Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Virus Evol ; 7(2): veab058, 2021.
Article in English | MEDLINE | ID: mdl-34532061

ABSTRACT

Human polyomaviruses are widespread in humans and can cause severe disease in immunocompromised individuals. To identify human genetic determinants of the humoral immune response against polyomaviruses, we performed genome-wide association studies and meta-analyses of qualitative and quantitative immunoglobulin G responses against BK polyomavirus (BKPyV), JC polyomavirus (JCPyV), Merkel cellpolyomavirus (MCPyV), WU polyomavirus (WUPyV), and human polyomavirus 6 (HPyV6) in 15,660 individuals of European ancestry from three independent studies. We observed significant associations for all tested viruses: JCPyV, HPyV6, and MCPyV associated with human leukocyte antigen class II variation, BKPyV and JCPyV with variants in FUT2, responsible for secretor status, MCPyV with variants in STING1, involved in interferon induction, and WUPyV with a functional variant in MUC1, previously associated with risk for gastric cancer. These results provide insights into the genetic control of a family of very prevalent human viruses, highlighting genes and pathways that play a modulating role in human humoral immunity.

2.
Nat Commun ; 9(1): 3078, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082721

ABSTRACT

Higher educational attainment (EA) is negatively associated with schizophrenia (SZ). However, recent studies found a positive genetic correlation between EA and SZ. We investigate possible causes of this counterintuitive finding using genome-wide association study results for EA and SZ (N = 443,581) and a replication cohort (1169 controls; 1067 cases) with deeply phenotyped SZ patients. We find strong genetic dependence between EA and SZ that cannot be explained by chance, linkage disequilibrium, or assortative mating. Instead, several genes seem to have pleiotropic effects on EA and SZ, but without a clear pattern of sign concordance. Using EA as a proxy phenotype, we isolate FOXO6 and SLITRK1 as novel candidate genes for SZ. Our results reveal that current SZ diagnoses aggregate over at least two disease subtypes: one part resembles high intelligence and bipolar disorder (BIP), while the other part is a cognitive disorder that is independent of BIP.


Subject(s)
Educational Status , Genetic Heterogeneity , Genome-Wide Association Study , Linkage Disequilibrium , Schizophrenia/genetics , Adolescent , Adult , Aged , Bipolar Disorder/genetics , Cognition Disorders/genetics , Female , Forkhead Transcription Factors/genetics , Gene Frequency , Humans , Male , Membrane Proteins/genetics , Middle Aged , Models, Genetic , Nerve Tissue Proteins/genetics , Phenotype , Polymorphism, Single Nucleotide , Reproducibility of Results , Young Adult
3.
Mol Psychiatry ; 23(2): 476-486, 2018 02.
Article in English | MEDLINE | ID: mdl-27922606

ABSTRACT

Despite high heritability of schizophrenia, genome-wide association studies (GWAS) have not yet revealed distinct combinations of single-nucleotide polymorphisms (SNPs), relevant for mental disease-related, quantifiable behavioral phenotypes. Here we propose an individual-based model to use genome-wide significant markers for extracting first genetic signatures of such behavioral continua. 'OTTO' (old Germanic=heritage) marks an individual characterized by a prominent phenotype and a particular load of phenotype-associated risk SNPs derived from GWAS that likely contributed to the development of his personal mental illness. This load of risk SNPs is shared by a small squad of 'similars' scattered under the genetically and phenotypically extremely heterogeneous umbrella of a schizophrenia end point diagnosis and to a variable degree also by healthy subjects. In a discovery sample of >1000 deeply phenotyped schizophrenia patients and several independent replication samples, including the general population, a gradual increase in the severity of 'OTTO's phenotype' expression is observed with an increasing share of 'OTTO's risk SNPs', as exemplified here by autistic and affective phenotypes. These data suggest a model in which the genetic contribution to dimensional behavioral traits can be extracted from combinations of GWAS SNPs derived from individuals with prominent phenotypes. Even though still in the 'model phase' owing to a world-wide lack of sufficiently powered, deeply phenotyped replication samples, the OTTO approach constitutes a conceptually novel strategy to delineate biological subcategories of mental diseases starting from GWAS findings and individual subjects.


Subject(s)
Genome-Wide Association Study/methods , Schizophrenia/genetics , Adult , Aged , Female , Genetic Heterogeneity , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide/genetics , Psychotic Disorders/genetics , Risk Factors
4.
Transl Psychiatry ; 7(10): e1247, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28994820

ABSTRACT

Ambra1 is linked to autophagy and neurodevelopment. Heterozygous Ambra1 deficiency induces autism-like behavior in a sexually dimorphic manner. Extraordinarily, autistic features are seen in female mice only, combined with stronger Ambra1 protein reduction in brain compared to males. However, significance of AMBRA1 for autistic phenotypes in humans and, apart from behavior, for other autism-typical features, namely early brain enlargement or increased seizure propensity, has remained unexplored. Here we show in two independent human samples that a single normal AMBRA1 genotype, the intronic SNP rs3802890-AA, is associated with autistic features in women, who also display lower AMBRA1 mRNA expression in peripheral blood mononuclear cells relative to female GG carriers. Located within a non-coding RNA, likely relevant for mRNA and protein interaction, rs3802890 (A versus G allele) may affect its stability through modification of folding, as predicted by in silico analysis. Searching for further autism-relevant characteristics in Ambra1+/- mice, we observe reduced interest of female but not male mutants regarding pheromone signals of the respective other gender in the social intellicage set-up. Moreover, altered pentylentetrazol-induced seizure propensity, an in vivo readout of neuronal excitation-inhibition dysbalance, becomes obvious exclusively in female mutants. Magnetic resonance imaging reveals mild prepubertal brain enlargement in both genders, uncoupling enhanced brain dimensions from the primarily female expression of all other autistic phenotypes investigated here. These data support a role of AMBRA1/Ambra1 partial loss-of-function genotypes for female autistic traits. Moreover, they suggest Ambra1 heterozygous mice as a novel multifaceted and construct-valid genetic mouse model for female autism.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autism Spectrum Disorder/genetics , Sex Characteristics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/metabolism , Brain/pathology , Female , Gene Knockdown Techniques , Humans , Leukocytes, Mononuclear/metabolism , Male , Mice, Transgenic , Phenotype , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Schizophrenia/complications , Schizophrenia/genetics , Seizures/complications , Seizures/genetics , Social Behavior , Species Specificity
5.
Mol Psychiatry ; 22(12): 1776-1784, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27502473

ABSTRACT

Autoantibodies of the IgG class against N-methyl-d-aspartate-receptor subunit NR1 (NMDAR1) were first described in anti-NMDAR encephalitis and seen as disease indicators. Recent work on together over 5000 individuals challenged this exclusive view by showing age-dependently up to >20% NMDAR1-autoantibody seroprevalence with comparable immunoglobulin class and titer distribution across health and disease. The key question therefore is to understand the properties of these autoantibodies, also in healthy carriers, in order to assess secondary complications and possible contributions to neuropsychiatric disease. Here, we believe we provide for human NMDAR1-autoantibodies the first comprehensive analysis of their target epitopes and functionality. We selected sera of representative carriers, healthy or diagnosed with very diverse conditions, that is, schizophrenia, age-related disorders like hypertension and diabetes, or anti-NMDAR encephalitis. We show that all positive sera investigated, regardless of source (ill or healthy donor) and immunoglobulin class, provoked NMDAR1 internalization in human-induced pluripotent stem cell-derived neurons and reduction of glutamate-evoked currents in NR1-1b/NR2A-expressing Xenopus oocytes. They displayed frequently polyclonal/polyspecific epitope recognition in the extracellular or intracellular NMDAR1 domains and some additionally in NR2A. We conclude that all circulating NMDAR1-autoantibodies have pathogenic potential regarding the whole spectrum of neuronal NMDAR-mediated effects upon access to the brain in situations of increased blood-brain-barrier permeability.


Subject(s)
Autoantibodies/blood , Epitopes/metabolism , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Receptors, N-Methyl-D-Aspartate/immunology , Aged , Animals , Endocytosis/physiology , Female , Fibroblasts/immunology , Glutamic Acid/metabolism , Humans , Induced Pluripotent Stem Cells/immunology , Male , Membrane Potentials/physiology , Mental Disorders/blood , Mental Disorders/immunology , Middle Aged , Nervous System Diseases/blood , Nervous System Diseases/immunology , Neurons/immunology , Oocytes , Xenopus laevis
6.
Acta Psychiatr Scand ; 134(3): 249-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27259062

ABSTRACT

OBJECTIVE: Erythropoietin (EPO) improves verbal memory and reverses subfield hippocampal volume loss across depression and bipolar disorder (BD). This study aimed to investigate with functional magnetic resonance imaging (fMRI) whether these effects were accompanied by functional changes in memory-relevant neuro-circuits in this cohort. METHOD: Eighty-four patients with treatment-resistant unipolar depression who were moderately depressed or BD in remission were randomized to eight weekly EPO (40 000 IU) or saline infusions in a double-blind, parallel-group design. Participants underwent whole-brain fMRI at 3T, mood ratings, and blood tests at baseline and week 14. During fMRI, participants performed a picture encoding task followed by postscan recall. RESULTS: Sixty-two patients had complete data (EPO: N = 32, saline: N = 30). EPO improved picture recall and increased encoding-related activity in dorsolateral prefrontal cortex (dlPFC) and temporo-parietal regions, but not in hippocampus. Recall correlated with activity in the identified dlPFC and temporo-parietal regions at baseline, and change in recall correlated with activity change in these regions from baseline to follow-up across the entire cohort. The effects of EPO were not correlated with change in mood, red blood cells, blood pressure, or medication. CONCLUSION: The findings highlight enhanced encoding-related dlPFC and temporo-parietal activity as key neuronal underpinnings of EPO-associated memory improvement.


Subject(s)
Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder/drug therapy , Erythropoietin/adverse effects , Mental Recall/drug effects , Adult , Depressive Disorder/diagnostic imaging , Depressive Disorder, Treatment-Resistant/diagnostic imaging , Double-Blind Method , Erythropoietin/pharmacology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Prefrontal Cortex/diagnostic imaging , Psychiatric Status Rating Scales , Treatment Outcome
7.
Psychol Med ; 46(8): 1679-91, 2016 06.
Article in English | MEDLINE | ID: mdl-26996196

ABSTRACT

BACKGROUND: Cognitive dysfunction in depression and bipolar disorder (BD) is insufficiently targeted by available treatments. Erythropoietin (EPO) increases neuroplasticity and may improve cognition in mood disorders, but the neuronal mechanisms of these effects are unknown. This functional magnetic resonance imaging (fMRI) study investigated the effects of EPO on neural circuitry activity during working memory (WM) performance. METHOD: Patients with treatment-resistant major depression, who were moderately depressed, or with BD in partial remission, were randomized to eight weekly infusions of EPO (40 000 IU) (N = 30) or saline (N = 26) in a double-blind, parallel-group design. Patients underwent fMRI, mood ratings and blood tests at baseline and week 14. During fMRI patients performed an n-back WM task. RESULTS: EPO improved WM accuracy compared with saline (p = 0.045). Whole-brain analyses revealed that EPO increased WM load-related activity in the right superior frontal gyrus (SFG) compared with saline (p = 0.01). There was also enhanced WM load-related deactivation of the left hippocampus in EPO-treated compared to saline-treated patients (p = 0.03). Across the entire sample, baseline to follow-up changes in WM performance correlated positively with changes in WM-related SFG activity and negatively with hippocampal response (r = 0.28-0.30, p < 0.05). The effects of EPO were not associated with changes in mood or red blood cells (p ⩾0.08). CONCLUSIONS: The present findings associate changes in WM-load related activity in the right SFG and left hippocampus with improved executive function in EPO-treated patients. CLINICAL TRIAL REGISTRATION: clinicaltrials.gov: NCT00916552.


Subject(s)
Bipolar Disorder/drug therapy , Brain/physiopathology , Cognitive Dysfunction/drug therapy , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Erythropoietin/therapeutic use , Executive Function , Adult , Bipolar Disorder/diagnostic imaging , Bipolar Disorder/physiopathology , Bipolar Disorder/psychology , Brain/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/psychology , Depressive Disorder, Treatment-Resistant/diagnostic imaging , Depressive Disorder, Treatment-Resistant/physiopathology , Depressive Disorder, Treatment-Resistant/psychology , Double-Blind Method , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Memory, Short-Term , Middle Aged , Spatial Memory , Treatment Outcome
8.
Mol Psychiatry ; 21(12): 1752-1767, 2016 12.
Article in English | MEDLINE | ID: mdl-26809838

ABSTRACT

Recombinant human erythropoietin (EPO) improves cognitive performance in neuropsychiatric diseases ranging from schizophrenia and multiple sclerosis to major depression and bipolar disease. This consistent EPO effect on cognition is independent of its role in hematopoiesis. The cellular mechanisms of action in brain, however, have remained unclear. Here we studied healthy young mice and observed that 3-week EPO administration was associated with an increased number of pyramidal neurons and oligodendrocytes in the hippocampus of ~20%. Under constant cognitive challenge, neuron numbers remained elevated until >6 months of age. Surprisingly, this increase occurred in absence of altered cell proliferation or apoptosis. After feeding a 15N-leucine diet, we used nanoscopic secondary ion mass spectrometry, and found that in EPO-treated mice, an equivalent number of neurons was defined by elevated 15N-leucine incorporation. In EPO-treated NG2-Cre-ERT2 mice, we confirmed enhanced differentiation of preexisting oligodendrocyte precursors in the absence of elevated DNA synthesis. A corresponding analysis of the neuronal lineage awaits the identification of suitable neuronal markers. In cultured neurospheres, EPO reduced Sox9 and stimulated miR124, associated with advanced neuronal differentiation. We are discussing a resulting working model in which EPO drives the differentiation of non-dividing precursors in both (NG2+) oligodendroglial and neuronal lineages. As endogenous EPO expression is induced by brain injury, such a mechanism of adult neurogenesis may be relevant for central nervous system regeneration.


Subject(s)
Erythropoietin/metabolism , Neurogenesis/drug effects , Oligodendroglia/drug effects , Animals , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Central Nervous System/metabolism , Cognition/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurogenesis/physiology , Neurons/metabolism , Oligodendroglia/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Recombinant Proteins/metabolism
10.
Transl Psychiatry ; 4: e362, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24548877

ABSTRACT

A recent publication reported an exciting polygenic effect of schizophrenia (SCZ) risk variants, identified by a large genome-wide association study (GWAS), on total brain and white matter volumes in schizophrenic patients and, even more prominently, in healthy subjects. The aim of the present work was to replicate and then potentially extend these findings. According to the original publication, polygenic risk scores-using single nucleotide polymorphism (SNP) information of SCZ GWAS-(polygenic SCZ risk scores; PSS) were calculated in 122 healthy subjects, enrolled in a structural magnetic resonance imaging (MRI) study. These scores were computed based on P-values and odds ratios available through the Psychiatric GWAS Consortium. In addition, polygenic white matter scores (PWM) were calculated, using the respective SNP subset in the original publication. None of the polygenic scores, either PSS or PWM, were found to be associated with total brain, white matter or gray matter volume in our replicate sample. Minor differences between the original and the present study that might have contributed to lack of reproducibility (but unlikely explain it fully), are number of subjects, ethnicity, age distribution, array technology, SNP imputation quality and MRI scanner type. In contrast to the original publication, our results do not reveal the slightest signal of association of the described sets of GWAS-identified SCZ risk variants with brain volumes in adults. Caution is indicated in interpreting studies building on polygenic risk scores without replication sample.


Subject(s)
Brain/anatomy & histology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Multifactorial Inheritance/genetics , Adult , Aged , Aged, 80 and over , Female , Gray Matter/anatomy & histology , Humans , Italy , Magnetic Resonance Imaging , Male , Middle Aged , Polymorphism, Single Nucleotide , White Matter/anatomy & histology , Young Adult
11.
Neuroscience ; 258: 218-27, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24269933

ABSTRACT

The cytokine erythropoietin (Epo) initiates adaptive cellular responses to both moderate environmental challenges and tissue damaging insults in various non-hematopoietic mammalian tissues including the nervous system. Neuroprotective and neuroregenerative functions of Epo in mammals are mediated through receptor-associated Janus kinase 2 and intracellular signaling cascades that modify the transcription of Epo-regulated genes. Signal transducers and activators of transcription (STAT) and phosphoinositol-3-kinase (PI3K) represent key components of two important Epo-induced transduction pathways. Our previous study on insects revealed neuroprotective and regenerative functions of recombinant human Epo (rhEpo) similar to those in mammalian nervous tissues. Here we demonstrate that rhEpo effectively rescues primary cultured locust brain neurons from apoptotic cell death induced by hypoxia or the chemical compound H-7. The Janus kinase inhibitor AG-490 and the STAT inhibitor sc-355797 abolished protective effects of rhEpo on locust brain neurons. In contrast, inhibition of PI3K with LY294002 had no effect on rhEpo-mediated neuroprotection. The results indicate that rhEpo mediates the protection of locust brain neurons through interference with apoptotic pathways by the activation of a Janus kinase-associated receptor and STAT transcription factor(s). The involvement of similar transduction pathways in mammals and insects for the mediation of neuroprotection and support of neural regeneration by Epo indicates that an Epo/Epo receptor-like signaling system with high structural and functional similarity exists in both groups of animals. Epo-like signaling involved in tissue protection appears to be an ancient beneficial function shared by vertebrates and invertebrates.


Subject(s)
Apoptosis/drug effects , Erythropoietin/pharmacology , Grasshoppers/drug effects , Neuroprotective Agents/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/toxicity , Animals , Apoptosis/physiology , Brain/drug effects , Brain/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Grasshoppers/physiology , Humans , Hypoxia/drug therapy , Hypoxia/physiopathology , Insect Proteins/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Morpholines/pharmacology , Neurons/drug effects , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology
12.
Mol Psychiatry ; 19(10): 1143-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23999527

ABSTRACT

In 2007, a multifaceted syndrome, associated with anti-NMDA receptor autoantibodies (NMDAR-AB) of immunoglobulin-G isotype, has been described, which variably consists of psychosis, epilepsy, cognitive decline and extrapyramidal symptoms. Prevalence and significance of NMDAR-AB in complex neuropsychiatric disease versus health, however, have remained unclear. We tested sera of 2817 subjects (1325 healthy, 1081 schizophrenic, 263 Parkinson and 148 affective-disorder subjects) for presence of NMDAR-AB, conducted a genome-wide genetic association study, comparing AB carriers versus non-carriers, and assessed their influenza AB status. For mechanistic insight and documentation of AB functionality, in vivo experiments involving mice with deficient blood-brain barrier (ApoE(-/-)) and in vitro endocytosis assays in primary cortical neurons were performed. In 10.5% of subjects, NMDAR-AB (NR1 subunit) of any immunoglobulin isotype were detected, with no difference in seroprevalence, titer or in vitro functionality between patients and healthy controls. Administration of extracted human serum to mice influenced basal and MK-801-induced activity in the open field only in ApoE(-/-) mice injected with NMDAR-AB-positive serum but not in respective controls. Seropositive schizophrenic patients with a history of neurotrauma or birth complications, indicating an at least temporarily compromised blood-brain barrier, had more neurological abnormalities than seronegative patients with comparable history. A common genetic variant (rs524991, P=6.15E-08) as well as past influenza A (P=0.024) or B (P=0.006) infection were identified as predisposing factors for NMDAR-AB seropositivity. The >10% overall seroprevalence of NMDAR-AB of both healthy individuals and patients is unexpectedly high. Clinical significance, however, apparently depends on association with past or present perturbations of blood-brain barrier function.


Subject(s)
Autoantibodies/blood , Blood-Brain Barrier/metabolism , Mood Disorders/metabolism , Parkinson Disease/metabolism , Receptors, N-Methyl-D-Aspartate/immunology , Schizophrenia/metabolism , Adult , Aged , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cerebral Cortex/metabolism , Endocytosis/physiology , Female , Genome-Wide Association Study , Humans , Influenza, Human/genetics , Influenza, Human/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mood Disorders/genetics , Neurons/metabolism , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics
13.
Transl Psychiatry ; 3: e254, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23632458

ABSTRACT

Claustrophobia, the well-known fear of being trapped in narrow/closed spaces, is often considered a conditioned response to traumatic experience. Surprisingly, we found that mutations affecting a single gene, encoding a stress-regulated neuronal protein, can cause claustrophobia. Gpm6a-deficient mice develop normally and lack obvious behavioral abnormalities. However, when mildly stressed by single-housing, these mice develop a striking claustrophobia-like phenotype, which is not inducible in wild-type controls, even by severe stress. The human GPM6A gene is located on chromosome 4q32-q34, a region linked to panic disorder. Sequence analysis of 115 claustrophobic and non-claustrophobic subjects identified nine variants in the noncoding region of the gene that are more frequent in affected individuals (P=0.028). One variant in the 3'untranslated region was linked to claustrophobia in two small pedigrees. This mutant mRNA is functional but cannot be silenced by neuronal miR124 derived itself from a stress-regulated transcript. We suggest that loosing dynamic regulation of neuronal GPM6A expression poses a genetic risk for claustrophobia.


Subject(s)
Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Phobic Disorders/genetics , Adult , Amygdala/chemistry , Animals , Behavior, Animal , Electroretinography , Female , Genetic Engineering/methods , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Psychological Tests , Reflex, Startle/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Psychological/genetics
14.
Fortschr Neurol Psychiatr ; 81(5): 240-2, 2013 May.
Article in German | MEDLINE | ID: mdl-23695788

ABSTRACT

Schizophrenias are diagnosed purely clinically. The biological basis for this clinical entity is still fully unknown. Genetic studies have revealed some interesting hints but have not led to the identification of actual disease genotypes. On the contrary, it has become more and more probable that widely differing genotype constellations together with manifold environmental factors can trigger schizophrenia according to the motto "many roads lead to Rome...". Thus, new strategies that allow a better insight into complex genotype-phenotype relationships, e. g. PGAS (phenotype-based genetic associations studies) are urgently needed. PGAS became possible on the basis of the GRAS data collection, the as yet largest worldwide phenotypical databank of schizophrenic patients. First PGAS proof-of-concept results on cognition or development-relevant genes are already available.


Subject(s)
Schizophrenia/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Phenotype , Schizophrenic Psychology
16.
Neuroscience ; 188: 95-108, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21600963

ABSTRACT

In addition to its function as a regulator of hematopoiesis, the cytokine erythropoietin (Epo) initiates adaptive cellular responses to both moderate environmental challenges and tissue damaging insults in various non-hematopoietic mammalian tissues. Epo's neuroprotective and neuroregenerative functions mediated through janus kinases (JAK)/signal transducers and activators of transcription (STAT) transduction pathways and regulation of Epo and Epo receptor expression in the nervous system by hypoxia inducible factor (HIF) have been documented in a variety of in vitro and in vivo studies and homologs of the human Epo gene are present in fish, amphibians and mammals. The present study reproduces the hallmarks of Epo-mediated mammalian neuroprotection in the grasshopper nervous system. Recombinant human Epo (rhEpo) increases the survival of dissociated grasshopper brain neurons under normoxic and hypoxic conditions and promotes the regeneration of neurites in vitro. In addition, reestablishment of sound source localization after unilateral tympanic nerve crush injury was accelerated and more complete after application of rhEpo, demonstrating in vivo support of auditory receptor cell axon regeneration. Immunoblots of central nervous tissue extracts from mouse, grasshopper, crayfish and leech labeled protein bands of ∼38 kDa, fitting to the molecular weight of Epo reported in earlier studies. These results indicate that a ligand/receptor system that shares structural and functional similarities with mammalian Epo and Epo receptor exerts neuroprotective and neuroregenerative effects in insects. With both upstream (HIF system) and downstream (JAK/STAT pathway) elements of the mammalian Epo system being present in insects and other invertebrates, Epo-like signaling involved in tissue protection appears to be an ancient beneficial function shared by vertebrates and invertebrates.


Subject(s)
Brain/physiology , Erythropoietin/metabolism , Grasshoppers/physiology , Nerve Regeneration/physiology , Neurons/physiology , Animals , Blotting, Western , Cell Survival/physiology , Humans , Receptors, Erythropoietin/metabolism , Recombinant Proteins
17.
Mol Psychiatry ; 16(1): 26-36, 1, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20479759

ABSTRACT

Neurodevelopmental abnormalities together with neurodegenerative processes contribute to schizophrenia, an etiologically heterogeneous, complex disease phenotype that has been difficult to model in animals. The neurodegenerative component of schizophrenia is best documented by magnetic resonance imaging (MRI), demonstrating progressive cortical gray matter loss over time. No treatment exists to counteract this slowly proceeding atrophy. The hematopoietic growth factor erythropoietin (EPO) is neuroprotective in animals. Here, we show by voxel-based morphometry in 32 human subjects in a placebo-controlled study that weekly high-dose EPO for as little as 3 months halts the progressive atrophy in brain areas typically affected in schizophrenia, including hippocampus, amygdala, nucleus accumbens, and several neocortical areas. Specifically, gray matter protection is highly associated with improvement in attention and memory functions. These findings suggest that a neuroprotective strategy is effective against common pathophysiological features of schizophrenic patients, and strongly encourage follow-up studies to optimize EPO treatment dose and duration.


Subject(s)
Brain/pathology , Erythropoietin/administration & dosage , Neuroprotective Agents/administration & dosage , Schizophrenia/pathology , Adult , Analysis of Variance , Atrophy/drug therapy , Attention/drug effects , Brain/drug effects , Double-Blind Method , Humans , Male , Memory/drug effects , Middle Aged , Recombinant Proteins , Schizophrenia/drug therapy , Treatment Outcome
18.
Transl Psychiatry ; 1: e45, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-22833191

ABSTRACT

Genotype-phenotype correlations of common monogenic diseases revealed that the degree of deviation of mutant genes from wild-type structure and function often predicts disease onset and severity. In complex disorders such as schizophrenia, the overall genetic risk is still often >50% but genotype-phenotype relationships are unclear. Recent genome-wide association studies (GWAS) replicated a risk for several single-nucleotide polymorphisms (SNPs) regarding the endpoint diagnosis of schizophrenia. The biological relevance of these SNPs, however, for phenotypes or severity of schizophrenia has remained obscure. We hypothesized that the GWAS 'top-10' should as single markers, but even more so upon their accumulation, display associations with lead features of schizophrenia, namely positive and negative symptoms, cognitive deficits and neurological signs (including catatonia), and/or with age of onset of the disease prodrome as developmental readout and predictor of disease severity. For testing this hypothesis, we took an approach complementary to GWAS, and performed a phenotype-based genetic association study (PGAS). We utilized the to our knowledge worldwide largest phenotypical database of schizophrenic patients (n>1000), the GRAS (Göttingen Research Association for Schizophrenia) Data Collection. We found that the 'top-10' GWAS-identified risk SNPs neither as single markers nor when explored in the sense of a cumulative genetic risk, have any predictive value for disease onset or severity in the schizophrenic patients, as demonstrated across all core symptoms. We conclude that GWAS does not extract disease genes of general significance in schizophrenia, but may yield, on a hypothesis-free basis, candidate genes relevant for defining disease subgroups.


Subject(s)
Genetic Association Studies , Schizophrenia/diagnosis , Schizophrenia/genetics , Severity of Illness Index , Adult , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Male , Polymorphism, Single Nucleotide/genetics , Risk
19.
Genes Brain Behav ; 9(6): 592-602, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20412316

ABSTRACT

Schizophrenia is a devastating disease that affects approximately 1% of the population across cultures. Its neurobiological underpinnings are still unknown. Accordingly, animal models of schizophrenia often lack construct validity. As concordance rate in monozygotic twins amounts to only 50%, environmental risk factors (e.g. neurotrauma, drug abuse, psychotrauma) likely act as necessary 'second hit' to trigger/drive the disease process in a genetically predisposed individual. Valid animal models would have to consider this genetic-environmental interaction. Based on this concept, we designed an experimental approach for modeling a schizophrenia-like phenotype in mice. As dysfunction in synaptic transmission plays a key role in schizophrenia, and complexin2 (CPLX2) gene expression is reduced in hippocampus of schizophrenic patients, we developed a mouse model with Cplx2 null mutation as genetic risk factor and a mild parietal neurotrauma, applied during puberty, as environmental 'second hit'. Several months after lesion, Cplx2 null mutants showed reduced pre-pulse inhibition, deficit of spatial learning and loss of inhibition after MK-801 challenge. These abnormalities were largely absent in lesioned wild-type mice and non-lesioned Cplx2 null mutants. Forced alternation in T-maze, object recognition, social interaction and elevated plus maze tests were unaltered in all groups. The previously reported mild motor phenotype of Cplx2 null mutants was accentuated upon lesion. MRI volumetrical analysis showed a decrease of hippocampal volume exclusively in lesioned Cplx2 null mutants. These findings provide suggestive evidence for the 'second hit' hypothesis of schizophrenia and may offer new tools for the development of advanced treatment strategies.


Subject(s)
Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Parietal Lobe/injuries , Schizophrenia/etiology , Schizophrenia/genetics , Animals , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Recognition, Psychology/physiology , Reflex, Startle/physiology , Risk Factors , Social Behavior , Space Perception/physiology
20.
Biol Lett ; 5(5): 589-92, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19515648

ABSTRACT

The ultrasonic vocalizations of mice are attracting increasing attention, because they have been recognized as an informative readout in genetically modified strains. In addition, the observation that male mice produce elaborate sequences of ultrasonic vocalizations ('song') when exposed to female mice or their scents has sparked a debate as to whether these sounds are--in terms of their structure and function--analogous to bird song. We conducted playback experiments with cycling female mice to explore the function of male mouse songs. Using a place preference design, we show that these vocalizations elicited approach behaviour in females. In contrast, the playback of whistle-like artificial control sounds did not evoke approach responses. Surprisingly, the females also did not respond to pup isolation calls. In addition, female responses did not vary in relation to reproductive cycle, i.e. whether they were in oestrus or not. Furthermore, our data revealed a rapid habituation of subjects to the experimental situation, which stands in stark contrast to other species' responses to courtship vocalizations. Nevertheless, our results clearly demonstrate that male mouse songs elicit females' interest.


Subject(s)
Mice/physiology , Sexual Behavior, Animal , Vocalization, Animal , Animals , Estrous Cycle , Female , Male , Mice, Inbred C57BL , Ultrasonics
SELECTION OF CITATIONS
SEARCH DETAIL
...