Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Brain ; 145(9): 3250-3263, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35775424

ABSTRACT

ADNP and POGZ are two top-ranking risk factors for autism spectrum disorder and intellectual disability, but how they are linked to these neurodevelopmental disorders is largely unknown. Both ADNP and POGZ are chromatin regulators, which could profoundly affect gene transcription and cellular function in the brain. Using post-mortem tissue from patients with autism spectrum disorder, we found diminished expression of ADNP and POGZ in the prefrontal cortex, a region highly implicated in neurodevelopmental disorders. To understand the functional role of these neurodevelopmental disorder risk factors, we used viral-based gene transfer to investigate how Adnp or Pogz deficiency in mouse prefrontal cortex affects behavioural, transcriptomic and synaptic function. Mice with prefrontal cortex deficiency of Adnp or Pogz exhibited specific impairment of cognitive task performance. RNA-sequencing revealed that Adnp or Pogz deficiency induced prominent upregulation of overlapping genes enriched in neuroinflammation, similar to the elevation of pro-inflammatory genes in humans with neurodevelopmental disorders. Concomitantly, Adnp or Pogz deficiency led to the significant increase of pro-phagocytic microglial activation in prefrontal cortex, as well as the significant decrease of glutamatergic transmission and postsynaptic protein expression. These findings have uncovered the convergent functions of two top risk factors for autism spectrum disorder and intellectual disability in prefrontal cortex, providing a mechanism linking chromatin, transcriptional and synaptic dysregulation to cognitive deficits associated with neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder , Cell Cycle Proteins , DNA-Binding Proteins , Homeodomain Proteins , Intellectual Disability , Nerve Tissue Proteins , Neurodevelopmental Disorders , Animals , Autism Spectrum Disorder/genetics , Cell Cycle Proteins/genetics , Chromatin , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Mice , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/metabolism , Risk Factors , Transposases/genetics , Transposases/metabolism
2.
Proc Natl Acad Sci U S A ; 117(46): 29069-29079, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139536

ABSTRACT

Chronic traumatic encephalopathy (CTE) is associated with repeated traumatic brain injuries (TBI) and is characterized by cognitive decline and the presence of neurofibrillary tangles (NFTs) of the protein tau in patients' brains. Here we provide direct evidence that cell-scale mechanical deformation can elicit tau abnormalities and synaptic deficits in neurons. Using computational modeling, we find that the early pathological loci of NFTs in CTE brains are regions of high deformation during injury. The mechanical energy associated with high-strain rate deformation alone can induce tau mislocalization to dendritic spines and synaptic deficits in cultured rat hippocampal neurons. These cellular changes are mediated by tau hyperphosphorylation and can be reversed through inhibition of GSK3ß and CDK5 or genetic deletion of tau. Together, these findings identify a mechanistic pathway that directly relates mechanical deformation of neurons to tau-mediated synaptic impairments and provide a possibly exploitable therapeutic pathway to combat CTE.


Subject(s)
Brain Injuries, Traumatic/metabolism , Chronic Traumatic Encephalopathy/metabolism , Dendritic Spines/metabolism , Neurons/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Brain Injuries, Traumatic/pathology , Chronic Traumatic Encephalopathy/pathology , Cyclin-Dependent Kinase 5/metabolism , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Humans , Male , Neurofibrillary Tangles/metabolism , Rats , tau Proteins/genetics
3.
J Neurophysiol ; 123(4): 1283-1294, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31891524

ABSTRACT

Sensory processing deficits are increasingly recognized as core symptoms of autism spectrum disorders (ASDs). However the molecular and circuit mechanisms that lead to sensory deficits are unknown. We show that two molecularly disparate mouse models of autism display similar deficits in sensory-evoked responses in the mouse olfactory system. We find that both Cntnap2- and Shank3-deficient mice of both sexes exhibit reduced response amplitude and trial-to-trial reliability during repeated odor presentation. Mechanistically, we show that both mouse models have weaker and fewer synapses between olfactory sensory nerve (OSN) terminals and olfactory bulb tufted cells and weaker synapses between OSN terminals and inhibitory periglomerular cells. Consequently, deficits in sensory processing provide an excellent candidate phenotype for analysis in ASDs.NEW & NOTEWORTHY The genetics of autism spectrum disorder (ASD) are complex. How the many risk genes generate the similar sets of symptoms that define the disorder is unknown. In particular, little is understood about the functional consequences of these genetic alterations. Sensory processing deficits are important aspects of the ASD diagnosis and may be due to unreliable neural circuits. We show that two mouse models of autism, Cntnap2- and Shank3-deficient mice, display reduced odor-evoked response amplitudes and reliability. These data suggest that altered sensory-evoked responses may constitute a circuit phenotype in ASDs.


Subject(s)
Autism Spectrum Disorder/physiopathology , Olfaction Disorders/physiopathology , Olfactory Bulb/physiopathology , Olfactory Nerve/physiopathology , Olfactory Perception/physiology , Perceptual Disorders/physiopathology , Synaptic Potentials/physiology , Animals , Calcium , Disease Models, Animal , Female , Male , Membrane Proteins/deficiency , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microscopy, Fluorescence, Multiphoton , Nerve Tissue Proteins/deficiency , Patch-Clamp Techniques , Phenotype
4.
Brain Behav Immun ; 81: 329-340, 2019 10.
Article in English | MEDLINE | ID: mdl-31255679

ABSTRACT

Synaptic deficits and neuronal dystrophy in the prefrontal cortex (PFC) are linked to behavioral and cognitive symptoms in depressed individuals. Preclinical studies indicate that chronic stress causes synaptic deficits on pyramidal neurons in the PFC that contribute to behavioral and cognitive impairments. Our recent work shows that chronic stress provokes microglia-mediated neuronal remodeling via neuronal colony stimulating factor (CSF)-1 signaling, leading to synaptic deficits and depressive-like behaviors. Other reports indicate that elevated corticosterone causes pyramidal neuron atrophy and microglia activation in the medial PFC, implicating glucocorticoid signaling in microglia-mediated neuronal remodeling following chronic stress. In this study, male mice were exposed to chronic unpredictable stress (CUS) and received daily administration of glucocorticoid receptor antagonist RU486 (25 mg/kg, i.p.). As expected, CUS exposure caused adrenal hypertrophy and elevated plasma corticosterone levels. Glucocorticoid receptor blockade prevented behavioral despair and cognitive impairments following CUS. Moreover, RU486 administration diminished CUS-induced CSF1 signaling in the PFC and reduced markers of phagocytosis on purified microglia. Confocal imaging in Thy1-GFP(M) mice showed that CUS increased microglia-mediated neuronal remodeling, and RU486 administration attenuated microglial engulfment of neuronal elements and prevented dendritic spine density deficits on pyramidal neurons following CUS. These results demonstrate that chronic stress-induced glucocorticoid signaling promotes CSF1 signaling and microglia-mediated neuronal remodeling in the medial PFC, which contributes to development of behavioral despair and cognitive impairments. This study presents primary evidence that neuroendocrine responses engage neuron-microglia interactions in the PFC; further implicating microglia in stress-induced neuronal remodeling, PFC dysfunction, and associated behavioral consequences.


Subject(s)
Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Stress, Psychological/metabolism , Animals , Behavior, Animal/drug effects , Brain/metabolism , Corticosterone/blood , Depression , Hippocampus/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Mifepristone/pharmacology , Neurons/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors
5.
Alzheimers Dement ; 12(5): 527-37, 2016 05.
Article in English | MEDLINE | ID: mdl-26555315

ABSTRACT

INTRODUCTION: Stress and corticotropin-releasing factor (CRF) have been implicated as mechanistically involved in Alzheimer's disease (AD), but agents that impact CRF signaling have not been carefully tested for therapeutic efficacy or long-term safety in animal models. METHODS: To test whether antagonism of the type-1 corticotropin-releasing factor receptor (CRFR1) could be used as a disease-modifying treatment for AD, we used a preclinical prevention paradigm and treated 30-day-old AD transgenic mice with the small-molecule, CRFR1-selective antagonist, R121919, for 5 months, and examined AD pathologic and behavioral end points. RESULTS: R121919 significantly prevented the onset of cognitive impairment in female mice and reduced cellular and synaptic deficits and beta amyloid and C-terminal fragment-ß levels in both genders. We observed no tolerability or toxicity issues in mice treated with R121919. DISCUSSION: CRFR1 antagonism presents a viable disease-modifying therapy for AD, recommending its advancement to early-phase human safety trials.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Cognition/physiology , Disease Models, Animal , Receptors, Corticotropin-Releasing Hormone , Synapses/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Humans , Mice , Mice, Transgenic , Pyrimidines , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics
6.
Neurobiol Dis ; 63: 129-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24326163

ABSTRACT

Microdeletion of the human CHRNA7 gene (α7 nicotinic acetylcholine receptor, nAChR) as well as dysfunction in N-methyl-d-aspartate receptors (NMDARs) have been associated with cortical dysfunction in a broad spectrum of neurodevelopmental and neuropsychiatric disorders including schizophrenia. However, the pathophysiological roles of synaptic vs. extrasynaptic NMDARs and their interactions with α7 nAChRs in cortical dysfunction remain largely uncharacterized. Using a combination of in vivo and in vitro models, we demonstrate that α7 nAChR gene deletion leads to specific loss of synaptic NMDARs and their coagonist, d-serine, as well as glutamatergic synaptic deficits in mouse cortex. α7 nAChR null mice had decreased cortical NMDAR expression and glutamatergic synapse formation during postnatal development. Similar reductions in NMDAR expression and glutamatergic synapse formation were revealed in cortical cultures lacking α7 nAChRs. Interestingly, synaptic, but not extrasynaptic, NMDAR currents were specifically diminished in cultured cortical pyramidal neurons as well as in acute prefrontal cortical slices of α7 nAChR null mice. Moreover, d-serine responsive synaptic NMDAR-mediated currents and levels of the d-serine synthetic enzyme serine racemase were both reduced in α7 nAChR null cortical pyramidal neurons. Our findings thus identify specific loss of synaptic NMDARs and their coagonist, d-serine, as well as glutamatergic synaptic deficits in α7 nAChR gene deletion models of cortical dysfunction, thereby implicating α7 nAChR-mediated control of synaptic NMDARs and serine racemase/d-serine pathways in cortical dysfunction underlying many neuropsychiatric and neurodevelopmental disorders, particularly those associated with deletion of human CHRNA7.


Subject(s)
Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/genetics , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/deficiency , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor/deficiency , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Disks Large Homolog 4 Protein , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Guanylate Kinases/metabolism , In Vitro Techniques , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, N-Methyl-D-Aspartate/genetics , Vesicular Glutamate Transport Protein 1/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics
7.
Eur J Neurosci ; 39(7): 1214-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24713000

ABSTRACT

In our previous studies, phosphorylation-dependent tau mislocalization to dendritic spines resulted in early cognitive and synaptic deficits. It is well known that amyloid beta (Aß) oligomers cause synaptic dysfunction by inducing calcineurin-dependent AMPA receptor (AMPAR) internalization. However, it is unknown whether Aß-induced synaptic deficits depend upon tau phosphorylation. It is also unknown whether changes in tau can cause calcineurin-dependent loss of AMPARs in synapses. Here, we show that tau mislocalizes to dendritic spines in cultured hippocampal neurons from APPSwe Alzheimer's disease (AD)-transgenic mice and in cultured rat hippocampal neurons treated with soluble Aß oligomers. Interestingly, Aß treatment also impairs synaptic function by decreasing the amplitude of miniature excitatory postsynaptic currents (mEPSCs). The above tau mislocalization and Aß-induced synaptic impairment are both diminished by the expression of AP tau, indicating that these events require tau phosphorylation. The phosphatase activity of calcineurin is important for AMPAR internalization via dephosphorylation of GluA1 residue S845. The effects of Aß oligomers on mEPSCs are blocked by the calcineurin inhibitor FK506. Aß-induced loss of AMPARs is diminished in neurons from knock-in mice expressing S845A mutant GluA1 AMPA glutamate receptor subunits. This finding suggests that changes in phosphorylation state at S845 are involved in this pathogenic cascade. Furthermore, FK506 rescues deficits in surface AMPAR clustering on dendritic spines in neurons cultured from transgenic mice expressing P301L tau proteins. Together, our results support the role of tau and calcineurin as two intermediate signaling molecules between Aß initiation and eventual synaptic dysfunction early in AD pathogenesis.


Subject(s)
Amyloid beta-Peptides/toxicity , Dendritic Spines/metabolism , Miniature Postsynaptic Potentials , Peptide Fragments/toxicity , Receptors, AMPA/metabolism , tau Proteins/metabolism , Animals , Calcineurin/metabolism , Calcineurin Inhibitors/pharmacology , Cells, Cultured , Dendritic Spines/drug effects , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials , Mice , Mutation , Phosphorylation , Protein Transport , Receptors, AMPA/genetics , Tacrolimus/pharmacology , tau Proteins/genetics
8.
Antioxidants (Basel) ; 12(3)2023 Mar 12.
Article in English | MEDLINE | ID: mdl-36978950

ABSTRACT

Owing to the strong antioxidant capacity of selenium (Se) in vivo, a variety of Se compounds have been shown to have great potential for improving the main pathologies and cognitive impairment in Alzheimer's disease (AD) models. However, the differences in the anti-AD effects and mechanisms of different Se compounds are still unclear. Theoretically, the absorption and metabolism of different forms of Se in the body vary, which directly determines the diversification of downstream regulatory pathways. In this study, low doses of Se-methylselenocysteine (SMC), selenomethionine (SeM), or sodium selenate (SeNa) were administered to triple transgenic AD (3× Tg-AD) mice for short time periods. AD pathology, activities of selenoenzymes, and metabolic profiles in the brain were studied to explore the similarities and differences in the anti-AD effects and mechanisms of the three Se compounds. We found that all of these Se compounds significantly increased Se levels and antioxidant capacity, regulated amino acid metabolism, and ameliorated synaptic deficits, thus improving the cognitive capacity of AD mice. Importantly, SMC preferentially increased the expression and activity of thioredoxin reductase and reduced tau phosphorylation by inhibiting glycogen synthase kinase-3 beta (GSK-3ß) activity. Glutathione peroxidase 1 (GPx1), the selenoenzyme most affected by SeM, decreased amyloid beta production and improved mitochondrial function. SeNa improved methionine sulfoxide reductase B1 (MsrB1) expression, reflected in AD pathology as promoting the expression of synaptic proteins and restoring synaptic deficits. Herein, we reveal the differences and mechanisms by which different Se compounds improve multiple pathologies of AD and provide novel insights into the targeted administration of Se-containing drugs in the treatment of AD.

9.
eNeuro ; 10(5)2023 05.
Article in English | MEDLINE | ID: mdl-37072176

ABSTRACT

SYNGAP1 haploinsufficiency in humans causes intellectual disability (ID). SYNGAP1 is highly expressed in cortical excitatory neurons and, reducing its expression in mice accelerates the maturation of excitatory synapses during sensitive developmental periods, restricts the critical period window for plasticity, and impairs cognition. However, its specific role in interneurons remains largely undetermined. In this study, we investigated the effects of conditional Syngap1 disruption in medial ganglionic eminence (MGE)-derived interneurons on hippocampal interneuron firing properties and excitatory synaptic inputs, as well as on pyramidal cell synaptic inhibition and synaptic integration. We show that conditional Syngap1 disruption in MGE-derived interneurons results in cell-specific impairment of firing properties of hippocampal Nkx2.1 fast-spiking interneurons, with enhancement of their AMPA receptor (AMPAR)-mediated excitatory synaptic inputs but compromised short-term plasticity. In contrast, regular-spiking Nkx2.1 interneurons are largely unaffected. These changes are associated with impaired pyramidal cell synaptic inhibition and enhanced summation of excitatory responses. Unexpectedly, we found that the Syngap1flox allele used in this study contains inverted loxP sites and that its targeted recombination in MGE-derived interneurons induces some cell loss during embryonic development and the reversible inversion of the sequence flanked by the loxP sites in postmitotic cells. Together, these results suggest that Syngap1 plays a role in cell-specific regulation of hippocampal interneuron function and inhibition of pyramidal cells in mice. However, because of our finding that the Syngap1flox allele used in this study contains inverted loxP sites, it will be important to further investigate interneuron function using a different Syngap1 conditional allele.


Subject(s)
Interneurons , Pyramidal Cells , Humans , Mice , Animals , Mice, Transgenic , Interneurons/physiology , Pyramidal Cells/physiology , Hippocampus/metabolism , Recombination, Genetic , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
10.
J Alzheimers Dis ; 87(2): 843-861, 2022.
Article in English | MEDLINE | ID: mdl-35404278

ABSTRACT

BACKGROUND: miR-34c has been found to be implicated in the pathological process of Alzheimer's disease, diabetes, and its complications. OBJECTIVE: To investigate the underlying mechanisms of miR-34c in the pathogenesis of diabetic encephalopathy (DE). METHODS: Diabetes mellitus rats were developed by incorporating a high-fat diet and streptozotocin injection. Morris water maze test and novel object recognition test were used to assess the cognitive function of rats. Expression of miR-34c were detected by fluorescence in situ hybridization and qRT-PCR. Immunofluorescence and western blot were used to evaluate synaptotagmin 1 (SYT1) and AdipoR2 or other proteins. Golgi staining was performed to investigate dendritic spine density. RESULTS: The increased miR-34c induced by advanced glycation end-products (AGEs) was mediated by ROS-JNK-p53 pathway, but not ROS-Rb-E2F1 pathway, in hippocampus of DE rats or in HT-22 cells. miR-34c negatively regulated the expression of SYT1, but not AdipoR2, in hippocampal neurons. miR-34c inhibitor rescued the AGE-induced decrease in the density of dendritic spines in primary hippocampal neurons. Administration of AM34c by the intranasal delivery increased the hippocampus levels of SYT1 and ameliorated the cognitive function in DE rats. The serum levels of miR-34c were increased in patients with DE comparing with normal controls. CONCLUSION: These results demonstrated that AGE-induced oxidative stress mediated increase of miR-34c through ROS-JNK-p53 pathway, resulting in synaptic deficits and cognitive decline by targeting SYT1 in DE, and the miR-34c/SYT1 axis could be considered as a novel therapeutic target for DE patients.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Diabetes Mellitus , MicroRNAs , Animals , Cognitive Dysfunction/genetics , Glycation End Products, Advanced/metabolism , Humans , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Reactive Oxygen Species/metabolism , Synaptotagmin I/genetics , Synaptotagmin I/metabolism , Tumor Suppressor Protein p53
11.
Neurotherapeutics ; 18(4): 2468-2483, 2021 10.
Article in English | MEDLINE | ID: mdl-34738197

ABSTRACT

Alzheimer's disease (AD) is conceptualized as a synaptic failure disorder in which loss of glutamatergic synapses is a major driver of cognitive decline. Thus, novel therapeutic strategies aimed at regenerating synapses may represent a promising approach to mitigate cognitive deficits in AD patients. At present, no disease-modifying drugs exist for AD, and approved therapies are palliative at best, lacking in the ability to reverse the synaptic failure. Here, we tested the efficacy of a novel synaptogenic small molecule, SPG302 - a 3rd-generation benzothiazole derivative that increases the density of axospinous glutamatergic synapses - in 3xTg-AD mice. Daily dosing of 3xTg-AD mice with SPG302 at 3 and 30 mg/kg (i.p.) for 4 weeks restored hippocampal synaptic density and improved cognitive function in hippocampal-dependent tasks. Mushroom and stubby spine profiles were increased by SPG302, and associated with enhanced expression of key postsynaptic proteins - including postsynaptic density protein 95 (PSD95), drebrin, and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) - and increased colocalization of PSD95 with synaptophysin. Notably, SPG302 proved efficacious in this model without modifying Aß and tau pathology. Thus, our study provides preclinical support for the idea that compounds capable of restoring synaptic density offer a viable strategy to reverse cognitive decline in AD.


Subject(s)
Alzheimer Disease , Cognition Disorders , Cognitive Dysfunction , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cognition , Cognition Disorders/pathology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Synapses/metabolism , Synapses/pathology , tau Proteins/metabolism
12.
Int Immunopharmacol ; 101(Pt B): 108335, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34781121

ABSTRACT

The accumulation of amyloid beta (Aß) in the brain is thought to be associated with cognitive deficits in Alzheimer's disease (AD). However, current methods to combat Aß neurotoxicity are still lacking. G protein-coupled receptor 17 (GPR17) has become a target for treating inflammation in brain diseases, but it is unclear whether it has a role in AD. Here, we investigated the effects of cangrelor, a GPR17 antagonist, on neurotoxicity and memory impairment induced by intracerebroventricular (i.c.v.) injection of Aß1-42 in mice. The behavior results showed that cangrelor (2.0 or 4.0 µg/mouse, i.c.v.) treatment reversed the deficits in memory and learning ability induced by Aß1-42 in mice. Importantly, we demonstrated for the first time that GPR17 expression in the hippocampus and frontal cortex is increased in response to Aß1-42 exposures. We also found that cangrelor treatment reduced the activity of ß-secretase 1 (BACE1) and the levels of soluble Aß1-42 in the hippocampus and frontal cortex. Meanwhile, cangrelor treatment suppressed oxidative stress induced by Aß1-42, as proved by reduced production of malondialdehyde (MDA), and increased glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), and promoted the expression of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Furthermore, cangrelor also suppressed Aß1-42-induced neuroinflammation, characterized by suppressed activation of microglia, decreased the levels of pro-inflammatory cytokines, and nuclear translocation of NF-κB p65, as well as ameliorated synaptic deficits by promoting the upregulation of synaptic proteins, and increasing the number of Golgi-Cox stained dendritic spines. These results suggest that cangrelor may reverse Aß1-42-induced cognition deficits via inhibiting oxidative stress, neuroinflammation, and synaptic dysfunction mediated by Nrf2/HO-1 and NF-κB signaling.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/drug therapy , Electrical Synapses/physiology , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Adenosine Monophosphate/therapeutic use , Animals , Heme Oxygenase-1/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction
13.
Environ Toxicol Pharmacol ; 76: 103350, 2020 May.
Article in English | MEDLINE | ID: mdl-32058320

ABSTRACT

Lead (Pb) is recognized as a potent inducer of synaptic toxicity generally associated with reduced synaptic transmission and increased neuronal fiber excitability, becoming an environmental risk for neurodegenerative processes. Despite numerous toxicological studies on Pb have been directed to the developing brain, attention concerning long-term consequences of pubertal chronic Pb exposure on neuronal activity is still lacking. Thus, we exposed 4-week-old male mice to 0.2 % lead acetate solution for one month, then, conducted behavioral tests or extracted brain homogenate from mice prefrontal cortex (PFC) and hippocampus at the age of 4, 13 and 16-month-old respectively. Our results showed that treated mice exhibited an evident increase in latency to reach platform following pubertal Pb exposure and aging. The increase of 8-OHdG revealed evident neural DNA oxidative damage across time upon pubertal Pb exposure. In the hippocampus of lead exposed mice at three age nodes, the expression of brain-derived neurotrophic factor precursor (proBDNF) increased, while that of mature BDNF (mBDNF), cAMP-response element binding protein (CREB) and phosphorylated CREB (pCREB) decreased compared with the control group. Furthermore, the expression of BACE1 protein and tau phosphorylation level in PFC and hippocampus increased, APP mRNAs in PFC and prolonged induction of BACE1 in hippocampus. Our results show that chronic Pb exposure from pubertal stage onward can either initiate divergent synaptic-related gene expression patterns in adulthood or trigger time-course of neurodegenerative profile within the PFC or hippocampus, which can contribute consistent deficits of cognition across subsequent age-nodes.


Subject(s)
Aging , DNA/metabolism , Gene Expression Regulation/drug effects , Lead/toxicity , Learning/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Female , Hippocampus/drug effects , Hippocampus/metabolism , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sexual Maturation , Synapses
14.
Cell Rep ; 33(8): 108420, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33238112

ABSTRACT

Neuronal endosomal dysfunction, the earliest known pathobiology specific to Alzheimer's disease (AD), is mediated by the aberrant activation of Rab5 triggered by APP-ß secretase cleaved C-terminal fragment (APP-ßCTF). To distinguish pathophysiological consequences specific to overactivated Rab5 itself, we activate Rab5 independently from APP-ßCTF in the PA-Rab5 mouse model. We report that Rab5 overactivation alone recapitulates diverse prodromal and degenerative features of AD. Modest neuron-specific transgenic Rab5 expression inducing hyperactivation of Rab5 comparable to that in AD brain reproduces AD-related Rab5-endosomal enlargement and mistrafficking, hippocampal synaptic plasticity deficits via accelerated AMPAR endocytosis and dendritic spine loss, and tau hyperphosphorylation via activated glycogen synthase kinase-3ß. Importantly, Rab5-mediated endosomal dysfunction induces progressive cholinergic neurodegeneration and impairs hippocampal-dependent memory. Aberrant neuronal Rab5-endosome signaling, therefore, drives a pathogenic cascade distinct from ß-amyloid-related neurotoxicity, which includes prodromal and neurodegenerative features of AD, and suggests Rab5 overactivation as a potential therapeutic target.


Subject(s)
Alzheimer Disease/genetics , Endosomes/metabolism , Neurodegenerative Diseases/genetics , rab5 GTP-Binding Proteins/metabolism , Alzheimer Disease/physiopathology , Animals , Disease Models, Animal , Humans , Mice , Neurodegenerative Diseases/physiopathology
15.
Cell Transplant ; 28(9-10): 1173-1182, 2019.
Article in English | MEDLINE | ID: mdl-31161783

ABSTRACT

Cerebral organoids are an emerging cutting-edge technology to model human brain development and neurodevelopmental disorders, for which mouse models exhibit significant limitations. In the human brain, synaptic connections define neural circuits, and synaptic deficits account for various neurodevelopmental disorders. Thus, harnessing the full power of cerebral organoids for human brain modeling requires the ability to visualize and analyze synapses in cerebral organoids. Previously, we devised an optimized method to generate human cerebral organoids, and showed that optimal organoids express mature-neuron markers, including synaptic proteins and neurotransmitter receptors and transporters. Here, we give evidence for synaptogenesis in cerebral organoids, via microscopical visualization of synapses. We also describe multiple approaches to quantitatively analyze synapses in cerebral organoids. Collectively, our work provides sufficient evidence for the possibility of modeling synaptogenesis and synaptic disorders in cerebral organoids, and may help advance the use of cerebral organoids in molecular neuroscience and studies of neurodevelopmental disorders such as autism.


Subject(s)
Cerebellum/metabolism , Models, Neurological , Neurodevelopmental Disorders/metabolism , Organoids/metabolism , Synapses/metabolism , Cerebellum/pathology , Humans , Nerve Tissue Proteins/metabolism , Neurodevelopmental Disorders/pathology , Organoids/pathology , Synapses/pathology
16.
J Alzheimers Dis ; 59(1): 223-239, 2017.
Article in English | MEDLINE | ID: mdl-28598851

ABSTRACT

Loss of synapse and synaptic dysfunction contribute importantly to cognitive impairment in Alzheimer's disease (AD). Mitochondrial dysfunction and oxidative stress are early pathological features in AD-affected brain. However, the effect of AD mitochondria on synaptogenesis remains to be determined. Using human trans-mitochondrial "cybrid" (cytoplasmic hybrid) neuronal cells whose mitochondria were transferred from platelets of patients with sporadic AD or age-matched non-AD subjects with relatively normal cognition, we provide the first evidence of mitochondrial dysfunction compromises synaptic development and formation of synapse in AD cybrid cells in response to chemical-induced neuronal differentiation. Compared to non-AD control cybrids, AD cybrid cells showed synaptic loss which was evidenced by a significant reduction in expression of two synaptic marker proteins: synaptophysin (presynaptic marker) and postsynaptic density protein-95, and neuronal proteins (MAP-2 and NeuN) upon neuronal differentiation. In parallel, AD-mediated synaptic deficits correlate to mitochondrial dysfunction and oxidative stress as well as activation of p38 MAP kinase. Notably, inhibition of p38 MAP kinase by pharmacological specific p38 inhibitor significantly increased synaptic density, improved mitochondrial function, and reduced oxidative stress. These results suggest that activation of p38 MAP kinase signaling pathway contributes to AD-mediated impairment in neurogenesis, possibly by inhibiting the neuronal differentiation. Our results provide new insight into the crosstalk of dysfunctional AD mitochondria to synaptic formation and maturation via activation of p38 MAP kinase. Therefore, blockade of p38 MAP kinase signal transduction could be a potential therapeutic strategy for AD by alleviating loss of synapses.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Mitochondria/pathology , Mitochondrial Diseases/etiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation , Disks Large Homolog 4 Protein , Electron Transport Complex IV/metabolism , Female , Humans , Hybrid Cells , Male , Membrane Potential, Mitochondrial , Mitochondria/ultrastructure , Mitochondrial Diseases/pathology , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Rhodamines/metabolism , Synapses/metabolism , Synapses/pathology , Synaptophysin/metabolism
17.
Front Cell Neurosci ; 9: 55, 2015.
Article in English | MEDLINE | ID: mdl-25767435

ABSTRACT

Autism spectrum disorders (ASDs) are genetically and clinically heterogeneous and lack effective medications to treat their core symptoms. Studies of syndromic ASDs caused by single gene mutations have provided insights into the pathophysiology of autism. Fragile X and Rett syndromes belong to the syndromic ASDs in which preclinical studies have identified rational targets for drug therapies focused on correcting underlying neural dysfunction. These preclinical discoveries are increasingly translating into exciting human clinical trials. Since there are significant molecular and neurobiological overlaps among ASDs, targeted treatments developed for fragile X and Rett syndromes may be helpful for autism of different etiologies. Here, we review the targeted pharmacological treatment of fragile X and Rett syndromes and discuss related issues in both preclinical studies and clinical trials of potential therapies for the diseases.

SELECTION OF CITATIONS
SEARCH DETAIL