Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 11.272
1.
BMC Cardiovasc Disord ; 24(1): 266, 2024 May 21.
Article En | MEDLINE | ID: mdl-38773462

BACKGROUND: Cardiopulmonary bypass (CPB) results in brain injury, which is primarily caused by inflammation. Ac2-26 protects against ischemic or hemorrhage brain injury. The present study was to explore the effect and mechanism of Ac2-26 on brain injury in CPB rats. METHODS: Forty-eight rats were randomized into sham, CPB, Ac, Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups. Rats in sham group only received anesthesia and in the other groups received standard CPB surgery. Rats in the sham and CPB groups received saline, and rats in the Ac, Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups received Ac2-26 immediately after CPB. Rats in the Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups were injected with shRNA, inhibitor and agonist of GSK3ß respectively. The neurological function score, brain edema and histological score were evaluated. The neuronal survival and hippocampal pyroptosis were assessed. The cytokines, activity of NF-κB, S100 calcium-binding protein ß(S100ß) and neuron-specific enolase (NSE), and oxidative were tested. The NLRP3, cleaved-caspase-1 and cleaved-gadermin D (GSDMD) in the brain were also detected. RESULTS: Compared to the sham group, all indicators were aggravated in rats that underwent CPB. Compared to the CPB group, Ac2-26 significantly improved neurological scores and brain edema and ameliorated pathological injury. Ac2-26 reduced the local and systemic inflammation, oxidative stress response and promoted neuronal survival. Ac2-26 reduced hippocampal pyroptosis and decreased pyroptotic proteins in brain tissue. The protection of Ac2-26 was notably lessened by shRNA and inhibitor of GSK3ß. The agonist of GSK3ß recovered the protection of Ac2-26 in presence of shRNA. CONCLUSIONS: Ac2-26 significantly improved neurological function, reduced brain injury via regulating inflammation, oxidative stress response and pyroptosis after CPB. The protective effect of Ac2-26 primarily depended on AKT1/ GSK3ß pathway.


Cardiopulmonary Bypass , Disease Models, Animal , Glycogen Synthase Kinase 3 beta , Proto-Oncogene Proteins c-akt , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Animals , Cardiopulmonary Bypass/adverse effects , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyroptosis/drug effects , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neurons/enzymology , Neuroprotective Agents/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Brain Edema/prevention & control , Brain Edema/metabolism , Brain Edema/enzymology , Brain Edema/pathology , Anti-Inflammatory Agents/pharmacology , Rats , S100 Calcium Binding Protein beta Subunit/metabolism , Inflammation Mediators/metabolism
2.
J Mol Neurosci ; 74(2): 44, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38630337

Plants are a valuable source of information for pharmacological research and new drug discovery. The present study aimed to evaluate the neuroprotective potential of the leaves of the medicinal plant Sterculia setigera. In vitro, the effect of Sterculia setigera leaves dry hydroethanolic extract (SSE) was tested on cultured cerebellar granule neurons (CGN) survival when exposed to hydrogen peroxide (H2O2) or 6-hydroxydopamine (6-OHDA), using the viability probe fluorescein diacetate (FDA), a lactate dehydrogenase (LDH) activity assay, an immunocytochemical staining against Gap 43, and the quantification of the expression of genes involved in apoptosis, necrosis, or oxidative stress. In vivo, the effect of intraperitoneal (ip) injection of SSE was assessed on the developing brain of 8-day-old Wistar rats exposed to ethanol neurotoxicity by measuring caspase-3 activity on cerebellum homogenates, the expression of some genes in tissue extracts, the thickness of cerebellar cortical layers and motor coordination. In vitro, SSE protected CGN against H2O2 and 6-OHDA-induced cell death at a dose of 10 µg/mL, inhibited the expression of genes Casp3 and Bad, and upregulated the expression of Cat and Gpx7. In vivo, SSE significantly blocked the deleterious effect of ethanol by reducing the activity of caspase-3, inhibiting the expression of Bax and Tp53, preventing the reduction of the thickness of the internal granule cell layer of the cerebellar cortex, and restoring motor functions. Sterculia setigera exerts neuroactive functions as claimed by traditional medicine and should be a good candidate for the development of a neuroprotective treatment against neurodegenerative diseases.


Cell Death , Ethanol , Neurons , Neuroprotective Agents , Plant Extracts , Plant Leaves , Sterculia , Animals , Rats , Caspase 3/metabolism , Ethanol/administration & dosage , Ethanol/chemistry , Ethanol/toxicity , Hydrogen Peroxide/toxicity , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Rats, Wistar , Sterculia/chemistry , Plant Leaves/chemistry , Plants, Medicinal/chemistry , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Lactate Dehydrogenases/metabolism , GAP-43 Protein/analysis , Apoptosis/genetics , Oxidative Stress/genetics , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/physiology , Male , Female , Cells, Cultured , Cell Death/drug effects , Gene Expression Regulation/drug effects , Phytochemicals/administration & dosage , Phytochemicals/analysis , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antioxidants/analysis , Antioxidants/chemistry , Antioxidants/pharmacology , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Liquid Chromatography-Mass Spectrometry , Secondary Metabolism
3.
J Virol ; 97(12): e0133823, 2023 Dec 21.
Article En | MEDLINE | ID: mdl-38009916

IMPORTANCE: Betacoronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mouse hepatitis virus (MHV), exploit the lysosomal exocytosis pathway for egress. However, whether all betacoronaviruses members use the same pathway to exit cells remains unknown. Here, we demonstrated that porcine hemagglutinating encephalomyelitis virus (PHEV) egress occurs by Arl8b-dependent lysosomal exocytosis, a cellular egress mechanism shared by SARS-CoV-2 and MHV. Notably, PHEV acidifies lysosomes and activates lysosomal degradative enzymes, while SARS-CoV-2 and MHV deacidify lysosomes and limit the activation of lysosomal degradative enzymes. In addition, PHEV release depends on V-ATPase-mediated lysosomal pH. Furthermore, this is the first study to evaluate ßCoV using lysosome for spreading through the body, and we have found that lysosome played a critical role in PHEV neural transmission and brain damage caused by virus infection in the central nervous system. Taken together, different betacoronaviruses could disrupt lysosomal function differently to exit cells.


Betacoronavirus 1 , Coronavirus Infections , Exocytosis , Lysosomes , Neurons , Animals , Mice , Betacoronavirus 1/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Lysosomes/virology , Murine hepatitis virus/metabolism , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Neurons/virology , SARS-CoV-2/metabolism , Swine/virology , Hydrogen-Ion Concentration , Vacuolar Proton-Translocating ATPases/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/transmission , Coronavirus Infections/virology
4.
PLoS Pathog ; 19(9): e1011487, 2023 09.
Article En | MEDLINE | ID: mdl-37747931

Select prion diseases are characterized by widespread cerebral plaque-like deposits of amyloid fibrils enriched in heparan sulfate (HS), a abundant extracellular matrix component. HS facilitates fibril formation in vitro, yet how HS impacts fibrillar plaque growth within the brain is unclear. Here we found that prion-bound HS chains are highly sulfated, and that the sulfation is essential for accelerating prion conversion in vitro. Using conditional knockout mice to deplete the HS sulfation enzyme, Ndst1 (N-deacetylase / N-sulfotransferase) from neurons or astrocytes, we investigated how reducing HS sulfation impacts survival and prion aggregate distribution during a prion infection. Neuronal Ndst1-depleted mice survived longer and showed fewer and smaller parenchymal plaques, shorter fibrils, and increased vascular amyloid, consistent with enhanced aggregate transit toward perivascular drainage channels. The prolonged survival was strain-dependent, affecting mice infected with extracellular, plaque-forming, but not membrane bound, prions. Live PET imaging revealed rapid clearance of recombinant prion protein monomers into the CSF of neuronal Ndst1- deficient mice, neuronal, further suggesting that HS sulfate groups hinder transit of extracellular prion protein monomers. Our results directly show how a host cofactor slows the spread of prion protein through the extracellular space and identify an enzyme to target to facilitate aggregate clearance.


Neurons , Prion Diseases , Prions , Sulfotransferases , Animals , Mice , Heparitin Sulfate/metabolism , Mice, Knockout , Neurons/enzymology , Prion Diseases/metabolism , Prion Proteins/genetics , Prions/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism
5.
Aging Cell ; 22(3): e13780, 2023 03.
Article En | MEDLINE | ID: mdl-36644807

The contribution of cellular senescence to the behavioral changes observed in the elderly remains elusive. Here, we observed that aging is associated with a decline in protein phosphatase 2A (PP2A) activity in the brains of zebrafish and mice. Moreover, drugs activating PP2A reversed age-related behavioral changes. We developed a transgenic zebrafish model to decrease PP2A activity in the brain through knockout of the ppp2r2c gene encoding a regulatory subunit of PP2A. Mutant fish exhibited the behavioral phenotype observed in old animals and premature accumulation of neural cells positive for markers of cellular senescence, including senescence-associated ß-galactosidase, elevated levels cdkn2a/b, cdkn1a, senescence-associated secretory phenotype gene expression, and an increased level of DNA damage signaling. The behavioral and cell senescence phenotypes were reversed in mutant fish through treatment with the senolytic ABT263 or diverse PP2A activators as well as through cdkn1a or tp53 gene ablation. Senomorphic function of PP2A activators was demonstrated in mouse primary neural cells with downregulated Ppp2r2c. We conclude that PP2A reduction leads to neural cell senescence thereby contributing to age-related behavioral changes and that PP2A activators have senotherapeutic properties against deleterious behavioral effects of brain aging.


Behavior, Animal , Brain , Cellular Senescence , Cognitive Aging , Neurons , Protein Phosphatase 2 , Senotherapeutics , Animals , Mice , Aniline Compounds/pharmacology , Animals, Genetically Modified , Behavior, Animal/drug effects , Behavior, Animal/physiology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism , Biomarkers/metabolism , Brain/enzymology , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cellular Senescence/physiology , Cognitive Aging/physiology , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , Gene Expression Regulation , Gene Knockout Techniques , Models, Animal , Mutation , Neurons/drug effects , Neurons/enzymology , Neurons/physiology , Primary Cell Culture , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Senotherapeutics/pharmacology , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish
6.
J Biol Chem ; 299(3): 102928, 2023 03.
Article En | MEDLINE | ID: mdl-36681123

Inositol pyrophosphates regulate diverse physiological processes; to better understand their functional roles, assessing their tissue-specific distribution is important. Here, we profiled inositol pyrophosphate levels in mammalian organs using an originally designed liquid chromatography-mass spectrometry (LC-MS) protocol and discovered that the gastrointestinal tract (GIT) contained the highest levels of diphosphoinositol pentakisphosphate (IP7) and its precursor inositol hexakisphosphate (IP6). Although their absolute levels in the GIT are diet dependent, elevated IP7 metabolism still exists under dietary regimens devoid of exogenous IP7. Of the major GIT cells, enteric neurons selectively express the IP7-synthesizing enzyme IP6K2. We found that IP6K2-knockout mice exhibited significantly impaired IP7 metabolism in the various organs including the proximal GIT. In addition, our LC-MS analysis displayed that genetic ablation of IP6K2 significantly impaired IP7 metabolism in the gut and duodenal muscularis externa containing myenteric plexus. Whole transcriptome analysis of duodenal muscularis externa further suggested that IP6K2 inhibition significantly altered expression levels of the gene sets associated with mature neurons, neural progenitor/stem cells, and glial cells, as well as of certain genes modulating neuronal differentiation and functioning, implying critical roles of the IP6K2-IP7 axis in developmental and functional regulation of the enteric nervous system. These results collectively reveal an unexpected role of mammalian IP7-a highly active IP6K2-IP7 pathway is conducive to the enteric nervous system.


Enteric Nervous System , Inositol Phosphates , Transcriptome , Animals , Mice , Diphosphates/analysis , Diphosphates/metabolism , Enteric Nervous System/growth & development , Enteric Nervous System/metabolism , Inositol Phosphates/analysis , Inositol Phosphates/metabolism , Mice, Knockout , Neurons/enzymology , Phosphotransferases (Phosphate Group Acceptor)/genetics , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Phytic Acid/metabolism , Gastrointestinal Tract/metabolism
7.
Science ; 378(6623): 983-989, 2022 12 02.
Article En | MEDLINE | ID: mdl-36454826

Neurons harbor high levels of single-strand DNA breaks (SSBs) that are targeted to neuronal enhancers, but the source of this endogenous damage remains unclear. Using two systems of postmitotic lineage specification-induced pluripotent stem cell-derived neurons and transdifferentiated macrophages-we show that thymidine DNA glycosylase (TDG)-driven excision of methylcytosines oxidized with ten-eleven translocation enzymes (TET) is a source of SSBs. Although macrophage differentiation favors short-patch base excision repair to fill in single-nucleotide gaps, neurons also frequently use the long-patch subpathway. Disrupting this gap-filling process using anti-neoplastic cytosine analogs triggers a DNA damage response and neuronal cell death, which is dependent on TDG. Thus, TET-mediated active DNA demethylation promotes endogenous DNA damage, a process that normally safeguards cell identity but can also provoke neurotoxicity after anticancer treatments.


DNA Breaks, Single-Stranded , DNA Demethylation , DNA Repair , Enhancer Elements, Genetic , Induced Pluripotent Stem Cells , Neurons , Thymine DNA Glycosylase , Cell Differentiation , Neurons/enzymology , 5-Methylcytosine/metabolism , Humans , Cell Transdifferentiation
8.
Nature ; 611(7937): 762-768, 2022 11.
Article En | MEDLINE | ID: mdl-36352228

The canonical model of striatal function predicts that animal locomotion is associated with the opposing regulation of protein kinase A (PKA) in direct and indirect pathway striatal spiny projection neurons (SPNs) by dopamine1-7. However, the precise dynamics of PKA in dorsolateral SPNs during locomotion remain to be determined. It is also unclear whether other neuromodulators are involved. Here we show that PKA activity in both types of SPNs is essential for normal locomotion. Using two-photon fluorescence lifetime imaging8-10 of a PKA sensor10 through gradient index lenses, we measured PKA activity within individual SPNs of the mouse dorsolateral striatum during locomotion. Consistent with the canonical view, dopamine activated PKA activity in direct pathway SPNs during locomotion through the dopamine D1 receptor. However, indirect pathway SPNs exhibited a greater increase in PKA activity, which was largely abolished through the blockade of adenosine A2A receptors. In agreement with these results, fibre photometry measurements of an adenosine sensor11 revealed an acute increase in extracellular adenosine during locomotion. Functionally, antagonism of dopamine or adenosine receptors resulted in distinct changes in SPN PKA activity, neuronal activity and locomotion. Together, our results suggest that acute adenosine accumulation interplays with dopamine release to orchestrate PKA activity in SPNs and proper striatal function during animal locomotion.


Adenosine , Corpus Striatum , Cyclic AMP-Dependent Protein Kinases , Dopamine , Locomotion , Neurons , Animals , Mice , Adenosine/metabolism , Corpus Striatum/cytology , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Locomotion/physiology , Neurons/enzymology , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptor, Adenosine A2A/metabolism
9.
Proc Natl Acad Sci U S A ; 119(37): e2120079119, 2022 09 13.
Article En | MEDLINE | ID: mdl-36067316

The extracellular protein Reelin, expressed by Cajal-Retzius (CR) cells at early stages of cortical development and at late stages by GABAergic interneurons, regulates radial migration and the "inside-out" pattern of positioning. Current models of Reelin functions in corticogenesis focus on early CR cell-derived Reelin in layer I. However, developmental disorders linked to Reelin deficits, such as schizophrenia and autism, are related to GABAergic interneuron-derived Reelin, although its role in migration has not been established. Here we selectively inactivated the Reln gene in CR cells or GABAergic interneurons. We show that CR cells have a major role in the inside-out order of migration, while CR and GABAergic cells sequentially cooperate to prevent invasion of cortical neurons into layer I. Furthermore, GABAergic cell-derived Reelin compensates some features of the reeler phenotype and is needed for the fine tuning of the layer-specific distribution of cortical neurons. In the hippocampus, the inactivation of Reelin in CR cells causes dramatic alterations in the dentate gyrus and mild defects in the hippocampus proper. These findings lead to a model in which both CR and GABAergic cell-derived Reelin cooperate to build the inside-out order of corticogenesis, which might provide a better understanding of the mechanisms involved in the pathogenesis of neuropsychiatric disorders linked to abnormal migration and Reelin deficits.


Cerebral Cortex , Nerve Tissue Proteins , Neurons , Reelin Protein , Animals , Cell Movement , Cerebral Cortex/cytology , Cerebral Cortex/embryology , GABAergic Neurons/enzymology , Hippocampus/embryology , Hippocampus/enzymology , Interneurons/enzymology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Reelin Protein/genetics , Reelin Protein/metabolism
10.
Neuroreport ; 33(11): 476-480, 2022 08 03.
Article En | MEDLINE | ID: mdl-35775322

Seizures induce brain region-dependent enhancements in microglia/macrophage activation. Neuronal subset-specific phosphatase and tensin homolog (PTEN) knockout (KO) mice display hyperactive mammalian target of rapamycin (mTOR) signaling in the hippocampus, cerebellum, and cortex followed by seizures that increase in severity with age. To determine if KO mice also exhibit alterations in the spatiotemporal activation pattern of microglia, we used flow cytometry to compare the percentage of major histocompatibility complex-II activated microglia/macrophages between KO and wildtype (WT) mice at 5, 10, and 15 weeks of age. At 5 weeks, microglia/macrophage activation was greater in the cortex, P < 0.001, cerebellum, P < 0.001, and hippocampus, P < 0.001, of KO compared to WT mice. At 10 weeks, activation was greatest in the cortex of KO mice, P < 0.001, in the cerebellum of WT mice, P < 0.001, but similar in the hippocampus, P > 0.05. By 15 weeks, activation in the hippocampus was more than 25 times greater in KO mice compared to WT mice, P < 0.001. We show that hyperactive mTOR signaling is associated with an altered spatiotemporal pattern of microglia/macrophage activation in the brain and induces an enhanced neuroimmune response in the hippocampus.


Brain , Macrophages , Microglia , Neurons , PTEN Phosphohydrolase , Animals , Brain/metabolism , Brain/pathology , Macrophage Activation , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurons/enzymology , Neurons/pathology , PTEN Phosphohydrolase/metabolism , Seizures/metabolism , Seizures/pathology , TOR Serine-Threonine Kinases/metabolism
11.
Science ; 377(6605): eabo0001, 2022 07 29.
Article En | MEDLINE | ID: mdl-35857622

In the 20th century, researchers studying animal and plant signaling pathways discovered a protein domain that is shared across diverse innate immune systems: the Toll/interleukin-1/resistance gene (TIR) domain. The TIR domain is found in several protein architectures and was defined as an adaptor that mediates protein-protein interactions in animal innate immunity and developmental signaling pathways. However, studies of nerve degeneration in animals-and subsequent breakthroughs in plant, bacterial, and archaeal systems-revealed that TIR domains possess enzymatic activities. We provide a synthesis of TIR functions and the role of various related TIR enzymatic products in evolutionarily diverse immune systems. These studies may ultimately guide interventions that would span the tree of life, from treating human neurodegenerative disorders and bacterial infections to preventing plant diseases.


Cell Death , Enzymes , Immune System , Immunity, Innate , Neurodegenerative Diseases , Animals , Enzymes/chemistry , Enzymes/metabolism , Evolution, Molecular , Humans , Immune System/enzymology , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/immunology , Neurons/enzymology , Protein Domains , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 119(30): e2203503119, 2022 07 26.
Article En | MEDLINE | ID: mdl-35867816

Women with polycystic ovary syndrome (PCOS) frequently experience decreased sexual arousal, desire, and sexual satisfaction. While the hypothalamus is known to regulate sexual behavior, the specific neuronal pathways affected in patients with PCOS are not known. To dissect the underlying neural circuitry, we capitalized on a robust preclinical animal model that reliably recapitulates all cardinal PCOS features. We discovered that female mice prenatally treated with anti-Müllerian hormone (PAMH) display impaired sexual behavior and sexual partner preference over the reproductive age. Blunted female sexual behavior was associated with increased sexual rejection and independent of sex steroid hormone status. Structurally, sexual dysfunction was associated with a substantial loss of neuronal nitric oxide synthase (nNOS)-expressing neurons in the ventromedial nucleus of the hypothalamus (VMH) and other areas of hypothalamic nuclei involved in social behaviors. Using in vivo chemogenetic manipulation, we show that nNOSVMH neurons are required for the display of normal sexual behavior in female mice and that pharmacological replenishment of nitric oxide restores normal sexual performance in PAMH mice. Our data provide a framework to investigate facets of hypothalamic nNOS neuron biology with implications for sexual disturbances in PCOS.


Nitric Oxide Synthase Type I , Nitric Oxide , Polycystic Ovary Syndrome , Sexual Behavior , Ventromedial Hypothalamic Nucleus , Animals , Anti-Mullerian Hormone/pharmacology , Disease Models, Animal , Female , Mating Preference, Animal , Mice , Neurons/drug effects , Neurons/enzymology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/physiopathology , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/metabolism
13.
Biochem Biophys Res Commun ; 616: 110-114, 2022 08 06.
Article En | MEDLINE | ID: mdl-35653825

Earlier it was shown that a group of extracellular low-specific metallopeptidases is present in the mammalian brain Kropotova and Mosevitsky (2016) [1]. These enzymes are weakly connected to the axonal ends of neurons. They were named Neuron bound Extracellular MetalloPeptidases (NEMP). The enzyme named NEMP3 turned out to be a unique exopeptidase that exhibits two activities: it removes the dipeptide from the N-end of the peptide, and it can also remove the tripeptide from the C-end of the peptide. Therefore, NEMP3 possesses the activities of dipeptidylaminopeptidase and of tripeptidylcarboxypeptidase. Mass spectrometry has revealed a homology of NEMP3 with DPP3 (DPP III, EC3.4.14.4), known as cytosolic dipeptidylaminopeptidase. We isolated DPP3 from rat and bovine liver and brain by the procedures used for this purpose by other authors. The effect of DPP3 on test peptides is the same as that of NEMP3. In particular, all DPP3 samples delete the tripeptide (AKF) from the C-end of the test peptide blocked at the N-end. The data obtained show that NEMP3 and DPP3 are the same protein (enzyme). Thus, DPP3 has two exopeptidase activities: the previously known activity of dipeptidylaminopeptidase and the activity of tripeptidylcarboxypeptidase discovered in this study. Another discovery is the extracellular activity of DPP 3 in the mammalian brain near synapses, which controls neuropeptides. DPP3 is involved in various processes, but in many cases its role remains to be clarified. The results obtained in this study will be useful for solving these questions.


Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Neuropeptides , Animals , Cattle , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Metalloproteases/metabolism , Neurons/enzymology , Neurons/metabolism , Neuropeptides/metabolism , Peptides/metabolism , Rats
14.
Proc Natl Acad Sci U S A ; 119(15): e2116844119, 2022 04 12.
Article En | MEDLINE | ID: mdl-35377811

In pathological or artificial conditions, memory can be formed as silenced engrams that are unavailable for retrieval by presenting conditioned stimuli but can be artificially switched into the latent state so that natural recall is allowed. However, it remains unclear whether such different states of engrams bear any physiological significance and can be switched through physiological mechanisms. Here, we show that an acute social reward experience switches the silent memory engram into the latent state. Conversely, an acute social stress causes transient forgetting via turning a latent memory engram into a silent state. Such emotion-driven bidirectional switching between latent and silent states of engrams is mediated through regulation of Rac1 activity­dependent reversible forgetting in the hippocampus, as stress-activated Rac1 suppresses retrieval, while reward recovers silenced memory under amnesia by inhibiting Rac1. Thus, data presented reveal hippocampal Rac1 activity as the basis for emotion-mediated switching between latent and silent engrams to achieve emotion-driven behavioral flexibility.


CA1 Region, Hippocampal , Mental Recall , Social Behavior , rac1 GTP-Binding Protein , Animals , CA1 Region, Hippocampal/enzymology , Cues , Mental Recall/physiology , Mice , Neurons/enzymology , Reward , rac1 GTP-Binding Protein/metabolism
15.
Nat Commun ; 13(1): 465, 2022 01 24.
Article En | MEDLINE | ID: mdl-35075108

Chromatin regulation is a key process in development but its contribution to the evolution of animals is largely unexplored. Chromatin is regulated by a diverse set of proteins, which themselves are tightly regulated in a cell/tissue-specific manner. Using the cnidarian Nematostella vectensis as a basal metazoan model, we explore the function of one such chromatin regulator, Lysine specific demethylase 1 (Lsd1). We generated an endogenously tagged allele and show that NvLsd1 expression is developmentally regulated and higher in differentiated neural cells than their progenitors. We further show, using a CRISPR/Cas9 generated mutant that loss of NvLsd1 leads to developmental abnormalities. This includes the almost complete loss of differentiated cnidocytes, cnidarian-specific neural cells, as a result of a cell-autonomous requirement for NvLsd1. Together this suggests that the integration of chromatin modifying proteins into developmental regulation predates the split of the cnidarian and bilaterian lineages and constitutes an ancient feature of animal development.


Cell Differentiation , Histone Demethylases/metabolism , Neurons/cytology , Neurons/enzymology , Sea Anemones/enzymology , Animals , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Histone Demethylases/genetics , Neurons/metabolism , Sea Anemones/embryology , Sea Anemones/metabolism
16.
Int J Biol Sci ; 18(2): 693-706, 2022.
Article En | MEDLINE | ID: mdl-35002518

The aggregation of amyloid-ß (Aß) peptides into oligomers and fibrils is a key pathological feature of Alzheimer's disease (AD). An increasing amount of evidence suggests that oligomeric Aß might be the major culprit responsible for various neuropathological changes in AD. Death-associated protein kinase 1 (DAPK1) is abnormally elevated in brains of AD patients and plays an important role in modulating tau homeostasis by regulating prolyl isomerase Pin1 phosphorylation. However, it remains elusive whether and how Aß species influence the function of DAPK1, and whether this may further affect the function and phosphorylation of tau in neurons. Herein, we demonstrated that Aß aggregates (both oligomers and fibrils) prepared from synthetic Aß42 peptides were able to upregulate DAPK1 protein levels and thereby its function through heat shock protein 90 (HSP90)-mediated protein stabilization. DAPK1 activation not only caused neuronal apoptosis, but also phosphorylated Pin1 at the Ser71 residue, leading to tau accumulation and phosphorylation at multiple AD-related sites in primary neurons. Both DAPK1 knockout (KO) and the application of a specific DAPK1 inhibitor could effectively protect primary neurons against Aß aggregate-induced cell death and tau dysregulation, corroborating the critical role of DAPK1 in mediating Aß aggregation-induced neuronal damage. Our study suggests a mechanistic link between Aß oligomerization and tau hyperphosphorylation mediated by DAPK1, and supports the role of DAPK1 as a promising target for early intervention in AD.


Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/enzymology , Death-Associated Protein Kinases/metabolism , Neurons/enzymology , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apoptosis/genetics , Brain/pathology , Death-Associated Protein Kinases/deficiency , Death-Associated Protein Kinases/genetics , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neurons/pathology , Peptide Fragments/genetics , Phosphorylation , tau Proteins/genetics , tau Proteins/metabolism
17.
Cell Mol Neurobiol ; 42(1): 305-309, 2022 Jan.
Article En | MEDLINE | ID: mdl-32623546

The recent outbreak of 2019 coronavirus disease (COVID-19), caused by a novel coronavirus, has now spread quickly worldwide. Like the severe acute respiratory syndrome coronavirus (SARS-CoV), this novel type of coronavirus, SARS-CoV-2, has been demonstrated to utilize angiotensin-converting enzyme 2 (ACE2) as an entry point to the cells. There is a growing body of reports indicating that COVID-19 patients, especially those in severe condition, exhibit neurological symptoms, thus supporting the possibility that SARS-CoV-2 could infect and damage neurons within the central nervous system in humans. Using human pluripotent stem cells-derived neurons, here we show the expression of ACE2 in human neurons via immunocytochemistry. From this perspective, we elaborate on the idea that the neuro-invasive potential of SARS-CoV-2 should be considered as a possible contributory factor, as well as a therapeutic target, for the severe respiratory symptoms in critical COVID-19 cases.


Angiotensin-Converting Enzyme 2/metabolism , COVID-19/enzymology , COVID-19/virology , Neurons/enzymology , Neurons/pathology , SARS-CoV-2/physiology , Humans , Models, Biological , Organ Specificity , Pluripotent Stem Cells/metabolism
18.
Protein Cell ; 13(1): 6-25, 2022 01.
Article En | MEDLINE | ID: mdl-33306168

The serine/threonine p21-activated kinases (PAKs), as main effectors of the Rho GTPases Cdc42 and Rac, represent a group of important molecular switches linking the complex cytoskeletal networks to broad neural activity. PAKs show wide expression in the brain, but they differ in specific cell types, brain regions, and developmental stages. PAKs play an essential and differential role in controlling neural cytoskeletal remodeling and are related to the development and fate of neurons as well as the structural and functional plasticity of dendritic spines. PAK-mediated actin signaling and interacting functional networks represent a common pathway frequently affected in multiple neurodevelopmental and neurodegenerative disorders. Considering specific small-molecule agonists and inhibitors for PAKs have been developed in cancer treatment, comprehensive knowledge about the role of PAKs in neural cytoskeletal remodeling will promote our understanding of the complex mechanisms underlying neurological diseases, which may also represent potential therapeutic targets of these diseases.


Cytoskeleton/enzymology , Nervous System Diseases/enzymology , Neurons/enzymology , Signal Transduction , p21-Activated Kinases/metabolism , Animals , Cytoskeleton/genetics , Humans , Nervous System Diseases/genetics , p21-Activated Kinases/genetics
19.
Endocrinology ; 163(2)2022 02 01.
Article En | MEDLINE | ID: mdl-34967909

Reproductive dysfunction in women has been linked to high caloric diet (HCD)-feeding and obesity. Central resistance to leptin and insulin have been shown to accompany diet-induced infertility in rodent studies, and we have previously shown that deleting suppressor of cytokine signaling 3, which is a negative regulator of leptin signaling, from all forebrain neurons partially protects mice from HCD-induced infertility. In this study, we were interested in exploring the role of protein tyrosine phosphatase 1B (PTP1B), which is a negative regulator of both leptin and insulin signaling, in the pathophysiology of HCD-induced obesity and infertility. To this end, we generated male and female neuron-specific PTP1B knockout mice and compared their body weight gain, food intake, glucose tolerance, and fertility relative to control littermates under both normal calorie diet and HCD feeding conditions. Both male and female mice with neuronal PTP1B deletion exhibited slower body weight gain in response to HCD feeding, yet only male knockout mice exhibited improved glucose tolerance compared with controls. Neuronal PTP1B deletion improved the time to first litter in HCD-fed mice but did not protect female mice from eventual HCD-induced infertility. While the mice fed a normal caloric diet remained fertile throughout the 150-day period of assessment, HCD-fed females became infertile after producing only a single litter, regardless of their genotype. These data show that neuronal PTP1B deletion is able to partially protect mice from HCD-induced obesity but is not a critical mediator of HCD-induced infertility.


Brain/enzymology , Infertility, Female/prevention & control , Neurons/enzymology , Obesity/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Animals , Crosses, Genetic , Energy Intake , Female , Infertility, Female/etiology , Male , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Obesity/enzymology , Obesity/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Sexual Maturation
20.
J Endocrinol ; 252(3): 167-177, 2022 01 13.
Article En | MEDLINE | ID: mdl-34854381

Recent evidence identifies a potent role for aerobic exercise to modulate the activity of hypothalamic neurons related to appetite; however, these studies have been primarily performed in male rodents. Since females have markedly different neuronal mechanisms regulating food intake, the current study aimed to determine the effects of acute treadmill exercise on hypothalamic neuron populations involved in regulating appetite in female mice. Mature, untrained female mice were exposed to acute sedentary, low- (10 m/min), moderate- (14 m/min), and high (18 m/min)-intensity treadmill exercise in a randomized crossover design. Mice were fasted 10 h before exercise, and food intake was monitored for 48 h after bouts. Immunohistochemical detection of cFOS was performed 3 h post-exercise to determine the changes in hypothalamic neuropeptide Y (NPY)/agouti-related peptide (AgRP), pro-opiomelanocortin (POMC), tyrosine hydroxylase (TH), and SIM1-expressing neuron activity concurrent with the changes in food intake. Additionally, stains for pSTAT3tyr705 and pERKthr202/tyr204 were performed to detect exercise-mediated changes in intracellular signaling. Briefly, moderate- and high-intensity exercises increased 24-h food intake by 5.9 and 19%, respectively, while low-intensity exercise had no effects. Furthermore, increases in NPY/AgRPARC, SIM1PVN, and TH neuron activity were observed 3 h after high-intensity exercise, with no effects on POMCARC neurons. While no effects of exercise on pERKthr202/tyr204 were observed, pSTAT3tyr705 was elevated specifically in NPY/AgRP neurons 3 h post-exercise. Overall, aerobic exercise increased the activity of several appetite-stimulating neuron populations in the hypothalamus of female mice, which may provide insight into previously reported sexual dimorphisms in post-exercise feeding.


Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Neuropeptide Y/metabolism , Physical Conditioning, Animal/physiology , Tyrosine 3-Monooxygenase/metabolism , Animals , Female , Mice , Neurons/enzymology
...