Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Autism ; 15(1): 14, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570876

ABSTRACT

BACKGROUND: SH3 and multiple ankyrin repeat domains protein 3 (SHANK3) monogenic mutations or deficiency leads to excessive stereotypic behavior and impaired sociability, which frequently occur in autism cases. To date, the underlying mechanisms by which Shank3 mutation or deletion causes autism and the part of the brain in which Shank3 mutation leads to the autistic phenotypes are understudied. The hypothalamus is associated with stereotypic behavior and sociability. p38α, a mediator of inflammatory responses in the brain, has been postulated as a potential gene for certain cases of autism occurrence. However, it is unclear whether hypothalamus and p38α are involved in the development of autism caused by Shank3 mutations or deficiency. METHODS: Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and immunoblotting were used to assess alternated signaling pathways in the hypothalamus of Shank3 knockout (Shank3-/-) mice. Home-Cage real-time monitoring test was performed to record stereotypic behavior and three-chamber test was used to monitor the sociability of mice. Adeno-associated viruses 9 (AAV9) were used to express p38α in the arcuate nucleus (ARC) or agouti-related peptide (AgRP) neurons. D176A and F327S mutations expressed constitutively active p38α. T180A and Y182F mutations expressed inactive p38α. RESULTS: We found that Shank3 controls stereotypic behavior and sociability by regulating p38α activity in AgRP neurons. Phosphorylated p38 level in hypothalamus is significantly enhanced in Shank3-/- mice. Consistently, overexpression of p38α in ARC or AgRP neurons elicits excessive stereotypic behavior and impairs sociability in wild-type (WT) mice. Notably, activated p38α in AgRP neurons increases stereotypic behavior and impairs sociability. Conversely, inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. In contrast, activated p38α in pro-opiomelanocortin (POMC) neurons does not affect stereotypic behavior and sociability in mice. LIMITATIONS: We demonstrated that SHANK3 regulates the phosphorylated p38 level in the hypothalamus and inactivated p38α in AgRP neurons significantly ameliorates autistic behaviors of Shank3-/- mice. However, we did not clarify the biochemical mechanism of SHANK3 inhibiting p38α in AgRP neurons. CONCLUSIONS: These results demonstrate that the Shank3 deficiency caused autistic-like behaviors by activating p38α signaling in AgRP neurons, suggesting that p38α signaling in AgRP neurons is a potential therapeutic target for Shank3 mutant-related autism.


Subject(s)
Autistic Disorder , Animals , Mice , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Hypothalamus/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Mitogen-Activated Protein Kinase 14/metabolism
3.
Neurobiol Dis ; 179: 106061, 2023 04.
Article in English | MEDLINE | ID: mdl-36870457

ABSTRACT

Mounting evidence indicates complex interaction between the immune system and the nervous system, challenging the traditional view about the immune privilege of the brain. Innate lymphoid cells (ILCs) and innate-like T cells are unique families of immune cells that functionally mirror traditional T cells but may function via antigen- and T cell antigen receptor (TCR)-independent mechanisms. Recent work indicates that various ILCs and innate-like T cell subsets are present in the brain barrier tissue, where they play important roles in regulating brain barrier integrity, brain homeostasis and cognitive function. In this review, we discuss recent advances in understanding the intricate roles for innate and innate-like lymphocytes in regulating brain and cognitive function.


Subject(s)
Immunity, Innate , Lymphocytes , T-Lymphocytes , Brain , Cognition
4.
Sleep Med ; 103: 146-158, 2023 03.
Article in English | MEDLINE | ID: mdl-36805914

ABSTRACT

Alzheimer's disease (AD) patients exhibit sleep and circadian disturbances prior to the onset of cognitive decline, and these disruptions worsen with disease severity. However, the molecular mechanisms behind sleep and circadian disruptions in AD patients are poorly understood. In this study, we investigated sleep pattern and circadian rhythms in Presenilin-1/2 conditional knockout (DKO) mice. Assessment of EEG and EMG recordings showed that DKO mice displayed increased NREM sleep time but not REM sleep during the dark phase compared to WT mice at the age of two months; at the age of six months, the DKO mice showed increased wakefulness periods and decreased total time spent in both NREM and REM sleep. WT exhibited time-of-day dependent modulation of contextual and cued memory. Compared with WT mice, 4-month-old DKO mice exhibited the deficiency regardless trained and tested in the same light/night phase or not. Particularly interesting was that DKO showed circadian modulation deficiency when trained in the resting period but not in the active period. Long noncoding RNAs (lncRNAs) are typically defined as transcripts longer than 200 nucleotides, and they have rhythmic expression in mammals. To date no study has investigated rhythmic lncRNA expression in Alzheimer's disease. We applied RNA-seq technology to profile hippocampus expression of lncRNAs in DKO mice during the light (/resting) and dark (/active) phases and performed gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses of the cis lncRNA targets. Expression alteration of lncRNAs associated with immune response and metallodipeptidase activity may contribute to the circadian disruptions of DKO mice. Especially we identified some LncRNAs which expression change oppositely between day and light in DKO mice compared to WT mice and are worthy to be studied further. Our results exhibited the circadian rhythm sleep disorders and a noteworthy time-of-day-dependent memory deficiency in AD model mice and provide a useful resource for studying the expression and function of lncRNAs during circadian disruptions in Alzheimer's disease.


Subject(s)
Alzheimer Disease , RNA, Long Noncoding , Sleep Disorders, Circadian Rhythm , Animals , Mice , Alzheimer Disease/genetics , Circadian Rhythm/genetics , Mammals/genetics , Mice, Knockout , RNA, Long Noncoding/genetics , Sleep/physiology , Sleep Disorders, Circadian Rhythm/genetics
5.
Behav Brain Res ; 418: 113652, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34758364

ABSTRACT

Impairments in activities of daily living (ADL) are common clinical symptoms of human Alzheimer's disease (AD). Describing the ADL in AD animal models might provide more insights into the mechanism/treatment of the disease. Here, we demonstrated that the forebrain presenilin 1(Psen1)/presenilin 2 (Psen2) conditional double knockout (DKO) mice exhibited deficits in nest building, marble burying and food burrowing starting at 3 months old and worsening at later ages. At 4 months of age, spontaneous activities in the home cage were also impaired in DKO mice, including physically demanding activities, habituation-like behaviors, and nourishment behaviors during the first two hours in the dark phase. These results indicated that loss of function of Psen1 and Psen2 in mice impaired a series of noncognitive behaviors in the early phase of neurodegeneration. This observation suggests that DKO mice are an ideal model for further mechanistic studies of Psen1 and Psen2 functions in regulating noncognitive behaviors.


Subject(s)
Behavior, Animal/physiology , Feeding Behavior/physiology , Mice, Knockout , Presenilin-1/genetics , Presenilin-2/genetics , Animals , Female , Male , Mice
6.
Cell Death Discov ; 7(1): 335, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34741025

ABSTRACT

Endogenous clocks generate rhythms in gene expression, which facilitates the organisms to cope through periodic environmental variations in accordance with 24-h light/dark time. A core question that needs to be elucidated is how such rhythms proliferate throughout the cells and regulate the dynamic physiology. In this study, we demonstrate the role of REGγ as a new regulator of circadian clock in mice, primary MEF, and SY5Y cells. Assessment of circadian conduct reveals a difference in circadian period, wheel mode, and the ability to acclimate the external light stimulus between WT and KO littermates. Compared to WT mice, REGγ KO mice attain the phase delay behavior upon light shock at early night. During the variation of 12/12 h light/dark (LD) exposure, levels of Per1, Per2, Cry1, Clock, Bmal1, and Rorα circadian genes in suprachiasmatic nucleus are significantly higher in REGγ KO than in WT mice, concomitant with remarkable changes in BMAL1 and PER2 proteins. In cultured cells depleted of REGγ, serum shock induces early response of the circadian genes Per1 and Per2 with the cyclic rhythm maintained. Mechanistic study indicates that REGγ directly degrades BMAL1 by the non-canonical proteasome pathway independent of ATP and ubiquitin. Silencing BMAL1 abrogates the changes in circadian genes in REGγ-deficient cells. However, inhibition of GSK-3ß, a known promoter for degradation of BMAL1, exacerbates the action of REGγ depletion. In conclusion, our findings define REGγ as a new factor, which functions as a rheostat of circadian rhythms to mitigate the levels of Per1 and Per2 via proteasome-dependent degradation of BMAL1.

7.
Neurobiol Aging ; 73: 41-49, 2019 01.
Article in English | MEDLINE | ID: mdl-30316051

ABSTRACT

MicroRNAs, small noncoding RNAs, not only regulate gene expression at the post-transcriptional level in a variety of physiological processes but also accompany the initiation and progression of a vast number of diseases, including dementia. While miR-125b has been shown to be aberrantly expressed in some dementia patients, its role in the pathological process remains ambiguous. Presenilin-1/2 conditional double knockout mice exhibit a range of symptoms, including impaired cognition and memory, increased tau phosphorylation, neuroinflammation, and apoptosis, and are therefore regarded as a useful dementia model. In the prefrontal cortices of double knockout mice, miR-125b was found to be abnormally increased in an age-dependent manner. We further verified the neural cell adhesion molecule (NCAM) as an miR-125b target using the dual luciferase reporter assay. The NCAM protein level was decreased when miR-125b was overexpressed (OE) in neuronal growth factor-induced differentiated PC12 cells, which further inhibited the neuronal growth factor-induced phosphorylation of glycogen synthase kinase 3 beta (GSK3ß) at the Ser9 site and ultimately increased the GSK3ß activity and tau phosphorylation. Moreover, on serum deprivation, high GSK3ß activity in differentiated miR-125b-OE PC12 cells induced increased caspase-3 activation. Finally, adeno-associated virus-mediated miR-125b overexpression in the prefrontal cortexes of wild-type C57B/L6 mice resulted in decreased dendritic spine density. In addition, similar to the in vitro data, elevated GSK3ß activity and hyperphosphorylation of the tau protein were confirmed. Taken together, our findings reveal a direct regulation of miR-125b on NCAM, which leads to further effects on downstream GSK3ß activity and tau phosphorylation and may contribute to the generation of neurofibrillary tangles in neuropathological progression.


Subject(s)
Dementia/genetics , Dementia/pathology , MicroRNAs/physiology , Neural Cell Adhesion Molecules/metabolism , Neurofibrillary Tangles/genetics , tau Proteins/metabolism , Aging/genetics , Aging/metabolism , Animals , Cells, Cultured , Dendritic Spines/pathology , Disease Progression , Gene Expression , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Neurofibrillary Tangles/pathology , PC12 Cells , Phosphorylation/genetics , Prefrontal Cortex/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...