Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
2.
Tissue Barriers ; : 2292461, 2023 Dec 14.
Article En | MEDLINE | ID: mdl-38095516

BACKGROUND: A number of peptide incretin receptor agonists (IRAs) show promise as therapeutics for Alzheimer's disease (AD) and Parkinson's disease (PD). Transport across the blood-brain barrier (BBB) is one way for IRAs to act directly within the brain. To determine which IRAs are high priority candidates for treating these disorders, we have studied their brain uptake pharmacokinetics. METHODS: We quantitatively measure the ability of four IRAs to cross the BBB. We injected adult male CD-1 mice intravenously with 125I- or 14C-labeled albiglutide, dulaglutide, DA5-CH, or tirzepatide and used multiple-time regression analyses to measure brain kinetics up to 1 hour. For those IRAs failing to enter the brain 1 h after intravenous injection, we also investigated their ability to enter over a longer time frame (i.e., 6 h). RESULTS: Albiglutide and dulaglutide had the fastest brain uptake rates within 1 hour. DA5-CH appears to enter the brain rapidly, reaching equilibrium quickly. Tirzepatide does not appear to cross the BBB within 1 h after iv injection but like albumin, did so slowly over 6 h, presumably via the extracellular pathways. CONCLUSIONS: We find that IRAs can cross the BBB by two separate processes; one that is fast and one that is slow. Three of the four IRAs investigated here have fast rates of transport and should be taken into consideration for testing as AD and PD therapeutics as they would have the ability to act quickly and directly on the brain as a whole.

4.
Alzheimers Dement ; 18(3): 478-497, 2022 03.
Article En | MEDLINE | ID: mdl-34647685

Medications for type 2 diabetes (T2DM) offer a promising path for discovery and development of effective interventions for dementia syndromes. A common feature of dementia syndromes is an energy failure due to reduced energy supply to neurons and is associated with synaptic loss and results in cognitive decline and behavioral changes. Among diabetes medications, glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) promote protective effects on vascular, microglial, and neuronal functions. In this review, we present evidence from animal models, imaging studies, and clinical trials that support developing GLP-1 RAs for dementia syndromes. The review examines how changes in brain energy metabolism differ in conditions of insulin resistance and T2DM from dementia and underscores the challenges that arise from the heterogeneity of dementia syndromes. The development of GLP-1 RAs as dementia therapies requires a deeper understanding of the regional changes in brain energy homeostasis guided by novel imaging biomarkers.


Dementia , Diabetes Mellitus, Type 2 , Animals , Brain/metabolism , Dementia/drug therapy , Dementia/metabolism , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Syndrome
5.
Biochem Pharmacol ; 180: 114187, 2020 10.
Article En | MEDLINE | ID: mdl-32755557

Among the more promising treatments proposed for Alzheimer's disease (AD) and Parkinson's disease (PD) are those reducing brain insulin resistance. The antidiabetics in the class of incretin receptor agonists (IRAs) reduce symptoms and brain pathology in animal models of AD and PD, as well as glucose utilization in AD cases and clinical symptoms in PD cases after their systemic administration. At least 9 different IRAs are showing promise as AD and PD therapeutics, but we still lack quantitative data on their relative ability to cross the blood-brain barrier (BBB) reaching the brain parenchyma. We consequently compared brain uptake pharmacokinetics of intravenous 125I-labeled IRAs in adult CD-1 mice over the course of 60 min. We tested single IRAs (exendin-4, liraglutide, lixisenatide, and semaglutide), which bind receptors for one incretin (glucagon-like peptide-1 [GLP-1]), and dual IRAs, which bind receptors for two incretins (GLP-1 and glucose-dependent insulinotropic polypeptide [GIP]), including unbranched, acylated, PEGylated, or C-terminally modified forms (Finan/Ma Peptides 17, 18, and 20 and Hölscher peptides DA3-CH and DA-JC4). The non-acylated and non-PEGylated IRAs (exendin-4, lixisenatide, Peptide 17, DA3-CH and DA-JC4) had significant rates of blood-to-brain influx (Ki), but the acylated IRAs (liraglutide, semaglutide, and Peptide 18) did not measurably cross the BBB. The brain influx of the non-acylated, non-PEGylated IRAs were not saturable up to 1 µg of these drugs and was most likely mediated by adsorptive transcytosis across brain endothelial cells, as observed for exendin-4. Of the non-acylated, non-PEGylated IRAs tested, exendin-4 and DA-JC4 were best able to cross the BBB based on their rate of brain influx, percentage reaching the brain that accumulated in brain parenchyma, and percentage of the systemic dose taken up per gram of brain tissue. Exendin-4 and DA-JC4 thus merit special attention as IRAs well-suited to enter the central nervous system (CNS), thus reaching areas pathologic in AD and PD.


Alzheimer Disease/metabolism , Brain/metabolism , Incretins/agonists , Incretins/metabolism , Parkinson Disease/metabolism , Alzheimer Disease/drug therapy , Amino Acid Sequence , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Exenatide/agonists , Exenatide/genetics , Exenatide/metabolism , Humans , Incretins/genetics , Male , Mice , Parkinson Disease/drug therapy
7.
J Neurol Sci ; 388: 97-102, 2018 05 15.
Article En | MEDLINE | ID: mdl-29627040

OBJECTIVES: To examine the relationship between homeostatic model of insulin resistance (HOMA-IR) and cognitive test performance among population≥60years in a national database. HYPOTHESIS: Higher insulin resistance is associated with lower cognitive test performance score in the population≥60years. PARTICIPANTS: We analyzed data from the National Health and Nutrition Examination Survey (NHANES) 1999-2000 and 2001-2002. MEASUREMENTS: Cognitive test performance was measured by the Digit Symbol Substitution (DSS) exercise score. The main independent variable was the homeostasis model assessment of insulin resistance (HOMA-IR). We used bivariate analysis and generalized linear model adjusting for age, gender, race, education, body mass index, and systolic and diastolic blood pressures; total cholesterol, low density lipoprotein (LDL), high density lipoprotein (HDL) and triglyceride levels; and physical activity, diabetes mellitus, stroke, and congestive heart failure. STATA 14 was used to analyze the data taking into consideration the design, strata and weight. RESULTS: Of the 1028 participants, 44% were male and 85% were white. The mean age was 70.0±0.28 (SE) years. Their average HOMA-IR was 3.6±0.14 and they had a mean of 49.2±0.8 correct DSS score in the cognitive test. Adjusting for the confounding variables, HOMA-IR was associated with decline in DSS score (B=-0.30, 95% confidence interval=-0.54 and -0.05, p=0.01). The model explained 44% of the variability of the DSS score (R2=0.44). Significant predictors of decline in DSS score were age, gender, race, and education (p=0.01). CONCLUSION: Insulin resistance as measured by HOMA-IR was independently associated with lower cognitive test performance score among elderly participants aged ≥60years. Longitudinal studies are needed to test the mechanism and the causal relationship.


Cognition , Cognitive Dysfunction/epidemiology , Insulin Resistance , Aged , Cognition/physiology , Cognitive Dysfunction/physiopathology , Cross-Sectional Studies , Female , Health Surveys , Humans , Male , Middle Aged , Neuropsychological Tests , Nutrition Surveys
8.
Neuropsychopharmacology ; 42(6): 1349-1360, 2017 May.
Article En | MEDLINE | ID: mdl-27986973

Dysbindin-1, a protein that regulates aspects of early and late brain development, has been implicated in the pathobiology of schizophrenia. As the functional roles of the three major isoforms of dysbindin-1, (A, B, and C) remain unknown, we generated a novel mutant mouse, dys-1A-/-, with selective loss of dysbindin-1A and investigated schizophrenia-related phenotypes in both males and females. Loss of dysbindin-1A resulted in heightened initial exploration and disruption in subsequent habituation to a novel environment, together with heightened anxiety-related behavior in a stressful environment. Loss of dysbindin-1A was not associated with disruption of either long-term (olfactory) memory or spontaneous alternation behavior. However, dys-1A-/- showed enhancement in delay-dependent working memory under high levels of interference relative to controls, ie, impairment in sensitivity to the disruptive effect of such interference. These findings in dys-1A-/- provide the first evidence for differential functional roles for dysbindin-1A vs dysbindin-1C isoforms among phenotypes relevant to the pathobiology of schizophrenia. Future studies should investigate putative sex differences in these phenotypic effects.


Attention/physiology , Behavior, Animal/physiology , Dysbindin/physiology , Memory, Short-Term/physiology , Schizophrenia/physiopathology , Animals , Disease Models, Animal , Female , Male , Memory, Long-Term/physiology , Mice , Mice, Transgenic , Olfactory Perception/physiology , Phenotype , Protein Isoforms
9.
Mol Neurobiol ; 54(3): 1699-1709, 2017 04.
Article En | MEDLINE | ID: mdl-26873854

Schizophrenia is a chronic debilitating neuropsychiatric disorder that affects about 1 % of the population. Dystrobrevin-binding protein 1 (DTNBP1 or dysbindin) is one of the Research Domain Constructs (RDoC) associated with cognition and is significantly reduced in the brain of schizophrenia patients. To further understand the molecular underpinnings of pathogenesis of schizophrenia, we have performed microarray analyses of the hippocampi from dysbindin knockout mice, and found that genes involved in the lipogenic pathway are suppressed. Moreover, we discovered that maturation of a master transcriptional regulator for lipid synthesis, sterol regulatory element binding protein-1 (SREBP1) is induced by neuronal activity, and is required for induction of the immediate early gene ARC (activity-regulated cytoskeleton-associated protein), necessary for synaptic plasticity and memory. We found that nuclear SREBP1 is dramatically reduced in dysbindin-1 knockout mice and postmortem brain tissues from human patients with schizophrenia. Furthermore, activity-dependent maturation of SREBP1 as well as ARC expression were attenuated in dysbindin-1 knockout mice, and these deficits were restored by an atypical antipsychotic drug, clozapine. Together, results indicate an important role of dysbindin-1 in neuronal activity induced SREBP1 and ARC, which could be related to cognitive deficits in schizophrenia.


Cognitive Dysfunction/metabolism , Dysbindin/deficiency , Neurons/metabolism , Schizophrenia/metabolism , Sterol Regulatory Element Binding Protein 1/biosynthesis , Aged , Aged, 80 and over , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/psychology , Dysbindin/genetics , Female , Gene Regulatory Networks/physiology , Humans , Longitudinal Studies , Male , Mice , Mice, Knockout , Organ Culture Techniques , PC12 Cells , Random Allocation , Rats , Schizophrenia/genetics , Schizophrenic Psychology , Sterol Regulatory Element Binding Protein 1/genetics
10.
Exp Neurol ; 288: 176-186, 2017 Feb.
Article En | MEDLINE | ID: mdl-27845037

Several single incretin receptor agonists that are approved for the treatment of type 2 diabetes mellitus (T2DM) have been shown to be neuroprotective in cell and animal models of neurodegeneration. Recently, a synthetic dual incretin receptor agonist, nicknamed "twincretin," was shown to improve upon the metabolic benefits of single receptor agonists in mouse and monkey models of T2DM. In the current study, the neuroprotective effects of twincretin are probed in cell and mouse models of mild traumatic brain injury (mTBI), a prevalent cause of neurodegeneration in toddlers, teenagers and the elderly. Twincretin is herein shown to have activity at two different receptors, dose-dependently increase levels of intermediates in the neurotrophic CREB pathway and enhance viability of human neuroblastoma cells exposed to toxic concentrations of glutamate and hydrogen peroxide, insults mimicking the inflammatory conditions in the brain post-mTBI. Additionally, twincretin is shown to improve upon the neurotrophic effects of single incretin receptor agonists in these same cells. Finally, a clinically translatable dose of twincretin, when administered post-mTBI, is shown to fully restore the visual and spatial memory deficits induced by mTBI, as evaluated in a mouse model of weight drop close head injury. These results establish twincretin as a novel neuroprotective agent and suggest that it may improve upon the effects of the single incretin receptor agonists via dual agonism.


Brain Injuries, Traumatic/drug therapy , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Incretins/therapeutic use , Neuroprotective Agents/therapeutic use , Receptors, Gastrointestinal Hormone/metabolism , Animals , Body Temperature/drug effects , Brain Injuries, Traumatic/complications , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/prevention & control , Mice , Mice, Inbred ICR , Neuroblastoma/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Gastrointestinal Hormone/agonists , Recognition, Psychology/drug effects , Signal Transduction/drug effects
11.
Nat Neurosci ; 19(11): 1442-1453, 2016 11.
Article En | MEDLINE | ID: mdl-27668389

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, ∼20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.


Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Multifactorial Inheritance/genetics , Schizophrenia/genetics , Brain/metabolism , Female , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide , Risk
12.
Neurochem Int ; 79: 65-9, 2014 Dec.
Article En | MEDLINE | ID: mdl-25445987

Oxidative stress resulting from the generation of reactive oxygen species has been proposed as an etiological factor in schizophrenia. The present study tests the hypothesis that oxidative stress can affect levels of dysbindin-1A, encoded by Dtnbp1, a genetic risk factor for schizophrenia, via its PEST domain. In vitro studies on SH-SY5Y cells indicate that oxidative stress triggers proteasomal degradation of dysbindin-1A, and that this requires interactions with its PEST domain, which may be a TRIM32 target. We specifically found (a) that oxidative stress induced in SH-SY5Y cells by 500 µM hydrogen peroxide reduced levels of full-length dysbindin-1, but did not reduce levels of that protein lacking its PEST domain and (b) that levels of full-length dysbindin-1, but not dysbindin-1 lacking its PEST domain, were higher in cells treated with the proteasome inhibitor MG132. Oxidative stress thus emerges as the first known cellular factor regulating dysbindin-1 isoforms with PEST domains. These findings are consistent with the previously noted fact that phosphorylation of PEST domains often marks proteins for proteasomal degradation, and raises the possibility that treatments reducing oxidative stress in the brain, especially during development, may lower schizophrenia risk.


Dystrophin-Associated Proteins/metabolism , Oxidative Stress/drug effects , Animals , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dysbindin , Leupeptins/pharmacology , Phosphorylation , Proteasome Endopeptidase Complex/drug effects , Rats , Reactive Oxygen Species/metabolism , Schizophrenia/metabolism
13.
Expert Rev Neurother ; 14(6): 583-8, 2014 Jun.
Article En | MEDLINE | ID: mdl-24852225

Delirium is 1.5 to 4.1 times as likely in intensive care unit patients when they are mechanically ventilated. While progress in treatment has occurred, delirium is still a major problem in mechanically ventilated patients. Based on studies of a murine mechanical ventilation model, we summarize evidence here for a novel mechanism by which such ventilation can quickly initiate brain damage likely to cause cognitive deficits expressed as delirium. That mechanism consists of aberrant vagal sensory input driving sustained dopamine D2 receptor (D2R) signaling in the hippocampal formation, which induces apoptosis in that brain area within 90 min without causing hypoxia, oxidative stress, or inflammatory responses. This argues for minimizing the duration and tidal volumes of mechanical ventilation and for more effectively reducing sustained D2R signaling than achieved with haloperidol alone. The latter might be accomplished by reducing D2R cell surface expression and D2R-mediated Akt inhibition by elevating protein expression of dysbindin-1C.


Brain Injuries/etiology , Delirium/etiology , Delirium/therapy , Respiration, Artificial/adverse effects , Humans
14.
J Biol Chem ; 289(20): 14291-300, 2014 May 16.
Article En | MEDLINE | ID: mdl-24713699

Post-mortem analysis has revealed reduced levels of the protein dysbindin in the brains of those suffering from the neurodevelopmental disorder schizophrenia. Consequently, mechanisms controlling the cellular levels of dysbindin and its interacting partners may participate in neurodevelopmental processes impaired in that disorder. To address this question, we studied loss of function mutations in the genes encoding dysbindin and its interacting BLOC-1 subunits. We focused on BLOC-1 mutants affecting synapse composition and function in addition to their established systemic pigmentation, hematological, and lung phenotypes. We tested phenotypic homogeneity and gene dosage effects in the mouse null alleles muted (Bloc1s5(mu/mu)) and dysbindin (Bloc1s8(sdy/sdy)). Transcripts of NMDA receptor subunits and GABAergic interneuron markers, as well as expression of BLOC-1 subunit gene products, were affected differently in the brains of Bloc1s5(mu/mu) and Bloc1s8(sdy/sdy) mice. Unlike Bloc1s8(sdy/sdy), elimination of one or two copies of Bloc1s5 generated indistinguishable pallidin transcript phenotypes. We conclude that monogenic mutations abrogating the expression of a protein complex subunit differentially affect the expression of other complex transcripts and polypeptides as well as their downstream effectors. We propose that the genetic disruption of different subunits of protein complexes and combinations thereof diversifies phenotypic presentation of pathway deficiencies, contributing to the wide phenotypic spectrum and complexity of neurodevelopmental disorders.


Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutant Proteins/metabolism , Mutation , Phenotype , Protein Subunits/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Animals , Dysbindin , Dystrophin-Associated Proteins , Hippocampus/metabolism , Humans , Mice , Mutant Proteins/genetics , Neurotransmitter Agents/metabolism , Pigmentation/genetics , Protein Subunits/genetics , Schizophrenia/etiology , Schizophrenia/genetics , Schizophrenia/metabolism , Transcription, Genetic/genetics , gamma-Aminobutyric Acid/metabolism
15.
Neurodegener Dis Manag ; 4(1): 31-40, 2014.
Article En | MEDLINE | ID: mdl-24640977

The prevalence of Alzheimer's disease is increasing rapidly in the absence of truly effective therapies. A promising strategy for developing such therapies is the treatment of brain insulin resistance, a common and early feature of Alzheimer's disease, closely tied to cognitive decline and capable of promoting many biological abnormalities in the disorder. The proximal cause of brain insulin resistance appears to be neuronal elevation in the serine phosphorylation of IRS-1, most likely due to amyloid-ß-triggered microglial release of proinflammatory cytokines. Preclinically, the first line of defense is behavior-lowering peripheral insulin resistance (e.g., physical exercise and a Mediterranean diet supplemented with foods rich in flavonoids, curcumin and ω-3 fatty acids). More potent remediation is required, however, at clinical stages. Fortunately, the US FDA-approved antidiabetics exenatide (Byetta; Amylin Pharmaceuticals, Inc., CA, USA) and liraglutide (Victoza; Novo Nordisk A/S, Bagsvaerd, Denmark) are showing much promise in reducing Alzheimer's disease pathology and in restoring normal brain insulin responsiveness and cognitive function.


Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Brain/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , Aging/drug effects , Aging/physiology , Alzheimer Disease/pathology , Animals , Brain/pathology , Brain/physiopathology , Humans , Hypoglycemic Agents/pharmacology , Models, Neurological , Nootropic Agents/pharmacology , Nootropic Agents/therapeutic use
16.
Alzheimers Dement ; 10(1 Suppl): S12-25, 2014 Feb.
Article En | MEDLINE | ID: mdl-24529520

Alzheimer's disease (AD) is an age-related neurodegenerative disease leading over the course of decades to the most common form of dementia. Many of its pathologic features and cognitive deficits may be due in part to brain insulin resistance recently demonstrated in the insulin receptor→insulin receptor substrate-1 (IRS-1) signaling pathway. The proximal cause of such resistance in AD dementia and amnestic mild cognitive impairment (aMCI) appears to be serine inhibition of IRS-1, a phenomenon likely due to microglial release of inflammatory cytokines triggered by oligomeric Aß. Studies on animal models of AD and on human brain tissue from MCI cases at high risk of AD dementia have shown that brain insulin resistance and many other pathologic features and symptoms of AD may be greatly reduced or even reversed by treatment with FDA-approved glucagon-like peptide-1 (GLP-1) analogs such as liraglutide (Victoza). These findings call attention to the need for further basic, translational, and clinical studies on GLP-1 analogs as promising AD therapeutics.


Alzheimer Disease , Brain/drug effects , Brain/metabolism , Glucagon-Like Peptide 1/therapeutic use , Insulin Resistance , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Humans
17.
Hippocampus ; 24(2): 204-13, 2014 Feb.
Article En | MEDLINE | ID: mdl-24446171

Genetic variants in DTNBP1 encoding the protein dysbindin-1 have often been associated with schizophrenia and with the cognitive deficits prominent in that disorder. Because impaired function of the hippocampus is thought to play a role in these memory deficits and because NMDAR-dependent synaptic plasticity in this region is a proposed biological substrate for some hippocampal-dependent memory functions in schizophrenia, we hypothesized that reduced dysbindin-1 expression would lead to impairments in NMDAR-dependent synaptic plasticity and in contextual fear conditioning. Acute slices from male mice carrying 0, 1, or 2 null mutant alleles of the Dtnbp1 gene were prepared, and field recordings from the CA1 striatum radiatum were obtained before and after tetanization of Schaffer collaterals of CA3 pyramidal cells. Mice homozygous for the null mutation in Dtnbp1 exhibited significantly reduced NMDAR-dependent synaptic potentiation compared to wild type mice, an effect that could be rescued by bath application of the NMDA receptor coagonist glycine (10 µM). Behavioral testing in adult mice revealed deficits in hippocampal memory processes. Homozygous null mice exhibited lower conditional freezing, without a change in the response to shock itself, indicative of a learning and memory deficit. Taken together, these results indicate that a loss of dysbindin-1 impairs hippocampal plasticity which may, in part, explain the role dysbindin-1 plays in the cognitive impairments of schizophrenia.


Carrier Proteins/metabolism , Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/genetics , Analysis of Variance , Animals , Biophysics , Carrier Proteins/genetics , Dysbindin , Dystrophin-Associated Proteins , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Freezing Reaction, Cataleptic/physiology , Hippocampus/cytology , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Synapses/physiology
18.
Genome Biol ; 14(8): R94, 2013 Aug 30.
Article En | MEDLINE | ID: mdl-24000956

The behavior of epigenetic mechanisms in the brain is obscured by tissue heterogeneity and disease-related histological changes. Not accounting for these confounders leads to biased results. We develop a statistical methodology that estimates and adjusts for celltype composition by decomposing neuronal and non-neuronal differential signal. This method provides a conceptual framework for deconvolving heterogeneous epigenetic data from postmortem brain studies. We apply it to find cell-specific differentially methylated regions between prefrontal cortex and hippocampus. We demonstrate the utility of the method on both Infinium 450k and CHARM data.


Algorithms , DNA Methylation , Genome, Human , Hippocampus/metabolism , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Prefrontal Cortex/metabolism , Autopsy , CpG Islands , Epigenesis, Genetic , Hippocampus/cytology , Humans , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Organ Specificity , Prefrontal Cortex/cytology
19.
Am J Respir Crit Care Med ; 188(6): 693-702, 2013 Sep 15.
Article En | MEDLINE | ID: mdl-23962032

RATIONALE: Critically ill patients frequently develop neuropsychological disturbances including acute delirium or memory impairment. The need for mechanical ventilation is a risk factor for these adverse events, but a mechanism that links lung stretch and brain injury has not been identified. OBJECTIVES: To identify the mechanisms that lead to brain dysfunction during mechanical ventilation. METHODS: Brains from mechanically ventilated mice were harvested, and signals of apoptosis and alterations in the Akt survival pathway were studied. These measurements were repeated in vagotomized or haloperidol-treated mice, and in animals intracerebroventricularly injected with selective dopamine-receptor blockers. Hippocampal slices were cultured and treated with micromolar concentrations of dopamine, with or without dopamine receptor blockers. Last, levels of dysbindin, a regulator of the membrane availability of dopamine receptors, were assessed in the experimental model and in brain samples from ventilated patients. MEASUREMENTS AND MAIN RESULTS: Mechanical ventilation triggers hippocampal apoptosis as a result of type 2 dopamine receptor activation in response to vagal signaling. Activation of these receptors blocks the Akt/GSK3ß prosurvival pathway and activates the apoptotic cascade, as demonstrated in vivo and in vitro. Vagotomy, systemic haloperidol, or intracerebroventricular raclopride (a type 2 dopamine receptor blocker) ameliorated this effect. Moreover, ventilation induced a concomitant change in the expression of dysbindin-1C. These results were confirmed in brain samples from ventilated patients. CONCLUSIONS: These results prove the existence of a pathogenic mechanism of lung stretch-induced hippocampal apoptosis that could explain the neurological changes in ventilated patients and may help to identify novel therapeutic approaches.


Apoptosis , Dopamine/metabolism , Hippocampus/pathology , Respiration, Artificial/adverse effects , Vagus Nerve/pathology , Ventilator-Induced Lung Injury/metabolism , Animals , Carrier Proteins/metabolism , Disease Models, Animal , Dysbindin , Dystrophin-Associated Proteins , Hippocampus/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Signal Transduction , Vagus Nerve/metabolism
20.
PLoS One ; 8(6): e65069, 2013.
Article En | MEDLINE | ID: mdl-23750231

Clinical, epidemiological, and genetic evidence suggest overlapping pathogenic mechanisms between autism spectrum disorder (ASD) and schizophrenia. We tested this hypothesis by asking if mutations in the ASD gene MECP2 which cause Rett syndrome affect the expression of genes encoding the schizophrenia risk factor dysbindin, a subunit of the biogenesis of lysosome-related organelles complex-1 (BLOC-1), and associated interacting proteins. We measured mRNA and protein levels of key components of a dysbindin interaction network by, quantitative real time PCR and quantitative immunohistochemistry in hippocampal samples of wild-type and Mecp2 mutant mice. In addition, we confirmed results by performing immunohistochemistry of normal human hippocampus and quantitative qRT-PCR of human inducible pluripotent stem cells (iPSCs)-derived human neurons from Rett syndrome patients. We defined the distribution of the BLOC-1 subunit pallidin in human and mouse hippocampus and contrasted this distribution with that of symptomatic Mecp2 mutant mice. Neurons from mutant mice and Rett syndrome patients displayed selectively reduced levels of pallidin transcript. Pallidin immunoreactivity decreased in the hippocampus of symptomatic Mecp2 mutant mice, a feature most prominent at asymmetric synapses as determined by immunoelectron microcopy. Pallidin immunoreactivity decreased concomitantly with reduced BDNF content in the hippocampus of Mecp2 mice. Similarly, BDNF content was reduced in the hippocampus of BLOC-1 deficient mice suggesting that genetic defects in BLOC-1 are upstream of the BDNF phenotype in Mecp2 deficient mice. Our results demonstrate that the ASD-related gene Mecp2 regulates the expression of components belonging to the dysbindin interactome and these molecular differences may contribute to synaptic phenotypes that characterize Mecp2 deficiencies and ASD.


Carrier Proteins/metabolism , Gene Expression Regulation , Hippocampus/cytology , Lectins/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Carrier Proteins/genetics , Computational Biology , Dysbindin , Dystrophin-Associated Proteins , Humans , Induced Pluripotent Stem Cells/cytology , Lectins/genetics , Methyl-CpG-Binding Protein 2/deficiency , Mice , Neurons/cytology , Protein Interaction Maps , RNA, Messenger/genetics , RNA, Messenger/metabolism
...