Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Brain Behav Immun ; 108: 135-147, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36323361

RESUMO

BACKGROUND: Circulating autoantibodies (AB) against brain-antigens, often deemed pathological, receive increasing attention. We assessed predispositions and seroprevalence/characteristics of 49 AB in > 7000 individuals. METHODS: Exploratory cross-sectional cohort study, investigating deeply phenotyped neuropsychiatric patients and healthy individuals of GRAS Data Collection for presence/characteristics of 49 brain-directed serum-AB. Predispositions were evaluated through GWAS of NMDAR1-AB carriers, analyses of immune check-point genotypes, APOE4 status, neurotrauma. Chi-square, Fisher's exact tests and logistic regression analyses were used. RESULTS: Study of N = 7025 subjects (55.8 % male; 41 ±â€¯16 years) revealed N = 1133 (16.13 %) carriers of any AB against 49 defined brain-antigens. Overall, age dependence of seroprevalence (OR = 1.018/year; 95 % CI [1.015-1.022]) emerged, but no disease association, neither general nor with neuropsychiatric subgroups. Males had higher AB seroprevalence (OR = 1.303; 95 % CI [1.144-1.486]). Immunoglobulin class (N for IgM:462; IgA:487; IgG:477) and titers were similar. Abundant were NMDAR1-AB (7.7 %). Low seroprevalence (1.25 %-0.02 %) was seen for most AB (e.g., amphiphysin, KCNA2, ARHGAP26, GFAP, CASPR2, MOG, Homer-3, KCNA1, GLRA1b, GAD65). Non-detectable were others. GWAS of NMDAR1-AB carriers revealed three genome-wide significant SNPs, two intergenic, one in TENM3, previously autoimmune disease-associated. Targeted analysis of immune check-point genotypes (CTLA4, PD1, PD-L1) uncovered effects on humoral anti-brain autoimmunity (OR = 1.55; 95 % CI [1.058-2.271]) and disease likelihood (OR = 1.43; 95 % CI [1.032-1.985]). APOE4 carriers (∼19 %) had lower seropositivity (OR = 0.766; 95 % CI [0.625-0.933]). Neurotrauma predisposed to NMDAR1-AB seroprevalence (IgM: OR = 1.599; 95 % CI [1.022-2.468]). CONCLUSIONS: Humoral autoimmunity against brain-antigens, frequent across health and disease, is predicted by age, gender, genetic predisposition, and brain injury. Seroprevalence, immunoglobulin class, or titers do not predict disease.


Assuntos
Autoanticorpos , Autoimunidade , Feminino , Humanos , Masculino , Apolipoproteína E4 , Estudos Transversais , Isotipos de Imunoglobulinas , Imunoglobulina M , Proteínas de Membrana , Proteínas do Tecido Nervoso , Estudos Soroepidemiológicos , Adulto , Pessoa de Meia-Idade
2.
Nature ; 604(7906): 502-508, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35396580

RESUMO

Schizophrenia has a heritability of 60-80%1, much of which is attributable to common risk alleles. Here, in a two-stage genome-wide association study of up to 76,755 individuals with schizophrenia and 243,649 control individuals, we report common variant associations at 287 distinct genomic loci. Associations were concentrated in genes that are expressed in excitatory and inhibitory neurons of the central nervous system, but not in other tissues or cell types. Using fine-mapping and functional genomic data, we identify 120 genes (106 protein-coding) that are likely to underpin associations at some of these loci, including 16 genes with credible causal non-synonymous or untranslated region variation. We also implicate fundamental processes related to neuronal function, including synaptic organization, differentiation and transmission. Fine-mapped candidates were enriched for genes associated with rare disruptive coding variants in people with schizophrenia, including the glutamate receptor subunit GRIN2A and transcription factor SP4, and were also enriched for genes implicated by such variants in neurodevelopmental disorders. We identify biological processes relevant to schizophrenia pathophysiology; show convergence of common and rare variant associations in schizophrenia and neurodevelopmental disorders; and provide a resource of prioritized genes and variants to advance mechanistic studies.


Assuntos
Estudo de Associação Genômica Ampla , Esquizofrenia , Alelos , Predisposição Genética para Doença/genética , Genômica , Humanos , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética
3.
Biol Psychiatry ; 91(1): 102-117, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34099189

RESUMO

BACKGROUND: Sex differences in incidence and/or presentation of schizophrenia (SCZ), major depressive disorder (MDD), and bipolar disorder (BIP) are pervasive. Previous evidence for shared genetic risk and sex differences in brain abnormalities across disorders suggest possible shared sex-dependent genetic risk. METHODS: We conducted the largest to date genome-wide genotype-by-sex (G×S) interaction of risk for these disorders using 85,735 cases (33,403 SCZ, 19,924 BIP, and 32,408 MDD) and 109,946 controls from the PGC (Psychiatric Genomics Consortium) and iPSYCH. RESULTS: Across disorders, genome-wide significant single nucleotide polymorphism-by-sex interaction was detected for a locus encompassing NKAIN2 (rs117780815, p = 3.2 × 10-8), which interacts with sodium/potassium-transporting ATPase (adenosine triphosphatase) enzymes, implicating neuronal excitability. Three additional loci showed evidence (p < 1 × 10-6) for cross-disorder G×S interaction (rs7302529, p = 1.6 × 10-7; rs73033497, p = 8.8 × 10-7; rs7914279, p = 6.4 × 10-7), implicating various functions. Gene-based analyses identified G×S interaction across disorders (p = 8.97 × 10-7) with transcriptional inhibitor SLTM. Most significant in SCZ was a MOCOS gene locus (rs11665282, p = 1.5 × 10-7), implicating vascular endothelial cells. Secondary analysis of the PGC-SCZ dataset detected an interaction (rs13265509, p = 1.1 × 10-7) in a locus containing IDO2, a kynurenine pathway enzyme with immunoregulatory functions implicated in SCZ, BIP, and MDD. Pathway enrichment analysis detected significant G×S interaction of genes regulating vascular endothelial growth factor receptor signaling in MDD (false discovery rate-corrected p < .05). CONCLUSIONS: In the largest genome-wide G×S analysis of mood and psychotic disorders to date, there was substantial genetic overlap between the sexes. However, significant sex-dependent effects were enriched for genes related to neuronal development and immune and vascular functions across and within SCZ, BIP, and MDD at the variant, gene, and pathway levels.


Assuntos
Transtorno Bipolar/genética , Transtorno Depressivo Maior , Transtornos Psicóticos , Esquizofrenia/genética , Caracteres Sexuais , Transtorno Depressivo Maior/genética , Células Endoteliais , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Transtornos Psicóticos/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Sulfurtransferases
4.
Mol Med ; 27(1): 120, 2021 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-34565332

RESUMO

BACKGROUND: Since fall 2019, SARS-CoV-2 spread world-wide, causing a major pandemic with estimated ~ 220 million subjects affected as of September 2021. Severe COVID-19 is associated with multiple organ failure, particularly of lung and kidney, but also grave neuropsychiatric manifestations. Overall mortality reaches > 2%. Vaccine development has thrived in thus far unreached dimensions and will be one prerequisite to terminate the pandemic. Despite intensive research, however, few treatment options for modifying COVID-19 course/outcome have emerged since the pandemic outbreak. Additionally, the substantial threat of serious downstream sequelae, called 'long COVID' and 'neuroCOVID', becomes increasingly evident. Among candidates that were suggested but did not yet receive appropriate funding for clinical trials is recombinant human erythropoietin. Based on accumulating experimental and clinical evidence, erythropoietin is expected to (1) improve respiration/organ function, (2) counteract overshooting inflammation, (3) act sustainably neuroprotective/neuroregenerative. Recent counterintuitive findings of decreased serum erythropoietin levels in severe COVID-19 not only support a relative deficiency of erythropoietin in this condition, which can be therapeutically addressed, but also made us coin the term 'hypoxia paradox'. As we review here, this paradox is likely due to uncoupling of physiological hypoxia signaling circuits, mediated by detrimental gene products of SARS-CoV-2 or unfavorable host responses, including microRNAs or dysfunctional mitochondria. Substitution of erythropoietin might overcome this 'hypoxia paradox' caused by deranged signaling and improve survival/functional status of COVID-19 patients and their long-term outcome. As supporting hints, embedded in this review, we present 4 male patients with severe COVID-19 and unfavorable prognosis, including predicted high lethality, who all profoundly improved upon treatment which included erythropoietin analogues. SHORT CONCLUSION: Substitution of EPO may-among other beneficial EPO effects in severe COVID-19-circumvent downstream consequences of the 'hypoxia paradox'. A double-blind, placebo-controlled, randomized clinical trial for proof-of-concept is warranted.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19/complicações , Eritropoetina/genética , Hipóxia/tratamento farmacológico , Pulmão/efeitos dos fármacos , COVID-19/genética , COVID-19/patologia , COVID-19/virologia , Eritropoetina/análogos & derivados , Eritropoetina/uso terapêutico , Humanos , Hipóxia/genética , Hipóxia/patologia , Hipóxia/virologia , Pulmão/patologia , Pulmão/virologia , Pandemias , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , SARS-CoV-2/efeitos dos fármacos , Síndrome de COVID-19 Pós-Aguda
5.
Mol Psychiatry ; 26(7): 3211-3222, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33824432

RESUMO

Considering the immense societal and personal costs and suffering associated with multiple drug use or "polytoxicomania", better understanding of environmental and genetic causes is crucial. While previous studies focused on single risk factors and selected drugs, effects of early-accumulated environmental risks on polytoxicomania were never addressed. Similarly, evidence of genetic susceptibility to particular drugs is abundant, while genetic predisposition to polytoxicomania is unexplored. We exploited the GRAS data collection, comprising information on N~2000 deep-phenotyped schizophrenia patients, to investigate effects of early-life environmental risk accumulation on polytoxicomania and additionally provide first genetic insight. Preadult accumulation of environmental risks (physical or sexual abuse, urbanicity, migration, cannabis, alcohol) was strongly associated with lifetime polytoxicomania (p = 1.5 × 10-45; OR = 31.4), preadult polytoxicomania with OR = 226.6 (p = 1.0 × 10-33) and adult polytoxicomania with OR = 17.5 (p = 3.4 × 10-24). Parallel accessibility of genetic data from GRAS patients and N~2100 controls for genome-wide association (GWAS) and phenotype-based genetic association studies (PGAS) permitted the creation of a novel multiple GWAS-PGAS approach. This approach yielded 41 intuitively interesting SNPs, potentially conferring liability to preadult polytoxicomania, which await replication upon availability of suitable deep-phenotyped cohorts anywhere world-wide. Concisely, juvenile environmental risk accumulation, including cannabis and alcohol as starter/gateway drugs, strongly predicts polytoxicomania during adolescence and adulthood. This pivotal message should launch more effective sociopolitical measures to prevent this deleterious psychiatric condition.


Assuntos
Estudo de Associação Genômica Ampla , Esquizofrenia , Adulto , Estudos de Associação Genética , Predisposição Genética para Doença/genética , Humanos , Polimorfismo de Nucleotídeo Único/genética
6.
Mol Psychiatry ; 26(6): 2471-2482, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32089545

RESUMO

Circulating autoantibodies (AB) of different immunoglobulin classes (IgM, IgA, and IgG), directed against the obligatory N-methyl-D-aspartate-receptor subunit NR1 (NMDAR1-AB), belong to the mammalian autoimmune repertoire, and appear with age-dependently high seroprevalence across health and disease. Upon access to the brain, they can exert NMDAR-antagonistic/ketamine-like actions. Still unanswered key questions, addressed here, are conditions of NMDAR1-AB formation/boosting, intraindividual persistence/course in serum over time, and (patho)physiological significance of NMDAR1-AB in modulating neuropsychiatric phenotypes. We demonstrate in a translational fashion from mouse to human that (1) serum NMDAR1-AB fluctuate upon long-term observation, independent of blood-brain barrier (BBB) perturbation; (2) a standardized small brain lesion in juvenile mice leads to increased NMDAR1-AB seroprevalence (IgM + IgG), together with enhanced Ig-class diversity; (3) CTLA4 (immune-checkpoint) genotypes, previously found associated with autoimmune disease, predispose to serum NMDAR1-AB in humans; (4) finally, pursuing our prior findings of an early increase in NMDAR1-AB seroprevalence in human migrants, which implicated chronic life stress as inducer, we independently replicate these results with prospectively recruited refugee minors. Most importantly, we here provide the first experimental evidence in mice of chronic life stress promoting serum NMDAR1-AB (IgA). Strikingly, stress-induced depressive-like behavior in mice and depression/anxiety in humans are reduced in NMDAR1-AB carriers with compromised BBB where NMDAR1-AB can readily reach the brain. To conclude, NMDAR1-AB may have a role as endogenous NMDAR antagonists, formed or boosted under various circumstances, ranging from genetic predisposition to, e.g., tumors, infection, brain injury, and stress, altogether increasing over lifetime, and exerting a spectrum of possible effects, also including beneficial functions.


Assuntos
Autoanticorpos , Lesões Encefálicas , Animais , Barreira Hematoencefálica , Camundongos , Receptores de N-Metil-D-Aspartato , Estudos Soroepidemiológicos , Estresse Psicológico
7.
EClinicalMedicine ; 22: 100345, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32510048

RESUMO

BACKGROUND: Recently, we reported a strong, disease-independent relationship between accumulated preadult environmental risks and violent aggression later in life. Risk factors were interchangeable, and migration was among the explored risks. Alarmed by these data, we assessed collected risk load in young 'healthy' refugees as a specific subgroup of current migration streams and evaluated first signals of behavioral abnormalities. METHODS: In 9 German refugee centers, n = 133 young refugees, not previously in contact with the health system, were recruited, many of them unaccompanied minors. Risk factors experienced apart from migration/refuge were carefully assessed: Traumatic experiences before/during/after flight (including war, genocide, human trafficking, torture, murder, slavery, terrorist attacks), urbanicity, physical and sexual abuse, problematic alcohol and cannabis use (lifetime). Evaluation comprised physical exam and psychopathology screening. FINDINGS: Refugees arrived in Germany via Eastern Mediterranean/Balkan route (34.6%), from Africa via Central Mediterranean route (39.1%), by plane (17.3%) or other routes, such as Western Mediterranean or Atlantic (9.0%). Flight reasons were war/expulsion (25.6%), persecution/threats to life (51.9%), economical/others (22.5%). On top of migration/refuge, 42.8% of subjects had ≥3 risk factors; only 4.5% of refugees had no additional risks. Global level of functioning and severity of psychopathology were strongly associated with number of accumulated risks (Jonckheere-Terpstra trend-test: p = 7.61 × 10-7 and p = 3.62 × 10-7, respectively). INTERPRETATION: Young refugees, arriving in hosting countries with alarming 'risk burden', should be considered as highly vulnerable towards development of global functional deficits, behavioral abnormalities, and neuropsychiatric disorders. Rapid proactive integration or sustainable support of those who will return to rebuild their countries are mandatory. FUNDING: The Max Planck Society supported this work.

8.
Mol Med ; 26(1): 58, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546125

RESUMO

In light of the present therapeutic situation in COVID-19, any measure to improve course and outcome of seriously affected individuals is of utmost importance. We recap here evidence that supports the use of human recombinant erythropoietin (EPO) for ameliorating course and outcome of seriously ill COVID-19 patients. This brief expert review grounds on available subject-relevant literature searched until May 14, 2020, including Medline, Google Scholar, and preprint servers. We delineate in brief sections, each introduced by a summary of respective COVID-19 references, how EPO may target a number of the gravest sequelae of these patients. EPO is expected to: (1) improve respiration at several levels including lung, brainstem, spinal cord and respiratory muscles; (2) counteract overshooting inflammation caused by cytokine storm/ inflammasome; (3) act neuroprotective and neuroregenerative in brain and peripheral nervous system. Based on this accumulating experimental and clinical evidence, we finally provide the research design for a double-blind placebo-controlled randomized clinical trial including severely affected patients, which is planned to start shortly.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/tratamento farmacológico , Síndrome da Liberação de Citocina/prevenção & controle , Eritropoetina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Pneumonia Viral/tratamento farmacológico , Medicamentos para o Sistema Respiratório/uso terapêutico , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/imunologia , Tronco Encefálico/virologia , COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/patologia , Síndrome da Liberação de Citocina/virologia , Método Duplo-Cego , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Pandemias , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/imunologia , Nervo Frênico/virologia , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Estudo de Prova de Conceito , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/uso terapêutico , Músculos Respiratórios/efeitos dos fármacos , Músculos Respiratórios/imunologia , Músculos Respiratórios/virologia , SARS-CoV-2 , Índice de Gravidade de Doença , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/virologia
9.
FASEB J ; 33(7): 8634-8647, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31090455

RESUMO

Reduced expression of 2'-3'-cyclic nucleotide 3'-phosphodiesterase (Cnp) in humans and mice causes white matter inflammation and catatonic signs. These consequences are experimentally alleviated by microglia ablation via colony-stimulating factor 1 receptor (CSF1R) inhibition using PLX5622. Here we address for the first time preclinical topics crucial for translation, most importantly 1) the comparison of 2 long-term PLX5622 applications (prevention and treatment) vs. 1 treatment alone, 2) the correlation of catatonic signs and executive dysfunction, 3) the phenotype of leftover microglia evading depletion, and 4) the role of intercellular interactions for efficient CSF1R inhibition. Based on our Cnp-/- mouse model and in vitro time-lapse imaging, we report the unexpected discovery that microglia surviving under PLX5622 display a highly inflammatory phenotype including aggressive premortal phagocytosis of oligodendrocyte precursor cells. Interestingly, ablating microglia in vitro requires mixed glial cultures, whereas cultured pure microglia withstand PLX5622 application. Importantly, 2 extended rounds of CSF1R inhibition are not superior to 1 treatment regarding any readout investigated (magnetic resonance imaging and magnetic resonance spectroscopy, behavior, immunohistochemistry). Catatonia-related executive dysfunction and brain atrophy of Cnp-/- mice fail to improve under PLX5622. To conclude, even though microglia depletion is temporarily beneficial and worth pursuing, complementary treatment strategies are needed for full and lasting recovery.-Fernandez Garcia-Agudo, L., Janova, H., Sendler, L. E., Arinrad, S., Steixner, A. A., Hassouna, I., Balmuth, E., Ronnenberg, A., Schopf, N., van der Flier, F. J., Begemann, M., Martens, H., Weber, M. S., Boretius, S., Nave, K.-A., Ehrenreich, H. Genetically induced brain inflammation by Cnp deletion transiently benefits from microglia depletion.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , Encéfalo/patologia , Encefalite/genética , Microglia/patologia , Deleção de Sequência/genética , Adulto , Animais , Encéfalo/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Compostos Orgânicos/farmacologia , Fenótipo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Deleção de Sequência/efeitos dos fármacos
10.
Mol Psychiatry ; 24(10): 1489-1501, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29426955

RESUMO

Autoantibodies of the IgG class against N-methyl-D-aspartate-receptor subunit-NR1 (NMDAR1-AB) were considered pathognomonic for anti-NMDAR encephalitis. This view has been challenged by the age-dependent seroprevalence (up to >20%) of functional NMDAR1-AB of all immunoglobulin classes found in >5000 individuals, healthy or affected by different diseases. These findings question a merely encephalitogenic role of NMDAR1-AB. Here, we show that NMDAR1-AB belong to the normal autoimmune repertoire of dogs, cats, rats, mice, baboons, and rhesus macaques, and are functional in the NMDAR1 internalization assay based on human IPSC-derived cortical neurons. The age dependence of seroprevalence is lost in nonhuman primates in captivity and in human migrants, raising the intriguing possibility that chronic life stress may be related to NMDAR1-AB formation, predominantly of the IgA class. Active immunization of ApoE-/- and ApoE+/+ mice against four peptides of the extracellular NMDAR1 domain or ovalbumin (control) leads to high circulating levels of specific AB. After 4 weeks, the endogenously formed NMDAR1-AB (IgG) induce psychosis-like symptoms upon MK-801 challenge in ApoE-/- mice, characterized by an open blood-brain barrier, but not in their ApoE+/+ littermates, which are indistinguishable from ovalbumin controls. Importantly, NMDAR1-AB do not induce any sign of inflammation in the brain. Immunohistochemical staining for microglial activation markers and T lymphocytes in the hippocampus yields comparable results in ApoE-/- and ApoE+/+ mice, irrespective of immunization against NMDAR1 or ovalbumin. These data suggest that NMDAR1-AB of the IgG class shape behavioral phenotypes upon access to the brain but do not cause brain inflammation on their own.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato/imunologia , Transtornos Mentais/imunologia , Receptores de N-Metil-D-Aspartato/imunologia , Adulto , Animais , Autoanticorpos/imunologia , Barreira Hematoencefálica , Encéfalo/imunologia , Gatos , Cães , Feminino , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Masculino , Camundongos , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Primatas , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Estudos Soroepidemiológicos
11.
Mol Psychiatry ; 24(10): 1549-1564, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29795411

RESUMO

Early exposure to negative environmental impact shapes individual behavior and potentially contributes to any mental disease. We reported previously that accumulated environmental risk markedly decreases age at schizophrenia onset. Follow-up of matched extreme group individuals (≤1 vs. ≥3 risks) unexpectedly revealed that high-risk subjects had >5 times greater probability of forensic hospitalization. In line with longstanding sociological theories, we hypothesized that risk accumulation before adulthood induces violent aggression and criminal conduct, independent of mental illness. We determined in 6 independent cohorts (4 schizophrenia and 2 general population samples) pre-adult risk exposure, comprising urbanicity, migration, physical and sexual abuse as primary, and cannabis or alcohol as secondary hits. All single hits by themselves were marginally associated with higher violent aggression. Most strikingly, however, their accumulation strongly predicted violent aggression (odds ratio 10.5). An epigenome-wide association scan to detect differential methylation of blood-derived DNA of selected extreme group individuals yielded overall negative results. Conversely, determination in peripheral blood mononuclear cells of histone-deacetylase1 mRNA as 'umbrella mediator' of epigenetic processes revealed an increase in the high-risk group, suggesting lasting epigenetic alterations. Together, we provide sound evidence of a disease-independent unfortunate relationship between well-defined pre-adult environmental hits and violent aggression, calling for more efficient prevention.


Assuntos
Agressão/psicologia , Violência/psicologia , Adolescente , Adulto , Experiências Adversas da Infância , Epigênese Genética/genética , Exposição à Violência/psicologia , Feminino , Histona Desacetilase 1/genética , Humanos , Masculino , Razão de Chances , Fatores de Risco , Esquizofrenia/epidemiologia , Esquizofrenia/genética
12.
Nat Med ; 24(6): 792-801, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29808008

RESUMO

Defining the environmental context in which genes enhance disease susceptibility can provide insight into the pathogenesis of complex disorders. We report that the intra-uterine environment modulates the association of schizophrenia with genomic risk (in this study, genome-wide association study-derived polygenic risk scores (PRSs)). In independent samples from the United States, Italy, and Germany, the liability of schizophrenia explained by PRS is more than five times greater in the presence of early-life complications (ELCs) compared with their absence. Patients with ELC histories have significantly higher PRS than patients without ELC histories, which is confirmed in additional samples from Germany and Japan. The gene set composed of schizophrenia loci that interact with ELCs is highly expressed in placenta, is differentially expressed in placentae from complicated in comparison with normal pregnancies, and is differentially upregulated in placentae from male compared with female offspring. Pathway analyses reveal that genes driving the PRS-ELC interaction are involved in cellular stress response; genes that do not drive such interaction implicate orthogonal biological processes (for example, synaptic function). We conclude that a subset of the most significant genetic variants associated with schizophrenia converge on a developmental trajectory sensitive to events that affect the placental response to stress, which may offer insights into sex biases and primary prevention.


Assuntos
Predisposição Genética para Doença , Placenta/patologia , Esquizofrenia/genética , Estudos de Casos e Controles , Feminino , Regulação da Expressão Gênica , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Acontecimentos que Mudam a Vida , Masculino , Herança Multifatorial/genética , Placenta/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Gravidez , Fatores de Risco , Caracteres Sexuais
13.
J Clin Invest ; 128(2): 734-745, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29252214

RESUMO

The underlying cellular mechanisms of catatonia, an executive "psychomotor" syndrome that is observed across neuropsychiatric diseases, have remained obscure. In humans and mice, reduced expression of the structural myelin protein CNP is associated with catatonic signs in an age-dependent manner, pointing to the involvement of myelin-producing oligodendrocytes. Here, we showed that the underlying cause of catatonic signs is the low-grade inflammation of white matter tracts, which marks a final common pathway in Cnp-deficient and other mutant mice with minor myelin abnormalities. The inhibitor of CSF1 receptor kinase signaling PLX5622 depleted microglia and alleviated the catatonic symptoms of Cnp mutants. Thus, microglia and low-grade inflammation of myelinated tracts emerged as the trigger of a previously unexplained mental condition. We observed a very high (25%) prevalence of individuals with catatonic signs in a deeply phenotyped schizophrenia sample (n = 1095). Additionally, we found the loss-of-function allele of a myelin-specific gene (CNP rs2070106-AA) associated with catatonia in 2 independent schizophrenia cohorts and also associated with white matter hyperintensities in a general population sample. Since the catatonic syndrome is likely a surrogate marker for other executive function defects, we suggest that microglia-directed therapies may be considered in psychiatric disorders associated with myelin abnormalities.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , Catatonia/patologia , Microglia/citologia , Bainha de Mielina/química , Adulto , Fatores Etários , Alelos , Animais , Encéfalo/patologia , Catatonia/prevenção & controle , Feminino , Genótipo , Humanos , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Oligodendroglia/citologia , Compostos Orgânicos/química , Fenótipo , Prevalência , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Esquizofrenia/genética , Substância Branca/patologia
14.
EMBO Mol Med ; 9(8): 1100-1116, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28623238

RESUMO

Matrix metalloproteinase 9 (MMP-9) has recently emerged as a molecule that contributes to pathological synaptic plasticity in schizophrenia, but explanation of the underlying mechanisms has been missing. In the present study, we performed a phenotype-based genetic association study (PGAS) in > 1,000 schizophrenia patients from the Göttingen Research Association for Schizophrenia (GRAS) data collection and found an association between the MMP-9 rs20544 C/T single-nucleotide polymorphism (SNP) located in the 3'untranslated region (UTR) and the severity of a chronic delusional syndrome. In cultured neurons, the rs20544 SNP influenced synaptic MMP-9 activity and the morphology of dendritic spines. We demonstrated that Fragile X mental retardation protein (FMRP) bound the MMP-9 3'UTR We also found dramatic changes in RNA structure folding and alterations in the affinity of FMRP for MMP-9 RNA, depending on the SNP variant. Finally, we observed greater sensitivity to psychosis-related locomotor hyperactivity in Mmp-9 heterozygous mice. We propose a novel mechanism that involves MMP-9-dependent changes in dendritic spine morphology and the pathophysiology of schizophrenia, providing the first mechanistic insights into the way in which the single base change in the MMP-9 gene (rs20544) influences gene function and results in phenotypic changes observed in schizophrenia patients.


Assuntos
Metaloproteinase 9 da Matriz/análise , Metaloproteinase 9 da Matriz/genética , Polimorfismo de Nucleotídeo Único , Esquizofrenia Paranoide/patologia , Sinapses/enzimologia , Regiões 3' não Traduzidas , Adolescente , Adulto , Idoso , Animais , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Feminino , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Estudos de Associação Genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios/citologia , Conformação de Ácido Nucleico , Ligação Proteica , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Adulto Jovem
15.
EMBO Mol Med ; 7(12): 1565-79, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26612855

RESUMO

Fragile X syndrome (FXS) is mostly caused by a CGG triplet expansion in the fragile X mental retardation 1 gene (FMR1). Up to 60% of affected males fulfill criteria for autism spectrum disorder (ASD), making FXS the most frequent monogenetic cause of syndromic ASD. It is unknown, however, whether normal variants (independent of mutations) in the fragile X gene family (FMR1, FXR1, FXR2) and in FMR2 modulate autistic features. Here, we report an accumulation model of 8 SNPs in these genes, associated with autistic traits in a discovery sample of male patients with schizophrenia (N = 692) and three independent replicate samples: patients with schizophrenia (N = 626), patients with other psychiatric diagnoses (N = 111) and a general population sample (N = 2005). For first mechanistic insight, we contrasted microRNA expression in peripheral blood mononuclear cells of selected extreme group subjects with high- versus low-risk constellation regarding the accumulation model. Thereby, the brain-expressed miR-181 species emerged as potential "umbrella regulator", with several seed matches across the fragile X gene family and FMR2. To conclude, normal variation in these genes contributes to the continuum of autistic phenotypes.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Proteínas de Ligação a RNA/genética , Transtorno Autístico/genética , Síndrome do Cromossomo X Frágil/fisiopatologia , Humanos , Masculino , MicroRNAs/biossíntese , Mutação , Polimorfismo de Nucleotídeo Único , Esquizofrenia/genética , Esquizofrenia/fisiopatologia
16.
Am J Hum Genet ; 97(5): 738-43, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26456283

RESUMO

The magnitude of the human antibody response to viral antigens is highly variable. To explore the human genetic contribution to this variability, we performed genome-wide association studies of the immunoglobulin G response to 14 pathogenic viruses in 2,363 immunocompetent adults. Significant associations were observed in the major histocompatibility complex region on chromosome 6 for influenza A virus, Epstein-Barr virus, JC polyomavirus, and Merkel cell polyomavirus. Using local imputation and fine mapping, we identified specific amino acid residues in human leucocyte antigen (HLA) class II proteins as the most probable causal variants underlying these association signals. Common HLA-DRß1 haplotypes showed virus-specific patterns of humoral-response regulation. We observed an overlap between variants affecting the humoral response to influenza A and EBV and variants previously associated with autoimmune diseases related to these viruses. The results of this study emphasize the central and pathogen-specific role of HLA class II variation in the modulation of humoral immune response to viral antigens in humans.


Assuntos
Doenças Autoimunes/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Imunidade Humoral/imunologia , Hospedeiro Imunocomprometido/imunologia , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/imunologia , Viroses/imunologia , Vírus/imunologia , Adulto , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Estudos de Casos e Controles , Feminino , Seguimentos , Estudo de Associação Genômica Ampla , Antígenos de Histocompatibilidade Classe II/química , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulina G/sangue , Masculino , Prognóstico , Conformação Proteica , Esquizofrenia/genética , Esquizofrenia/patologia , Viroses/genética , Viroses/virologia
17.
BMC Psychiatry ; 15: 115, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25968177

RESUMO

BACKGROUND: Behavioral phenotypical continua from health to disease suggest common underlying mechanisms with quantitative rather than qualitative differences. Until recently, autism spectrum disorders and schizophrenia were considered distinct nosologic entities. However, emerging evidence contributes to the blurring of symptomatic and genetic boundaries between these conditions. The present study aimed at quantifying behavioral phenotypes shared by autism spectrum disorders and schizophrenia to prepare the ground for biological pathway analyses. METHODS: Specific items of the Positive and Negative Syndrome Scale were employed and summed up to form a dimensional autism severity score (PAUSS). The score was created in a schizophrenia sample (N = 1156) and validated in adult high-functioning autism spectrum disorder (ASD) patients (N = 165). To this end, the Autism Diagnostic Observation Schedule (ADOS), the Autism (AQ) and Empathy Quotient (EQ) self-rating questionnaires were applied back to back with the newly developed PAUSS. RESULTS: PAUSS differentiated between ASD, schizophrenia and a disease-control sample and substantially correlated with the Autism Diagnostic Observation Schedule. Patients with ADOS scores ≥12 obtained highest, those with scores <7 lowest PAUSS values. AQ and EQ were not found to vary dependent on ADOS diagnosis. ROC curves for ADOS and PAUSS resulted in AuC values of 0.9 and 0.8, whereas AQ and EQ performed at chance level in the prediction of ASD. CONCLUSIONS: This work underscores the convergence of schizophrenia negative symptoms and autistic phenotypes. PAUSS evolved as a measure capturing the continuous nature of autistic behaviors. The definition of extreme-groups based on the dimensional PAUSS may permit future investigations of genetic constellations modulating autistic phenotypes.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/diagnóstico , Fenótipo , Escalas de Graduação Psiquiátrica , Esquizofrenia/diagnóstico , Adulto , Estudos de Casos e Controles , Erros de Diagnóstico , Empatia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Psicometria , Sensibilidade e Especificidade , Índice de Gravidade de Doença , Inquéritos e Questionários , Adulto Jovem
18.
Bipolar Disord ; 16(8): 881-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24964373

RESUMO

OBJECTIVES: Over 12% of patients with bipolar disorder exhibit rapid cycling. The underlying biological mechanisms of this extreme form of bipolar disease are still unknown. This study aimed at replicating and extending findings of our previously published case report, where an involvement of prostaglandin synthesis-related genes in rapid cycling was first proposed. METHODS: Psychopathological follow-up of the reported case was performed under cessation of celecoxib treatment. In a prospective observational study, patients with bipolar disorder (n = 47; of these, four had rapid cycling) or with monopolar depression (n = 97) were recruited over a period of three years. Repeated psychopathology measurements were conducted using standard instruments. Peripheral blood mononuclear cells (PBMC) were obtained during as many consecutive episodes as possible and processed for mRNA isolation and quantitative real-time reverse transcriptase polymerase chain reaction for prostaglandin D2 synthase (PTGDS), aldo-ketoreductase family 1, member C3 (AKR1C3), cyclooxygenase-2 (PAN means all splice variants) (COX2PAN ), prostaglandin-endoperoxide synthase 2 (PTGS2), and purinergic receptor P2X, ligand-gated ion channel 7 (P2RX7). RESULTS: The follow-up of our original case of a patient with rapid cycling who had shown impressive psychopathological improvement under celecoxib revealed complete loss of this effect upon discontinuation of the COX2 inhibitor. Episode-specific gene expression measurements in PBMC of four newly recruited rapid cycling patients confirmed the higher expression of PTGDS in depressive compared to manic phases. Additionally, higher relative expression of PTGS2/COX2PAN was found. No comparable alterations were observable in samples available from the remaining 43 patients with bipolar disorder and the 97 monopolar depressed patients, emphasizing the advantages of the rapid cycling condition with its rapid and frequent shifts for identification of gene expression changes. CONCLUSIONS: This study supports a role for prostaglandins in rapid cycling and advocates the cyclooxygenase cascade as a treatment target in this condition.


Assuntos
Transtorno Bipolar/sangue , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/metabolismo , Oxirredutases Intramoleculares/metabolismo , Leucócitos Mononucleares/metabolismo , Lipocalinas/metabolismo , Idoso , Antidepressivos/uso terapêutico , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/fisiopatologia , Celecoxib , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Feminino , Seguimentos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Oxirredutases Intramoleculares/genética , Leucócitos Mononucleares/efeitos dos fármacos , Lipocalinas/genética , Masculino , Pessoa de Meia-Idade , Pirazóis/farmacologia , Pirazóis/uso terapêutico , RNA Mensageiro/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico
19.
Ann Neurol ; 76(1): 82-94, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24853231

RESUMO

OBJECTIVE: We previously reported an unexpectedly high seroprevalence (~10%) of N-methyl-D-aspartate-receptor subunit-NR1 (NMDAR1) autoantibodies (AB) in healthy and neuropsychiatrically ill subjects (N = 2,817). This finding challenges an unambiguous causal relationship of serum AB with brain disease. To test whether similar results would be obtained for other brain antigen-directed AB previously connected with pathological conditions, we systematically screened serum samples of 4,236 individuals. METHODS: Serum samples of healthy (n = 1,703) versus neuropsychiatrically ill subjects (schizophrenia, affective disorders, stroke, Parkinson disease, amyotrophic lateral sclerosis, personality disorder; total n = 2,533) were tested. For analysis based on indirect immunofluorescence, we used biochip mosaics of frozen brain sections (rat, monkey) and transfected HEK293 cells expressing respective recombinant target antigens. RESULTS: Seroprevalence of all screened AB was comparable in healthy and ill individuals. None of them, however, reached the abundance of NMDAR1 AB (again ~10%; immunoglobulin [Ig] G ~1%). Appreciable frequency was noted for AB against amphiphysin (2.0%), ARHGAP26 (1.3%), CASPR2 (0.9%), MOG (0.8%), GAD65 (0.5%), Ma2 (0.5%), Yo (0.4%), and Ma1 (0.4%), with titers and Ig class distribution similar among groups. All other AB were found in ≤0.1% of individuals (anti-AMPAR-1/2, AQP4, CV2, Tr/DNER, DPPX-IF1, GABAR-B1/B2, GAD67, GLRA1b, GRM1, GRM5, Hu, LGl1, recoverin, Ri, ZIC4). The predominant Ig class depended on antigen location, with intracellular epitopes predisposing to IgG (chi-square = 218.91, p = 2.8 × 10(-48) ). INTERPRETATION: To conclude, the brain antigen-directed AB tested here are comparably detectable in healthy subjects and the disease groups studied here, thus questioning an upfront pathological role of these serum AB.


Assuntos
Autoanticorpos/sangue , Transtornos Mentais/sangue , Transtornos Mentais/epidemiologia , Doenças do Sistema Nervoso/sangue , Doenças do Sistema Nervoso/epidemiologia , Adulto , Idoso , Animais , Autoanticorpos/biossíntese , Feminino , Alemanha/epidemiologia , Células HEK293 , Haplorrinos , Humanos , Masculino , Programas de Rastreamento , Transtornos Mentais/imunologia , Pessoa de Meia-Idade , Doenças do Sistema Nervoso/imunologia , Ratos , Receptores de N-Metil-D-Aspartato/imunologia , Valores de Referência , Estudos Soroepidemiológicos
20.
EMBO Mol Med ; 6(5): 662-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24648499

RESUMO

The X-chromosomal MECP2/Mecp2 gene encodes methyl-CpG-binding protein 2, a transcriptional activator and repressor regulating many other genes. We discovered in male FVB/N mice that mild (~50%) transgenic overexpression of Mecp2 enhances aggression. Surprisingly, when the same transgene was expressed in C57BL/6N mice, transgenics showed reduced aggression and social interaction. This suggests that Mecp2 modulates aggressive social behavior. To test this hypothesis in humans, we performed a phenotype-based genetic association study (PGAS) in >1000 schizophrenic individuals. We found MECP2 SNPs rs2239464 (G/A) and rs2734647 (C/T; 3'UTR) associated with aggression, with the G and C carriers, respectively, being more aggressive. This finding was replicated in an independent schizophrenia cohort. Allele-specific MECP2 mRNA expression differs in peripheral blood mononuclear cells by ~50% (rs2734647: C > T). Notably, the brain-expressed, species-conserved miR-511 binds to MECP2 3'UTR only in T carriers, thereby suppressing gene expression. To conclude, subtle MECP2/Mecp2 expression alterations impact aggression. While the mouse data provides evidence of an interaction between genetic background and mild Mecp2 overexpression, the human data convey means by which genetic variation affects MECP2 expression and behavior.


Assuntos
Agressão , Predisposição Genética para Doença , Proteína 2 de Ligação a Metil-CpG/biossíntese , MicroRNAs/metabolismo , Animais , Estudos de Coortes , Perfilação da Expressão Gênica , Estudos de Associação Genética , Humanos , Leucócitos Mononucleares , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA