, Boston, MA 02118
| | - Meghan L Smith
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
| | - Péter Szentkúti
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Anthony J Rosellini
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
| | | | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Sandro Galea
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
| | - Paula P Schnurr
- Executive Division, National Center for PTSD, White River Junction, Vermont
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | | | - Henrik T Sørensen
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
12
|
Yu Z, Kisner A, Bhatt A, Polter AM, Marvar PJ. Central amygdala angiotensin type 1 receptor (Agtr1) expressing neurons contribute to fear extinction. Neuropharmacology 2023; 229:109460. [PMID: 36801399 DOI: 10.1016/j.neuropharm.2023.109460] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 01/12/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023]
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.
Collapse
Affiliation(s)
- Zhe Yu
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Amy Bhatt
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Abigail M Polter
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Paul J Marvar
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA; Department of Psychiatry and Behavioral Sciences, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
13
|
Gomes-de-Souza L, Santana FG, Duarte JO, Barretto-de-Souza L, Crestani CC. Angiotensinergic neurotransmission in the bed nucleus of the stria terminalis is involved in cardiovascular responses to acute restraint stress in rats. Pflugers Arch 2023; 475:517-526. [PMID: 36715761 DOI: 10.1007/s00424-023-02791-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023]
Abstract
The brain angiotensin II acting via AT1 receptors is a prominent mechanism involved in physiological and behavioral responses during aversive situations. The AT2 receptor has also been implicated in stress responses, but its role was less explored. Despite these pieces of evidence, the brain sites related to control of the changes during aversive threats by the brain renin-angiotensin system (RAS) are poorly understood. The bed nucleus of the stria terminalis (BNST) is a limbic structure related to the cardiovascular responses by stress, and components of the RAS system were identified in this forebrain region. Therefore, we investigated the role of angiotensinergic neurotransmission present within the BNST acting via local AT1 and AT2 receptors in cardiovascular responses evoked by an acute session of restraint stress in rats. For this, rats were subjected to bilateral microinjection of either the angiotensin-converting enzyme inhibitor captopril, the selective AT1 receptor antagonist losartan, or the selective AT2 receptor antagonist PD123319 before they underwent the restraint stress session. We observed that BNST treatment with captopril reduced the decrease in tail skin temperature evoked by restraint stress, without affecting the pressor and tachycardic responses. Local AT2 receptor antagonism within the BNST reduced both the tachycardia and the drop in tail skin temperature during restraint. Bilateral microinjection of losartan into the BNST did not affect the restraint-evoked cardiovascular changes. Taken together, these data indicate an involvement of BNST angiotensinergic neurotransmission acting via local AT2 receptors in cardiovascular responses during stressful situations.
Collapse
Affiliation(s)
- Lucas Gomes-de-Souza
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Flávia G Santana
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Josiane O Duarte
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Lucas Barretto-de-Souza
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil.
| |
Collapse
|
14
|
Zhang R, Zhao W, Qi Z, Xu T, Zhou F, Becker B. Angiotensin II Regulates the Neural Expression of Subjective Fear in Humans: A Precision Pharmaco-Neuroimaging Approach. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:262-270. [PMID: 36174930 DOI: 10.1016/j.bpsc.2022.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/23/2022] [Accepted: 09/19/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Rodent models and pharmacological neuroimaging studies in humans have been used to test novel pharmacological agents to reduce fear. However, these strategies are limited with respect to determining process-specific effects on the actual subjective experience of fear, which represents the key symptom that motivates patients to seek treatment. In this study, we used a novel precision pharmacological functional magnetic resonance imaging approach based on process-specific neuroaffective signatures to determine effects of the selective angiotensin II type 1 receptor (AT1R) antagonist losartan on the subjective experience of fear. METHODS In a double-blind, placebo-controlled, randomized pharmacological functional magnetic resonance imaging design, healthy participants (N = 87) were administered 50 mg losartan or placebo before they underwent an oddball paradigm that included neutral, novel, and fear oddballs. Effects of losartan on brain activity and connectivity as well as on process-specific multivariate neural signatures were examined. RESULTS AT1R blockade selectively reduced neurofunctional reactivity to fear-inducing visual oddballs in terms of attenuating dorsolateral prefrontal activity and amygdala-ventral anterior cingulate communication. Neurofunctional decoding further demonstrated fear-specific effects in that AT1R blockade reduced the neural expression of subjective fear but not of threat or nonspecific negative affect and did not influence reactivity to novel oddballs. CONCLUSIONS These results show a specific role of the AT1R in regulating the subjective fear experience and demonstrate the feasibility of a precision pharmacological functional magnetic resonance imaging approach to the affective characterization of novel receptor targets for fear in humans.
Collapse
Affiliation(s)
- Ran Zhang
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Ministry of Education, Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Weihua Zhao
- Ministry of Education, Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyu Qi
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Ministry of Education, Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Ting Xu
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Ministry of Education, Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Feng Zhou
- Faculty of Psychology, Southwest University, ChongQing, China; Key Laboratory of Cognition and Personality, Ministry of Education, ChongQing, China.
| | - Benjamin Becker
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Ministry of Education, Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
15
|
The Angiotensin Antagonist Losartan Modulates Social Reward Motivation and Punishment Sensitivity via Modulating Midbrain-Striato-Frontal Circuits. J Neurosci 2023; 43:472-483. [PMID: 36639890 PMCID: PMC9864573 DOI: 10.1523/jneurosci.1114-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/12/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Social deficits and dysregulations in dopaminergic midbrain-striato-frontal circuits represent transdiagnostic symptoms across psychiatric disorders. Animal models suggest that interactions between the dopamine (DA) and renin-angiotensin system (RAS) may modulate learning and reward-related processes. The present study therefore examined the behavioral and neural effects of the Angiotensin II type 1 receptor (AT1R) antagonist losartan on social reward and punishment processing in humans. A preregistered randomized double-blind placebo-controlled between-subject pharmacological design was combined with a social incentive delay (SID) functional MRI (fMRI) paradigm during which subjects could avoid social punishment or gain social reward. Healthy volunteers received a single-dose of losartan (50 mg, n = 43, female = 17) or placebo (n = 44, female = 20). We evaluated reaction times (RTs) and emotional ratings as behavioral and activation and functional connectivity as neural outcomes. Relative to placebo, losartan modulated the reaction time and arousal differences between social punishment and social reward. On the neural level the losartan-enhanced motivational salience of social rewards was accompanied by stronger ventral striatum-prefrontal connectivity during reward anticipation. Losartan increased the reward-neutral difference in the ventral tegmental area (VTA) and attenuated VTA associated connectivity with the bilateral insula in response to punishment during the outcome phase. Thus, losartan modulated approach-avoidance motivation and emotional salience during social punishment versus social reward via modulating distinct core nodes of the midbrain-striato-frontal circuits. The findings document a modulatory role of the renin-angiotensin system in these circuits and associated social processes, suggesting a promising treatment target to alleviate social dysregulations.SIGNIFICANCE STATEMENT Social deficits and anhedonia characterize several mental disorders and have been linked to the midbrain-striato-frontal circuits of the brain. Based on initial findings from animal models we here combine the pharmacological blockade of the Angiotensin II type 1 receptor (AT1R) via losartan with functional MRI (fMRI) to demonstrate that AT1R blockade enhances the motivational salience of social rewards and attenuates the negative impact of social punishment via modulating the communication in the midbrain-striato-frontal circuits in humans. The findings demonstrate for the first time an important role of the AT1R in social reward processing in humans and render the AT1R as promising novel treatment target for social and motivational deficits in mental disorders.
Collapse
|
16
|
Ponomareva OY, Fenster RJ, Ressler KJ. Enhancing Fear Extinction: Pharmacological Approaches. Curr Top Behav Neurosci 2023; 64:289-305. [PMID: 37584834 DOI: 10.1007/7854_2023_443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Extinction is the process by which the memory of a learned conditioned association decreases over time and with introduction of new associations. It is a vital part of fear learning, and it is critical to recovery in multiple fear-related disorders, including Specific and Social Phobias, Panic Disorder, Obsessive Compulsive Disorder (OCD), and Posttraumatic Stress Disorder (PTSD). The process of extinction is also the underlying mechanism for recovery in gold-standard therapies for PTSD, including prolonged exposure, cognitive processing therapy, eye movement desensitization and procession, as well as other empirically-based paradigms. Pharmacological modulators of extinction are thus promising targets for treatment of fear-related disorders. We focus here on emerging psychopharmacological treatments to facilitate extinction: D-cycloserine, scopolamine, losartan, ketamine, and 3,4-methylenedioxymethamphetamine. We also provide an overview of recent advances in molecular pathways that show promise as targets for extincion and inhibitory learning, including pathways related to cannabinoid, brain-derived neurotrophic factor, hypothalamic-pituitary-adrenal signaling, and promising work in neurosteroid compounds.
Collapse
|
17
|
Craske MG, Sandman CF, Stein MB. How can neurobiology of fear extinction inform treatment? Neurosci Biobehav Rev 2022; 143:104923. [DOI: 10.1016/j.neubiorev.2022.104923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
|
18
|
Seligowski AV, Misganaw B, Duffy LA, Ressler KJ, Guffanti G. Leveraging Large-Scale Genetics of PTSD and Cardiovascular Disease to Demonstrate Robust Shared Risk and Improve Risk Prediction Accuracy. Am J Psychiatry 2022; 179:814-823. [PMID: 36069022 PMCID: PMC9633348 DOI: 10.1176/appi.ajp.21111113] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Individuals with posttraumatic stress disorder (PTSD) are significantly more likely to be diagnosed with cardiovascular disease (CVD) (e.g., myocardial infarction, stroke). The evidence for this link is so compelling that the National Institutes of Health convened a working group to determine gaps in the literature, including the need for large-scale genomic studies to identify shared genetic risk. The aim of the present study was to address some of these gaps by utilizing PTSD and CVD genome-wide association study (GWAS) summary statistics in a large biobank sample to determine the shared genetic risk of PTSD and CVD. METHODS A large health care biobank data set was used (N=36,412), combined with GWAS summary statistics from publicly available large-scale PTSD and CVD studies. Disease phenotypes (e.g., PTSD) were collected from electronic health records. De-identified genetic data from the biobank were genotyped using Illumina SNP array. Summary statistics data sets were processed with the following quality-control criteria: 1) SNP heritability h2 >0.05, 2) compute z-statistics (z=beta/SE or z=log(OR)/SE), 3) filter nonvariable SNPs (0 RESULTS Significant genetic correlations were found between PTSD and CVD (rG=0.24, SE=0.06), and Mendelian randomization analyses indicated a potential causal link from PTSD to hypertension (β=0.20, SE=0.04), but not the reverse. PTSD summary statistics significantly predicted PTSD diagnostic status (R2=0.27), and this was significantly improved by incorporating summary statistics from CVD and major depressive disorder (R2=1.30). Further, pathway enrichment analyses indicated that genetic variants involved in shared PTSD-CVD risk included those involved in postsynaptic structure, synapse organization, and interleukin-7-mediated signaling pathways. CONCLUSIONS The results from this study suggest that PTSD and CVD may share genetic risk. Further, these results implicate PTSD as a risk factor leading to the development of hypertension and coronary artery disease. Additional research is needed to determine the clinical utility of these findings.
Collapse
Affiliation(s)
- Antonia V. Seligowski
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Burook Misganaw
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | | | - Kerry J. Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Guia Guffanti
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| |
Collapse
|
19
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
20
|
Seligowski AV, Webber TK, Marvar PJ, Ressler KJ, Philip NS. Involvement of the brain-heart axis in the link between PTSD and cardiovascular disease. Depress Anxiety 2022; 39:663-674. [PMID: 35708302 PMCID: PMC9588548 DOI: 10.1002/da.23271] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/22/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
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.
Collapse
Affiliation(s)
- Antonia V. Seligowski
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | | | | | - Kerry J. Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Noah S. Philip
- VA RR&D Center for Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School, of Brown University, Providence, RI, USA
| |
Collapse
|
21
|
Gasparyan A, Navarro D, Navarrete F, Manzanares J. Pharmacological strategies for post-traumatic stress disorder (PTSD): From animal to clinical studies. Neuropharmacology 2022; 218:109211. [PMID: 35973598 DOI: 10.1016/j.neuropharm.2022.109211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 01/27/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a disabling psychiatric condition with a critical familiar, personal, and social impact. Patients diagnosed with PTSD show various symptoms, including anxiety, depression, psychotic episodes, and sleep disturbances, complicating their therapeutic management. Only sertraline and paroxetine, two selective serotonin reuptake inhibitors, are approved by different international agencies to treat PTSD. In addition, these drugs are generally combined with psychotherapy to achieve positive results. However, these pharmacological strategies present limited efficacy. Nearly half of the PTSD patients do not experience remission of symptoms, possibly due to the high prevalence of psychiatric comorbidities. Therefore, in clinical practice, other off-label medications are common, even though the effectiveness of these drugs needs to be further investigated. In this line, antipsychotics, antiepileptics, adrenergic blockers, benzodiazepines, and other emerging pharmacological agents have aroused interest as potential therapeutic tools to improve some specific symptoms of PTSD. Thus, this review is focused on the most widely used drugs for the pharmacological treatment of PTSD with a translational approach, including clinical and preclinical studies, to emphasize the need to develop safer and more effective medications.
Collapse
Affiliation(s)
- Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain; Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Departamento de Medicina Clínica, Universidad Miguel Hernández, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain; Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Departamento de Medicina Clínica, Universidad Miguel Hernández, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain; Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Departamento de Medicina Clínica, Universidad Miguel Hernández, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550, Alicante, Spain; Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Departamento de Medicina Clínica, Universidad Miguel Hernández, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
22
|
Nord CL, Garfinkel SN. Interoceptive pathways to understand and treat mental health conditions. Trends Cogn Sci 2022; 26:499-513. [PMID: 35466044 DOI: 10.1016/j.tics.2022.03.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
Abstract
An increasing recognition that brain and body are dynamically coupled has enriched our scientific understanding of mental health conditions. Peripheral signals interact centrally to influence how we think and feel, generating our sense of the internal condition of the body, a process known as interoception. Disruptions to this interoceptive system may contribute to clinical conditions, including anxiety, depression, and psychosis. After reviewing the nature of interoceptive disturbances in mental health conditions, this review focuses on interoceptive pathways of existing and putative mental health treatments. Emerging clinical interventions may target novel peripheral treatment mechanisms. Future treatment development requires forward- and back-translation to uncover and target specific interoceptive processes in mental health to elucidate their efficacy relative to interventions targeting other factors.
Collapse
Affiliation(s)
- Camilla L Nord
- MRC Cognition and Brain Sciences Unit, 15 Chaucer Rd, Cambridge CB2 7EF, UK.
| | - Sarah N Garfinkel
- Institute of Cognitive Neuroscience, UCL, Alexandra House, 17-19 Queen Square, London WC1N 3AZ, UK.
| |
Collapse
|
23
|
Correa BHM, Becari L, Peliky Fontes MA, Simões-e-Silva AC, Kangussu LM. Involvement of the Renin-Angiotensin System in Stress: State of the Art and Research Perspectives. Curr Neuropharmacol 2022; 20:1212-1228. [PMID: 34554902 PMCID: PMC9886820 DOI: 10.2174/1570159x19666210719142300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/19/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Along with other canonical systems, the renin-angiotensin system (RAS) has shown important roles in stress. This system is a complex regulatory proteolytic cascade composed of various enzymes, peptides, and receptors. Besides the classical (ACE/Ang II/AT1 receptor) and the counter-regulatory (ACE2/Ang-(1-7)/Mas receptor) RAS axes, evidence indicates that nonclassical components, including Ang III, Ang IV, AT2 and AT4, can also be involved in stress. OBJECTIVE AND METHODS This comprehensive review summarizes the current knowledge on the participation of RAS components in different adverse environmental stimuli stressors, including air jet stress, cage switch stress, restraint stress, chronic unpredictable stress, neonatal isolation stress, and post-traumatic stress disorder. RESULTS AND CONCLUSION In general, activation of the classical RAS axis potentiates stress-related cardiovascular, endocrine, and behavioral responses, while the stimulation of the counter-regulatory axis attenuates these effects. Pharmacological modulation in both axes is optimistic, offering promising perspectives for stress-related disorders treatment. In this regard, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers are potential candidates already available since they block the classical axis, activate the counter-regulatory axis, and are safe and efficient drugs.
Collapse
Affiliation(s)
- Bernardo H. M. Correa
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Luca Becari
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Marco Antônio Peliky Fontes
- Department of Physiology & Biophysics - Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Ana Cristina Simões-e-Silva
- Department of Pediatrics, Faculty of Medicine, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucas M. Kangussu
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; ,Address correspondence to this author at the Department of Morphology, Biological Sciences Institute – Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; Tel: (+55-31) 3409-2772; E-mail:
| |
Collapse
|
24
|
Angiotensin antagonist inhibits preferential negative memory encoding via decreasing hippocampus activation and its coupling with amygdala. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:970-978. [DOI: 10.1016/j.bpsc.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022]
|
25
|
Anjos PAR, Marchette RCN, Kremer R, Granzotto N, Alves TM, Fadanni GP, Mazur FG, Anton EL, da Silva-Santos JE, Linder ÁE, Izídio GS. The influence of chromosome 4 on high ethanol consumption and blood pressure. Alcohol 2022; 102:1-10. [PMID: 35500756 DOI: 10.1016/j.alcohol.2022.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022]
Abstract
The Spontaneously Hypertensive Rats (SHR) strain was developed through selective breeding for high systolic blood pressure. In our laboratory, we established a congenic rat strain named SHR.Lewis-Anxrr16 (SLA16). The SLA16 rat strain is genetically identical to the SHR except for the inserted Anxrr16 region in chromosome 4. Our objective was to evaluate the influence of this genomic region on ethanol consumption and blood pressure. First, we exposed SHR and SLA16 male and female rats to ethanol consumption. Results showed that, regardless of strain, females consumed more ethanol than males during forced (10% v/v) and spontaneous ethanol consumption (SEC; 2.5-20% v/v). Then, females from both strains were used to evaluate sensitivity to ethanol. No strain differences in the loss of righting reflex were observed after ethanol treatment (3 g/kg, 20% w/v, intraperitoneal [i.p.]). But, in the triple test, female SHR rats presented lower sensitivity to the ethanol (1.2 g/kg, 14% w/v, i.p.). Surprisingly, female SHR rats also presented higher blood pressure after SEC (10% v/v). Finally, losartan treatment was effective in decreasing the blood pressure of female rats of both strains, but had specific effects on SHR ethanol consumption. Our data suggest that SLA16 female rats consume less ethanol (10%), are more sensitive to its effects, and present lower blood pressure than SHR female rats. We demonstrated that the Anxrr16 locus in chromosome 4 is a genetic candidate to explain high ethanol consumption and blood pressure, at least in females.
Collapse
Affiliation(s)
| | - Renata Cristina Nunes Marchette
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Neurobiology of Addiction Section, Integrative Neuroscience Branch, National Institute on Drug Abuse Intramural Program, Baltimore, MD, United States
| | - Rafael Kremer
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Medicine - Federal University of Fronteira Sul, Passo Fundo, Rio Grande do Sul, Brazil
| | - Natalli Granzotto
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Thalita Mello Alves
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Guilherme Pasetto Fadanni
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Fernando Gabriel Mazur
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Elaine Leocádia Anton
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Áurea Elizabeth Linder
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Geison Souza Izídio
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
26
|
Sfera A, Osorio C, Rahman L, Zapata-Martín del Campo CM, Maldonado JC, Jafri N, Cummings MA, Maurer S, Kozlakidis Z. PTSD as an Endothelial Disease: Insights From COVID-19. Front Cell Neurosci 2021; 15:770387. [PMID: 34776871 PMCID: PMC8586713 DOI: 10.3389/fncel.2021.770387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 virus, the etiologic agent of COVID-19, has affected almost every aspect of human life, precipitating stress-related pathology in vulnerable individuals. As the prevalence rate of posttraumatic stress disorder in pandemic survivors exceeds that of the general and special populations, the virus may predispose to this disorder by directly interfering with the stress-processing pathways. The SARS-CoV-2 interactome has identified several antigens that may disrupt the blood-brain-barrier by inducing premature senescence in many cell types, including the cerebral endothelial cells. This enables the stress molecules, including angiotensin II, endothelin-1 and plasminogen activator inhibitor 1, to aberrantly activate the amygdala, hippocampus, and medial prefrontal cortex, increasing the vulnerability to stress related disorders. This is supported by observing the beneficial effects of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in both posttraumatic stress disorder and SARS-CoV-2 critical illness. In this narrative review, we take a closer look at the virus-host dialog and its impact on the renin-angiotensin system, mitochondrial fitness, and brain-derived neurotrophic factor. We discuss the role of furin cleaving site, the fibrinolytic system, and Sigma-1 receptor in the pathogenesis of psychological trauma. In other words, learning from the virus, clarify the molecular underpinnings of stress related disorders, and design better therapies for these conditions. In this context, we emphasize new potential treatments, including furin and bromodomains inhibitors.
Collapse
Affiliation(s)
- Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Patton State Hospital, San Bernardino, CA, United States
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Leah Rahman
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Jose Campo Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Zisis Kozlakidis
- International Agency For Research On Cancer (IARC), Lyon, France
| |
Collapse
|
27
|
Stein MB, Jain S, Simon NM, West JC, Marvar PJ, Bui E, He F, Benedek DM, Cassano P, Griffith JL, Howlett J, Malgaroli M, Melaragno A, Seligowski AV, Shu IW, Song S, Szuhany K, Taylor CT, Ressler KJ. Randomized, Placebo-Controlled Trial of the Angiotensin Receptor Antagonist Losartan for Posttraumatic Stress Disorder. Biol Psychiatry 2021; 90:473-481. [PMID: 34275593 DOI: 10.1016/j.biopsych.2021.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/20/2021] [Accepted: 05/03/2021] [Indexed: 11/17/2022]
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.
Collapse
Affiliation(s)
- Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, California; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California.
| | - Sonia Jain
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California
| | - Naomi M Simon
- NYU Grossman School of Medicine and NYU Langone Health, New York, New York
| | - James C West
- Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Eric Bui
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
| | - Feng He
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California
| | - David M Benedek
- Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Paolo Cassano
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Jonathan Howlett
- Department of Psychiatry, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California
| | - Matteo Malgaroli
- NYU Grossman School of Medicine and NYU Langone Health, New York, New York
| | - Andrew Melaragno
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
| | - Antonia V Seligowski
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - I-Wei Shu
- Department of Psychiatry, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California
| | - Suzan Song
- George Washington University, Washington, DC
| | - Kristin Szuhany
- NYU Grossman School of Medicine and NYU Langone Health, New York, New York
| | - Charles T Taylor
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts.
| |
Collapse
|
28
|
Failure of Losartan in a Clinical Trial for Posttraumatic Stress Disorder: Lack of Efficacy or Spotlight on the Power of Placebo? Biol Psychiatry 2021; 90:432-433. [PMID: 34503672 DOI: 10.1016/j.biopsych.2021.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 11/22/2022]
|
29
|
Iftikhar A, Islam M, Shepherd S, Jones S, Ellis I. Is RAS the Link Between COVID-19 and Increased Stress in Head and Neck Cancer Patients? Front Cell Dev Biol 2021; 9:714999. [PMID: 34336866 PMCID: PMC8320172 DOI: 10.3389/fcell.2021.714999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/25/2021] [Indexed: 01/04/2023] Open
Abstract
The COVID-19 pandemic emerged as a largely unexplained outbreak of pneumonia cases, in Wuhan City, China and rapidly spread across the world. By 11th March 2020, WHO declared it as a global pandemic. The resulting restrictions, to contain its spread, demanded a momentous change in the lifestyle of the general population as well as cancer patients. This augmented negative effects on the mental health of patients with head and neck cancer (HNC), who already battle with the stress of cancer diagnosis and treatment. The causative agent of COVID-19, SARS-CoV2, gains entry through the Angiotensin converting enzyme 2 (ACE2) receptor, which is a component of the Renin Angiotensin System (RAS). RAS has been shown to influence cancer and stress such that it can have progressive and suppressive effects on both. This review provides an overview of SARS-CoV2, looks at how the RAS provides a mechanistic link between stress, cancer and COVID-19 and the probable activation of the RAS axis that increase stress (anxiogenic) and tumor progression (tumorigenic), when ACE2 is hijacked by SARS-CoV2. The mental health crises brought about by this pandemic have been highlighted in many studies. The emerging links between cancer and stress make it more important than ever before to assess the stress burden of cancer patients and expand the strategies for its management.
Collapse
Affiliation(s)
| | | | | | | | - Ian Ellis
- Unit of Cell and Molecular Biology, The Dental School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
30
|
Yoo S, Stremlau M, Pinto A, Woo H, Curtis O, van Praag H. Effects of Combined Anti-Hypertensive and Statin Treatment on Memory, Fear Extinction, Adult Neurogenesis, and Angiogenesis in Adult and Middle-Aged Mice. Cells 2021; 10:1778. [PMID: 34359946 PMCID: PMC8304131 DOI: 10.3390/cells10071778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023] Open
Abstract
Hyperlipidemia and hypertension are modifiable risk factors for cognitive decline. About 25% of adults over age 65 use both antihypertensives (AHTs) and statins to treat these conditions. Recent research in humans suggests that their combined use may delay or prevent dementia onset. However, it is not clear whether and how combination treatment may benefit brain function. To begin to address this question, we examined effects of atorvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, and Captopril, an angiotensin-converting enzyme inhibitor (ACEI), administration on memory function, anxiety-like behavior, adult hippocampal neurogenesis and angiogenesis in adult and middle-aged male C57Bl/6J mice. In adult mice (3-months-old) combination (combo) treatment, as well as administration of each compound individually, for six weeks, accelerated memory extinction in contextual fear conditioning. However, pattern separation in the touchscreen-based location discrimination test, a behavior linked to adult hippocampal neurogenesis, was unchanged. In addition, dentate gyrus (DG) neurogenesis and vascularization were unaffected. In middle-aged mice (10-months-old) combo treatment had no effect on spatial memory in the Morris water maze, but did reduce anxiety in the open field test. A potential underlying mechanism may be the modest increase in new hippocampal neurons (~20%) in the combo as compared to the control group. DG vascularization was not altered. Overall, our findings suggest that statin and anti-hypertensive treatment may serve as a potential pharmacotherapeutic approach for anxiety, in particular for post-traumatic stress disorder (PTSD) patients who have impairments in extinction of aversive memories.
Collapse
Affiliation(s)
- Seungwoo Yoo
- Stiles-Nicholson Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA; (S.Y.); (A.P.); (H.W.); (O.C.)
| | | | - Alejandro Pinto
- Stiles-Nicholson Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA; (S.Y.); (A.P.); (H.W.); (O.C.)
| | - Hyewon Woo
- Stiles-Nicholson Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA; (S.Y.); (A.P.); (H.W.); (O.C.)
| | - Olivia Curtis
- Stiles-Nicholson Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA; (S.Y.); (A.P.); (H.W.); (O.C.)
| | - Henriette van Praag
- Stiles-Nicholson Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA; (S.Y.); (A.P.); (H.W.); (O.C.)
- National Institute on Aging (NIA), Baltimore, MD 21224, USA;
| |
Collapse
|
31
|
Molecular signaling pathways underlying schizophrenia. Schizophr Res 2021; 232:33-41. [PMID: 34010744 DOI: 10.1016/j.schres.2021.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/16/2021] [Accepted: 05/02/2021] [Indexed: 12/19/2022]
Abstract
The molecular pathophysiological mechanisms underlying schizophrenia have remained unknown, and no treatment exists for primary prevention. We used Ingenuity Pathway Analysis to analyze canonical and causal pathways in two different datasets, including patients from Finland and USA. The most significant findings in canonical pathway analysis were observed for glutamate receptor signaling, hepatic fibrosis, and glycoprotein 6 (GP6) pathways in the Finnish dataset, and GP6 and hepatic fibrosis pathways in the US dataset. In data-driven causal pathways, ADCYAP1, ADAMTS, and CACNA genes were involved in the majority of the top 10 pathways differentiating patients and controls in both Finnish and US datasets. Results from a Finnish nation-wide database showed that the risk of schizophrenia relapse was 41% lower among first-episode patients during the use of losartan, the master regulator of an ADCYAP1, ADAMTS, and CACNA-related pathway, compared to those time periods when the same individual did not use the drug. The results from the two independent datasets suggest that the GP6 signaling pathway, and the ADCYAP1, ADAMTS, and CACNA-related purine, oxidative stress, and glutamatergic signaling pathways are among primary pathophysiological alterations in schizophrenia among patients with European ancestry. While no reproducible dopaminergic alterations were observed, the results imply that agents such as losartan, and ADCYAP1/PACAP -deficit alleviators, such as metabotropic glutamate 2/3 agonist MGS0028 and 5-HT7 antagonists - which have shown beneficial effects in an experimental Adcyap1-/- mouse model for schizophrenia - could be potential treatments even before the full manifestation of illness involving dopaminergic abnormalities.
Collapse
|
32
|
Mehranfard D, Linares A, Chabbra A, Campos G, de Souza AMA, Ji H, West C, Sandberg K, Speth RC. Preliminary study of ovariectomy and chronic losartan-induced alterations in brain AT 1 receptors. Brain Res 2021; 1766:147520. [PMID: 33991491 DOI: 10.1016/j.brainres.2021.147520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 11/19/2022]
Abstract
Women who undergo oophorectomy prior to the age of natural menopause have a higher risk of neurological and psychological impairment. Treatment with the angiotensin receptor blocker (ARB) losartan for 10 weeks following ovariectomy of Long-Evans rats at 3 months of age reduced the ovariectomy-induced cognitive decrements. Following completion of the behavioral experiments, (Campos et al., 2019), the brains were harvested for preliminary receptor autoradiographic studies of AT1 receptor (AT1R) binding in selected brain regions using quantitative densitometric analysis of autoradiograms of 125I-sarcosine1, isoleucine8 angiotensin II binding. Four of the brain regions (amygdala, ventral subiculum, piriform cortex, and cingulate cortex) are associated with cognitive and emotional behavior while one (lateral hypothalamus) is associated with homeostasis. The density of AT1R varied by region: ventral subiculum > amygdala and cingulate cortex, and piriform cortex > cingulate cortex. Losartan treatment decreased AT1R binding in the ventral subiculum of sham and ovariectomized rats by 41.6%, and 46% in the piriform cortex of the sham rats, but tended to increase AT1R binding in the piriform cortex and cingulate cortex 77% and 107%, respectively, in the ovariectomized rats. AT1R binding did not differ significantly between intact male and sham-vehicle female rats among surveyed brain regions. These results suggest that losartan-induced changes in brain AT1R expression may contribute to the reduced anxiety-like behavior and memory impairments seen in ovariectomized rats, but replication of these observations will be needed to determine the extent to which brain AT1R changes mediate the adverse behavioral effects of ovariectomy.
Collapse
Affiliation(s)
- Danial Mehranfard
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Andrea Linares
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Alesa Chabbra
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Glenda Campos
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Aline M A de Souza
- Department of Medicine, School of Medicine, Georgetown University, Washington, DC, United States
| | - Hong Ji
- Department of Medicine, School of Medicine, Georgetown University, Washington, DC, United States
| | - Crystal West
- Department of Biology, Appalachian State University, Kannapolis, NC, United States
| | - Kathryn Sandberg
- Department of Medicine, School of Medicine, Georgetown University, Washington, DC, United States
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States; Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, United States.
| |
Collapse
|
33
|
Saavedra JM. Angiotensin Receptor Blockers Are Not Just for Hypertension Anymore. Physiology (Bethesda) 2021; 36:160-173. [PMID: 33904788 DOI: 10.1152/physiol.00036.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Beyond blood pressure control, angiotensin receptor blockers reduce common injury mechanisms, decreasing excessive inflammation and protecting endothelial and mitochondrial function, insulin sensitivity, the coagulation cascade, immune responses, cerebrovascular flow, and cognition, properties useful to treat inflammatory, age-related, neurodegenerative, and metabolic disorders of many organs including brain and lung.
Collapse
Affiliation(s)
- Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
34
|
Attilio PJ, Snapper DM, Rusnak M, Isaac A, Soltis AR, Wilkerson MD, Dalgard CL, Symes AJ. Transcriptomic Analysis of Mouse Brain After Traumatic Brain Injury Reveals That the Angiotensin Receptor Blocker Candesartan Acts Through Novel Pathways. Front Neurosci 2021; 15:636259. [PMID: 33828448 PMCID: PMC8019829 DOI: 10.3389/fnins.2021.636259] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/19/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) results in complex pathological reactions, where the initial lesion is followed by secondary inflammation and edema. Our laboratory and others have reported that angiotensin receptor blockers (ARBs) have efficacy in improving recovery from traumatic brain injury in mice. Treatment of mice with a subhypotensive dose of the ARB candesartan results in improved functional recovery, and reduced pathology (lesion volume, inflammation and gliosis). In order to gain a better understanding of the molecular mechanisms through which candesartan improves recovery after controlled cortical impact injury (CCI), we performed transcriptomic profiling on brain regions after injury and drug treatment. We examined RNA expression in the ipsilateral hippocampus, thalamus and hypothalamus at 3 or 29 days post injury (dpi) treated with either candesartan (0.1 mg/kg) or vehicle. RNA was isolated and analyzed by bulk mRNA-seq. Gene expression in injured and/or candesartan treated brain region was compared to that in sham vehicle treated mice in the same brain region to identify genes that were differentially expressed (DEGs) between groups. The most DEGs were expressed in the hippocampus at 3 dpi, and the number of DEGs reduced with distance and time from the lesion. Among pathways that were differentially expressed at 3 dpi after CCI, candesartan treatment altered genes involved in angiogenesis, interferon signaling, extracellular matrix regulation including integrins and chromosome maintenance and DNA replication. At 29 dpi, candesartan treatment reduced the expression of genes involved in the inflammatory response. Some changes in gene expression were confirmed in a separate cohort of animals by qPCR. Fewer DEGs were found in the thalamus, and only one in the hypothalamus at 3 dpi. Additionally, in the hippocampi of sham injured mice, 3 days of candesartan treatment led to the differential expression of 384 genes showing that candesartan in the absence of injury had a powerful impact on gene expression specifically in the hippocampus. Our results suggest that candesartan has broad actions in the brain after injury and affects different processes at acute and chronic times after injury. These data should assist in elucidating the beneficial effect of candesartan on recovery from TBI.
Collapse
Affiliation(s)
- Peter J. Attilio
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Dustin M. Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Milan Rusnak
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Akira Isaac
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Anthony R. Soltis
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Matthew D. Wilkerson
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L. Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Aviva J. Symes
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
35
|
The renin-angiotensin system in PTSD: a replication and extension. Neuropsychopharmacology 2021; 46:750-755. [PMID: 33318633 PMCID: PMC8026983 DOI: 10.1038/s41386-020-00923-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/08/2022]
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.
Collapse
|
36
|
Wang Y, Takeshita H, Yamamoto K, Huang Y, Wang C, Nakajima T, Nozato Y, Fujimoto T, Yokoyama S, Hongyo K, Nakagami F, Akasaka H, Takami Y, Takeya Y, Sugimoto K, Rakugi H. A pressor dose of angiotensin II has no influence on the angiotensin-converting enzyme 2 and other molecules associated with SARS-CoV-2 infection in mice. FASEB J 2021; 35:e21419. [PMID: 33566370 PMCID: PMC7995007 DOI: 10.1096/fj.202100016r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
In the early phase of the Coronavirus disease 2019 (COVID‐19) pandemic, it was postulated that the renin‐angiotensin‐system inhibitors (RASi) increase the infection risk. This was primarily based on numerous reports, which stated that the RASi could increase the organ Angiotensin‐converting enzyme 2 (ACE2), the receptor of Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), in rodents. RASi can theoretically antagonize the potential influence of angiotensin II (Ang II) on ACE2. However, while Ang II decreases the ACE2 levels in cultured cells, there is little evidence that supports this phenomenon in living animals. In this study, we tested whether Ang II or Ang II combined with its antagonist would alter the ACE2 and other molecules associated with the infection of SARS‐CoV‐2. Male C57BL6/J mice were administered vehicle, Ang II (400 ng/kg/min), or Ang II with losartan (10 mg/kg/min) for 2 weeks. ACE2 knockout mice were used as a negative control for the ACE2 assay. We found that both Ang II, which elevated blood pressure by 30 mm Hg, and Ang II with losartan, had no effect on the expression or protein activity of ACE2 in the lung, left ventricle, kidney, and ileum. Likewise, these interventions had no effect on the expression of Transmembrane Protease Serine 2 (TMPRSS2) and Furin, proteases that facilitate the virus‐cell fusion, and the expression or activity of Tumor Necrosis Factor α‐Convertase (TACE) that cleaves cell‐surface ACE2. Collectively, physiological concentrations of Ang II do not modulate the molecules associated with SARS‐CoV‐2 infection. These results support the recent observational studies suggesting that the use of RASi is not a risk factor for COVID‐19.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hikari Takeshita
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yibin Huang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Cheng Wang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tsuneo Nakajima
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Taku Fujimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Serina Yokoyama
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuhiro Hongyo
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Futoshi Nakagami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Takeya
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ken Sugimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
37
|
Garfinkel SN. Disgust: Internal Context Matters. Curr Biol 2021; 31:R120-R122. [PMID: 33561407 DOI: 10.1016/j.cub.2020.11.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Novel research suggests the rhythms in our stomachs can influence our response to disgusting stimuli. Monetary reward will only encourage people to engage with disgusting stimuli after a peripherally acting drug alters gastric signals.
Collapse
Affiliation(s)
- Sarah N Garfinkel
- Institute of Cognitive Neuroscience, University College London, London WC1N 3AZ, UK.
| |
Collapse
|
38
|
Marvar PJ, Andero R, Hurlemann R, Lago TR, Zelikowsky M, Dabrowska J. Limbic Neuropeptidergic Modulators of Emotion and Their Therapeutic Potential for Anxiety and Post-Traumatic Stress Disorder. J Neurosci 2021; 41:901-910. [PMID: 33472824 PMCID: PMC7880296 DOI: 10.1523/jneurosci.1647-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Paul J Marvar
- Department of Pharmacology & Physiology, Department of Psychiatry and Behavioral Sciences, George Washington Institute for Neuroscience, George Washington University, Washington, DC, 20037
| | - Raül Andero
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain, 08193. Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain, 28029. ICREA, Pg. Lluís Companys 23, Barcelona, Spain, 08010
| | - Rene Hurlemann
- Department of Psychiatry, School of Medicine & Health Sciences, and Research Center Neurosensory Science, University of Oldenburg, Oldenburg, 26129, Germany
| | - Tiffany R Lago
- Department of Psychiatry, Veterans Administration Boston Healthcare System, Boston, Massachusetts, 02130
| | - Moriel Zelikowsky
- Department of Neurobiology and Anatomy, University of Utah, School of Medicine, Salt Lake City, Utah, 84112
| | - Joanna Dabrowska
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, 60064
| |
Collapse
|
39
|
Stein MB, Levey DF, Cheng Z, Wendt FR, Harrington K, Pathak GA, Cho K, Quaden R, Radhakrishnan K, Girgenti MJ, Ho YLA, Posner D, Aslan M, Duman RS, Zhao H, Polimanti R, Concato J, Gelernter J. Genome-wide association analyses of post-traumatic stress disorder and its symptom subdomains in the Million Veteran Program. Nat Genet 2021; 53:174-184. [PMID: 33510476 PMCID: PMC7972521 DOI: 10.1038/s41588-020-00767-x] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 12/15/2020] [Indexed: 01/30/2023]
Abstract
We conducted genome-wide association analyses of over 250,000 participants of European (EUR) and African (AFR) ancestry from the Million Veteran Program using electronic health record-validated post-traumatic stress disorder (PTSD) diagnosis and quantitative symptom phenotypes. Applying genome-wide multiple testing correction, we identified three significant loci in European case-control analyses and 15 loci in quantitative symptom analyses. Genomic structural equation modeling indicated tight coherence of a PTSD symptom factor that shares genetic variance with a distinct internalizing (mood-anxiety-neuroticism) factor. Partitioned heritability indicated enrichment in several cortical and subcortical regions, and imputed genetically regulated gene expression in these regions was used to identify potential drug repositioning candidates. These results validate the biological coherence of the PTSD syndrome, inform its relationship to comorbid anxiety and depressive disorders and provide new considerations for treatment.
Collapse
Affiliation(s)
- Murray B Stein
- VA San Diego Healthcare System, Psychiatry Service, San Diego, CA, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA.
| | - Daniel F Levey
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zhongshan Cheng
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Frank R Wendt
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Harrington
- VA Boston Healthcare System, Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Gita A Pathak
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Cho
- VA Boston Healthcare System, Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Rachel Quaden
- VA Boston Healthcare System, Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
| | - Krishnan Radhakrishnan
- Clinical Epidemiology Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
- College of Medicine, University of Kentucky, Lexington, KY, USA
- Office of the Director, Center for Behavioral Health Statistics and Quality, Substance Abuse and Mental Health Services Administration, Rockville, MD, USA
| | - Matthew J Girgenti
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yuk-Lam Anne Ho
- VA Boston Healthcare System, Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
| | - Daniel Posner
- VA Boston Healthcare System, Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
| | - Mihaela Aslan
- Clinical Epidemiology Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ronald S Duman
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hongyu Zhao
- Clinical Epidemiology Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Renato Polimanti
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - John Concato
- Clinical Epidemiology Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Joel Gelernter
- VA Connecticut Healthcare System, Psychiatry Service, West Haven, CT, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
40
|
SKF83959, an agonist of phosphatidylinositol-linked dopamine receptors, prevents renewal of extinguished conditioned fear and facilitates extinction. Brain Res 2020; 1749:147136. [PMID: 32980332 DOI: 10.1016/j.brainres.2020.147136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Fear-related anxiety disorders, such as social phobia and post-traumatic stress disorder, are partly explained by an uncontrollable state of fear. An emerging literature suggests dopamine receptor-1 (D1 receptor) in the amygdala is involved in the regulation of fear memory. An early study has reported that amygdaloid D1 receptor (D1R) is not coupled to the classic cAMP-dependent signal transduction. Here, we investigated whether SKF83959, a typical D1R agonist that mainly activates a D1-like receptor-dependent phosphatidylinositol (PI) signal pathway, facilitates fear extinction and reduces the return of extinguished fear. Interestingly, long-term loss of fearful memories can be induced through a combination of SKF83959 (1 mg/kg/day, i.p., once daily for one week) pharmacotherapy and extinction training. Furthermore, sub-chronic administration of SKF83959 after fear conditioning reduced fear renewal and reinstatement in the mice. We found that the activation D1R and PI signaling in the amygdala was responsible for the effect of SKF83959 on fear extinction. Additionally, SKF83959 significantly promoted the elevation of brain-derived neurotrophic factor (BDNF) expression, possibly by the cAMP response element binding protein (CREB) -directed gene transcription. Given the beneficial effects on extinction, SKF83959 may emerge as a candidate pharmacological approach for improving cognitive-behavioral therapy on fear-related anxiety disorders.
Collapse
|
41
|
Mohite S, Sanches M, Teixeira AL. Exploring the Evidence Implicating the Renin-Angiotensin System (RAS) in the Physiopathology of Mood Disorders. Protein Pept Lett 2020; 27:449-455. [PMID: 31868144 DOI: 10.2174/0929866527666191223144000] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023]
Abstract
Mood disorders include Major Depressive Disorder (MDD), Bipolar Disorder (BD) and variations of both. Mood disorders has a public health significance with high comorbidity, suicidal mortality and economic burden on the health system. Research related to mood disorders has evolved over the years to relate it with systemic conditions. The Renin Angiotensin System (RAS) has been noticed to play major physiological roles beyond renal and cardiovascular systems. Recent studies have linked RAS not only with neuro-immunological processes, but also with psychiatric conditions like mood and anxiety disorders. In this comprehensive review, we integrated basic and clinical studies showing the associations between RAS and mood disorders. Animal studies on mood disorders models - either depression or mania - were focused on the reversal of behavioral and/or cognitive symptoms through the inhibition of RAS components like the Angiotensin- Converting Enzyme (ACE), Angiotensin II Type 1 receptor (AT1) or Mas receptors. ACE polymorphisms, namely insertion-deletion (I/D), were linked to mood disorders and suicidal behavior. Hypertension was associated with neurocognitive deficits in mood disorders, which reversed with RAS inhibition. Low levels of RAS components (renin activity or aldosterone) and mood symptoms improvement with ACE inhibitors or AT1 blockers were also observed in mood disorders. Overall, this review reiterates the strong and under-researched connection between RAS and mood disorders.
Collapse
Affiliation(s)
- Satyajit Mohite
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Marsal Sanches
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Antonio L Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77054, United States
| |
Collapse
|
42
|
de Melo LA, Almeida-Santos AF. Neuropsychiatric Properties of the ACE2/Ang-(1-7)/Mas Pathway: A Brief Review. Protein Pept Lett 2020; 27:476-483. [PMID: 31868143 DOI: 10.2174/0929866527666191223143230] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/02/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
The current pharmacological strategies for the management of anxiety disorders and depression, serious conditions which are gaining greater prevalence worldwide, depend on only two therapeutic classes of mood-stabilizing drugs: Serotonin Reuptake Inhibitors (SSRIs) and Serotonin-Norepinephrine Reuptake Inhibitors (SNRIs). Although first line agents with proven efficacy, their clinical success in the management of anxiety disorders and depression is still considered highly complex due to the multifaceted nature of such conditions. Several studies have shown a possible therapeutic target could be found in the form of the Angiotensin-Converting Enzyme [ACE] type 2 (ACE2), Angiotensin [Ang]-(1-7) and Mas receptor pathway of the Renin- Angiotensin System (RAS), which as will be discussed, has been described to exhibit promising therapeutic properties for the management of anxiety disorders and depression. In this article, the literature to describe recent findings related to the role of the RAS in anxiety and depression disorders was briefly revised. The literature used covers a time range from 1988 to 2019 and were acquired from the National Center for Biotechnology Information's (NCBI) PubMed search engine. The results demonstrated in this review are promising and encourage the development of new research for the treatment of anxiety and depression disorders focusing on the RAS. In conclusion, the ACE2/Ang-(1-7)/Mas pathway may exhibit anxiolytic and anti-depressive effects through many possible biochemical mechanisms both centrally and peripherally, and result in highly promising mental health benefits which justifies further investigation into this system as a possible new therapeutic target in the management of neuropsychiatric disorders, including any as of yet undescribed risk-benefit analysis compared to currently-implemented pharmacological strategies.
Collapse
Affiliation(s)
- Leonardo Augusto de Melo
- Nucleo de Neurociencias, Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Flávia Almeida-Santos
- Nucleo de Neurociencias, Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
43
|
Swiercz AP, Iyer L, Yu Z, Edwards A, Prashant NM, Nguyen BN, Horvath A, Marvar PJ. Evaluation of an angiotensin Type 1 receptor blocker on the reconsolidation of fear memory. Transl Psychiatry 2020; 10:363. [PMID: 33110066 PMCID: PMC7591922 DOI: 10.1038/s41398-020-01043-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 09/11/2020] [Accepted: 10/07/2020] [Indexed: 12/30/2022] Open
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.
Collapse
Affiliation(s)
- Adam P. Swiercz
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - Laxmi Iyer
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - Zhe Yu
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - Allison Edwards
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - N. M. Prashant
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - Bryan N. Nguyen
- grid.253615.60000 0004 1936 9510Computational Biology Institute, George Washington University, Washington, DC 20052 USA
| | - Anelia Horvath
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA
| | - Paul J. Marvar
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, George Washington University, Washington, DC 20052 USA ,grid.253615.60000 0004 1936 9510Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC 20052 USA
| |
Collapse
|
44
|
de Kloet AD, Cahill KM, Scott KA, Krause EG. Overexpression of angiotensin converting enzyme 2 reduces anxiety-like behavior in female mice. Physiol Behav 2020; 224:113002. [PMID: 32525008 PMCID: PMC7503770 DOI: 10.1016/j.physbeh.2020.113002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 01/19/2023]
Abstract
Accumulating evidence has revealed an intricate role for the renin-angiotensin system (RAS) in the progression or alleviation of stress-related disorders. Along these lines, the 'pro-stress' actions of angiotensin-II (Ang-II) are largely thought to be mediated by the angiotensin type-1a receptor (AT1aR). On the other hand, a counter regulatory limb of the RAS that depends on the conversion of Ang-II to angiotensin-(1-7) by angiotensin-converting enzyme 2 (ACE2) has been postulated to exert stress-dampening actions. We have previously found that augmenting ACE2 activity is potently anxiolytic and blunts stress-induced activation of the hypothalamic-pituitary-adrenal (HPA) axis in male mice. Whether increasing ACE2 activity also relieves stress and anxiety in females has not yet been determined. Consequently, this series of experiments tests the hypothesis that augmenting ACE2 expression is anxiolytic and dampens the activity of the HPA axis in female mice. Using the Cre-LoxP system, we generated female mice that were homo-, heterozygous or wild-type for a mutated allele resulting in ubiquitous overexpression of ACE2. Next, we used qPCR to determine that levels of ACE2 mRNA isolated from central and peripheral tissues was dependent on genotype. That is, mice homo- and heterozygous for the ACE2 overexpression had significantly greater levels of ACE2 mRNA relative to littermate matched wild-type controls. Interestingly, anxiety-like behavior as determined by the elevated plus maze, light-dark box and novelty-induced hypophagia tests was also affected by genotype. Specifically, ACE2 overexpression significantly decreased anxiety-like behavior in paradigms dependent on approach-avoidance conflict and novelty; however, locomotor activity was similar amongst the genotypes. Final experiments measured plasma corticosterone to evaluate whether increasing ACE2 alters basal and/or stress-induced HPA axis activity. In contrast to what was previously found in males, increasing ACE2 expression had no effect on plasma corticosterone under basal conditions or subsequent to an acute restraint challenge. Collectively, these results suggest that increasing ACE2 expression potently elicits anxiolysis in female mice without altering HPA axis activity.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| | - Karlena M Cahill
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Karen A Scott
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
45
|
Shkreli L, Woud ML, Ramsbottom R, Rupietta AE, Waldhauser GT, Kumsta R, Reinecke A. Angiotensin involvement in trauma processing-exploring candidate neurocognitive mechanisms of preventing post-traumatic stress symptoms. Neuropsychopharmacology 2020; 45:507-514. [PMID: 31655485 PMCID: PMC6969172 DOI: 10.1038/s41386-019-0553-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 11/09/2022]
Abstract
The angiotensin-II antagonist losartan is a promising candidate that has enhanced extinction in a post-traumatic stress disorder (PTSD) animal model and was related to reducing PTSD symptom development in humans. Here, we investigate the neurocognitive mechanisms underlying these results, testing the effect of losartan on data-driven and contextual processing of traumatic material, mechanisms proposed to be relevant for PTSD development. In a double-blind between-subject design, 40 healthy participants were randomised to a single oral dose of losartan (50 mg) or placebo, 1 h before being exposed to distressing films as a trauma analogue while heart rate (HR) was measured. Peritraumatic processing was investigated using blurry picture stimuli from the films, which transformed into clear images. Data-driven processing was measured by the level of blurriness at which contents were recognised. Contextual processing was measured as the amount of context information retrieved when describing the pictures' contents. Negative-matched control images were used to test perceptual processing of peripheral trauma-cues. Post-traumatic stress symptoms were assessed via self-report questionnaires after analogue trauma and an intrusion diary completed over 4 days following the experiment. Compared to placebo, losartan facilitated contextual processing and enhanced detail perception in the negative-match pictures. During the films, the losartan group recorded lower HR and higher HR variability, reflecting lower autonomic stress responses. We discuss potential mechanisms of losartan in preventing PTSD symptomatology, including the role of reduced arousal and increased contextual processing during trauma exposure, as well as increased threat-safety differentiation when encountering peripheral trauma-cues in the aftermaths of traumatic events.
Collapse
Affiliation(s)
- Lorika Shkreli
- 0000000121885934grid.5335.0Department of Psychiatry, University of Cambridge, Cambridge, UK ,0000 0004 0490 981Xgrid.5570.7Department of Clinical Psychology and Psychotherapy, Ruhr-Universität Bochum, Bochum, Germany ,0000 0004 1936 8948grid.4991.5Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX37JX UK
| | - Marcella Lydia Woud
- 0000 0004 0490 981Xgrid.5570.7Department of Clinical Psychology and Psychotherapy, Ruhr-Universität Bochum, Bochum, Germany
| | - Roger Ramsbottom
- 0000 0001 0726 8331grid.7628.bFaculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Aleksandra Ewa Rupietta
- 0000 0004 0490 981Xgrid.5570.7Department of Clinical Psychology and Psychotherapy, Ruhr-Universität Bochum, Bochum, Germany ,0000 0004 0490 981Xgrid.5570.7Department of Neuropsychology, Ruhr-Universität Bochum, Bochum, Germany
| | - Gerd Thomas Waldhauser
- 0000 0004 0490 981Xgrid.5570.7Department of Neuropsychology, Ruhr-Universität Bochum, Bochum, Germany
| | - Robert Kumsta
- 0000 0004 0490 981Xgrid.5570.7Genetic Psychology, Ruhr-Universität Bochum, Bochum, Germany
| | - Andrea Reinecke
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX37JX, UK.
| |
Collapse
|
46
|
Hammoud MZ, Foa EB, Milad MR. Oestradiol, threat conditioning and extinction, post-traumatic stress disorder, and prolonged exposure therapy: A common link. J Neuroendocrinol 2020; 32:e12800. [PMID: 31595559 DOI: 10.1111/jne.12800] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/12/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022]
Abstract
The accumulating evidence regarding the impact of estradiol on learning and memory synergized studies to examine its influence on enhancing animal's ability to quell fear and anxiety. In this review, we first provide a foundational platform regarding the impact of oestradiol on cellular mechanisms of learning and memory and we review recent advances from rodent and human data showing that oestrogen enhances extinction learning across species. We then propose clinical application to these data. We discuss the potential role of oestradiol variance on the aetiology, maintenance and treatment for post-traumatic stress disorder. Specifically, we argue that the use of oestradiol as an adjunct to prolonged exposure (PE) therapy for PTSD may provide a new treatment approach for enhancing the efficacy of PE in women with PTSD. This could advance our understanding of the mechanisms of PTSD and help tailor sex-specific treatments for this disorder.
Collapse
Affiliation(s)
- Mira Z Hammoud
- Department of Psychiatry, New York University Medical Center, New York, NY, USA
| | - Edna B Foa
- Department of Psychiatry, Center for the Treatment and Study of Anxiety, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohammed R Milad
- Department of Psychiatry, New York University Medical Center, New York, NY, USA
| |
Collapse
|
47
|
Salmani H, Hosseini M, Baghcheghi Y, Moradi-Marjaneh R, Mokhtari-Zaer A. Losartan modulates brain inflammation and improves mood disorders and memory impairment induced by innate immune activation: The role of PPAR-γ activation. Cytokine 2020; 125:154860. [PMID: 31574424 DOI: 10.1016/j.cyto.2019.154860] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 12/19/2022]
Abstract
In recent years, the role of angiotensin II (Ang II) and Ang II type 1 receptor (AT1) in the crosstalk between the immune system and the central nervous system has received more attention. The present study aimed to investigate the role of losartan, an AT1 receptor blocker, in the modulation of long-lasting adverse effects of repeated systemic lipopolysaccharide (LPS) injection in the brain function. For this purpose, 110 male BALB/c mice were administrated LPS (250 µg/kg) intraperitoneally (i.p.) for seven consecutive days. Mice were i.p. injected with losartan (1 and 3 mg/kg) three days before and during the LPS injection. To determine the role of PPAR-γ activation in the protective actions of losartan, GW9662, a PPAR-γ antagonist, was also co-administrated with losartan. Then, behavioral tests, including Morris water maze (MWM), novel object recognition test, passive avoidance, forced swim test (FST), elevated plus maze, and marble burying task, were conducted. The results demonstrated that losartan improved learning and memory impairment, attenuated anxiety-like behaviors, modulated brain inflammation and oxidative stress, and decreased amyloid-β accumulation. Losartan was unable to improve hippocampal BDNF and IL-10 levels as well as the retention trial in the MWM task and depressive-like behaviors. In addition, the PPAR-γ antagonist did not significantly influence the beneficial effects of losartan. Our findings suggest that AT1R blockade can protect the brain against most long-lasting hallmark effects of systemic inflammation. Also, based on the results, the beneficial actions of losartan were not mediated through PPAR-γ activation.
Collapse
Affiliation(s)
- Hossein Salmani
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Yousef Baghcheghi
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Moradi-Marjaneh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Mokhtari-Zaer
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Angiotensin II Type 2 Receptor-Expressing Neurons in the Central Amygdala Influence Fear-Related Behavior. Biol Psychiatry 2019; 86:899-909. [PMID: 31420088 DOI: 10.1016/j.biopsych.2019.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/29/2022]
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.
Collapse
|
49
|
Stout DM, Risbrough VB. Angiotensin II Signaling and Fear Extinction: Translational Evidence and Novel Receptor Targets. Biol Psychiatry 2019; 86:874-876. [PMID: 31753101 DOI: 10.1016/j.biopsych.2019.09.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Daniel M Stout
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, California; Department of Psychiatry, University of California San Diego, San Diego, California
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, California; Department of Psychiatry, University of California San Diego, San Diego, California.
| |
Collapse
|
50
|
Zhou F, Geng Y, Xin F, Li J, Feng P, Liu C, Zhao W, Feng T, Guastella AJ, Ebstein RP, Kendrick KM, Becker B. Human Extinction Learning Is Accelerated by an Angiotensin Antagonist via Ventromedial Prefrontal Cortex and Its Connections With Basolateral Amygdala. Biol Psychiatry 2019; 86:910-920. [PMID: 31471037 DOI: 10.1016/j.biopsych.2019.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/27/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Deficient extinction learning and threat adaptation in the ventromedial prefrontal cortex (vmPFC)-amygdala circuitry strongly impede the efficacy of exposure-based interventions in anxiety disorders. Recent animal models suggest a regulatory role of the renin-angiotensin system in both these processes. Against this background, the present randomized placebo-controlled pharmacologic functional magnetic resonance imaging experiment aimed at determining the extinction enhancing potential of the angiotensin II type 1 receptor antagonist losartan (LT) in humans. METHODS Seventy healthy male subjects underwent Pavlovian threat conditioning and received single-dose LT (50 mg) or placebo administration before extinction. Psychophysiological threat reactivity (skin conductance response) and neural activity during extinction served as primary outcomes. Psychophysiological interaction, voxelwise mediation, and novel multivariate pattern classification analyses were used to determine the underlying neural mechanisms. RESULTS LT significantly accelerated the decline of the psychophysiological threat response during within-session extinction learning. On the neural level, the acceleration was accompanied and critically mediated by threat-specific enhancement of vmPFC activation. Furthermore, LT enhanced vmPFC-basolateral amygdala coupling and attenuated the neural threat expression, particularly in the vmPFC, during early extinction. CONCLUSIONS Overall the results indicate that LT facilitates within-session threat memory extinction by augmenting threat-specific encoding in the vmPFC and its regulatory control over the amygdala. The findings document a pivotal role of angiotensin regulation of extinction learning in humans and suggest that adjunct LT administration has the potential to facilitate the efficacy of exposure-based interventions in anxiety disorders.
Collapse
Affiliation(s)
- Feng Zhou
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Yayuan Geng
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Xin
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Jialin Li
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Pan Feng
- Faculty of Psychology, Southwest University, Chongqing, China; Key Laboratory of Cognition and Personality, Ministry of Education, Southwest University, Chongqing, China
| | - Congcong Liu
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Weihua Zhao
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingyong Feng
- Faculty of Psychology, Southwest University, Chongqing, China; Key Laboratory of Cognition and Personality, Ministry of Education, Southwest University, Chongqing, China
| | - Adam J Guastella
- Autism Clinic for Translational Research, Brain and Mind Centre, Central Clinical School, Faculty of Medicine, University of Sydney, Camperdown, Australia; Youth Mental Health Unit, Brain and Mind Centre, Central Clinical School, Faculty of Medicine, University of Sydney, Camperdown, Australia
| | - Richard P Ebstein
- China Center for Behavior Economics and Finance, Southwestern University of Finance and Economics, Chengdu, China
| | - Keith M Kendrick
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Benjamin Becker
- Clinical Hospital of Chengdu Brain Science Institute and Ministry of Education (MOE) Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|