Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Glia ; 70(4): 650-660, 2022 04.
Article in English | MEDLINE | ID: mdl-34936134

ABSTRACT

Previous studies have implicated several brain cell types in schizophrenia (SCZ), but the genetic impact of astrocytes is unknown. Considering their high complexity in humans, astrocytes are likely key determinants of neurodevelopmental diseases, such as SCZ. Human induced pluripotent stem cell (hiPSC)-derived astrocytes differentiated from five monozygotic twin pairs discordant for SCZ and five healthy subjects were studied for alterations related to high genetic risk and clinical manifestation of SCZ in astrocyte transcriptomics, neuron-astrocyte co-cultures, and in humanized mice. We found gene expression and signaling pathway alterations related to synaptic dysfunction, inflammation, and extracellular matrix components in SCZ astrocytes, and demyelination in SCZ astrocyte transplanted mice. While Ingenuity Pathway Analysis identified SCZ disease and synaptic transmission pathway changes in SCZ astrocytes, the most consistent findings were related to collagen and cell adhesion associated pathways. Neuronal responses to glutamate and GABA differed between astrocytes from control persons, affected twins, and their unaffected co-twins and were normalized by clozapine treatment. SCZ astrocyte cell transplantation to the mouse forebrain caused gene expression changes in synaptic dysfunction and inflammation pathways of mouse brain cells and resulted in behavioral changes in cognitive and olfactory functions. Differentially expressed transcriptomes and signaling pathways related to synaptic functions, inflammation, and especially collagen and glycoprotein 6 pathways indicate abnormal extracellular matrix composition in the brain as one of the key characteristics in the etiology of SCZ.


Subject(s)
Induced Pluripotent Stem Cells , Schizophrenia , Animals , Astrocytes/metabolism , Genetic Predisposition to Disease/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Prosencephalon/metabolism , Schizophrenia/genetics
2.
Mol Psychiatry ; 25(12): 3432-3441, 2020 12.
Article in English | MEDLINE | ID: mdl-31455857

ABSTRACT

Psychopathy is an extreme form of antisocial behavior, with about 1% prevalence in the general population, and 10-30% among incarcerated criminal offenders. Although the heritability of severe antisocial behavior is up to 50%, the genetic background is unclear. The underlying molecular mechanisms have remained unknown but several previous studies suggest that abnormal glucose metabolism and opioidergic neurotransmission contribute to violent offending and psychopathy. Here we show using iPSC-derived cortical neurons and astrocytes from six incarcerated extremely antisocial and violent offenders, three nonpsychopathic individuals with substance abuse, and six healthy controls that there are robust alterations in the expression of several genes and immune response-related molecular pathways which were specific for psychopathy. In neurons, psychopathy was associated with marked upregulation of RPL10P9 and ZNF132, and downregulation of CDH5 and OPRD1. In astrocytes, RPL10P9 and MT-RNR2 were upregulated. Expression of aforementioned genes explained 30-92% of the variance of psychopathic symptoms. The gene expression findings were confirmed with qPCR. These genes may be relevant to the lack of empathy and emotional callousness seen in psychopathy, since several studies have linked these genes to autism and social interaction.


Subject(s)
Antisocial Personality Disorder , Criminals , Aggression , Antisocial Personality Disorder/genetics , Emotions , Empathy , Humans
3.
Mol Psychiatry ; 25(12): 3455-3456, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31570776

ABSTRACT

A correction to this paper has been published and can be accessed via a link at the top of the paper.

4.
Glia ; 68(3): 589-599, 2020 03.
Article in English | MEDLINE | ID: mdl-31670864

ABSTRACT

Alzheimer's disease (AD) is a common dementia affecting a vast number of individuals and significantly impairing quality of life. Despite extensive research in animal models and numerous promising treatment trials, there is still no curative treatment for AD. Astrocytes, the most common cell type of the central nervous system, have been shown to play a role in the major AD pathologies, including accumulation of amyloid plaques, neuroinflammation, and oxidative stress. Here, we show that inflammatory stimulation leads to metabolic activation of human astrocytes and reduces amyloid secretion. On the other hand, the activation of oxidative metabolism leads to increased reactive oxygen species production especially in AD astrocytes. While healthy astrocytes increase glutathione (GSH) release to protect the cells, Presenilin-1-mutated AD patient astrocytes do not. Thus, chronic inflammation is likely to induce oxidative damage in AD astrocytes. Activation of NRF2, the major regulator of cellular antioxidant defenses, encoded by the NFE2L2 gene, poses several beneficial effects on AD astrocytes. We report here that the activation of NRF2 pathway reduces amyloid secretion, normalizes cytokine release, and increases GSH secretion in AD astrocytes. NRF2 induction also activates the metabolism of astrocytes and increases the utilization of glycolysis. Taken together, targeting NRF2 in astrocytes could be a potent therapeutic strategy in AD.


Subject(s)
Alzheimer Disease/metabolism , Antioxidants/pharmacology , Astrocytes/metabolism , NF-E2-Related Factor 2/metabolism , Presenilin-1/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Animals , Astrocytes/drug effects , Disease Models, Animal , Humans , Inflammation/metabolism , Plaque, Amyloid/metabolism
5.
Nat Commun ; 10(1): 3933, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477693

ABSTRACT

It has remained unclear why schizophrenia typically manifests after adolescence and which neurobiological mechanisms are underlying the cascade leading to the actual onset of the illness. Here we show that the use of induced pluripotent stem cell-derived neurons of monozygotic twins from pairs discordant for schizophrenia enhances disease-specific signal by minimizing genetic heterogeneity. In proteomic and pathway analyses, clinical illness is associated especially with altered glycosaminoglycan, GABAergic synapse, sialylation, and purine metabolism pathways. Although only 12% of all 19,462 genes are expressed differentially between healthy males and females, up to 61% of the illness-related genes are sex specific. These results on sex-specific genes are replicated in another dataset. This implies that the pathophysiology differs between males and females, and may explain why symptoms appear after adolescence when the expression of many sex-specific genes change, and suggests the need for sex-specific treatments.


Subject(s)
Gene Expression Profiling/methods , Proteome/genetics , Proteomics/methods , Schizophrenia/genetics , Adolescent , Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Diseases in Twins/genetics , Diseases in Twins/metabolism , Female , Humans , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteome/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Sex Factors , Twins, Monozygotic/genetics
6.
Stem Cell Res ; 31: 181-185, 2018 08.
Article in English | MEDLINE | ID: mdl-30099334

ABSTRACT

A double mutation (KM670/671NL) in amyloid precursor protein gene (APP) is causative for familial Alzheimer's disease and has been shown to increase the total Aß burden. Here we report the generation and characterization of an iPSC line from a fAD patient carrying APP KM670/671NL. The generated iPSCs retained the mutation, expressed pluripotency markers, showed a normal karyotype and differentiated into all three germ layers. This iPSC line can be used, for example, in disease modeling and mechanistic studies. Resource table.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Induced Pluripotent Stem Cells/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Humans , Mutation , Sweden
SELECTION OF CITATIONS
SEARCH DETAIL
...