Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.955
Filter
1.
J Clin Psychiatry ; 85(2)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836865

ABSTRACT

Objectives: Women veterans are more likely than men veterans to receive medications that Department of Veterans Affairs clinical practice guidelines recommend against to treat posttraumatic stress disorder (PTSD). To understand this difference, we examined potential confounders in incident prescribing of guideline discordant medications (GDMs) in veterans with PTSD.Methods: Veterans receiving care for PTSD during 2020 were identified using Veterans Health Administration administrative data. PTSD diagnosis was established by the presence of at least 1 ICD-10 coded outpatient encounter or inpatient hospitalization during the calendar year 2020. Incident GDM prescribing was assessed during 2021, including benzodiazepines, antipsychotics, select anticonvulsants, and select antidepressants. Log-binomial regression was used to estimate the difference in risk for GDM initiation between men and women, adjusted for patient, prescriber, and facility-level covariates, and to identify key confounding variables.Results: Of 704,699 veterans with PTSD, 16.9% of women and 10.1% of men initiated a GDM, an increased risk of 67% for women [relative risk (RR) = 1.67; 95% CI, 1.65-1.70]. After adjustment, the gender difference decreased to 1.22 (95% CI, 1.20-1.24) in a fully specified model. Three key confounding variables were identified: bipolar disorder (RR = 1.60; 95% CI, 1.57-1.63), age (<40 years: RR = 1.20 [1.18-1.22]; 40-54 years: RR = 1.13 [1.11-1.16]; ≥65 years: RR = 0.64 [0.62-0.65]), and count of distinct psychiatric medications prescribed in the prior year (RR = 1.14; 1.13-1.14).Conclusions: Women veterans with PTSD were 67% more likely to initiate a GDM, where more than half of this effect was explained by bipolar disorder, age, and prior psychiatric medication. After adjustment, women veterans remained at 22% greater risk for an incident GDM, suggesting that other factors remain unidentified and warrant further investigation.


Subject(s)
Stress Disorders, Post-Traumatic , United States Department of Veterans Affairs , Veterans , Humans , Female , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/drug therapy , Male , Veterans/statistics & numerical data , Veterans/psychology , Middle Aged , United States/epidemiology , Adult , Sex Factors , United States Department of Veterans Affairs/statistics & numerical data , Practice Guidelines as Topic , Aged , Practice Patterns, Physicians'/statistics & numerical data , Guideline Adherence/statistics & numerical data , Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use
2.
Expert Rev Neurother ; 24(6): 575-584, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38771657

ABSTRACT

INTRODUCTION: Post-traumatic stress disorder (PTSD) is a disabling psychiatric condition with a worldwide prevalence between 6% and 9%, and more common in the female than in the male sex. The aim of this paper is to review and comment on the different factors that might explain the discrepancies in the pharmacological management of women and men. AREAS COVERED: The available literature shows that there exists a vulnerability of women to develop PTSD that may depend on neurobiological as well as environmental/cultural factors. These variables might influence the clinical picture, the outcome and the response to specific treatments, given their consequences on the pharmacokinetics of commonly prescribed drugs. Women suffering from PTSD are more prone to consult and receive more prescriptions of psychotropic drugs than men. However, it is evident that the particular stages of a women's life such as pregnancy or breastfeeding might require a specific evaluation and care. EXPERT OPINION: It is necessary to explore the pharmacokinetics of compounds highlighting sex-related differences, and their safety during pregnancy and lactation. Taking care of differences between women and men should represent a main focus of research, while being a primary target towards a really tailored pharmacological treatment of PTSD.


Subject(s)
Psychotropic Drugs , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/drug therapy , Female , Male , Psychotropic Drugs/therapeutic use , Pregnancy , Sex Characteristics , Sex Factors
3.
Cochrane Database Syst Rev ; 5: CD013613, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38767196

ABSTRACT

BACKGROUND: Acute traumatic stress symptoms may develop in people who have been exposed to a traumatic event. Although they are usually self-limiting in time, some people develop post-traumatic stress disorder (PTSD), a severe and debilitating condition. Pharmacological interventions have been proposed for acute symptoms to act as an indicated prevention measure for PTSD development. As many individuals will spontaneously remit, these interventions should balance efficacy and tolerability. OBJECTIVES: To assess the efficacy and acceptability of early pharmacological interventions for prevention of PTSD in adults experiencing acute traumatic stress symptoms. SEARCH METHODS: We searched the Cochrane Common Mental Disorders Controlled Trial Register (CCMDCTR), CENTRAL, MEDLINE, Embase and two other databases. We checked the reference lists of all included studies and relevant systematic reviews. The search was last updated on 23 January 2023. SELECTION CRITERIA: We included randomised controlled trials on adults exposed to any kind of traumatic event and presenting acute traumatic stress symptoms, without restriction on their severity. We considered comparisons of any medication with placebo, or with another medication. We excluded trials that investigated medications as an augmentation to psychotherapy. DATA COLLECTION AND ANALYSIS: We used standard Cochrane methodological procedures. Using a random-effects model, we analysed dichotomous data as risk ratios (RR) and calculated the number needed to treat for an additional beneficial/harmful outcome (NNTB/NNTH). We analysed continuous data as mean differences (MD) or standardised mean differences (SMD). Our primary outcomes were PTSD severity and dropouts due to adverse events. Secondary outcomes included PTSD rate, functional disability and quality of life. MAIN RESULTS: We included eight studies that considered four interventions (escitalopram, hydrocortisone, intranasal oxytocin, temazepam) and involved a total of 779 participants. The largest trial contributed 353 participants and the next largest, 120 and 118 participants respectively. The trials enrolled participants admitted to trauma centres or emergency departments. The risk of bias in the included studies was generally low except for attrition rate, which we rated as high-risk. We could meta-analyse data for two comparisons: escitalopram versus placebo (but limited to secondary outcomes) and hydrocortisone versus placebo. One study compared escitalopram to placebo at our primary time point of three months after the traumatic event. There was inconclusive evidence of any difference in terms of PTSD severity (mean difference (MD) on the Clinician-Administered PTSD Scale (CAPS, score range 0 to 136) -11.35, 95% confidence interval (CI) -24.56 to 1.86; 1 study, 23 participants; very low-certainty evidence), dropouts due to adverse events (no participant left the study early due to adverse events; 1 study, 31 participants; very low-certainty evidence) and PTSD rates (RR 0.59, 95% CI 0.03 to 13.08; NNTB 37, 95% CI NNTB 15 to NNTH 1; 1 study, 23 participants; very low-certainty evidence). The study did not assess functional disability or quality of life. Three studies compared hydrocortisone to placebo at our primary time point of three months after the traumatic event. We found inconclusive evidence on whether hydrocortisone was more effective in reducing the severity of PTSD symptoms compared to placebo (MD on CAPS -7.53, 95% CI -25.20 to 10.13; I2 = 85%; 3 studies, 136 participants; very low-certainty evidence) and whether it reduced the risk of developing PTSD (RR 0.47, 95% CI 0.09 to 2.38; NNTB 14, 95% CI NNTB 8 to NNTH 5; I2 = 36%; 3 studies, 136 participants; very low-certainty evidence). Evidence on the risk of dropping out due to adverse events is inconclusive (RR 3.19, 95% CI 0.13 to 75.43; 2 studies, 182 participants; low-certainty evidence) and it is unclear whether hydrocortisone might improve quality of life (MD on the SF-36 (score range 0 to 136, higher is better) 19.70, 95% CI -1.10 to 40.50; 1 study, 43 participants; very low-certainty evidence). No study assessed functional disability. AUTHORS' CONCLUSIONS: This review provides uncertain evidence regarding the use of escitalopram, hydrocortisone, intranasal oxytocin and temazepam for people with acute stress symptoms. It is therefore unclear whether these pharmacological interventions exert a positive or negative effect in this population. It is important to note that acute traumatic stress symptoms are often limited in time, and that the lack of data prevents the careful assessment of expected benefits against side effects that is therefore required. To yield stronger conclusions regarding both positive and negative outcomes, larger sample sizes are required. A common operational framework of criteria for inclusion and baseline assessment might help in better understanding who, if anyone, benefits from an intervention. As symptom severity alone does not provide the full picture of the impact of exposure to trauma, assessment of quality of life and functional impairment would provide a more comprehensive picture of the effects of the interventions. The assessment and reporting of side effects may facilitate a more comprehensive understanding of tolerability.


Subject(s)
Bias , Randomized Controlled Trials as Topic , Stress Disorders, Post-Traumatic , Stress Disorders, Traumatic, Acute , Humans , Stress Disorders, Post-Traumatic/prevention & control , Stress Disorders, Post-Traumatic/drug therapy , Adult , Stress Disorders, Traumatic, Acute/prevention & control , Quality of Life , Citalopram/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Placebos/therapeutic use
4.
Subst Abuse Treat Prev Policy ; 19(1): 25, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702783

ABSTRACT

BACKGROUND: There is little study of lifetime trauma exposure among individuals engaged in medication treatment for opioid use disorder (MOUD). A multisite study provided the opportunity to examine the prevalence of lifetime trauma and differences by gender, PTSD status, and chronic pain. METHODS: A cross-sectional study examined baseline data from participants (N = 303) enrolled in a randomized controlled trial of a mind-body intervention as an adjunct to MOUD. All participants were stabilized on MOUD. Measures included the Trauma Life Events Questionnaire (TLEQ), the Brief Pain Inventory (BPI), and the Posttraumatic Stress Disorder Checklist (PCL-5). Analyses involved descriptive statistics, independent sample t-tests, and linear and logistic regression. RESULTS: Participants were self-identified as women (n = 157), men (n = 144), and non-binary (n = 2). Fifty-seven percent (n = 172) self-reported chronic pain, and 41% (n = 124) scored above the screening cut-off for PTSD. Women reported significantly more intimate partner violence (85%) vs 73%) and adult sexual assault (57% vs 13%), while men reported more physical assault (81% vs 61%) and witnessing trauma (66% vs 48%). Men and women experienced substantial childhood physical abuse, witnessed intimate partner violence as children, and reported an equivalent exposure to accidents as adults. The number of traumatic events predicted PTSD symptom severity and PTSD diagnostic status. Participants with chronic pain, compared to those without chronic pain, had significantly more traumatic events in childhood (85% vs 75%). CONCLUSION: The study found a high prevalence of lifetime trauma among people in MOUD. Results highlight the need for comprehensive assessment and mental health services to address trauma among those in MOUD treatment. TRIAL REGISTRATION: NCT04082637.


Subject(s)
Chronic Pain , Opioid-Related Disorders , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/drug therapy , Chronic Pain/drug therapy , Chronic Pain/epidemiology , Female , Male , Cross-Sectional Studies , Adult , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/epidemiology , Sex Factors , Middle Aged , Psychological Trauma/epidemiology
5.
Chem Biol Interact ; 396: 111035, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38703807

ABSTRACT

Early life stress (ELS) can cause long-term changes by epigenetic factors, especially histone acetylation modification, playing a crucial role, affect normal cognition, mood, and behavior, and increase susceptibility to post-traumatic stress disorder (PTSD) in adulthood. It has been found that paeoniflorin (PF) can cross the blood-brain barrier to exert anti-PTSD effects on adult PTSD rats. However, whether PF can alleviate the harmful effects caused by ELS in adulthood has not yet been reported. Therefore, to explore the relationship between ELS and PTSD susceptibility in adulthood and its mechanism, in this study, SPS was used as a stressor of ELS, and the mathematical tool Z-normalization was employed as an evaluation criterion of behavioral resilience susceptibility. To investigate the regulatory mechanism of PF on histone acetylation in the hippocampus and amygdala of ELS rats in adulthood, using changes in HATs/HDACs as the entry point, meanwhile, the epigenetic marks (H3K9 and H4K12) in the key brain regions of ELS (hippocampus and amygdala) were evaluated, and the effects of PF on behavioral representation and PTSD susceptibility were observed. This study found that ELS lead to a series of PTSD-like behaviors in adulthood and caused imbalance of HATs/HDACs ratio in the hippocampus and amygdala, which confirms that ELS is an important risk factor for the development of PTSD in adulthood. In addition, paeoniflorin may improve ELS-induced PTSD-like behaviors and reduce the susceptibility of ELS rats to develop PTSD in adulthood by modulating the HATs/HDACs ratio in the hippocampus and amygdala.


Subject(s)
Amygdala , Glucosides , Hippocampus , Histones , Monoterpenes , Stress Disorders, Post-Traumatic , Stress, Psychological , Animals , Glucosides/pharmacology , Glucosides/therapeutic use , Monoterpenes/pharmacology , Monoterpenes/therapeutic use , Hippocampus/metabolism , Hippocampus/drug effects , Acetylation/drug effects , Amygdala/metabolism , Amygdala/drug effects , Histones/metabolism , Rats , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Male , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Rats, Sprague-Dawley , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism
6.
Curr Opin Psychiatry ; 37(4): 277-281, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38726805

ABSTRACT

PURPOSE OF REVIEW: The pace of psychedelic treatments continues to increase. Regulation and coherent clinical guidance have not been established. A philosophical divide limits effective resolution of a practice delivery quandary: is this primarily a pharmacological or psychotherapeutic intervention? RECENT FINDINGS: Lykos (formerly MAPS) has submitted its new drug application (NDA) request to the FDA for 3-4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for PTSD and is expecting a response by the summer of 2024. Australia endorsed psilocybin and MDMA for regulated use in 2023. Multiple phase II and III clinical trials are also being conducted in the United States and Europe to study the use of psilocybin. Currently, Colorado and Oregon have legalized psilocybin in different manners. In Colorado, plants containing psilocybin, ibogaine, dimethyltryptamine (DMT) and mescaline (other than peyote) are now legal to possess, share and cultivate. Guidelines for regulated treatment with psilocybin containing mushrooms are in process with service delivery to begin early in 2025. In Oregon, clients must complete a preparation session with a licensed facilitator before consuming psilocybin products at a licensed service center. A prescription is not required. It is expected that other states will follow suit with a ballot measure likely in Massachusetts this year. Additionally, in the United States, the DEA, state boards, pharmaceutical distributors, and professional liability carriers all share mounting concerns about the in-home use of compounded ketamine used as a psychedelic therapeutic via remote prescribing. SUMMARY: Psychedelic treatments are rapidly entering the mainstream of medical care delivery in the United States. Clinical guidelines are urgently needed to ensure well tolerated practice and coherent regulation. The delivery of this guidance is limited by a core philosophical disagreement. Resolution of this conflict will be needed to deliver coherent clinical guidelines. Current research and clinical experience provide a solid foundation for practical clinical guidance and the introduction of psychedelics into healthcare.


Subject(s)
Hallucinogens , Hallucinogens/therapeutic use , Humans , Psilocybin/therapeutic use , Psychotherapy/methods , United States , N-Methyl-3,4-methylenedioxyamphetamine/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy
7.
Transl Psychiatry ; 14(1): 209, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796504

ABSTRACT

Stress affects the brain and alters its neuroarchitecture and function; these changes can be severe and lead to psychiatric disorders. Recent evidence suggests that astrocytes and microglia play an essential role in the stress response by contributing to the maintenance of cerebral homeostasis. These cells respond rapidly to all stimuli that reach the brain, including stressors. Here, we used a recently validated rodent model of post-traumatic stress disorder in which rats can be categorized as resilient or vulnerable after acute inescapable footshock stress. We then investigated the functional, molecular, and morphological determinants of stress resilience and vulnerability in the prefrontal cortex, focusing on glial and neuronal cells. In addition, we examined the effects of a single subanesthetic dose of ketamine, a fast-acting antidepressant recently approved for the treatment of resistant depression and proposed for other stress-related psychiatric disorders. The present results suggest a prompt glial cell response and activation of the NF-κB pathway after acute stress, leading to an increase in specific cytokines such as IL-18 and TNF-α. This response persists in vulnerable individuals and is accompanied by a significant change in the levels of critical glial proteins such as S100B, CD11b, and CX43, brain trophic factors such as BDNF and FGF2, and proteins related to dendritic arborization and synaptic architecture such as MAP2 and PSD95. Administration of ketamine 24 h after the acute stress event rescued many of the changes observed in vulnerable rats, possibly contributing to support brain homeostasis. Overall, our results suggest that pivotal events, including reactive astrogliosis, changes in brain trophic factors, and neuronal damage are critical determinants of vulnerability to acute traumatic stress and confirm the therapeutic effect of acute ketamine against the development of stress-related psychiatric disorders.


Subject(s)
Astrocytes , Disease Models, Animal , Ketamine , Microglia , Stress Disorders, Post-Traumatic , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microglia/drug effects , Microglia/metabolism , Male , Rats , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Stress, Psychological/metabolism , Rats, Sprague-Dawley , NF-kappa B/metabolism
8.
Genes (Basel) ; 15(4)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38674453

ABSTRACT

Post-traumatic stress disorder (PTSD) is the most common psychiatric disorder after a catastrophic event; however, the efficacious treatment options remain insufficient. Increasing evidence suggests that cannabidiol (CBD) exhibits optimal therapeutic effects for treating PTSD. To elucidate the cell-type-specific transcriptomic pathology of PTSD and the mechanisms of CBD against this disease, we conducted single-nucleus RNA sequencing (snRNA-seq) in the hippocampus of PTSD-modeled mice and CBD-treated cohorts. We constructed a mouse model by adding electric foot shocks following exposure to single prolonged stress (SPS+S) and tested the freezing time, anxiety-like behavior, and cognitive behavior. CBD was administrated before every behavioral test. The PTSD-modeled mice displayed behaviors resembling those of PTSD in all behavioral tests, and CBD treatment alleviated all of these PTSD-like behaviors (n = 8/group). Three mice with representative behavioral phenotypes were selected from each group for snRNA-seq 15 days after the SPS+S. We primarily focused on the excitatory neurons (ExNs) and inhibitory neurons (InNs), which accounted for 68.4% of the total cell annotations. A total of 88 differentially upregulated genes and 305 differentially downregulated genes were found in the PTSD mice, which were found to exhibit significant alterations in pathways and biological processes associated with fear response, synaptic communication, protein synthesis, oxidative phosphorylation, and oxidative stress response. A total of 63 overlapping genes in InNs were identified as key genes for CBD in the treatment of PTSD. Subsequent Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the anti-PTSD effect of CBD was related to the regulation of protein synthesis, oxidative phosphorylation, oxidative stress response, and fear response. Furthermore, gene set enrichment analysis (GSEA) revealed that CBD also enhanced retrograde endocannabinoid signaling in ExNs, which was found to be suppressed in the PTSD group. Our research may provide a potential explanation for the pathogenesis of PTSD and facilitate the discovery of novel therapeutic targets for drug development. Moreover, it may shed light on the therapeutic mechanisms of CBD.


Subject(s)
Cannabidiol , Disease Models, Animal , Hippocampus , Stress Disorders, Post-Traumatic , Transcriptome , Animals , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Mice , Cannabidiol/pharmacology , Male , Gene Expression Profiling , Mice, Inbred C57BL
9.
J Ethnopharmacol ; 330: 118270, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38685368

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Cheonwangbosimdan (CWBSD), a herbal medicine traditionally used for anxiety, insomnia, depression, and heart palpitations, has been reported to have anti-anxiety, antidepressant, cognitive improvement, and neuroprotective effects. AIM OF THE STUDY: The purpose of this study was to determine if CWBSD could affect post-traumatic stress disorder (PTSD)-like behaviors because it has prioritized clinical use over mechanism study. MATERIALS AND METHODS: A single prolonged stress (SPS) mouse model, a well-established animal model of PTSD, was used to investigate whether standardized CWBSD could mitigate PTSD-like behaviors through robust behavioral tests, including the elevated plus-maze test and marble burying test for measuring anxiety-like behaviors, the splash test, forced swimming test, and tail suspension test for evaluating depression-like behaviors, and the Y-maze test and novel object recognition test for assessing cognitive function. Additionally, a fear extinction test was employed to determine whether CWBSD might reverse fear memory extinction deficits. Amygdala tissue was isolated from SPS-treated mouse brain and subjected to Western blotting or quantitative PCR to explore mechanisms by which CWBSD could mitigate PTSD-like behaviors. RESULTS: CWBSD ameliorated emotional impairments and cognitive dysfunction in an SPS-induced PTSD-like mouse model. It also mitigated deficits in abnormal fear memory extinction. Protein expression levels of N-methyl-D-aspartate (NMDA) receptor subunit 2B (GluN2B) and phosphorylation levels of Ca2+/calmodulin-dependent protein kinase II in the amygdala were increased in SPS model mice and normalized by CWBSD. Additionally, co-administration of CWBSD and GluN2B-containing NMDA receptor antagonist, ifenprodil, at each sub-effective dose promoted fear memory extinction. CONCLUSIONS: CWBSD can alleviate SPS-induced PTSD-like behaviors by normalizing GluN2B-containing NMDA receptor activity in the amygdala. Therefore, CWBSD could be a promising candidate for PTSD treatment with fewer adverse effects and better efficacy than existing therapies.


Subject(s)
Behavior, Animal , Disease Models, Animal , Receptors, N-Methyl-D-Aspartate , Stress Disorders, Post-Traumatic , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Stress Disorders, Post-Traumatic/metabolism , Male , Mice , Behavior, Animal/drug effects , Mice, Inbred C57BL , Fear/drug effects , Amygdala/drug effects , Amygdala/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Anxiety/drug therapy , Anxiety/psychology
10.
Behav Brain Res ; 467: 115005, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38641178

ABSTRACT

BACKGROUND: Post-traumatic stress disorder (PTSD) refers to a chronic impairing psychiatric disorder occurring after exposure to the severe traumatic event. Studies have demonstrated that medicinal cannabis oil plays an important role in neuroprotection, but the mechanism by which it exerts anti-PTSD effects remains unclear. METHODS: The chronic complex stress (CCS) simulating the conditions of long voyage stress for 4 weeks was used to establish the PTSD mice model. After that, behavioral tests were used to evaluate PTSD-like behaviors in mice. Mouse brain tissue index was detected and hematoxylin-eosin staining was used to assess pathological changes in the hippocampus. The indicators of cell apoptosis and the BDNF/TRPC6 signaling activation in the mice hippocampus were detected by western blotting or real-time quantitative reverse transcription PCR experiments. RESULTS: We established the PTSD mice model induced by CCS, which exhibited significant PTSD-like phenotypes, including increased anxiety-like and depression-like behaviors. Medicinal cannabis oil treatment significantly ameliorated PTSD-like behaviors and improved brain histomorphological abnormalities in CCS mice. Mechanistically, medicinal cannabis oil reduced CCS-induced cell apoptosis and enhanced the activation of BDNF/TRPC6 signaling pathway. CONCLUSIONS: We constructed a PTSD model with CCS and medicinal cannabis oil that significantly improved anxiety-like and depressive-like behaviors in CCS mice, which may play an anti-PTSD role by stimulating the BDNF/TRPC6 signaling pathway.


Subject(s)
Anxiety , Brain-Derived Neurotrophic Factor , Depression , Disease Models, Animal , Hippocampus , Signal Transduction , Stress Disorders, Post-Traumatic , TRPC6 Cation Channel , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Mice , Signal Transduction/drug effects , Anxiety/drug therapy , Anxiety/metabolism , Male , Depression/drug therapy , Depression/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , TRPC6 Cation Channel/metabolism , Behavior, Animal/drug effects , Medical Marijuana/pharmacology , Mice, Inbred C57BL , Apoptosis/drug effects , Plant Oils/pharmacology , Plant Oils/administration & dosage , Stress, Psychological/drug therapy , Stress, Psychological/metabolism
11.
Article in English | MEDLINE | ID: mdl-38467326

ABSTRACT

Post-traumatic stress disorder (PTSD) is a mental illness that can occur in individuals who have experienced trauma. Current treatments for PTSD, typically serotonin reuptake inhibitors, have limited effectiveness for patients and often cause serious adverse effects. Therefore, a novel class of treatment with better pharmacological profile is necessary. D-Pinitol has been reported to be effective for depression and anxiety disorders, but there are no reports associated with PTSD. In the present study, we investigated the effects of D-pinitol in a mouse model of PTSD induced by a single prolonged stress (SPS) protocol. We examined the therapeutic effects of D-pinitol on emotional and cognitive impairments in the SPS mouse model. We also investigated the effects of D-pinitol on fear memory formation. Mineralocorticoid receptor transactivation assay, Western blot, and quantitative PCR were employed to investigate how D-pinitol exerts its pharmacological activities. D-Pinitol ameliorated PTSD-like behaviors in a SPS mouse model. D-Pinitol also normalized the increased mRNA expression levels and protein levels of the mineralocorticoid receptor in the amygdala. A mineralocorticoid receptor agonist reversed the effects of D-pinitol on fear extinction and recall, and the antagonistic property of D-pinitol against the mineralocorticoid receptor was confirmed in vitro. Our findings suggest that D-pinitol could serve as a potential therapeutic agent for PTSD due to its antagonistic effect on the mineralocorticoid receptor.


Subject(s)
Inositol/analogs & derivatives , Stress Disorders, Post-Traumatic , Mice , Humans , Animals , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Fear/physiology , Extinction, Psychological , Receptors, Mineralocorticoid/metabolism , Receptors, Mineralocorticoid/therapeutic use , Disease Models, Animal , Stress, Psychological/psychology
13.
Indian J Pharmacol ; 56(1): 20-27, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38454585

ABSTRACT

AIMS AND OBJECTIVES: Posttraumatic stress disorder (PTSD) is a complex neuropsychiatric pathophysiology with an unmet need for safe, effective, and sustainable therapeutic modalities. Thus, the present study evaluated the effects of Withaniasomnifera (WS, Ashwagandha) on an experimental model of PTSD in rats. MATERIALS AND METHODS: Wistar rats (200-250 g) were used and time-dependent sensitization (TDS) was used as the experimental model of PTSD. Standardized WS root extract (100 and 300 mg/kg, p.o. for 15 days) was administered with TDS and their effects were observed on neurobehavioral (anxiety) and brain cytokines, corticosterone, and oxidative stress markers. RESULTS: Exposure to TDS resulted in anxiogenic behavior in the elevated plus maze (EPM) test, i.e., reductions in open arm entries and open arm time, as compared to the control group. Pretreatment with WS extract (100 and 300 mg/kg × 14 days) attenuated the TDS-induced anxiogenic activity in a dose-related manner, and these WS effects were comparable to those seen after the comparator drug fluoxetine (10 mg/kg). Assay of brain homogenates showed that TDS also resulted in elevations in brain interleukin-6 and reduction in corticosterone levels in both the hippocampus and prefrontal cortex (PFC), which were reversed after WS pretreatments. Further, WS pretreatment also reversed the TDS-induced changes in brain oxidative stress markers, namely elevated malondialdehyde and reduced glutathione levels in both the hippocampus and PFC. CONCLUSION: These results suggest that WS could have potential as a therapeutic agent for treating PTSD by attenuating anxiogenesis, neuroimmune axis activation, and oxidative stress.


Subject(s)
Stress Disorders, Post-Traumatic , Withania , Rats , Animals , Stress Disorders, Post-Traumatic/drug therapy , Rats, Wistar , Corticosterone , Antioxidants/pharmacology
14.
Eur Psychiatry ; 67(1): e24, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38450651

ABSTRACT

BACKGROUND: Considering the recently growing number of potentially traumatic events in Europe, the European Psychiatric Association undertook a study to investigate clinicians' treatment choices for post-traumatic stress disorder (PTSD). METHODS: The case-based analysis included 611 participants, who correctly classified the vignette as a case of PTSD, from Central/ Eastern Europe (CEE) (n = 279), Southern Europe (SE) (n = 92), Northern Europe (NE) (n = 92), and Western Europe (WE) (N = 148). RESULTS: About 82% woulduse antidepressants (sertraline being the most preferred one). Benzodiazepines and antipsychotics were significantly more frequently recommended by participants from CEE (33 and 4%, respectively), compared to participants from NE (11 and 0%) and SE (9% and 3%). About 52% of clinicians recommended trauma-focused cognitive behavior therapy and 35% psychoeducation, irrespective of their origin. In the latent class analysis, we identified four distinct "profiles" of clinicians. In Class 1 (N = 367), psychiatrists would less often recommend any antidepressants. In Class 2 (N = 51), clinicians would recommend trazodone and prolonged exposure therapy. In Class 3 (N = 65), they propose mirtazapine and eye movement desensitization reprocessing therapy. In Class 4 (N = 128), clinicians propose different types of medications and cognitive processing therapy. About 50.1% of participants in each region stated they do not adhere to recognized treatment guidelines. CONCLUSIONS: Clinicians' decisions for PTSD are broadly similar among European psychiatrists, but regional differences suggest the need for more dialogue and education to harmonize practice across Europe and promote the use of guidelines.


Subject(s)
Cognitive Behavioral Therapy , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Psychiatrists , Europe , Antidepressive Agents/therapeutic use
15.
Am J Ther ; 31(2): e141-e154, 2024.
Article in English | MEDLINE | ID: mdl-38518271

ABSTRACT

BACKGROUND: After becoming notorious for its use as a party drug in the 1980s, 3,4-methylenedioxy-methampetamine (MDMA), also known by its street names "molly" and "ecstasy," has emerged as a powerful treatment for post-traumatic stress disorder (PTSD). AREAS OF UNCERTAINTY: There are extensive data about the risk profile of MDMA. However, the literature is significantly biased. Animal models demonstrating neurotoxic or adverse effects used doses well beyond the range that would be expected in humans (up to 40 mg/kg in rats compared with roughly 1-2 mg/kg in humans). Furthermore, human samples often comprise recreational users who took other substances in addition to MDMA, in uncontrolled settings. THERAPEUTIC ADVANCES: Phase III clinical trials led by the Multidisciplinary Association for Psychedelic Studies (MAPS) have shown that MDMA-assisted psychotherapy has an effect size of d = 0.7-0.91, up to 2-3 times higher than the effect sizes of existing antidepressant treatments. 67%-71% of patients who undergo MDMA-assisted psychotherapy no longer meet the diagnostic criteria for PTSD within 18 weeks. We also describe other promising applications of MDMA-assisted psychotherapy for treating alcohol use disorder, social anxiety, and other psychiatric conditions. LIMITATIONS: Thus far, almost all clinical trials on MDMA have been sponsored by a single organization, MAPS. More work is needed to determine whether MDMA-assisted therapy is more effective than existing nonpharmacological treatments such as cognitive behavioral therapy. CONCLUSIONS: Phase III trials suggest that MDMA is superior to antidepressant medications for treating PTSD. Now that MAPS has officially requested the Food and Drug Administration to approve MDMA as a treatment for PTSD, legal MDMA-assisted therapy may become available as soon as 2024.


Subject(s)
Hallucinogens , Methamphetamine , N-Methyl-3,4-methylenedioxyamphetamine , Stress Disorders, Post-Traumatic , Animals , Humans , Rats , Antidepressive Agents/therapeutic use , Clinical Trials, Phase III as Topic , Hallucinogens/therapeutic use , Methamphetamine/therapeutic use , N-Methyl-3,4-methylenedioxyamphetamine/therapeutic use , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Primary Health Care , Psychotherapy , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology
16.
Nord J Psychiatry ; 78(4): 353-361, 2024 May.
Article in English | MEDLINE | ID: mdl-38451197

ABSTRACT

BACKGROUND: Research on long-term pharmacotherapy for trauma-affected refugees is scarce. The purpose of this follow-up study of a randomised trial was to investigate the effects of sertraline compared to venlafaxine in combination with psychotherapy, 6 and 18 months after end of trial. METHOD: The primary outcome was PTSD symptoms, measured by the Harvard Trauma Questionnaire (HTQ). The secondary outcomes included: Hopkins Symptom Checklist-25 (HSCL-25), somatisation items of the Symptoms Checklist-90 (SCL), pain on a visual analogue scale, well-being on the WHO-5, Sheehan Disability Scale, Hamilton Depression and Anxiety scales and Global Assessment of Functioning. Moreover, the shorter version of the Recent Life Events (IRLE) was adopted to obtain information regarding the patients' treatment and life events between the follow-up periods. RESULTS: Out of 195 patients eligible for intention-to-treat analyses during trial, 116 participated in the 6-month follow-up and 97 participated in the 18-month follow-up. The results of our intention-to-treat analyses revealed no significant long-term differences between the groups on the primary outcome assessing PTSD symptoms (HTQ). For the secondary outcomes significant differences were found at the 18-month follow-up in favour of venlafaxine assessing symptoms of anxiety, depression and somatisation (HSCL-25 and SCL), although only in intention-to-treat and not per-protocol analyses. CONCLUSIONS: No conclusions could be drawn due to conflicting results between our intention-to-treat and per-protocol analyses.


Subject(s)
Psychotherapy , Refugees , Sertraline , Stress Disorders, Post-Traumatic , Venlafaxine Hydrochloride , Humans , Venlafaxine Hydrochloride/therapeutic use , Venlafaxine Hydrochloride/administration & dosage , Female , Adult , Stress Disorders, Post-Traumatic/therapy , Stress Disorders, Post-Traumatic/drug therapy , Male , Follow-Up Studies , Psychotherapy/methods , Refugees/psychology , Sertraline/therapeutic use , Combined Modality Therapy , Middle Aged , Psychological Trauma/therapy , Psychological Trauma/drug therapy , Treatment Outcome , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/therapeutic use
17.
Child Adolesc Psychiatr Clin N Am ; 33(2): 193-205, 2024 04.
Article in English | MEDLINE | ID: mdl-38395505

ABSTRACT

Trauma exposure significantly impacts sleep in children. Nightmares are common. Evidence-based therapies are superior to medications but may not always be available in acute settings. No FDA-approved medications exist for the treatment of trauma-related sleep disturbances in youth. The evidence-base for the use of medications is largely based on case reports, retrospective chart reviews, clinical opinion, and adult studies. This evidence is reviewed for a number of medications, including prazosin, trazodone, alpha-2 agonists, quetiapine, and others.


Subject(s)
Sleep Wake Disorders , Stress Disorders, Post-Traumatic , Transients and Migrants , Adult , Child , Humans , Adolescent , Retrospective Studies , Stress Disorders, Post-Traumatic/drug therapy , Sleep , Prazosin/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/etiology
18.
J Clin Psychiatry ; 85(1)2024 02 14.
Article in English | MEDLINE | ID: mdl-38353645

ABSTRACT

Objective: Given the high rate of comorbid posttraumatic stress disorder (PTSD) and cannabis use, it is critical that further research be conducted to address the associated benefits and risks of cannabis use in this population. This systematic review evaluated evidence on the effects of cannabis and cannabinoids on PTSD symptoms and PTSD clusters.Data Sources: A systematic search of PubMed, PsycINFO, and EMBASE databases was performed using terms related to cannabis, cannabinoids, and PTSD. Peer-reviewed studies available online in English and published from January 1990 through February 2023 were considered.Study Selection: Included studies were experimental or observational in design, were conducted in cannabis-using patients with PTSD, used validated measures of PTSD, and were published in English.Data Extraction: Extracted information included study aims, study design, sample size and sex, comparator group, cannabis-related characteristics, psychometric instruments, and relevant clinical findings regarding overall PTSD symptoms and cluster symptoms.Results: Fourteen studies were included, 3 in a comorbid PTSD and cannabis use disorder (CUD) sample and 11 in a non-CUD sample. Of the 10 studies examining overall PTSD symptoms in a non-CUD sample, 5 suggested benefits associated with cannabis use and 5 suggested no effect or worsening of symptoms. Four studies reported benefits of cannabis for cluster B- and E-related symptoms in a non-CUD sample. All 3 studies in cannabis-using patients with a comorbid PTSD and CUD diagnosis reported risks for worsening of overall symptoms.Conclusions: This review did not find major benefits of cannabinoids in improving overall PTSD symptoms. Some benefits with regard to cluster B and E symptoms were observed. Some risks with regard to worsening suicidal ideation and violent behavior were also reported. Individuals with a comorbid CUD diagnosis may be at greater risk for negative cannabis-related PTSD outcomes. More experimental studies are needed to determine the causal effects of cannabis and cannabinoids in PTSD.


Subject(s)
Cannabinoids , Cannabis , Hallucinogens , Stress Disorders, Post-Traumatic , Humans , Cannabinoids/adverse effects , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/epidemiology , Syndrome
19.
Drugs ; 84(2): 149-164, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38413493

ABSTRACT

Post-traumatic stress disorder (PTSD) is a debilitating disorder inflicting high degrees of symptomatic and socioeconomic burdens. The development of PTSD results from a cascade of events with contributions from multiple processes and the underlying pathophysiology is complex, involving neurotransmitters, neurocircuitry, and neuroanatomical pathways. Presently, only two medications are US FDA-approved for the treatment of PTSD, both selective serotonin reuptake inhibitors (SSRIs). However, the complex underlying pathophysiology suggests a number of alternative pathways and mechanisms that may be targets for potential drug development. Indeed, investigations and drug development are proceeding in a number of these alternative, non-serotonergic pathways in an effort to improve the management of PTSD. In this manuscript, the authors introduce novel and emerging treatments for PTSD, including drugs in various stages of development and clinical testing (BI 1358894, BNC-210, PRAX-114, JZP-150, LU AG06466, NYV-783, PH-94B, SRX246, TNX-102), established agents and known compounds being investigated for their utility in PTSD (brexpiprazole, cannabidiol, doxasoin, ganaxolone, intranasal neuropeptide Y, intranasal oxytocin, tianeptine oxalate, verucerfont), and emerging psychedelic interventions (ketamine, MDMA-assisted psychotherapy, psilocybin-assisted psychotherapy), with an aim to examine and integrate these agents into the underlying pathophysiological frameworks of trauma-related disorders.


Subject(s)
Ketamine , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/drug therapy , Psychotherapy/methods , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Ketamine/therapeutic use
20.
Psychiatry Res ; 334: 115764, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38350291

ABSTRACT

Sleep disturbances in posttraumatic stress disorder (PTSD) are a potential target for improving PTSD severity with pharmacotherapy. TNX-102 SL is a bedtime sublingual formulation of cyclobenzaprine with potent binding and antagonist activity at 5-HT2A, α1-adrenergic, H1 histaminergic, and M1 muscarinic receptors, which play roles in the pharmacological management of sleep disturbances. This Phase 3 trial evaluated the efficacy and safety of TNX-102 SL in patients with military-related PTSD. Early and sustained improvements in sleep were associated with TNX-102 SL treatment by PROMIS Sleep Disturbance scale and Clinician Administered PTSD Scale (CAPS-5) "sleep disturbance" item, establishing a sleep quality benefit. Primary analysis comparing change from baseline in CAPS-5 total severity between TNX-102 SL and placebo at week 12 was not significant; however, week 4 was associated with an improvement. Secondary analyses showed TNX-102 SL treatment was associated with benefits on the Clinician Global Impression of Improvement at week 4 and the Patient Global Impression of Change at week 12. Time since trauma exposure was a discriminator of CAPS-5 treatment response in the subgroup ≤ 9 years since the index event. This study provides preliminary evidence that TNX-102 SL is well-tolerated and may promote recovery from PTSD by addressing sleep-related symptoms.


Subject(s)
Amitriptyline/analogs & derivatives , Military Personnel , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/diagnosis , Sleep , Treatment Outcome , Double-Blind Method
SELECTION OF CITATIONS
SEARCH DETAIL
...