Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Neuroimage ; 289: 120542, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38369167

ABSTRACT

MRI-guided neuro interventions require rapid, accurate, and reproducible segmentation of anatomical brain structures for identification of targets during surgical procedures and post-surgical evaluation of intervention efficiency. Segmentation algorithms must be validated and cleared for clinical use. This work introduces a methodology for shape-constrained deformable brain segmentation, describes the quantitative validation used for its clinical clearance, and presents a comparison with manual expert segmentation and FreeSurfer, an open source software for neuroimaging data analysis. ClearPoint Maestro is software for fully-automatic brain segmentation from T1-weighted MRI that combines a shape-constrained deformable brain model with voxel-wise tissue segmentation within the cerebral hemispheres and the cerebellum. The performance of the segmentation was validated in terms of accuracy and reproducibility. Segmentation accuracy was evaluated with respect to training data and independently traced ground truth. Segmentation reproducibility was quantified and compared with manual expert segmentation and FreeSurfer. Quantitative reproducibility analysis indicates superior performance compared to both manual expert segmentation and FreeSurfer. The shape-constrained methodology results in accurate and highly reproducible segmentation. Inherent point based-correspondence provides consistent target identification ideal for MRI-guided neuro interventions.


Subject(s)
Algorithms , Software , Humans , Reproducibility of Results , Brain/diagnostic imaging , Magnetic Resonance Imaging/methods , Image Processing, Computer-Assisted/methods
2.
J Stroke ; 25(1): 160-168, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36746385

ABSTRACT

BACKGROUND AND PURPOSE: Nelonemdaz (Neu2000) has both selective antagonism against 2B subunit of N-methyl-D-aspartate receptor and antioxidant activity. This drug provides sufficient evidence of neuroprotection in acute cerebral ischemia/reperfusion models. This phase III trial aims to determine this effect in patients. DESIGN: The Rescue on Reperfusion Damage in Cerebral Infarction by Nelonemdaz is a multicenter, double-blinded clinical trial. A total of 496 patients will be randomly assigned into the nelonemdaz (a total of 5,250 mg divided by 10 times for 5 days) and placebo groups. Patients will be included if they have an acute ischemic stroke (National Institutes of Health Stroke Scale score ≥8) caused by intracranial large vessel occlusion in the anterior circulation (Alberta Stroke Program Early CT Score ≥4), and if they are expected to undergo endovascular thrombectomy within 12 hours after stroke onset. ENDPOINTS: The primary endpoint is a favorable shift in the modified Rankin Scale (mRS) score at 90 days after the first dose of drug. The data will be analyzed by the Cochran-Mantel-Haenszel shift test. The secondary endpoints include functional independence (mRS 0-2) at 35 and 90 days, the favorable shift of mRS at 35 days, the proportion of mRS 0 at 35 and 90 days, and the occurrence rates of symptomatic intracranial hemorrhage within 7 days. CONCLUSION: This trial will clarify the efficacy and safety of nelonemdaz in patients with acute ischemic stroke and endovascular thrombectomy. This study has been registered at ClinicalTrials. gov (NCT05041010).

3.
Stroke ; 53(11): 3250-3259, 2022 11.
Article in English | MEDLINE | ID: mdl-36065810

ABSTRACT

BACKGROUND: Nelonemdaz is a multitarget neuroprotectant that selectively blocks N-methyl-D-aspartate receptors and scavenges free radicals, as proven in preclinical ischemia-reperfusion studies. We aimed to evaluate the safety and efficacy of nelonemdaz in patients with acute ischemic stroke receiving endovascular reperfusion therapy. METHODS: This phase II randomized trial involved participants with large-artery occlusion in the anterior circulation at baseline who received endovascular reperfusion therapy <8 hours from symptom onset at 7 referral stroke centers in South Korea between October 29, 2016, and June 1, 2020. Two hundred thirteen patients were screened and 209 patients were randomly assigned at a 1:1:1 ratio using a computer-generated randomization system. Patients were divided into 3 groups based on the medication received-placebo, low-dose (2750 mg) nelonemdaz, and high-dose (5250 mg) nelonemdaz. The primary outcome was the proportion of patients with modified Rankin Scale scores of 0-2 at 12 weeks. RESULTS: Two hundred eight patients were assigned to the placebo (n=70), low-dose (n=71), and high-dose (n=67) groups. The groups had similar baseline characteristics. The primary outcome was achieved in 183 patients, and it did not differ among the groups (33/61 [54.1%], 40/65 [61.5%], and 36/57 [63.2%] patients; P=0.5578). The common odds ratio (90% CI) indicating a favorable shift in the modified Rankin Scale scores at 12 weeks was 1.55 (0.92-2.60) between the placebo and low-dose groups and 1.61 (0.94-2.76) between the placebo and high-dose groups. No serious adverse events were reported. CONCLUSIONS: The study arms showed no significant difference in the proportion of patients achieving modified Rankin Scale scores of 0-2 at 12 weeks. Nevertheless, nelonemdaz-treated patients showed a favorable tendency toward achieving these scores at 12 weeks, without serious adverse effects. Thus, a large-scale phase III trial is warranted. REGISTRATION: URL: https://clinicaltrials.gov; Unique identifier: NCT02831088.


Subject(s)
Brain Ischemia , Endovascular Procedures , Ischemic Stroke , Neuroprotective Agents , Stroke , Humans , Brain Ischemia/drug therapy , Brain Ischemia/surgery , Brain Ischemia/diagnosis , Thrombectomy/adverse effects , Ischemic Stroke/drug therapy , Ischemic Stroke/surgery , Stroke/drug therapy , Stroke/surgery , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate , Endovascular Procedures/adverse effects , Treatment Outcome , Reperfusion
4.
Surg Endosc ; 36(3): 1876-1886, 2022 03.
Article in English | MEDLINE | ID: mdl-33825018

ABSTRACT

BACKGROUND: The obesity rate is projected to reach 50% by 2030. Obesity may be modifiable prior to elective colorectal surgery, but there is no opportunity for weight loss when patients present for urgent/emergency operations. The impact of obesity focused on urgent/emergent colorectal operations has not been fully characterized. The study aim was to determine outcomes of obese patients who undergo urgent/emergency colorectal surgery and differences when compared with elective outcomes. METHODS: This is a retrospective cohort study of 30-day outcomes for normal (BMI 18.5-25), obese (BMI 30-39.9), and morbidly obese (BMI > 40) patients in the Michigan Surgical Quality Collaborative between 1/1/2009 and 12/31/2018. Propensity score weighting was used to derive adjusted rates for overall morbidity, mortality, and specific complications. Primary outcome was postoperative complications (any morbidity). RESULTS: The study included 5268 urgent/emergency and 10,414 elective colorectal surgery patients. Postoperative complications were significantly more common in morbidly obese and obese than the normal BMI group for both urgent/emergency (morbidly obese 42.76% vs 33.75%, p = 0.003; obese 36.46% vs 33.75%, p = 0.043) and elective (morbidly obese 18.17% vs 13.36%, p = 0.004; obese 15.45% vs 13.36%, p = 0.011) operations. Surgical site infections are were significantly more common in morbidly obese and obese BMI groups as compared to normal BMI for both urgent/emergency and elective cases. Mortality was significantly higher in the morbidly obese (14.93% vs 11.44%, p = 0.013) but not obese BMI groups as compared to the normal BMI group for urgent/emergency cases. Mortality for all groups undergoing elective operations was < 1% and with no significant differences. CONCLUSIONS: Morbid obesity and obesity are associated with complications that are largely driven by surgical site infections after both urgent/emergency and elective colorectal surgery. Obesity is a risk factor difficult to modify prior to urgent/emergency surgery. Managing complications related to obesity after colorectal surgery will be a continued challenge with projected increasing obesity rates.


Subject(s)
Colorectal Neoplasms , Obesity, Morbid , Body Mass Index , Colorectal Neoplasms/complications , Humans , Obesity, Morbid/complications , Obesity, Morbid/epidemiology , Obesity, Morbid/surgery , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Retrospective Studies , Treatment Outcome
5.
Front Neurosci ; 14: 579953, 2020.
Article in English | MEDLINE | ID: mdl-33192266

ABSTRACT

Interest in excitotoxicity expanded following its implication in the pathogenesis of ischemic brain injury in the 1980s, but waned subsequent to the failure of N-methyl-D-aspartate (NMDA) antagonists in high profile clinical stroke trials. Nonetheless there has been steady progress in elucidating underlying mechanisms. This review will outline the historical path to current understandings of excitotoxicity in the ischemic brain, and suggest that this knowledge should be leveraged now to develop neuroprotective treatments for stroke.

6.
Brain Res ; 1748: 147122, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32919984

ABSTRACT

Free radicals are downstream mediators of several cytotoxic cascades contributing to ischemic brain injury. Molecular hydrogen (H2) is an antioxidant potentially useful in the treatment of stroke. Hydrogen is easy to deliver, biologically non-toxic and diffuses freely through all biological structures including the blood-brain barrier and cellular membranes. This study evaluated the efficacy of hydrogen treatments in a rat stroke model compared to vehicle-treated controls using multiparametric MRI and neurological tests. Additionally, comparison of H2 treatment alone was made with H2 combined with minocycline (H2M) treatment (12 rats per group). The primary findings were: i) H2 therapy reduced infarct volume in both H2 and H2M groups compared to controls at 1 and 7 days after stroke, and ii) both H2 and H2M improved neurologic functional recovery on day 7. The secondary outcomes were: iii) H2M treatment attenuated post-stroke hyperperfusion in the hyperacute phase, and iv) H2M markedly minimized white matter injury. In conclusion, this is the first study to use MRI to longitudinally study H2 and H2M treatment on ischemic stroke and the first study to compare H2 treatment combined with another potential stroke therapeutic (H2M).


Subject(s)
Brain/diagnostic imaging , Hydrogen/therapeutic use , Ischemic Stroke/therapy , Minocycline/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Disease Models, Animal , Ischemic Stroke/diagnostic imaging , Male , Multiparametric Magnetic Resonance Imaging , Rats , Rats, Sprague-Dawley , Recovery of Function , Tourette Syndrome , Water
7.
BMC Neurol ; 20(1): 293, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32758167

ABSTRACT

BACKGROUND: The blood-brain barrier has been a hindrance to developing blood-based diagnostic tests for dementias, as it limits the appearance of brain biomarkers in the blood. Our aim was to see if the natural opening of the blood-brain barrier induced by ischemic stroke would increase serum levels of inflammatory biomarkers known to be elevated in the brains of patients with Alzheimer's disease and other neurodegenerative dementias. METHODS: Forty-three patients with acute ischemic stroke presenting to Stony Brook University Hospital were prospectively enrolled in the study. Eight of these patients were clinically diagnosed as having an underlying neurodegenerative dementia. Blood was drawn acutely within 72 h of stroke symptom onset, and serum levels of the classic inflammatory biomarkers, interleukin-6 (IL-6) and C-reactive protein (CRP) were measured, along with levels of S100B protein (S100B) and complement C3 (CC3). RESULTS: Serum levels of IL-6 and CRP in patients with acute ischemic stroke and underlying dementia (AIS + D) were significantly higher (p = 0.002 and 0.003, respectively) than in patients with acute ischemic stroke alone (AIS). Serum levels of S100B and CC3 did not differ significantly between the groups. CONCLUSIONS: This study supports the possibility that opening of the blood-brain barrier may enhance the blood appearance of brain tissue markers of inflammation associated with neurodegenerative dementia. Further study is warranted to test this possibility, given the recent emergence of methods to open the blood-brain barrier for diagnostic or therapeutic purposes.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/pathology , Dementia/diagnosis , Stroke/pathology , Aged , Aged, 80 and over , Biomarkers/blood , Brain/metabolism , C-Reactive Protein/metabolism , Dementia/blood , Female , Humans , Inflammation/blood , Interleukin-6/blood , Male , Middle Aged , Prospective Studies , S100 Calcium Binding Protein beta Subunit/metabolism
8.
Neuropharmacology ; 148: 40-49, 2019 04.
Article in English | MEDLINE | ID: mdl-30557566

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is known to promote fear learning as well as avoidant behavioral responses to chronic social defeat stress, but, conversely, this peptide can also have antidepressant effects and can reduce depressant-like symptoms such as social avoidance. The purpose of this study was to use a variety of approaches to determine whether BDNF acting on tropomyosin receptor kinase B (TrkB) promotes or prevents avoidant phenotypes in hamsters and mice that have experienced acute social defeat stress. We utilized systemic and brain region-dependent manipulation of BDNF signaling before or immediately following social defeat stress in Syrian hamsters, TrkBF616A knock-in mice, and C57Bl/6J mice and measured the subsequent behavioral response to a novel opponent. Systemic TrkB receptor agonists reduced, and TrkB receptor antagonists enhanced, behavioral responses to social defeat in hamsters and mice. In the neural circuit that we have shown mediates defeat-induced behavioral responses, BDNF in the basolateral amygdala, but not the nucleus accumbens, also reduced social avoidant phenotypes. Conversely, knockdown in the basolateral amygdala of TrkB signaling in TrkBF616A mice enhanced defeat-induced social avoidance. These data demonstrate that systemic administration of BDNF-TrkB drugs at the time of social defeat alters the behavioral response to the defeat stressor. These drugs appear to act, at least in part, in the basolateral amygdala and not the nucleus accumbens. These findings were generalizable to two rodent species with very different social structures and, within mice, to a variety of strains providing converging evidence that BDNF-TrkB signaling reduces anxiety- and depression-like symptoms following short-term social stress.


Subject(s)
Avoidance Learning/physiology , Basolateral Nuclear Complex/physiology , Brain-Derived Neurotrophic Factor/physiology , Membrane Glycoproteins/physiology , Nucleus Accumbens/physiology , Protein-Tyrosine Kinases/physiology , Social Behavior , Stress, Psychological/psychology , Animals , Avoidance Learning/drug effects , Basolateral Nuclear Complex/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cricetinae , Gene Knock-In Techniques , Gene Knockdown Techniques , Male , Membrane Glycoproteins/agonists , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Microinjections , Nucleus Accumbens/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics
9.
Trials ; 19(1): 375, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30005644

ABSTRACT

BACKGROUND: The potential of neuroprotective agents should be revisited in the era of endovascular thrombectomy (EVT) for acute large-artery occlusion because their preclinical effects have been optimized for ischemia and reperfusion injury. Neu2000, a derivative of sulfasalazine, is a multi-target neuroprotectant. It selectively blocks N-methyl-D-aspartate receptors and scavenges for free radicals. This trial aimed to determine whether neuroprotectant administration before EVT is safe and leads to a more favorable outcome. METHODS: This trial is a phase-II, multicenter, three-arm, randomized, double-blinded, placebo-controlled, blinded-endpoint drug trial that enrolled participants aged ≥ 19 years undergoing an EVT attempt less than 8 h from symptom onset, with baseline National Institutes of Health Stroke Scale (NIHSS) score ≥ 8, Alberta Stroke Program Early CT score ≥ 6, evidence of large-artery occlusion, and at least moderate collaterals on computed tomography angiography. EVT-attempted patients are randomized into control, low-dose (2.75 g), and high-dose (5.25 g) Neu2000KWL over 5 days. Seventy participants per group are enrolled for 90% power, assuming that the treatment group has a 28.4% higher proportion of participants with functional independence than the placebo group. The primary outcome, based on intention-to-treat criteria is the improvement of modified Rankin Scale (mRS) scores at 3 months using a dichotomized model. Safety outcomes include symptomatic intracranial hemorrhage within 5 days. Secondary outcomes are distributional change of mRS, mean differences in NIHSS score, proportion of NIHSS score 0-2, and Barthel Index > 90 at 1 and 4 weeks, and 3 months. DISCUSSION: The trial results may provide information on new therapeutic options as multi-target neuroprotection might mitigate reperfusion injury in patients with acute ischemic stroke before EVT. TRIAL REGISTRATION: ClinicalTrials.gov, ID: NCT02831088 . Registered on 13 July 2016.


Subject(s)
Brain Ischemia/therapy , Endovascular Procedures , Fluorobenzenes/therapeutic use , Neuroprotective Agents/therapeutic use , Salicylates/therapeutic use , Stroke/therapy , Thrombectomy/methods , meta-Aminobenzoates/therapeutic use , Brain Ischemia/diagnosis , Brain Ischemia/physiopathology , Clinical Trials, Phase II as Topic , Disability Evaluation , Double-Blind Method , Endovascular Procedures/adverse effects , Fluorobenzenes/adverse effects , Humans , Multicenter Studies as Topic , Neuroprotective Agents/adverse effects , Prospective Studies , Randomized Controlled Trials as Topic , Recovery of Function , Republic of Korea , Salicylates/adverse effects , Stroke/diagnosis , Stroke/physiopathology , Thrombectomy/adverse effects , Time Factors , Treatment Outcome , meta-Aminobenzoates/adverse effects
10.
Biol Psychiatry ; 83(3): 284-295, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29054677

ABSTRACT

BACKGROUND: Molecular mechanisms underlying psychological sequelae of exposure to stressful experiences, such as posttraumatic stress disorder (PTSD) and depression, are not well understood. METHODS: Using convergent evidence from animal and human transcriptomic and genomic studies, we aimed to identify genetic mechanisms underlying depression and anxiety after traumatic experiences. RESULTS: From a transcriptome-wide analysis in mice, we found the Ppm1f gene to be differentially expressed in the amygdala and medial prefrontal cortex (mPFC) a week after immobilization stress. Next, we found that PPM1F messenger RNA levels in human blood were downregulated in cases with symptoms of comorbid PTSD and depression and consistently in cases with anxiety symptoms in a separate human dataset. Furthermore, we showed that a genetic variant of PPM1F, rs17759843, was associated with comorbid PTSD and depression and with PPM1F expression in both human brain and blood. Given prior reported mechanistic links between PPM1F and CAMK2 (CAMKII), we examined blood messenger RNA level of CAMK2G in humans and found it to be lower in cases with comorbid PTSD and depression. We also found that PPM1F protein levels and colocalization with CAMK2G were altered in amygdala and mPFC of male mice. Additionally, we found that a systemic dose of corticosterone blocked the depressive-like phenotype elicited by stress in female mice. Lastly, corticosterone rescued the anxiety-like phenotype and messenger RNA levels of Ppm1f in amygdala and mPFC in male mice and in mPFC of female mice. CONCLUSIONS: Taken together, our data suggest a mechanistic pathway involving PPM1F and CAMK2G in stress- and trauma-related manifestation of anxiety and depression across species.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Depressive Disorder/metabolism , Phosphoprotein Phosphatases/metabolism , Prefrontal Cortex/metabolism , Stress Disorders, Post-Traumatic/metabolism , Stress, Psychological/metabolism , Animals , Anxiety/epidemiology , Behavior, Animal/physiology , Comorbidity , Depressive Disorder/epidemiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Stress Disorders, Post-Traumatic/epidemiology , Stress, Psychological/epidemiology , Transcriptome/genetics
11.
J Steroid Biochem Mol Biol ; 174: 225-233, 2017 11.
Article in English | MEDLINE | ID: mdl-28964927

ABSTRACT

Accumulating evidence suggests that expression of aromatase, the enzyme responsible for the conversion of androgens to estrogens, is transiently upregulated in rat stroke models. It was further suggested that increased aromatase expression is linked to neuroinflammation and that it is neuroprotective in females. Our goal was to investigate aromatase upregulation in male rats subjected to experimental stroke in relationship to neuroinflammation, infarct and response to treatment with different putative neuroprotective agents. Intact male rats were subjected to transient (90min) middle cerebral artery occlusion (MCAO) and administered selfotel (N-methyl-d-aspartic acid (NMDA) receptor competitive antagonist), TPEN (a zinc chelator), a combination of the two drugs or vehicle, injected immediately after reperfusion. Animals were killed 14days after MCAO and consecutive brain sections used to measure aromatase expression, cerebral infarct volume and neuroinflammation. Quantitative immunohistochemistry (IHC) demonstrated increased brain aromatase expression in the peri-infarct area relative to contralesional area, which was partially abrogated by neuroprotective agents. There was no correlation between aromatase expression in the peri-infarct zone and infarct volume, which was reduced by neuroprotective agents. Microglial activation, measured by quantitative autoradiography, was positively correlated with infarct and inversely correlated with aromatase expression in the peri-infarct zone. Our findings indicate that focal ischemia upregulates brain aromatase in the male rat brain at 14days post surgery, which is within the time frame documented in females. However, the lack of negative correlation between aromatase expression and infarct volume and lack of positive correlation between microgliosis and aromatase do not support a major role for aromatase as a mediator of neuroprotection or a causal relationship between microglial activation and increased aromatase expression in male focal ischemia.


Subject(s)
Aromatase/metabolism , Brain/metabolism , Brain/pathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Animals , Brain/drug effects , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Humans , Isoquinolines/pharmacology , Male , Pipecolic Acids/pharmacology , Rats , Rats, Long-Evans
12.
Physiol Behav ; 177: 264-269, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28511867

ABSTRACT

Although Syrian hamsters are thought to be naturally solitary, recent evidence from our laboratory demonstrates that hamsters may actually prefer social contact. Hamsters increase their preference for a location associated with an agonistic encounter regardless of whether they have "won" or "lost". It has also been reported that social housing as well as exposure to intermittent social defeat or to a brief footshock stressor increase food intake and body mass in hamsters. By contrast, it has also been suggested that housing hamsters in social isolation causes anxiety-induced anorexia and reductions in body mass selectively in females. The purpose of this study was to determine the physiological consequences of housing hamsters in social isolation versus in social groups. Male and female hamsters were housed singly or in stable groups of 5 for 4weeks after which they were weighed and trunk blood was collected. In addition, fat pads and thymus and adrenal glands were extracted and weighed. Serum and fecal cortisol were measured using an enzyme-linked immunoassay. Housing condition had no effect on serum or fecal cortisol, but socially housed hamsters displayed modest thymus gland involution. Socially housed females weighed more than did any other group, and socially housed females and males had more fat than did socially isolated hamsters. No wounding or tissue damage occurred in grouped hamsters. Overall, these data suggest that Syrian hamsters tolerate both stable social housing and social isolation in the laboratory although social housing is associated with some alteration in stress-related and bioenergetic measures.


Subject(s)
Housing, Animal , Hydrocortisone/metabolism , Social Isolation/psychology , Stress, Psychological/metabolism , Adaptation, Psychological , Adiposity , Adrenal Glands/anatomy & histology , Animals , Body Weight , Dominance-Subordination , Female , Male , Mesocricetus , Organ Size , Sex Characteristics , Thymus Gland/anatomy & histology
13.
Psychoneuroendocrinology ; 75: 100-109, 2017 01.
Article in English | MEDLINE | ID: mdl-27810703

ABSTRACT

Histone acetylation has emerged as a critical factor regulating learning and memory both during and after exposure to stressful stimuli. There are drugs that we now know affect histone acetylation that are already in use in clinical populations. The current study uses these drugs to examine the consequences of acutely increasing or decreasing histone acetylation during exposure to social stress. Using an acute model of social defeat in Syrian hamsters, we systemically and site-specifically administered drugs that alter histone acetylation and measured subsequent behavior and immediate-early gene activity. We found that systemic administration of a histone deacetylase inhibitor enhances social stress-induced behavioral responses in males and females. We also found that systemic administration completely blocks defeat-induced neuronal activation, as measured by Fos-immunoreactivity, in the infralimbic cortex, but not in the amygdala, after a mild social defeat stressor. Lastly, we demonstrated that site-specific administration of histone deacetylase inhibitors in the infralimbic region of the prefrontal cortex, but not in the basolateral amygdala, mimics the systemic effect. Conversely, decreasing acetylation by inhibiting histone acetyltransferases in the infralimbic cortex reduces behavioral responses to defeat. This is the first demonstration that acute pharmacological manipulation of histone acetylation during social defeat alters subsequent behavioral responses in both males and females. These results reveal that even systemic administration of drugs that alter histone acetylation can significantly alter behavioral responses to social stress and highlight the importance of the infralimbic cortex in mediating this effect.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Behavior, Animal/drug effects , Histone Deacetylase Inhibitors/pharmacology , Prefrontal Cortex/drug effects , Social Behavior , Stress, Psychological/physiopathology , Animals , Cricetinae , Female , Male , Mesocricetus
14.
Nat Commun ; 7: 13149, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27767183

ABSTRACT

Molecular characterization of neuron populations, particularly those controlling threat responses, is essential for understanding the cellular basis of behaviour and identifying pharmacological agents acting selectively on fear-controlling circuitry. Here we demonstrate a comprehensive workflow for identification of pharmacologically tractable markers of behaviourally characterized cell populations. Thy1-eNpHR-, Thy1-Cre- and Thy1-eYFP-labelled neurons of the BLA consistently act as fear inhibiting or 'Fear-Off' neurons during behaviour. We use cell-type-specific optogenetics and chemogenetics (DREADDs) to modulate activity in this population during behaviour to block or enhance fear extinction. Dissociated Thy1-eYFP neurons are isolated using FACS. RNA sequencing identifies genes strongly upregulated in RNA of this population, including Ntsr2, Dkk3, Rspo2 and Wnt7a. Pharmacological manipulation of neurotensin receptor 2 confirms behavioural effects observed in optogenetic and chemogenetic experiments. These experiments identify and validate Ntsr2-expressing neurons within the BLA, as a putative 'Fear-Off' population.


Subject(s)
Basolateral Nuclear Complex/metabolism , Extinction, Psychological/physiology , Fear/physiology , Memory/physiology , Thy-1 Antigens/genetics , Adaptor Proteins, Signal Transducing , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/drug effects , Clozapine/analogs & derivatives , Clozapine/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Gene Expression Regulation , Genes, Reporter , Injections, Intraventricular , Integrases/genetics , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Memory/drug effects , Mice , Oligopeptides/pharmacology , Optogenetics/methods , Piperidines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sequence Analysis, RNA , Stereotaxic Techniques , Thrombospondins/genetics , Thrombospondins/metabolism , Thy-1 Antigens/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
15.
Autism ; 19(5): 622-36, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25096930

ABSTRACT

Social communication impairments are a core deficit in autism spectrum disorder. Social communication deficit is also an early indicator of autism spectrum disorder and a factor in long-term outcomes. Thus, this symptom domain represents a critical treatment target. Identifying reliable and valid outcome measures for social communication across a range of treatment approaches is essential. Autism Speaks engaged a panel of experts to evaluate the readiness of available measures of social communication for use as outcome measures in clinical trials. The panel held monthly conference calls and two face-to-face meetings over 14 months. Key criteria used to evaluate measures included the relevance to the clinical target, coverage of the symptom domain, and psychometric properties (validity and reliability, as well as evidence of sensitivity to change). In all, 38 measures were evaluated and 6 measures were considered appropriate for use, with some limitations. This report discusses the relative strengths and weaknesses of existing social communication measures for use in clinical trials and identifies specific areas in need of further development.


Subject(s)
Autism Spectrum Disorder/psychology , Communication , Outcome Assessment, Health Care , Psychometrics/instrumentation , Social Perception , Social Skills , Autism Spectrum Disorder/therapy , Humans , Reproducibility of Results , Social Behavior , Treatment Outcome
16.
Neuron ; 84(3): 554-63, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25442934

ABSTRACT

Several large pharmaceutical companies have selectively downsized their neuroscience research divisions, reflecting a growing view that developing drugs to treat brain diseases is more difficult and often more time-consuming and expensive than developing drugs for other therapeutic areas, and thus represents a weak area for investment. These withdrawals reduce global neuroscience translational capabilities and pose a serious challenge to society's interests in ameliorating the impact of nervous system diseases. While the path forward ultimately lies in improving understandings of disease mechanisms, many promising therapeutic approaches have already been identified, and rebalancing the underlying risk/reward calculus could help keep companies engaged in making CNS drugs. One way to do this that would not require upfront funding is to change the policies that regulate market returns for the most-needed breakthrough drugs. The broader neuroscience community including clinicians and patients should convene to develop and advocate for such policy changes.


Subject(s)
Central Nervous System Agents/therapeutic use , Health Policy , Health Services Needs and Demand , Motivation , Nervous System Diseases/drug therapy , Animals , Health Policy/economics , Health Policy/trends , Humans
17.
Prog Mol Biol Transl Sci ; 122: 169-92, 2014.
Article in English | MEDLINE | ID: mdl-24484701

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its single transmembrane receptor, tropomysin-related kinase B (TrkB), are essential for adult synaptic plasticity and the formation of memories. However, there are regional and task-dependent differences underlying differential mechanisms of BDNF-TrkB function in the formation of these memories. Additionally, the BDNF pathway has been implicated in several psychiatric disorders including posttraumatic stress disorder, phobia, and panic disorder. Gaining a better understanding of this pathway and the neurobiology of memory through fundamental research may be helpful to identify effective prevention and treatment approaches both for diseases of memory deficit as well as in cases of enhanced aversive memory, such as in anxiety disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation , Memory/physiology , Mental Disorders/metabolism , Mental Disorders/pathology , Neuronal Plasticity/physiology , Receptor, trkB/metabolism , Adult , Brain-Derived Neurotrophic Factor/genetics , Humans , Mental Disorders/genetics , Receptor, trkB/genetics , Signal Transduction
18.
Biol Psychiatry ; 75(11): 864-72, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24094510

ABSTRACT

BACKGROUND: The current effective treatment options for posttraumatic stress disorder (PTSD) are limited, and therefore the need to explore new treatment strategies is critical. Pharmacological inhibition of the renin-angiotensin system is a common approach to treat hypertension, and emerging evidence highlights the importance of this pathway in stress and anxiety. A recent clinical study from our laboratory provides evidence supporting a role for the renin-angiotensin system in the regulation of the stress response in patients diagnosed with PTSD. METHODS: With an animal model of PTSD and the selective angiotensin receptor type 1 (AT1) antagonist losartan, we investigated the acute and long-term effects of AT1 receptor inhibition on fear memory and baseline anxiety. After losartan treatment, we performed classical Pavlovian fear conditioning pairing auditory cues with footshocks and examined extinction behavior, gene expression changes in the brain, as well as neuroendocrine and cardiovascular responses. RESULTS: After cued fear conditioning, both acute and 2-week administration of losartan enhanced the consolidation of extinction memory but had no effect on fear acquisition, baseline anxiety, blood pressure, and neuroendocrine stress measures. Gene expression changes in the brain were also altered in mice treated with losartan for 2 weeks, in particular reduced amygdala AT1 receptor and bed nucleus of the stria terminalis c-Fos messenger RNA levels. CONCLUSIONS: These data suggest that AT1 receptor antagonism enhances the extinction of fear memory and therefore might be a beneficial therapy for PTSD patients who have impairments in extinction of aversive memories.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Losartan/pharmacology , Memory/drug effects , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Disease Models, Animal , Losartan/therapeutic use , Male , Mice , Mice, Inbred C57BL , Stress Disorders, Post-Traumatic/drug therapy
19.
J Neurosci ; 33(25): 10396-404, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785152

ABSTRACT

Research has identified distinct neuronal circuits within the basolateral amygdala (BLA) that differentially mediate fear expression versus inhibition; however, molecular markers of these populations remain unknown. Here we examine whether optogenetic activation of a cellular subpopulation, which may correlate with the physiologically identified extinction neurons in the BLA, would differentially support fear conditioning versus fear inhibition/extinction. We first molecularly characterized Thy1-channelrhodopsin-2 (Thy1-ChR2-EYFP)-expressing neurons as a subpopulation of glutamatergic pyramidal neurons within the BLA. Optogenetic stimulation of these neurons inhibited a subpopulation of medial central amygdala neurons and shunted excitation from the lateral amygdala. Brief activation of these neurons during fear training disrupted later fear memory in male mice. Optogenetic activation during unreinforced stimulus exposure enhanced extinction retention, but had no effect on fear expression, locomotion, or open-field behavior. Together, these data suggest that the Thy1-expressing subpopulation of BLA pyramidal neurons provide an important molecular and pharmacological target for inhibiting fear and enhancing extinction and for furthering our understanding of the molecular mechanisms of fear processing.


Subject(s)
Amygdala/physiology , Fear/physiology , Inhibition, Psychological , Neurons/physiology , Thy-1 Antigens/physiology , Amygdala/cytology , Animals , Channelrhodopsins , Conditioning, Psychological , Cues , Extinction, Psychological , Glutamic Acid/physiology , Immunohistochemistry , Lasers , Locomotion/physiology , Mice , Motor Activity/physiology , Patch-Clamp Techniques , Photic Stimulation , Thy-1 Antigens/genetics
20.
Front Physiol ; 4: 112, 2013.
Article in English | MEDLINE | ID: mdl-23720635

ABSTRACT

Huntington's disease (HD) is a neurodegenerative condition characterized by severe neuronal loss in the cortex and striatum that leads to motor and behavioral deficits. Excitotoxicity is thought to be involved in HD and several studies have indicated that NMDA receptor (NMDAR) overactivation can play a role in the selective neuronal loss found in HD. Interestingly, a small subset of striatal neurons (less than 1% of the overall population) is found to be spared in post-mortem HD brains. These neurons are medium-sized aspiny interneurons that highly express the neuronal isoform of nitric oxide synthase (nNOS). Intriguingly, neurons expressing large amounts of nNOS [hereafter indicated as nNOS(+) neurons] show reduced vulnerability to NMDAR-mediated excitotoxicity. Mechanisms underlying this reduced vulnerability are still largely unknown and may shed some light on pathogenic mechanisms involved in HD. One untested possibility is that nNOS(+) neurons possess fewer or less functioning NMDARs. Employing single cell calcium imaging we challenged this hypothesis and found that cultured striatal nNOS(+) neurons show NMDAR-evoked responses that are identical to the ones observed in the overall population of neurons that express lower levels of nNOS [nNOS(-) neurons]. NMDAR-dependent deregulation of intraneuronal Ca(2+) is known to generate high levels of reactive oxygen species of mitochondrial origin (mt-ROS), a crucial step in the excitotoxic cascade. With confocal imaging and dihydrorhodamine (DHR; a ROS-sensitive probe) we compared mt-ROS levels generated by NMDAR activation in nNOS(+) and (-) cultured striatal neurons. DHR experiments revealed that nNOS(+) neurons failed to produce significant amounts of mt-ROS in response to NMDA exposure, thereby providing a potential mechanism for their reduced vulnerability to excitotoxicity and decreased vulnerability in HD.

SELECTION OF CITATIONS
SEARCH DETAIL
...