Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146158

ABSTRACT

Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.


Subject(s)
Apoptosis , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Aging , Animals , Apoptosis/drug effects , Axons/metabolism , Behavior, Animal , Brain/cytology , Brain/metabolism , Cells, Cultured , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Spinal Cord/metabolism , Staurosporine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
2.
Cell ; 166(1): 13-5, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27368096

ABSTRACT

Mutations in the presenilins that cause familial Alzheimer's disease alter the activity of these proteases to increase generation of an aggregation-prone isoform of the amyloid ß-peptide (Aß). How these mutations do so has been unclear. Sannerud et al. now show that regulation of subcellular localization plays a central role, advancing our understanding of the cell biology of Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Presenilins/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Humans , Protein Transport
3.
Nature ; 574(7778): 359-364, 2019 10.
Article in English | MEDLINE | ID: mdl-31619788

ABSTRACT

The mechanisms that extend lifespan in humans are poorly understood. Here we show that extended longevity in humans is associated with a distinct transcriptome signature in the cerebral cortex that is characterized by downregulation of genes related to neural excitation and synaptic function. In Caenorhabditis elegans, neural excitation increases with age and inhibition of excitation globally, or in glutamatergic or cholinergic neurons, increases longevity. Furthermore, longevity is dynamically regulated by the excitatory-inhibitory balance of neural circuits. The transcription factor REST is upregulated in humans with extended longevity and represses excitation-related genes. Notably, REST-deficient mice exhibit increased cortical activity and neuronal excitability during ageing. Similarly, loss-of-function mutations in the C. elegans REST orthologue genes spr-3 and spr-4 elevate neural excitation and reduce the lifespan of long-lived daf-2 mutants. In wild-type worms, overexpression of spr-4 suppresses excitation and extends lifespan. REST, SPR-3, SPR-4 and reduced excitation activate the longevity-associated transcription factors FOXO1 and DAF-16 in mammals and worms, respectively. These findings reveal a conserved mechanism of ageing that is mediated by neural circuit activity and regulated by REST.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Longevity , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Aging , Animals , Brain/cytology , Brain/metabolism , Caenorhabditis elegans , Forkhead Transcription Factors/metabolism , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA Interference , RNA-Binding Proteins/metabolism
4.
Mol Psychiatry ; 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37938767

ABSTRACT

Neurodevelopmental changes and impaired stress resistance have been implicated in the pathogenesis of bipolar disorder (BD), but the underlying regulatory mechanisms are unresolved. Here we describe a human cerebral organoid model of BD that exhibits altered neural development, elevated neural network activity, and a major shift in the transcriptome. These phenotypic changes were reproduced in cerebral organoids generated from iPS cell lines derived in different laboratories. The BD cerebral organoid transcriptome showed highly significant enrichment for gene targets of the transcriptional repressor REST. This was associated with reduced nuclear REST and REST binding to target gene recognition sites. Reducing the oxygen concentration in organoid cultures to a physiological range ameliorated the developmental phenotype and restored REST expression. These effects were mimicked by treatment with lithium. Reduced nuclear REST and derepression of REST targets genes were also observed in the prefrontal cortex of BD patients. Thus, an impaired cellular stress response in BD cerebral organoids leads to altered neural development and transcriptional dysregulation associated with downregulation of REST. These findings provide a new model and conceptual framework for exploring the molecular basis of BD.

5.
Cell ; 135(5): 907-18, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-19041753

ABSTRACT

Genomic instability and alterations in gene expression are hallmarks of eukaryotic aging. The yeast histone deacetylase Sir2 silences transcription and stabilizes repetitive DNA, but during aging or in response to a DNA break, the Sir complex relocalizes to sites of genomic instability, resulting in the desilencing of genes that cause sterility, a characteristic of yeast aging. Using embryonic stem cells, we show that mammalian Sir2, SIRT1, represses repetitive DNA and a functionally diverse set of genes across the mouse genome. In response to DNA damage, SIRT1 dissociates from these loci and relocalizes to DNA breaks to promote repair, resulting in transcriptional changes that parallel those in the aging mouse brain. Increased SIRT1 expression promotes survival in a mouse model of genomic instability and suppresses age-dependent transcriptional changes. Thus, DNA damage-induced redistribution of SIRT1 and other chromatin-modifying proteins may be a conserved mechanism of aging in eukaryotes.


Subject(s)
Aging/genetics , Chromatin/metabolism , Genomic Instability , Sirtuins/genetics , Animals , Brain/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells , Gene Knockout Techniques , Humans , Lymphoma/metabolism , Mice , Molecular Sequence Data , Oxidative Stress , Sirtuin 1 , Specific Pathogen-Free Organisms , Thymus Neoplasms/metabolism , Yeasts/cytology , Yeasts/metabolism
6.
Nucleic Acids Res ; 49(10): e58, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33693773

ABSTRACT

We present barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel insitu analyses (BOLORAMIS), a reverse transcription-free method for spatially-resolved, targeted, in situ RNA identification of single or multiple targets. BOLORAMIS was demonstrated on a range of cell types and human cerebral organoids. Singleplex experiments to detect coding and non-coding RNAs in human iPSCs showed a stem-cell signature pattern. Specificity of BOLORAMIS was found to be 92% as illustrated by a clear distinction between human and mouse housekeeping genes in a co-culture system, as well as by recapitulation of subcellular localization of lncRNA MALAT1. Sensitivity of BOLORAMIS was quantified by comparing with single molecule FISH experiments and found to be 11%, 12% and 35% for GAPDH, TFRC and POLR2A, respectively. To demonstrate BOLORAMIS for multiplexed gene analysis, we targeted 96 mRNAs within a co-culture of iNGN neurons and HMC3 human microglial cells. We used fluorescence in situ sequencing to detect error-robust 8-base barcodes associated with each of these genes. We then used this data to uncover the spatial relationship among cells and transcripts by performing single-cell clustering and gene-gene proximity analyses. We anticipate the BOLORAMIS technology for in situ RNA detection to find applications in basic and translational research.


Subject(s)
Gene Expression Profiling/methods , In Situ Hybridization, Fluorescence/methods , Oligonucleotides/chemistry , RNA/analysis , Single-Cell Analysis/methods , Animals , Cell Line , Humans , Mice
7.
J Am Soc Nephrol ; 32(8): 1974-1986, 2021 08.
Article in English | MEDLINE | ID: mdl-34078664

ABSTRACT

BACKGROUND: CKD is associated with the loss of functional nephr ons, leading to increased mechanical and metabolic stress in the remaining cells, particularly for cells constituting the filtration barrier, such as podocytes. The failure of podocytes to mount an adequate stress response can lead to further nephron loss and disease progression. However, the mechanisms that regulate this degenerative process in the kidney are unknown. METHODS: We combined in vitro, in vivo, and organ-on-chip approaches to identify the RE1-silencing transcription factor (REST), a repressor of neuronal genes during embryonic development, as a central regulator of podocyte adaptation to injury and aging. RESULTS: Mice with a specific deletion of REST in podocytes exhibit albuminuria, podocyte apoptosis, and glomerulosclerosis during aging, and exhibit increased vulnerability to renal injury. This phenotype is mediated, in part, by the effects of REST on the podocyte cytoskeleton that promote resistance to mechanical stressors and augment podocyte survival. Finally, REST expression is upregulated in human podocytes during aging, consistent with a conserved mechanism of stress resistance. CONCLUSIONS: These results suggest REST protects the kidney from injury and degeneration during aging, with potentially important therapeutic implications.


Subject(s)
Adaptation, Physiological/genetics , Aging/physiology , Podocytes/pathology , Podocytes/physiology , Repressor Proteins/genetics , Stress, Physiological/genetics , Adult , Aged , Aged, 80 and over , Albuminuria/genetics , Animals , Apoptosis/genetics , Cell Line , Cell Survival , Cytoskeleton/physiology , Gene Expression Regulation/genetics , Homeostasis/genetics , Humans , Mice , Phenotype , Repressor Proteins/metabolism , Sclerosis , Young Adult
8.
Mol Cell ; 50(2): 172-84, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23499007

ABSTRACT

The Uba6 (E1)-Use1 (E2) ubiquitin transfer cascade is a poorly understood alternative arm of the ubiquitin proteasome system (UPS) and is required for mouse embryonic development, independent of the canonical Uba1-E2-E3 pathway. Loss of neuronal Uba6 during embryonic development results in altered patterning of neurons in the hippocampus and the amygdala, decreased dendritic spine density, and numerous behavioral disorders. The levels of the E3 ubiquitin ligase Ube3a (E6-AP) and Shank3, both linked with dendritic spine function, are elevated in the amygdala of Uba6-deficient mice, while levels of the Ube3a substrate Arc are reduced. Uba6 and Use1 promote proteasomal turnover of Ube3a in mouse embryo fibroblasts (MEFs) and catalyze Ube3a ubiquitylation in vitro. These activities occur in parallel with an independent pathway involving Uba1-UbcH7, but in a spatially distinct manner in MEFs. These data reveal an unanticipated role for Uba6 in neuronal development, spine architecture, mouse behavior, and turnover of Ube3a.


Subject(s)
Amygdala/abnormalities , CA3 Region, Hippocampal/abnormalities , Qc-SNARE Proteins/deficiency , Ubiquitin-Activating Enzymes/deficiency , Ubiquitination , Amygdala/enzymology , Amygdala/pathology , Animals , Body Weight , CA3 Region, Hippocampal/enzymology , CA3 Region, Hippocampal/pathology , Cells, Cultured , Dendritic Spines/pathology , Embryonic Development , Energy Metabolism , Female , Genes, Lethal , Learning Disabilities/metabolism , Locomotion , Male , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/metabolism , Oxygen Consumption , Protein Stability , Protein Structure, Tertiary , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , SNARE Proteins , Social Behavior , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins
9.
Nature ; 507(7493): 448-54, 2014 03 27.
Article in English | MEDLINE | ID: mdl-24670762

ABSTRACT

Human neurons are functional over an entire lifetime, yet the mechanisms that preserve function and protect against neurodegeneration during ageing are unknown. Here we show that induction of the repressor element 1-silencing transcription factor (REST; also known as neuron-restrictive silencer factor, NRSF) is a universal feature of normal ageing in human cortical and hippocampal neurons. REST is lost, however, in mild cognitive impairment and Alzheimer's disease. Chromatin immunoprecipitation with deep sequencing and expression analysis show that REST represses genes that promote cell death and Alzheimer's disease pathology, and induces the expression of stress response genes. Moreover, REST potently protects neurons from oxidative stress and amyloid ß-protein toxicity, and conditional deletion of REST in the mouse brain leads to age-related neurodegeneration. A functional orthologue of REST, Caenorhabditis elegans SPR-4, also protects against oxidative stress and amyloid ß-protein toxicity. During normal ageing, REST is induced in part by cell non-autonomous Wnt signalling. However, in Alzheimer's disease, frontotemporal dementia and dementia with Lewy bodies, REST is lost from the nucleus and appears in autophagosomes together with pathological misfolded proteins. Finally, REST levels during ageing are closely correlated with cognitive preservation and longevity. Thus, the activation state of REST may distinguish neuroprotection from neurodegeneration in the ageing brain.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Oxidative Stress , Repressor Proteins/metabolism , Aged , Aged, 80 and over , Aging/genetics , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Autophagy , Brain/cytology , Brain/metabolism , Brain/pathology , Caenorhabditis elegans Proteins/metabolism , Cell Death/genetics , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Cognition , Cognitive Dysfunction/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression Regulation , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Longevity , Mice , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , Phagosomes , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcription Factors/metabolism , Up-Regulation , Wnt Signaling Pathway , Young Adult
10.
Mol Cell ; 40(2): 333-44, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20965426

ABSTRACT

Aging is the outcome of a balance between damage and repair. The rate of aging and the appearance of age-related pathology are modulated by stress response and repair pathways that gradually decline, including the proteostasis and DNA damage repair networks and mitochondrial respiratory metabolism. Highly conserved insulin/IGF-1, TOR, and sirtuin signaling pathways in turn control these critical cellular responses. The coordinated action of these signaling pathways maintains cellular and organismal homeostasis in the face of external perturbations, such as changes in nutrient availability, temperature, and oxygen level, as well as internal perturbations, such as protein misfolding and DNA damage. Studies in model organisms suggest that changes in signaling can augment these critical stress response systems, increasing life span and reducing age-related pathology. The systems biology of stress response signaling thus provides a new approach to the understanding and potential treatment of age-related diseases.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Signal Transduction , Stress, Physiological/physiology , Animals , Humans , Models, Biological , Neoplasms/physiopathology , Neurodegenerative Diseases/physiopathology , Oxidative Stress/physiology
12.
Nature ; 480(7377): 372-5, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22056987

ABSTRACT

A formidable challenge in neural repair in the adult central nervous system (CNS) is the long distances that regenerating axons often need to travel in order to reconnect with their targets. Thus, a sustained capacity for axon regeneration is critical for achieving functional restoration. Although deletion of either phosphatase and tensin homologue (PTEN), a negative regulator of mammalian target of rapamycin (mTOR), or suppressor of cytokine signalling 3 (SOCS3), a negative regulator of Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, in adult retinal ganglion cells (RGCs) individually promoted significant optic nerve regeneration, such regrowth tapered off around 2 weeks after the crush injury. Here we show that, remarkably, simultaneous deletion of both PTEN and SOCS3 enables robust and sustained axon regeneration. We further show that PTEN and SOCS3 regulate two independent pathways that act synergistically to promote enhanced axon regeneration. Gene expression analyses suggest that double deletion not only results in the induction of many growth-related genes, but also allows RGCs to maintain the expression of a repertoire of genes at the physiological level after injury. Our results reveal concurrent activation of mTOR and STAT3 pathways as key for sustaining long-distance axon regeneration in adult CNS, a crucial step towards functional recovery.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , PTEN Phosphohydrolase/deficiency , Suppressor of Cytokine Signaling Proteins/deficiency , Animals , Axons/pathology , Cell Growth Processes/genetics , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Nerve Crush , Optic Nerve/cytology , Optic Nerve/growth & development , Optic Nerve/pathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , PTEN Phosphohydrolase/genetics , Retinal Ganglion Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
14.
Nature ; 464(7288): 529-35, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20336135

ABSTRACT

During the past century, treatments for the diseases of youth and middle age have helped raise life expectancy significantly. However, cognitive decline has emerged as one of the greatest health threats of old age, with nearly 50% of adults over the age of 85 afflicted with Alzheimer's disease. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain ageing. Recent advances in the biology of ageing in model organisms, together with molecular and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline.


Subject(s)
Aging/pathology , Brain/physiopathology , Cognition Disorders/physiopathology , Animals , Autophagy , Brain/physiology , Epigenesis, Genetic/physiology , Humans , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Mitochondria/pathology , Oxidative Stress/physiology , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 107(32): 14164-9, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660724

ABSTRACT

Dysregulation of autophagy, a cellular catabolic mechanism essential for degradation of misfolded proteins, has been implicated in multiple neurodegenerative diseases. However, the mechanisms that lead to the autophagy dysfunction are still not clear. Based on the results of a genome-wide screen, we show that reactive oxygen species (ROS) serve as common mediators upstream of the activation of the type III PI3 kinase, which is critical for the initiation of autophagy. Furthermore, ROS play an essential function in the induction of the type III PI3 kinase and autophagy in response to amyloid beta peptide, the main pathogenic mediator of Alzheimer's disease (AD). However, lysosomal blockage also caused by Abeta is independent of ROS. In addition, we demonstrate that autophagy is transcriptionally down-regulated during normal aging in the human brain. Strikingly, in contrast to normal aging, we observe transcriptional up-regulation of autophagy in the brains of AD patients, suggesting that there might be a compensatory regulation of autophagy. Interestingly, we show that an AD drug and an AD drug candidate have inhibitory effects on autophagy, raising the possibility that decreasing input into the lysosomal system may help to reduce cellular stress in AD. Finally, we provide a list of candidate drug targets that can be used to safely modulate levels of autophagy without causing cell death.


Subject(s)
Aging/genetics , Alzheimer Disease/pathology , Autophagy/genetics , Brain/physiology , Genome-Wide Association Study , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Brain/pathology , Gene Expression Regulation , Humans , Lysosomes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism
17.
Nat Commun ; 14(1): 7030, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919281

ABSTRACT

Many aging individuals accumulate the pathology of Alzheimer's disease (AD) without evidence of cognitive decline. Here we describe an integrated neurodegeneration checkpoint response to early pathological changes that restricts further disease progression and preserves cognitive function. Checkpoint activation is mediated by the REST transcriptional repressor, which is induced in cognitively-intact aging humans and AD mouse models at the onset of amyloid ß-protein (Aß) deposition and tau accumulation. REST induction is mediated by the unfolded protein response together with ß-catenin signaling. A consequence of this response is the targeting of REST to genes involved in key pathogenic pathways, resulting in downregulation of gamma secretase, tau kinases, and pro-apoptotic proteins. Deletion of REST in the 3xTg and J20 AD mouse models accelerates Aß deposition and the accumulation of misfolded and phosphorylated tau, leading to neurodegeneration and cognitive decline. Conversely, viral-mediated overexpression of REST in the hippocampus suppresses Aß and tau pathology. Thus, REST mediates a neurodegeneration checkpoint response with multiple molecular targets that may protect against the onset of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Animals , Humans , Mice , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Disease Models, Animal , Mice, Transgenic , tau Proteins/metabolism
18.
Nat Neurosci ; 11(4): 476-87, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18344994

ABSTRACT

Intrinsic antioxidant defenses are important for neuronal longevity. We found that in rat neurons, synaptic activity, acting via NMDA receptor (NMDAR) signaling, boosted antioxidant defenses by making changes to the thioredoxin-peroxiredoxin (Prx) system. Synaptic activity enhanced thioredoxin activity, facilitated the reduction of overoxidized Prxs and promoted resistance to oxidative stress. Resistance was mediated by coordinated transcriptional changes; synaptic NMDAR activity inactivated a previously unknown Forkhead box O target gene, the thioredoxin inhibitor Txnip. Conversely, NMDAR blockade upregulated Txnip in vivo and in vitro, where it bound thioredoxin and promoted vulnerability to oxidative damage. Synaptic activity also upregulated the Prx reactivating genes Sesn2 (sestrin 2) and Srxn1 (sulfiredoxin), via C/EBPbeta and AP-1, respectively. Mimicking these expression changes was sufficient to strengthen antioxidant defenses. Trans-synaptic stimulation of synaptic NMDARs was crucial for boosting antioxidant defenses; chronic bath activation of all (synaptic and extrasynaptic) NMDARs induced no antioxidative effects. Thus, synaptic NMDAR activity may influence the progression of pathological processes associated with oxidative damage.


Subject(s)
Antioxidants/metabolism , Oxidative Stress/physiology , Peroxiredoxins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Expression Regulation/physiology , Mice , Neurons/metabolism , Nuclear Proteins , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Peroxidases , Proteins/metabolism , Rats , Signal Transduction/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Transcription, Genetic/physiology
19.
Curr Opin Neurobiol ; 72: 91-100, 2022 02.
Article in English | MEDLINE | ID: mdl-34689041

ABSTRACT

The aging brain is shaped by many structural and functional alterations. Recent cross-disciplinary efforts have uncovered powerful and integrated adaptive mechanisms that promote brain health and prevent functional decline during aging. Here, we review some of the most robust adaptive mechanisms and how they can be engaged to protect, and restore the aging brain.


Subject(s)
Brain , Aging , Brain/physiology , Humans
20.
Sci Adv ; 8(13): eabj8658, 2022 04.
Article in English | MEDLINE | ID: mdl-35353567

ABSTRACT

The age-related cognitive decline of normal aging is exacerbated in neurodegenerative diseases including Alzheimer's disease (AD). However, it remains unclear whether age-related cognitive regulators in AD pathologies contribute to life span. Here, we show that C/EBPß, an Aß and inflammatory cytokine-activated transcription factor that promotes AD pathologies via activating asparagine endopeptidase (AEP), mediates longevity in a gene dose-dependent manner in neuronal C/EBPß transgenic mice. C/EBPß selectively triggers inhibitory GABAnergic neuronal degeneration by repressing FOXOs and up-regulating AEP, leading to aberrant neural excitation and cognitive dysfunction. Overexpression of CEBP-2 or LGMN-1 (AEP) in Caenorhabditis elegans neurons but not muscle stimulates neural excitation and shortens life span. CEBP-2 or LGMN-1 reduces daf-2 mutant-elongated life span and diminishes daf-16-induced longevity. C/EBPß and AEP are lower in humans with extended longevity and inversely correlated with REST/FOXO1. These findings demonstrate a conserved mechanism of aging that couples pathological cognitive decline to life span by the neuronal C/EBPß/AEP pathway.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Longevity/genetics , Mice , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL