Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Brain ; 144(6): 1833-1852, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33880508

ABSTRACT

Amyloid-ß precursor protein (APP) is sequentially cleaved by secretases and generates amyloid-ß, the major components in senile plaques in Alzheimer's disease. APP is upregulated in human Alzheimer's disease brains. However, the molecular mechanism of how APP contributes to Alzheimer's disease pathogenesis remains incompletely understood. Here we show that truncated APP C586-695 fragment generated by δ-secretase directly binds to CCAAT/enhancer-binding protein beta (CEBPB), an inflammatory transcription factor, and enhances its transcriptional activity, escalating Alzheimer's disease-related gene expression and pathogenesis. The APP C586-695 fragment, but not full-length APP, strongly associates with CEBPB and elicits its nuclear translocation and augments the transcriptional activities on APP itself, MAPT (microtubule-associated protein tau), δ-secretase and inflammatory cytokine mRNA expression, finally triggering Alzheimer's disease pathology and cognitive disorder in a viral overexpression mouse model. Blockade of δ-secretase cleavage of APP by mutating the cleavage sites reduces its stimulatory effect on CEBPB, alleviating amyloid pathology and cognitive dysfunctions. Clearance of APP C586-695 from 5xFAD mice by antibody administration mitigates Alzheimer's disease pathologies and restores cognitive functions. Thus, in addition to the sequestration of amyloid-ß, APP implicates in Alzheimer's disease pathology by activating CEBPB upon δ-secretase cleavage.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation/physiology , Aged , Animals , Cysteine Endopeptidases/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
2.
Proc Natl Acad Sci U S A ; 114(34): 9188-9193, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28790189

ABSTRACT

The prenatal period of cortical development is important for the establishment of neural circuitry and functional connectivity of the brain; however, the molecular mechanisms underlying this process remain unclear. Here we report that disruption of the actin-cytoskeletal network in the developing mouse prefrontal cortex alters dendritic morphogenesis and synapse formation, leading to enhanced formation of fear-related memory in adulthood. These effects are mediated by a brain-enriched microRNA, miR-9, through its negative regulation of diaphanous homologous protein 1 (Diap1), a key organizer of the actin cytoskeletal assembly. Our findings not only revealed important regulation of dendritogenesis and synaptogenesis during early brain development but also demonstrated a tight link between these early developmental events and cognitive functions later in life.


Subject(s)
Cognition , MicroRNAs/metabolism , Neurogenesis , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Formins , Gene Expression Regulation, Developmental , Male , Memory , Mice , MicroRNAs/genetics
3.
Proc Natl Acad Sci U S A ; 114(40): 10773-10778, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923922

ABSTRACT

BDNF/TrkB neurotrophic signaling is essential for dopaminergic neuronal survival, and the activities are reduced in the substantial nigra (SN) of Parkinson's disease (PD). However, whether α-Syn (alpha-synuclein) aggregation, a hallmark in the remaining SN neurons in PD, accounts for the neurotrophic inhibition remains elusive. Here we show that α-Syn selectively interacts with TrkB receptors and inhibits BDNF/TrkB signaling, leading to dopaminergic neuronal death. α-Syn binds to the kinase domain on TrkB, which is negatively regulated by BDNF or Fyn tyrosine kinase. Interestingly, α-Syn represses TrkB lipid raft distribution, decreases its internalization, and reduces its axonal trafficking. Moreover, α-Syn also reduces TrkB protein levels via up-regulation of TrkB ubiquitination. Remarkably, dopamine's metabolite 3,4-Dihydroxyphenylacetaldehyde (DOPAL) stimulates the interaction between α-Syn and TrkB. Accordingly, MAO-B inhibitor rasagiline disrupts α-Syn/TrkB complex and rescues TrkB neurotrophic signaling, preventing α-Syn-induced dopaminergic neuronal death and restoring motor functions. Hence, our findings demonstrate a noble pathological role of α-Syn in antagonizing neurotrophic signaling, providing a molecular mechanism that accounts for its neurotoxicity in PD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dopaminergic Neurons/pathology , Membrane Glycoproteins/metabolism , Parkinson Disease/pathology , Receptor, trkB/metabolism , alpha-Synuclein/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Death , Cells, Cultured , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Parkinson Disease/genetics , Parkinson Disease/metabolism , Receptor, trkB/genetics , Signal Transduction , alpha-Synuclein/genetics
4.
Proc Natl Acad Sci U S A ; 114(5): 1183-1188, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096359

ABSTRACT

The abnormal aggregation of fibrillar α-synuclein in Lewy bodies plays a critical role in the pathogenesis of Parkinson's disease. However, the molecular mechanisms regulating α-synuclein pathological effects are incompletely understood. Here we show that α-synuclein binds phosphoinositide-3 kinase enhancer L (PIKE-L) in a phosphorylation-dependent manner and sequesters it in Lewy bodies, leading to dopaminergic cell death via AMP-activated protein kinase (AMPK) hyperactivation. α-Synuclein interacts with PIKE-L, an AMPK inhibitory binding partner, and this action is increased by S129 phosphorylation through AMPK and is decreased by Y125 phosphorylation via Src family kinase Fyn. A pleckstrin homology (PH) domain in PIKE-L directly binds α-synuclein and antagonizes its aggregation. Accordingly, PIKE-L overexpression decreases dopaminergic cell death elicited by 1-methyl-4-phenylpyridinium (MPP+), whereas PIKE-L knockdown elevates α-synuclein oligomerization and cell death. The overexpression of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or α-synuclein induces greater dopaminergic cell loss and more severe motor defects in PIKE-KO and Fyn-KO mice than in wild-type mice, and these effects are attenuated by the expression of dominant-negative AMPK. Hence, our findings demonstrate that α-synuclein neutralizes PIKE-L's neuroprotective actions in synucleinopathies, triggering dopaminergic neuronal death by hyperactivating AMPK.


Subject(s)
Adenylate Kinase/metabolism , Dopaminergic Neurons/metabolism , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Lewy Bodies/metabolism , alpha-Synuclein/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Aged , Aged, 80 and over , Animals , Cell Death , Dopaminergic Neurons/ultrastructure , Enzyme Activation , GTP Phosphohydrolases/deficiency , GTP-Binding Proteins/chemistry , GTPase-Activating Proteins/chemistry , Humans , MPTP Poisoning/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Phosphorylation , Pleckstrin Homology Domains , Protein Aggregation, Pathological , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/metabolism
5.
Nature ; 500(7464): 593-7, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23892778

ABSTRACT

Mammalian pre-implantation development is a complex process involving dramatic changes in the transcriptional architecture. We report here a comprehensive analysis of transcriptome dynamics from oocyte to morula in both human and mouse embryos, using single-cell RNA sequencing. Based on single-nucleotide variants in human blastomere messenger RNAs and paternal-specific single-nucleotide polymorphisms, we identify novel stage-specific monoallelic expression patterns for a significant portion of polymorphic gene transcripts (25 to 53%). By weighted gene co-expression network analysis, we find that each developmental stage can be delineated concisely by a small number of functional modules of co-expressed genes. This result indicates a sequential order of transcriptional changes in pathways of cell cycle, gene regulation, translation and metabolism, acting in a step-wise fashion from cleavage to morula. Cross-species comparisons with mouse pre-implantation embryos reveal that the majority of human stage-specific modules (7 out of 9) are notably preserved, but developmental specificity and timing differ between human and mouse. Furthermore, we identify conserved key members (or hub genes) of the human and mouse networks. These genes represent novel candidates that are likely to be key in driving mammalian pre-implantation development. Together, the results provide a valuable resource to dissect gene regulatory mechanisms underlying progressive development of early mammalian embryos.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Sequence Analysis, RNA , Single-Cell Analysis , Alleles , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Cycle/genetics , Embryo, Mammalian/cytology , Gene Expression Profiling , Humans , Mice , Morula/cytology , Morula/metabolism , Oocytes/cytology , Oocytes/metabolism
6.
Proc Natl Acad Sci U S A ; 112(43): 13354-9, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460015

ABSTRACT

Neural stem cells (NSCs) in the adult mammalian central nervous system (CNS) hold the key to neural regeneration through proper activation, differentiation, and maturation, to establish nascent neural networks, which can be integrated into damaged neural circuits to repair function. However, the CNS injury microenvironment is often inhibitory and inflammatory, limiting the ability of activated NSCs to differentiate into neurons and form nascent circuits. Here we report that neurotrophin-3 (NT3)-coupled chitosan biomaterial, when inserted into a 5-mm gap of completely transected and excised rat thoracic spinal cord, elicited robust activation of endogenous NSCs in the injured spinal cord. Through slow release of NT3, the biomaterial attracted NSCs to migrate into the lesion area, differentiate into neurons, and form functional neural networks, which interconnected severed ascending and descending axons, resulting in sensory and motor behavioral recovery. Our study suggests that enhancing endogenous neurogenesis could be a novel strategy for treatment of spinal cord injury.


Subject(s)
Cellular Microenvironment/physiology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurotrophin 3/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Analysis of Variance , Animals , Chitosan/therapeutic use , Electromyography , Evoked Potentials/physiology , Fluorescence , Immunohistochemistry , Microscopy, Immunoelectron , Neural Stem Cells/physiology , Neurotrophin 3/therapeutic use , Rats
7.
Proc Natl Acad Sci U S A ; 112(43): 13360-5, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460053

ABSTRACT

Spinal cord injury (SCI) is considered incurable because axonal regeneration in the central nervous system (CNS) is extremely challenging, due to harsh CNS injury environment and weak intrinsic regeneration capability of CNS neurons. We discovered that neurotrophin-3 (NT3)-loaded chitosan provided an excellent microenvironment to facilitate nerve growth, new neurogenesis, and functional recovery of completely transected spinal cord in rats. To acquire mechanistic insight, we conducted a series of comprehensive transcriptome analyses of spinal cord segments at the lesion site, as well as regions immediately rostral and caudal to the lesion, over a period of 90 days after SCI. Using weighted gene coexpression network analysis (WGCNA), we established gene modules/programs corresponding to various pathological events at different times after SCI. These objective measures of gene module expression also revealed that enhanced new neurogenesis and angiogenesis, and reduced inflammatory responses were keys to conferring the effect of NT3-chitosan on regeneration.


Subject(s)
Cellular Microenvironment/physiology , Neurotrophin 3/pharmacology , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Animals , Chitosan/therapeutic use , Computational Biology/methods , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling/methods , Microarray Analysis , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Neurotrophin 3/therapeutic use , Polymerase Chain Reaction , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/genetics , Spinal Cord Injuries/genetics
8.
Proc Natl Acad Sci U S A ; 111(20): 7444-9, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799701

ABSTRACT

The studies on the exact lineage composition of NG2 expressing progenitors in the forebrain have been controversial. A number of studies have revealed the heterogeneous nature of postnatal NG2 cells. However, NG2 cells found in embryonic dates are far less understood. Our study indicates that early NG2 progenitors from a ventral origin (i.e., before embryonic day 16.5) tangentially migrate out of the medial ganglionic eminence and give rise to interneurons in deep layers of the dorsal cerebral cortex. The majority of myelinating oligodendrocytes found in both cortical gray and white matters are, in contrast, derived from NG2 progenitors with a neonatal subventricular zone origin. Our lineage tracing data reflect the heterogeneous nature of NG2 progenitor populations and define the relationship between lineage divergence and spatiotemporal origins. Beyond the typical lineage tracing studies of NG2(+) cells, by costaining with lineage-specific markers, our study addresses the origins of heterogeneity and its implications in the differentiation potentials of NG2(+) progenitors.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation , Interneurons/cytology , Neuroglia/cytology , Prosencephalon/cytology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cerebral Cortex/embryology , Glial Fibrillary Acidic Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Oligodendroglia/cytology , Stem Cells/cytology
9.
Proc Natl Acad Sci U S A ; 110(41): 16562-7, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24023061

ABSTRACT

Deposition of insoluble protein aggregates is a hallmark of neurodegenerative diseases. The universal presence of ß-amyloid and tau in Alzheimer's disease (AD) has facilitated advancement of the amyloid cascade and tau hypotheses that have dominated AD pathogenesis research and therapeutic development. However, the underlying etiology of the disease remains to be fully elucidated. Here we report a comprehensive study of the human brain-insoluble proteome in AD by mass spectrometry. We identify 4,216 proteins, among which 36 proteins accumulate in the disease, including U1-70K and other U1 small nuclear ribonucleoprotein (U1 snRNP) spliceosome components. Similar accumulations in mild cognitive impairment cases indicate that spliceosome changes occur in early stages of AD. Multiple U1 snRNP subunits form cytoplasmic tangle-like structures in AD but not in other examined neurodegenerative disorders, including Parkinson disease and frontotemporal lobar degeneration. Comparison of RNA from AD and control brains reveals dysregulated RNA processing with accumulation of unspliced RNA species in AD, including myc box-dependent-interacting protein 1, clusterin, and presenilin-1. U1-70K knockdown or antisense oligonucleotide inhibition of U1 snRNP increases the protein level of amyloid precursor protein. Thus, our results demonstrate unique U1 snRNP pathology and implicate abnormal RNA splicing in AD pathogenesis.


Subject(s)
Alternative Splicing/physiology , Alzheimer Disease/physiopathology , Brain/metabolism , Proteome/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/metabolism , Alternative Splicing/genetics , Blotting, Western , Chromatography, Liquid , Fluorescent Antibody Technique , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Proteome/genetics , Proteomics , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
10.
Neurobiol Dis ; 77: 155-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25779967

ABSTRACT

Disturbances in the sleep/wake cycle are prevalent in patients with Rett syndrome (RTT). We sought to determine whether the circadian system is disrupted in a RTT model, Mecp2(-/y) mice. We found that MeCP2 mutants showed decreased strength and precision of daily rhythms of activity coupled with extremely fragmented sleep. The central circadian clock (suprachiasmatic nucleus) exhibited significant reduction in the number of neurons expressing vasoactive intestinal peptide (VIP) as well as compromised spontaneous neural activity. The molecular clockwork was disrupted both centrally in the SCN and in peripheral organs, indicating a general disorganization of the circadian system. Disruption of the molecular clockwork was observed in fibroblasts of RTT patients. Finally, MeCP2 mutant mice were vulnerable to circadian disruption as chronic jet lag accelerated mortality. Our finds suggest an integral role of MeCP2 in the circadian timing system and provides a possible mechanistic explanation for the sleep/wake distrubances observed in RTT patients. The work raises the possibility that RTT patients may benefit from a temporally structured environment.


Subject(s)
Chronobiology Disorders/etiology , Disease Models, Animal , Rett Syndrome/complications , Action Potentials/genetics , Animals , Cells, Cultured , Chronobiology Disorders/genetics , Embryo, Mammalian , Gene Expression Regulation/genetics , In Vitro Techniques , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Mutation/genetics , Patch-Clamp Techniques , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Rett Syndrome/genetics , Sleep Wake Disorders/etiology
11.
Diabetologia ; 57(1): 236-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24078059

ABSTRACT

AIMS/HYPOTHESIS: Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus (ARC) regulate energy homeostasis by secreting α-melanocyte-stimulating hormone (α-MSH), derived from POMC precursor, in response to leptin signalling. Expression of Pomc is subject to multiple modes of regulation, including epigenetic regulation. Methyl-CpG-binding protein 2 (MeCP2), a nuclear protein essential for neuronal function, interacts with promoters to influence gene expression. We aim to address whether MeCP2 regulates hypothalamic Pomc expression and to investigate the role of epigenetics, particularly DNA methylation, in this process. METHODS: We generated a mouse line with MeCP2 specifically deleted in POMC neurons (Mecp2 flox/y /Pomc-Cre [PKO]) and characterised its metabolic phenotypes. We examined the DNA methylation pattern of the Pomc promoter and its impact on hypothalamic gene expression. We also studied the requirement of MeCP2 for, and the effects of, DNA methylation on Pomc promoter activity using luciferase assays. RESULTS: PKO mice are overweight, with increased fat mass resulting from increased food intake and respiratory exchange ratio. PKO mice also exhibit elevated plasma leptin. Deletion of MeCP2 in POMC neurons leads to increased DNA methylation of the hypothalamic Pomc promoter and reduced Pomc expression. Furthermore, in vitro studies show that hypermethylation of the Pomc promoter reduces its transcriptional activity and reveal a functional synergy between MeCP2 and cAMP responsive element binding protein 1 (CREB1) in positively regulating the Pomc promoter. CONCLUSIONS/INTERPRETATION: Our results demonstrate that MeCP2 positively regulates Pomc expression in the hypothalamus. Absence of MeCP2 in POMC neurons leads to increased DNA methylation of the Pomc promoter, which, in turn, downregulates Pomc expression, leading to obesity in mice with an accentuating degree of leptin resistance.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Methyl-CpG-Binding Protein 2/deficiency , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Obesity/metabolism
12.
Adv Sci (Weinh) ; : e2308711, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38881531

ABSTRACT

Understanding the liver stem cells (LSCs) holds great promise for new insights into liver diseases and liver regeneration. However, the heterogenicity and plasticity of liver cells have made it controversial. Here, by employing single-cell RNA-sequencing technology, transcriptome features of Krt19+ bile duct lineage cells isolated from Krt19CreERT; Rosa26R-GFP reporter mouse livers are examined. Distinct biliary epithelial cells which include adult LSCs, as well as their downstream hepatocytes and cholangiocytes are identified. Importantly, a novel cell surface LSCs marker, CD63, as well as CD56, which distinguished active and quiescent LSCs are discovered. Cell expansion and bi-potential differentiation in culture demonstrate the stemness ability of CD63+ cells in vitro. Transplantation and lineage tracing of CD63+ cells confirm their contribution to liver cell mass in vivo upon injury. Moreover, CD63+CD56+ cells are proved to be activated LSCs with vigorous proliferation ability. Further studies confirm that CD63+CD56- quiescent LSCs express VEGFR2 and FGFR1, and they can be activated to proliferation and differentiation through combination of growth factors: VEGF-A and bFGF. These findings define an authentic adult liver stem cells compartment, make a further understanding of fate regulation on LSCs, and highlight its contribution to liver during pathophysiologic processes.

13.
Cancer Cell ; 8(6): 479-84, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16338661

ABSTRACT

The decatenation checkpoint normally delays entry into mitosis until chromosomes have been disentangled through the action of topoisomerase II. We have found that the decatenation checkpoint is highly inefficient in mouse embryonic stem cells, mouse neural progenitor cells, and human CD34+ hematopoietic progenitor cells. Checkpoint efficiency increased when embryonic stem cells were induced to differentiate, which suggests that the deficiency is a feature of the undifferentiated state. Embryonic stem cells completed cell division in the presence of entangled chromosomes, which resulted in severe aneuploidy in the daughter cells. The decatenation checkpoint deficiency is likely to increase the rates of chromosome aberrations in progenitor cells, stem cells, and cancer stem cells.


Subject(s)
Genes, cdc , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Chromosome Aberrations , Diketopiperazines , Etoposide/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Neurons/drug effects , Piperazines/pharmacology , Stem Cells/drug effects , Time Factors
14.
Proc Natl Acad Sci U S A ; 107(42): 18161-6, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20921386

ABSTRACT

MicroRNAs (miRNAs) are a class of small, noncoding RNAs that function as posttranscriptional regulators of gene expression. Many miRNAs are expressed in the developing brain and regulate multiple aspects of neural development, including neurogenesis, dendritogenesis, and synapse formation. Rett syndrome (RTT) is a progressive neurodevelopmental disorder caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MECP2). Although Mecp2 is known to act as a global transcriptional regulator, miRNAs that are directly regulated by Mecp2 in the brain are not known. Using massively parallel sequencing methods, we have identified miRNAs whose expression is altered in cerebella of Mecp2-null mice before and after the onset of severe neurological symptoms. In vivo genome-wide analyses indicate that promoter regions of a significant fraction of dysregulated miRNA transcripts, including a large polycistronic cluster of brain-specific miRNAs, are DNA-methylated and are bound directly by Mecp2. Functional analysis demonstrates that the 3' UTR of messenger RNA encoding Brain-derived neurotrophic factor (Bdnf) can be targeted by multiple miRNAs aberrantly up-regulated in the absence of Mecp2. Taken together, these results suggest that dysregulation of miRNAs may contribute to RTT pathoetiology and also may provide a valuable resource for further investigations of the role of miRNAs in RTT.


Subject(s)
Disease Models, Animal , Genome-Wide Association Study , Methyl-CpG-Binding Protein 2/physiology , MicroRNAs/genetics , Rett Syndrome/genetics , 3' Untranslated Regions , Animals , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic , Rett Syndrome/metabolism
15.
Proc Natl Acad Sci U S A ; 107(6): 2687-92, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133810

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a cognate ligand for the tyrosine kinase receptor B (TrkB) receptor, mediates neuronal survival, differentiation, synaptic plasticity, and neurogenesis. However, BDNF has a poor pharmacokinetic profile that limits its therapeutic potential. Here we report the identification of 7,8-dihydroxyflavone as a bioactive high-affinity TrkB agonist that provokes receptor dimerization and autophosphorylation and activation of downstream signaling. 7,8-Dihydroxyflavone protected wild-type, but not TrkB-deficient, neurons from apoptosis. Administration of 7,8-dihydroxyflavone to mice activated TrkB in the brain, inhibited kainic acid-induced toxicity, decreased infarct volumes in stroke in a TrkB-dependent manner, and was neuroprotective in an animal model of Parkinson disease. Thus, 7,8-dihydroxyflavone imitates BDNF and acts as a robust TrkB agonist, providing a powerful therapeutic tool for the treatment of various neurological diseases.


Subject(s)
Apoptosis/drug effects , Flavones/pharmacology , Neurons/drug effects , Receptor, trkB/agonists , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Flavones/chemistry , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Molecular Structure , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Protein Multimerization/drug effects , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction/drug effects
16.
Biomaterials ; 292: 121919, 2023 01.
Article in English | MEDLINE | ID: mdl-36455486

ABSTRACT

Activation of endogenous neurogenesis by bioactive materials enables restoration of sensory/motor function after complete spinal cord injury (SCI) via formation of new relay neural circuits. The underlying wiring logic of newborn neurons in adult central nervous system (CNS) is unknown. Here, we report neurotrophin3-loaded chitosan biomaterial substantially recovered bladder function after SCI. Multiple neuro-circuitry tracing technologies using pseudorabies virus (PRV), rabies virus (RV), and anterograde adeno-associated virus (AAV), demonstrated that newborn neurons were integrated into the micturition neural circuits and reconnected higher brain centers and lower spinal cord centers to control voiding, and participated in the restoration of the lower urinary tract function, even in the absence of long-distance axonal regeneration. Opto- and chemo-genetic studies further supported the notion that the supraspinal control of the lower urinary tract function was partially recovered. Our data demonstrated that regenerated relay neurons could be properly integrated into disrupted long-range neural circuits to restore function of adult CNS.


Subject(s)
Herpesvirus 1, Suid , Spinal Cord Injuries , Animals , Humans , Infant, Newborn , Urinary Bladder , Spinal Cord Injuries/therapy , Neurons , Spinal Cord
17.
J Neurosci ; 31(49): 17800-10, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22159096

ABSTRACT

Aging-related cognitive declines are well documented in humans and animal models. Yet the synaptic and molecular mechanisms responsible for cognitive aging are not well understood. Here we demonstrated age-dependent deficits in long-term synaptic plasticity and loss of dendritic spines in the hippocampus of aged Fisher 344 rats, which were closely associated with reduced histone acetylation, upregulation of histone deacetylase (HDAC) 2, and decreased expression of a histone acetyltransferase. Further analysis showed that one of the key genes affected by such changes was the brain-derived neurotrophic factor (Bdnf) gene. Age-dependent reductions in H3 and H4 acetylation were detected within multiple promoter regions of the Bdnf gene, leading to a significant decrease in BDNF expression and impairment of downstream signaling in the aged hippocampus. These synaptic and signaling deficits could be rescued by enhancing BDNF and trkB expression via HDAC inhibition or by directly activating trkB receptors with 7,8-dihydroxyflavone, a newly identified, selective agonist for trkB. Together, our findings suggest that age-dependent declines in chromatin histone acetylation and the resulting changes in BDNF expression and signaling are key mechanisms underlying the deterioration of synaptic function and structure in the aging brain. Furthermore, epigenetic or pharmacological enhancement of BDNF-trkB signaling could be a promising strategy for reversing cognitive aging.


Subject(s)
Aging/physiology , Brain-Derived Neurotrophic Factor/genetics , Epigenomics/methods , Gene Expression Regulation/physiology , Neuronal Plasticity/physiology , Signal Transduction/genetics , Analysis of Variance , Animals , Biophysics , Brain-Derived Neurotrophic Factor/metabolism , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Electric Stimulation , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/drug effects , Hippocampus/physiology , Hippocampus/ultrastructure , Histone Acetyltransferases/metabolism , Histone Deacetylase 2/metabolism , In Vitro Techniques , Male , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Rats , Rats, Inbred F344 , Receptor, trkB/metabolism , Signal Transduction/drug effects , Silver Staining/methods
18.
J Neurosci ; 31(22): 8306-19, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21632951

ABSTRACT

The endoribonuclease, Dicer, is indispensable for generating the majority of mature microRNAs (miRNAs), which are posttranscriptional regulators of gene expression involved in a wide range of developmental and pathological processes in the mammalian CNS. Although functions of Dicer-dependent miRNA pathways in neurons and oligodendrocytes have been extensively investigated, little is known about the role of Dicer in astrocytes. Here, we report the effect of Cre-loxP-mediated conditional deletion of Dicer selectively from postnatal astroglia on brain development. Dicer-deficient mice exhibited normal motor development and neurological morphology before postnatal week 5. Thereafter, mutant mice invariably developed a rapidly fulminant neurological decline characterized by ataxia, severe progressive cerebellar degeneration, seizures, uncontrollable movements, and premature death by postnatal week 9-10. Integrated transcription profiling, histological, and functional analyses of cerebella showed that deletion of Dicer in cerebellar astrocytes altered the transcriptome of astrocytes to be more similar to an immature or reactive-like state before the onset of neurological symptoms or morphological changes. As a result, critical and mature astrocytic functions including glutamate uptake and antioxidant pathways were substantially impaired, leading to massive apoptosis of cerebellar granule cells and degeneration of Purkinje cells. Collectively, our study demonstrates the critical involvement of Dicer in normal astrocyte maturation and maintenance. Our findings also reveal non-cell-autonomous roles of astrocytic Dicer-dependent pathways in regulating proper neuronal functions and implicate that loss of or dysregulation of astrocytic Dicer-dependent pathways may be involved in neurodegeneration and other neurological disorders.


Subject(s)
Astrocytes/metabolism , Astrocytes/physiology , Cerebellum/growth & development , Cerebellum/pathology , Nerve Degeneration/physiopathology , Psychomotor Disorders/genetics , Psychomotor Disorders/pathology , Ribonuclease III/physiology , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/physiology , Glial Fibrillary Acidic Protein , Glutamic Acid/metabolism , In Vitro Techniques , Integrases/genetics , Male , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques/methods , Psychomotor Disorders/metabolism , Psychomotor Disorders/physiopathology , Purkinje Cells/pathology , Ribonuclease III/genetics , Signal Transduction/genetics , Signal Transduction/physiology
19.
Nat Chem Biol ; 6(9): 645-51, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20657584

ABSTRACT

Mechanistic studies of O-GlcNAc glycosylation have been limited by an inability to monitor the glycosylation stoichiometries of proteins obtained from cells. Here we describe a powerful method to visualize the O-GlcNAc-modified protein subpopulation using resolvable polyethylene glycol mass tags. This approach enables rapid quantification of in vivo glycosylation levels on endogenous proteins without the need for protein purification, advanced instrumentation or expensive radiolabels. In addition, it establishes the glycosylation state (for example, mono-, di-, tri-) of proteins, providing information regarding overall O-GlcNAc site occupancy that cannot be obtained using mass spectrometry. Finally, we apply this strategy to rapidly assess the complex interplay between glycosylation and phosphorylation and discover an unexpected reverse 'yin-yang' relationship on the transcriptional repressor MeCP2 that was undetectable by traditional methods. We anticipate that this mass-tagging strategy will advance our understanding of O-GlcNAc glycosylation, as well as other post-translational modifications and poorly understood glycosylation motifs.


Subject(s)
Acetylglucosamine/analysis , Acetylglucosamine/metabolism , Polyethylene Glycols/chemistry , Protein Processing, Post-Translational , Acetylglucosamine/chemistry , Glycosylation , Kinetics , Mass Spectrometry , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Phosphorylation , Polyethylene Glycols/analysis , Protein Processing, Post-Translational/genetics , Transcription, Genetic
20.
Front Aging Neurosci ; 14: 891644, 2022.
Article in English | MEDLINE | ID: mdl-35813950

ABSTRACT

Background: Exercise plays an essential role in improving motor symptoms in Parkinson's disease (PD), but the underlying mechanism in the central nervous system remains unclear. Methods: Motor ability was observed after 12-week treadmill exercise on a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. RNA-sequencing on four brain regions (cerebellum, cortex, substantia nigra (SN), and striatum) from control animals, MPTP-induced PD, and MPTP-induced PD model treated with exercise for 12 weeks were performed. Transcriptional networks on the four regions were further identified by an integrative network biology approach. Results: The 12-week treadmill exercise significantly improved the motor ability of an MPTP-induced mouse model of PD. RNA-seq analysis showed SN and striatum were remarkably different among individual region's response to exercise in the PD model. Especially, synaptic regulation pathways about axon guidance, synapse assembly, neurogenesis, synaptogenesis, transmitter transport-related pathway, and synaptic regulation genes, including Neurod2, Rtn4rl2, and Cd5, were upregulated in SN and striatum. Lastly, immunofluorescence staining revealed that exercise rescued the loss of TH+ synapses in the striatal region in PD mice, which validates the key role of synaptic regulation pathways in exercise-induced protective effects in vivo. Conclusion: SN and striatum are important brain regions in which critical transcriptional changes, such as in synaptic regulation pathways, occur after the exercise intervention on the PD model.

SELECTION OF CITATIONS
SEARCH DETAIL