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.
Front Cell Dev Biol ; 9: 669798, 2021.
Article in English | MEDLINE | ID: mdl-34307355

ABSTRACT

Many neurological disorders exhibit sex differences and sex-specific therapeutic responses. Unfortunately, significant amounts of studies investigating molecular and cellular mechanisms underlying these neurological disorders use primary cell cultures with undetermined sexes; and this may be a source for contradictory results among different studies and impair the validity of study conclusion. Herein, we comprehensively compared sexual dimorphism of gene expression in primary neurons, astrocytes, and microglia derived from neonatal mouse brains. We found that overall sexually dimorphic gene numbers were relatively low in these primary cells, with microglia possessing the most (264 genes), neurons possessing the medium (69 genes), and astrocytes possessing the least (30 genes). KEGG analysis indicated that sexually dimorphic genes in these three cell types were strongly enriched for the immune system and immune-related diseases. Furthermore, we identified that sexually dimorphic genes shared by these primary cells dominantly located on the Y chromosome, including Ddx3y, Eif2s3y, Kdm5d, and Uty. Finally, we demonstrated that overexpression of Eif2s3y increased synaptic transmission specifically in male neurons and caused autism-like behaviors specifically in male mice. Together, our results demonstrate that the sex of primary cells should be considered when these cells are used for studying the molecular mechanism underlying neurological disorders with sex-biased susceptibility, especially those related to immune dysfunction. Moreover, our findings indicate that dysregulation of sexually dimorphic genes on the Y chromosome may also result in autism and possibly other neurological disorders, providing new insights into the genetic driver of sex differences in neurological disorders.

2.
Exp Mol Med ; 51(7): 1-11, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337748

ABSTRACT

Focal cortical dysplasia type II (FCDII) is a cerebral cortex malformation characterized by local cortical structure disorganization, neuronal dysmorphology, and refractory epilepsy. Brain somatic mutations in several genes involved in the PI3K/AKT/mTOR pathway are associated with FCDII, but they are only found in a proportion of patients with FCDII. The genetic causes underlying the development FCDII in other patients remain unclear. Here, we carried out whole exome sequencing and targeted sequencing in paired brain-blood DNA from patients with FCDII and identified a brain somatic doublet mutation c.(A104T, C105A) in the Ras homolog, mTORC1 binding (RHEB) gene, which led to the RHEB p.Y35L mutation in one patient with FCDII. This RHEB mutation carrier had a dramatic increase of ribosomal protein S6 phosphorylation, indicating mTOR activation in the region of the brain lesion. The RHEB p.Y35L mutant protein had increased GTPλS-binding activity compared with wild-type RHEB. Overexpression of the RHEB p.Y35L variant in cultured cells also resulted in elevated S6 phosphorylation compared to wild-type RHEB. Importantly, in utero electroporation of the RHEB p.Y35L variant in mice induced S6 phosphorylation, cytomegalic neurons, dysregulated neuron migration, abnormal electroencephalogram, and seizures, all of which are found in patients with FCDII. Rapamycin treatment rescued abnormal electroencephalograms and alleviated seizures in these mice. These results demonstrate that brain somatic mutations in RHEB are also responsible for the pathogenesis of FCDII, indicating that aberrant activation of mTOR signaling is a primary driver and potential drug target for FCDII.


Subject(s)
Epilepsy/genetics , Malformations of Cortical Development, Group I/genetics , Phosphatidylinositol 3-Kinases/genetics , Ras Homolog Enriched in Brain Protein/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Brain/diagnostic imaging , Brain/pathology , Epilepsy/diagnostic imaging , Epilepsy/pathology , Female , Humans , Male , Malformations of Cortical Development, Group I/diagnostic imaging , Malformations of Cortical Development, Group I/pathology , Mice , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Ras Homolog Enriched in Brain Protein/metabolism , Sequence Alignment , TOR Serine-Threonine Kinases/metabolism , Exome Sequencing
3.
Biol Psychiatry ; 86(3): 171-184, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30292394

ABSTRACT

BACKGROUND: Although synaptic impairment is a prerequisite to cognitive deficiencies in Alzheimer's disease (AD), mechanisms underlying the dysregulation of essential synaptic scaffolding components and their integrity remain elusive. RPS23RG1 is a newly identified protein implicated in AD. However, the physiological function of RPS23RG1 has yet to be determined. METHODS: We investigated the role of RPS23RG1 in maintaining synaptic structure and function in cell cultures and in Rps23rg1 knockout mice and determined whether targeting RPS23RG1-mediated pathways has therapeutic potential in APP/PS1 AD model mice. RESULTS: Deletion of the Rps23rg1 gene resulted in severe memory deficits and impairment of postsynaptic structure and function, with marked reductions in postsynaptic densities-93 and -95 (PSD-93 and PSD-95) levels. RPS23RG1 interacted with PSD-93/PSD-95 through its intracellular domain, consequently sequestering PSD-93/PSD-95 from murine double minute 2-mediated ubiquitination and degradation, thereby maintaining synaptic function. Restoration of PSD-93/PS-D95 levels reversed synaptic and memory deficits in Rps23rg1 knockout mice. We further observed attenuated RPS23RG1 expression in human AD, which positively correlated with PSD-93/PSD-95 levels. Importantly, an RPS23RG1-derived peptide comprising a unique PSD-93/PSD-95 interaction motif rescued synaptic and cognitive defects in Rps23rg1 knockout and AD mouse models. CONCLUSIONS: Our results reveal a role for RPS23RG1 in maintaining synaptic integrity and function and provide a new mechanism for synaptic dysfunction in AD pathogenesis. This demonstrates that RPS23RG1-mediated pathways show good therapeutic potential in AD intervention.


Subject(s)
Cognitive Dysfunction/metabolism , Disks Large Homolog 4 Protein/metabolism , Neuronal Plasticity , Ribosomal Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Gene Expression Regulation , Gene Knockdown Techniques , Hippocampus/metabolism , Humans , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Ribosomal Proteins/genetics
4.
Neuron ; 97(5): 1023-1031.e7, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29518356

ABSTRACT

Mutations in triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to increased Alzheimer's disease (AD) risk. Neurobiological functions of TREM2 and its pathophysiological ligands remain elusive. Here we found that TREM2 directly binds to ß-amyloid (Aß) oligomers with nanomolar affinity, whereas AD-associated TREM2 mutations reduce Aß binding. TREM2 deficiency impairs Aß degradation in primary microglial culture and mouse brain. Aß-induced microglial depolarization, K+ inward current induction, cytokine expression and secretion, migration, proliferation, apoptosis, and morphological changes are dependent on TREM2. In addition, TREM2 interaction with its signaling adaptor DAP12 is enhanced by Aß, regulating downstream phosphorylation of SYK and GSK3ß. Our data demonstrate TREM2 as a microglial Aß receptor transducing physiological and AD-related pathological effects associated with Aß.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Membrane Glycoproteins/metabolism , Microglia/physiology , Receptors, Immunologic/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Brain/pathology , Cells, Cultured , Double-Blind Method , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/pathology , Protein Binding/physiology
5.
Exp Mol Med ; 49(12): e405, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29611543

ABSTRACT

Genetic mutations in triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to a variety of neurodegenerative diseases including Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia and Parkinson's disease. In the brain, TREM2 is highly expressed on the cell surface of microglia, where it can transduce signals to regulate microglial functions such as phagocytosis. To date, mechanisms underlying intracellular trafficking of TREM2 remain elusive. Mutations in the presenilin 1 (PS1) catalytic subunit of the γ-secretase complex have been associated with increased generation of the amyloidogenic Aß (amyloid-ß) 42 peptide through cleavage of the Aß precursor amyloid precursor protein. Here we found that TREM2 interacts with PS1 in a manner independent of γ-secretase activity. Mutations in TREM2 alter its subcellular localization and affects its interaction with PS1. Upregulation of PS1 reduces, whereas downregulation of PS1 increases, steady-state levels of cell surface TREM2. Furthermore, PS1 overexpression results in attenuated phagocytic uptake of Aß by microglia, which is reversed by TREM2 overexpression. Our data indicate a novel role for PS1 in regulating TREM2 intracellular trafficking and pathophysiological function.


Subject(s)
Membrane Glycoproteins/metabolism , Presenilin-1/metabolism , Receptors, Immunologic/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Biotinylation , Cell Line , HEK293 Cells , Humans , Immunoprecipitation , Membrane Glycoproteins/genetics , Mice , Mutation/genetics , Phagocytosis/genetics , Phagocytosis/physiology , Presenilin-1/genetics , Protein Transport/genetics , Protein Transport/physiology , Receptors, Immunologic/genetics
6.
Mol Neurobiol ; 54(6): 4189-4200, 2017 08.
Article in English | MEDLINE | ID: mdl-27324899

ABSTRACT

TMEM59L is a newly identified brain-specific membrane-anchored protein with unknown functions. Herein we found that both TMEM59L and its homolog, TMEM59, are localized in Golgi and endosomes. However, in contrast to a ubiquitous and relatively stable temporal expression of TMEM59, TMEM59L expression was limited in neurons and increased during development. We also found that both TMEM59L and TMEM59 interacted with ATG5 and ATG16L1, and that overexpression of them triggered cell autophagy. However, overexpression of TMEM59L induced intrinsic caspase-dependent apoptosis more dramatically than TMEM59. In addition, downregulation of TMEM59L prevented neuronal cell death and caspase-3 activation caused by hydrogen peroxide insults and reduced the lipidation of LC3B. Finally, we found that AAV-mediated knockdown of TMEM59L in mice significantly ameliorated caspase-3 activation, increased mouse duration in the open arm during elevated plus maze test, reduced mouse immobility time during forced swim test, and enhanced mouse memory during Y-maze and Morris water maze tests. Together, our study indicates that TMEM59L is a pro-apoptotic neuronal protein involved in animal behaviors such as anxiety, depression, and memory, and that TMEM59L downregulation protects neurons against oxidative stress.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Neurons/metabolism , Oxidative Stress , Animals , Anxiety/pathology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , Autophagy-Related Proteins , Behavior, Animal , Carrier Proteins/metabolism , Caspase 3/metabolism , Dependovirus/metabolism , Depression/pathology , Down-Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Hydrogen Peroxide/toxicity , Memory/drug effects , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neuroprotection/drug effects , Oxidative Stress/drug effects , Protein Binding/drug effects , Protein Transport/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...