Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 54(3): 1699-1709, 2017 04.
Article in English | MEDLINE | ID: mdl-26873854

ABSTRACT

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.


Subject(s)
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
2.
Acta Neuropathol ; 128(5): 679-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25107476

ABSTRACT

Neuronal insulin signaling abnormalities have been associated with Alzheimer's disease (AD). However, the specificity of this association and its underlying mechanisms have been unclear. This study investigated the expression of abnormal serine phosphorylation of insulin receptor substrate 1 (IRS1) in 157 human brain autopsy cases that included AD, tauopathies, α-synucleinopathies, TDP-43 proteinopathies, and normal aging. IRS1-pS(616), IRS1-pS(312) and downstream target Akt-pS(473) measures were most elevated in AD but were also significantly increased in the tauopathies: Pick's disease, corticobasal degeneration and progressive supranuclear palsy. Double immunofluorescence labeling showed frequent co-expression of IRS1-pS(616) with pathologic tau in neurons and dystrophic neurites. To further investigate an association between tau and abnormal serine phosphorylation of IRS1, we examined the presence of abnormal IRS1-pS(616) expression in pathological tau-expressing transgenic mice and demonstrated that abnormal IRS1-pS(616) frequently co-localizes in tangle-bearing neurons. Conversely, we observed increased levels of hyperphosphorylated tau in the high-fat diet-fed mouse, a model of insulin resistance. These results provide confirmation and specificity that abnormal phosphorylation of IRS1 is a pathological feature of AD and other tauopathies, and provide support for an association between insulin resistance and abnormal tau as well as amyloid-ß.


Subject(s)
Alzheimer Disease/pathology , Brain/metabolism , Insulin Receptor Substrate Proteins/metabolism , Serine/metabolism , Tauopathies/pathology , Age Factors , Aged , Aged, 80 and over , Analysis of Variance , Animals , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Phosphorylation/genetics , TDP-43 Proteinopathies/pathology , alpha-Synuclein/metabolism , tau Proteins/genetics , tau Proteins/metabolism
3.
Neurobiol Dis ; 67: 79-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24686304

ABSTRACT

Insulin resistance and other features of the metabolic syndrome are increasingly recognized for their effects on cognitive health. To ascertain mechanisms by which this occurs, we fed mice a very high fat diet (60% kcal by fat) for 17days or a moderate high fat diet (HFD, 45% kcal by fat) for 8weeks and examined changes in brain insulin signaling responses, hippocampal synaptodendritic protein expression, and spatial working memory. Compared to normal control diet mice, cerebral cortex tissues of HFD mice were insulin-resistant as evidenced by failed activation of Akt, S6 and GSK3ß with ex-vivo insulin stimulation. Importantly, we found that expression of brain IPMK, which is necessary for mTOR/Akt signaling, remained decreased in HFD mice upon activation of AMPK. HFD mouse hippocampus exhibited increased expression of serine-phosphorylated insulin receptor substrate 1 (IRS1-pS(616)), a marker of insulin resistance, as well as decreased expression of PSD-95, a scaffolding protein enriched in post-synaptic densities, and synaptopodin, an actin-associated protein enriched in spine apparatuses. Spatial working memory was impaired as assessed by decreased spontaneous alternation in a T-maze. These findings indicate that HFD is associated with telencephalic insulin resistance and deleterious effects on synaptic integrity and cognitive behaviors.


Subject(s)
Brain/metabolism , Dendrites/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance , Spatial Memory/physiology , Synapses/metabolism , Animals , Hyperglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Rats , Signal Transduction
4.
J Clin Invest ; 122(4): 1339-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22476196

ABSTRACT

Defective brain insulin signaling has been suggested to contribute to the cognitive deficits in patients with Alzheimer's disease (AD). Although a connection between AD and diabetes has been suggested, a major unknown is the mechanism(s) by which insulin resistance in the brain arises in individuals with AD. Here, we show that serine phosphorylation of IRS-1 (IRS-1pSer) is common to both diseases. Brain tissue from humans with AD had elevated levels of IRS-1pSer and activated JNK, analogous to what occurs in peripheral tissue in patients with diabetes. We found that amyloid-ß peptide (Aß) oligomers, synaptotoxins that accumulate in the brains of AD patients, activated the JNK/TNF-α pathway, induced IRS-1 phosphorylation at multiple serine residues, and inhibited physiological IRS-1pTyr in mature cultured hippocampal neurons. Impaired IRS-1 signaling was also present in the hippocampi of Tg mice with a brain condition that models AD. Importantly, intracerebroventricular injection of Aß oligomers triggered hippocampal IRS-1pSer and JNK activation in cynomolgus monkeys. The oligomer-induced neuronal pathologies observed in vitro, including impaired axonal transport, were prevented by exposure to exendin-4 (exenatide), an anti-diabetes agent. In Tg mice, exendin-4 decreased levels of hippocampal IRS-1pSer and activated JNK and improved behavioral measures of cognition. By establishing molecular links between the dysregulated insulin signaling in AD and diabetes, our results open avenues for the investigation of new therapeutics in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Insulin/physiology , Peptides/therapeutic use , Venoms/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Animals , Antibodies, Monoclonal/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Exenatide , Female , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hypoglycemic Agents/pharmacology , Infliximab , MAP Kinase Signaling System/drug effects , Macaca fascicularis , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurons/drug effects , Neurons/metabolism , Peptides/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Rats , Venoms/pharmacology
5.
J Clin Invest ; 122(4): 1316-38, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22476197

ABSTRACT

While a potential causal factor in Alzheimer's disease (AD), brain insulin resistance has not been demonstrated directly in that disorder. We provide such a demonstration here by showing that the hippocampal formation (HF) and, to a lesser degree, the cerebellar cortex in AD cases without diabetes exhibit markedly reduced responses to insulin signaling in the IR→IRS-1→PI3K signaling pathway with greatly reduced responses to IGF-1 in the IGF-1R→IRS-2→PI3K signaling pathway. Reduced insulin responses were maximal at the level of IRS-1 and were consistently associated with basal elevations in IRS-1 phosphorylated at serine 616 (IRS-1 pS6¹6) and IRS-1 pS6³6/6³9. In the HF, these candidate biomarkers of brain insulin resistance increased commonly and progressively from normal cases to mild cognitively impaired cases to AD cases regardless of diabetes or APOE ε4 status. Levels of IRS-1 pS6¹6 and IRS-1 pS6³6/6³9 and their activated kinases correlated positively with those of oligomeric Aß plaques and were negatively associated with episodic and working memory, even after adjusting for Aß plaques, neurofibrillary tangles, and APOE ε4. Brain insulin resistance thus appears to be an early and common feature of AD, a phenomenon accompanied by IGF-1 resistance and closely associated with IRS-1 dysfunction potentially triggered by Aß oligomers and yet promoting cognitive decline independent of classic AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Cognition Disorders/etiology , Insulin Receptor Substrate Proteins/physiology , Insulin Resistance , Insulin-Like Growth Factor I/pharmacology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Apolipoprotein E4/genetics , Brain/drug effects , Brain/pathology , Cerebellar Cortex/metabolism , Cerebellar Cortex/pathology , Cognition Disorders/metabolism , Diabetes Complications/complications , Drug Resistance , Female , Glucose/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins/chemistry , Insulin Receptor Substrate Proteins/genetics , Insulin-Like Growth Factor I/physiology , Male , Middle Aged , Phosphorylation , Phosphoserine/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , Signal Transduction
6.
Hippocampus ; 22(2): 230-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21049487

ABSTRACT

Genetic studies have associated deficient function of the serine/threonine kinase Akt1 with schizophrenia. This disorder is associated with developmental, structural, and functional abnormalities of the hippocampus that could be traced to abnormal Akt1 function. To establish a closer connection between Akt1 and hippocampal function, mice with a selective deletion of Akt1 (Akt1(-/-) mice) were examined for physiological and behavioral outcomes dependent on the hippocampus and associated with schizophrenia. Genetic deletion of Akt1 was associated with both impaired proliferative capacity of adult-born hippocampal progenitors and hippocampal long-term potentiation, indicating deficient functions of this brain region associated with neuroplasticity. Moreover, Akt1(-/-) mice demonstrated impairments in contextual fear conditioning and recall of spatial learning, behaviors known to selectively involve the hippocampus. Akt1(-/-) mice also showed reduced prepulse inhibition of the acoustic startle response, a sensorimotor gating response that is perturbed in schizophrenia. Postmortem tissue samples from patients with schizophrenia showed significant reductions of phosphorylated Akt levels in hilar neurons of the dentate gyrus, the neurogenic zone of the hippocampus. Taken together, these results implicate the Akt1 isoform in regulating hippocampal neuroplasticity and cognition and in contributing to the etiology of schizophrenia.


Subject(s)
Hippocampus/metabolism , Learning/physiology , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-akt/metabolism , Schizophrenia/metabolism , Aged , Aged, 80 and over , Animals , Behavior, Animal/physiology , Cell Proliferation , Conditioning, Classical/physiology , Fear , Female , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Reflex, Startle/physiology , Schizophrenia/genetics , Schizophrenia/physiopathology , Spatial Behavior/physiology
7.
Proc Natl Acad Sci U S A ; 108(43): E962-70, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21969553

ABSTRACT

DTNBP1 (dystrobrevin binding protein 1) is a leading candidate susceptibility gene in schizophrenia and is associated with working memory capacity in normal subjects. In schizophrenia, the encoded protein dystrobrevin-binding protein 1 (dysbindin-1) is often reduced in excitatory cortical limbic synapses. We found that reduced dysbindin-1 in mice yielded deficits in auditory-evoked response adaptation, prepulse inhibition of startle, and evoked γ-activity, similar to patterns in schizophrenia. In contrast to the role of dysbindin-1 in glutamatergic transmission, γ-band abnormalities in schizophrenia are most often attributed to disrupted inhibition and reductions in parvalbumin-positive interneuron (PV cell) activity. To determine the mechanism underlying electrophysiological deficits related to reduced dysbindin-1 and the potential role of PV cells, we examined PV cell immunoreactivity and measured changes in net circuit activity using voltage-sensitive dye imaging. The dominant circuit impact of reduced dysbindin-1 was impaired inhibition, and PV cell immunoreactivity was reduced. Thus, this model provides a link between a validated candidate gene and an auditory endophenotypes. Furthermore, these data implicate reduced fast-phasic inhibition as a common underlying mechanism of schizophrenia-associated intermediate phenotypes.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Evoked Potentials, Auditory/physiology , Limbic System/metabolism , Schizophrenia/genetics , Synapses/metabolism , Animals , Dysbindin , Dystrophin-Associated Proteins , Electrophysiology , Evoked Potentials, Auditory/genetics , Female , Genotype , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Parvalbumins
8.
Arch Neurol ; 68(11): 1398-403, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21747004

ABSTRACT

OBJECTIVE: To determine the correspondence of in vivo quantitative estimates of brain uptake of fluorine 18-labeled flutemetamol with immunohistochemical estimates of amyloid levels in patients who underwent previous biopsy. DESIGN: Cross-sectional study of ¹8F-flutemetamol positron emission tomography (PET) findings in patients with prior cortical biopsy specimen stained for the presence or absence of amyloid plaques. SETTING: University hospital. Patients Seven patients who previously had a prior right frontal cortical biopsy at the site of ventriculoperitoneal placement for presumed normal pressure hydrocephalus were recruited. Inclusion criteria included an adequate biopsy specimen for detection and quantification of ß-amyloid pathology and age older than 50 years. Intervention All patients underwent an ¹8F-flutemetamol PET scan. MAIN OUTCOME MEASURES: Quantitative measures of ¹8F-flutemetamol uptake (standardized uptake value ratio, a ratio of mean target cortex activity divided by that in a cerebellar reference region) were made at a location contralateral to the biopsy site and compared with estimates of amyloid load based on immunohistochemical and histological staining. RESULTS: There was complete agreement between visual reads of ¹8F-flutemetamol PET scans (3 blinded readers with majority rule) and histology. A regression model, including time from biopsy as a covariate, demonstrated a significant relationship (P = .01) between ¹8F-flutemetamol uptake and percentage of area of amyloid measured by a monoclonal antibody raised against amyloid (NAB228). Similar results were found with the amyloid-specific monoclonal antibody 4G8 and Thioflavin S. CONCLUSION: To our knowledge, these data are the first to demonstrate the concordance of ¹8F-flutemetamol PET imaging with histopathology, supporting its sensitivity to detect amyloid and potential use in the study and detection of Alzheimer disease.


Subject(s)
Benzothiazoles , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Fluorine Radioisotopes , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/pathology , Positron-Emission Tomography , Aged , Aged, 80 and over , Aniline Compounds , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Positron-Emission Tomography/methods , Thiazoles
9.
PLoS One ; 6(3): e16886, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21390302

ABSTRACT

BACKGROUND: An increasing number of studies report associations between variation in DTNBP1, a top candidate gene in schizophrenia, and both the clinical symptoms of the disorder and its cognitive deficits. DTNBP1 encodes dysbindin-1, reduced levels of which have been found in synaptic fields of schizophrenia cases. This study determined whether such synaptic reductions are isoform-specific. METHODOLOGY/PRINCIPAL FINDINGS: Using Western blotting of tissue fractions, we first determined the synaptic localization of the three major dysbindin-1 isoforms (A, B, and C). All three were concentrated in synaptosomes of multiple brain areas, including auditory association cortices in the posterior half of the superior temporal gyrus (pSTG) and the hippocampal formation (HF). Tests on the subsynaptic tissue fractions revealed that each isoform is predominantly, if not exclusively, associated with synaptic vesicles (dysbindin-1B) or with postsynaptic densities (dysbindin-1A and -1C). Using Western blotting on pSTG (n = 15) and HF (n = 15) synaptosomal fractions from schizophrenia cases and their matched controls, we discovered that synaptic dysbindin-1 is reduced in an isoform-specific manner in schizophrenia without changes in levels of synaptophysin or PSD-95. In pSTG, about 92% of the schizophrenia cases displayed synaptic dysbindin-1A reductions averaging 48% (p = 0.0007) without alterations in other dysbindin-1 isoforms. In the HF, by contrast, schizophrenia cases displayed normal levels of synaptic dysbindin-1A, but 67% showed synaptic reductions in dysbindin-1B averaging 33% (p = 0.0256), while 80% showed synaptic reductions in dysbindin-1C averaging 35% (p = 0.0171). CONCLUSIONS/SIGNIFICANCE: Given the distinctive subsynaptic localization of dysbindin-1A, -1B, and -1C across brain regions, the observed pSTG reductions in dysbindin-1A are postsynaptic and may promote dendritic spine loss with consequent disruption of auditory information processing, while the noted HF reductions in dysbindin-1B and -1C are both presynaptic and postsynaptic and could promote deficits in spatial working memory.


Subject(s)
Carrier Proteins/metabolism , Schizophrenia/metabolism , Synapses/metabolism , Aged , Animals , Antibody Affinity , Autopsy , Case-Control Studies , Demography , Dysbindin , Dystrophin-Associated Proteins , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Mice , Protein Isoforms/metabolism , Schizophrenia/pathology , Subcellular Fractions/metabolism , Synapses/pathology , Synaptosomes/metabolism , Temporal Lobe/metabolism , Temporal Lobe/pathology
10.
Ann Neurol ; 67(4): 462-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437581

ABSTRACT

OBJECTIVE: Olfactory dysfunction is common in Alzheimer disease (AD) and other neurodegenerative diseases. Paired helical filament (PHF)-tau, alpha-synuclein, and amyloid-beta lesions occur early and severely in cerebral regions of the olfactory system, and they have also been observed in olfactory epithelium (OE). However, their frequency, abundance, and disease specificity, and the relationships of OE pathology to brain pathology have not been established. METHODS: We investigated the pathological expression of amyloid-beta, PHFtau, alpha-synuclein, and TDP-43 in postmortem OE of 79 cases with AD, 63 cases with various other neurodegenerative diseases, and 45 neuropathologically normal cases. RESULTS: Amyloid-beta was present as punctate and small patchy aggregates in 71% of AD cases, compared with 22% of normal cases and 14% of cases with other diseases, and in greater amounts in AD than in either of the other 2 diagnostic categories. PHFtau was evident in dystrophic neurites in 55% of cases with AD, 34% with normal brains, and 39% with other neurodegenerative diseases, also at higher densities in AD. alpha-Synuclein was present in dystrophic neurites in 7 cases, 6 of which also had cerebral Lewy bodies. Pathological TDP-43 inclusions were not observed in the OE in any cases. Amyloid-beta and to a lesser degree, PHFtau ratings in OE significantly correlated with cortical Abeta and PHFtau lesion ratings in the brain. INTERPRETATION: These data demonstrate that AD pathology in the OE is present in the majority of cases with pathologically verified AD and correlates with brain pathology. Future work may assess the utility of amyloid-beta and PHFtau measurement in OE as a biomarker for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/metabolism , Olfactory Mucosa/metabolism , Olfactory Mucosa/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Analysis of Variance , DNA-Binding Proteins/metabolism , Female , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Male , Neurofibrillary Tangles/pathology , Statistics as Topic , alpha-Synuclein/metabolism
11.
Hum Mol Genet ; 15(20): 3041-54, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16980328

ABSTRACT

Variations in the gene encoding the novel protein dysbindin-1 (DTNBP1) are among the most commonly reported genetic variations associated with schizophrenia. Recent studies show that those variations are also associated with cognitive functioning in carriers with and without psychiatric diagnoses, suggesting a general role for dysbindin-1 in cognition. Such a role could stem from the protein's known ability to affect neuronal glutamate release. How dysbindin-1 might affect glutamate release nevertheless remains unknown without the discovery of the protein's neuronal binding partners and its subcellular locus of action. We demonstrate here that snapin is a binding partner of dysbindin-1 in vitro and in the brain. Tissue fractionation of whole mouse brains and human hippocampal formations revealed that both dysbindin-1 and snapin are concentrated in tissue enriched in synaptic vesicle membranes and less commonly in postsynaptic densities. It is not detected in presynaptic tissue fractions lacking synaptic vesicles. Consistent with that finding, immunoelectron microscopy showed that dysbindin-1 is located in (i) synaptic vesicles of axospinous terminals in the dentate gyrus inner molecular layer and CA1 stratum radiatum and in (ii) postsynaptic densities and microtubules of dentate hilus neurons and CA1 pyramidal cells. The labeled synapses are often asymmetric with thick postsynaptic densities suggestive of glutamatergic synapses, which are likely to be derived from dentate mossy cells and CA3 pyramidal cells. The function of dysbindin-1 in presynaptic, postsynaptic and microtubule locations may all be related to known functions of snapin.


Subject(s)
Brain/cytology , Carrier Proteins/metabolism , Synaptic Vesicles/chemistry , Vesicular Transport Proteins/metabolism , Aged , Aged, 80 and over , Animals , Brain/metabolism , Brain Chemistry , COS Cells , Carrier Proteins/analysis , Chlorocebus aethiops , Dysbindin , Dystrophin-Associated Proteins , Female , Hippocampus/chemistry , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , Schizophrenia/metabolism , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Vesicular Transport Proteins/analysis
12.
Schizophr Res ; 77(2-3): 229-39, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15946825

ABSTRACT

Olfactory deficits, observed in schizophrenia, may be associated with a disruption of synaptic transmission in the olfactory system. Using immunohistochemistry and optical densitometry, we assessed the integrity of the synaptic connection between olfactory receptor neurons and olfactory bulb target neurons in schizophrenia by comparing the level of eight proteins, expressed in the olfactory bulb glomeruli, among schizophrenia and control subjects. In schizophrenia, no change was observed in the levels of OMP, GAP43 and NCAM, proteins expressed by olfactory receptor neurons, suggesting an intact innervation of the olfactory bulb by these neurons. This was supported by the absence of change in calbindin level, which has been shown to decrease after the destruction of the olfactory epithelium. The level of synaptophysin, a pre-synaptic protein, was also unchanged. These findings suggested that axons of olfactory receptor neurons establish synapses with their olfactory bulb targets in schizophrenia. The absence of change in the level of poorly phosphorylated neurofilament of moderate and high molecular weight (NFM/HP) suggested no lack of dendritic innervation despite a previously seen reduction of glomerular MAP2 level in schizophrenia subjects. This and above findings were consistent with the absence of change in the level of beta-tubulin III, a protein expressed by neurons of both olfactory epithelium and bulb. Finally, we noted no significant decrease in trkB level, a neurotrophin receptor involved in the olfactory epithelium maintenance. This study showed no evidence of major structural alteration of the synapse between the olfactory epithelium and bulb in schizophrenia.


Subject(s)
Nerve Tissue Proteins/metabolism , Olfactory Bulb/pathology , Olfactory Mucosa/innervation , Olfactory Receptor Neurons/pathology , Schizophrenia/pathology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Axons/metabolism , Biomarkers , Case-Control Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neuronal Plasticity , Olfactory Bulb/metabolism , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Prospective Studies , Schizophrenia/metabolism , Statistics, Nonparametric , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...