Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Neurodegener ; 13(1): 24, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671492

ABSTRACT

BACKGROUND: Adult neurogenesis occurs in the subventricular zone (SVZ) and the subgranular zone of the dentate gyrus in the hippocampus. The neuronal stem cells in these two neurogenic niches respond differently to various physiological and pathological stimuli. Recently, we have found that the decrement of carboxypeptidase E (CPE) with aging impairs the maturation of brain-derived neurotrophic factor (BDNF) and neurogenesis in the SVZ. However, it remains unknown whether these events occur in the hippocampus, and what the role of CPE is in the adult hippocampal neurogenesis in the context of Alzheimer's disease (AD). METHODS: In vivo screening was performed to search for miRNA mimics capable of upregulating CPE expression and promoting neurogenesis in both neurogenic niches. Among these, two agomirs were further assessed for their effects on hippocampal neurogenesis in the context of AD. We also explored whether these two agomirs could ameliorate behavioral symptoms and AD pathology in mice, using direct intracerebroventricular injection or by non-invasive intranasal instillation. RESULTS: Restoration of CPE expression in the hippocampus improved BDNF maturation and boosted adult hippocampal neurogenesis. By screening the miRNA mimics targeting the 5'UTR region of Cpe gene, we developed two agomirs that were capable of upregulating CPE expression. The two agomirs significantly rescued adult neurogenesis and cognition, showing multiple beneficial effects against the AD-associated pathologies in APP/PS1 mice. Of note, noninvasive approach via intranasal delivery of these agomirs improved the behavioral and neurocognitive functions of APP/PS1 mice. CONCLUSIONS: CPE may regulate adult hippocampal neurogenesis via the CPE-BDNF-TrkB signaling pathway. This study supports the prospect of developing miRNA agomirs targeting CPE as biopharmaceuticals to counteract aging- and disease-related neurological decline in human brains.


Subject(s)
Alzheimer Disease , Carboxypeptidase H , Hippocampus , Memory Disorders , Neurogenesis , Up-Regulation , Animals , Neurogenesis/drug effects , Neurogenesis/physiology , Alzheimer Disease/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Carboxypeptidase H/genetics , Carboxypeptidase H/biosynthesis , Mice , Memory Disorders/genetics , Memory Disorders/etiology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , MicroRNAs/genetics , MicroRNAs/biosynthesis , Male , Mice, Transgenic , Mice, Inbred C57BL , Disease Models, Animal
2.
Biochem Cell Biol ; 97(4): 446-453, 2019 08.
Article in English | MEDLINE | ID: mdl-30508384

ABSTRACT

Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents, and metastatic OS is the major cause of OS-related death. Carboxypeptidase E (CPE) is known to be highly expressed in some cancer types, and its N-terminal truncated form, CPE-ΔN, is implicated in tumor metastasis and poor prognosis. In this study, we investigated the effect of CPE-ΔN on cell migration, invasiveness, and the epithelial-mesenchymal transition (EMT) of OS cells, and illustrated the molecular mechanisms. We first constructed CPE-ΔN overexpressing human OS cell lines (143B and U2OS cells), and found that ectopic CPE-ΔN expression in OS cells enhanced cell migration and invasiveness, and promoted the EMT process. Further, overexpression of CPE-ΔN increased the levels of c-myc and nuclear ß-catenin in OS cells, which suggested the CPE-ΔN promotes activation of the Wnt-ß-catenin pathway in OS cells. Treatment with ß-catenin small interfering RNA (siRNA) inhibited the migration and invasiveness of CPE-ΔN-overexpressing cells, and reduced the expression of E-cadherin. Together, these results suggest that CPE-ΔN promotes migration, invasiveness, and the EMT of OS cells via the Wnt-ß-catenin signaling pathway.


Subject(s)
Carboxypeptidase H/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Neoplasm Invasiveness , Osteosarcoma/metabolism , Osteosarcoma/pathology , Wnt Signaling Pathway , Carboxypeptidase H/biosynthesis , Humans , Osteosarcoma/enzymology , Tumor Cells, Cultured
3.
Endocrinology ; 154(9): 3284-93, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23825125

ABSTRACT

Prolonged chronic stress causing elevated plasma glucocorticoids leads to neurodegeneration. Adaptation to stress (allostasis) through neuroprotective mechanisms can delay this process. Studies on hippocampal neurons have identified carboxypeptidase E (CPE) as a novel neuroprotective protein that acts extracellularly, independent of its enzymatic activity, although the mechanism of action is unclear. Here, we aim to determine if CPE plays a neuroprotective role in allostasis in mouse hippocampus during chronic restraint stress (CRS), and the molecular mechanisms involved. Quantitative RT-PCR/in situ hybridization and Western blots were used to assay for mRNA and protein. After mild CRS (1 h/d for 7 d), CPE protein and mRNA were significantly elevated in the hippocampal CA3 region, compared to naïve littermates. In addition, luciferase reporter assays identified a functional glucocorticoid regulatory element within the cpe promoter that mediated the up-regulation of CPE expression in primary hippocampal neurons following dexamethasone treatment, suggesting that circulating plasma glucocorticoids could evoke a similar effect on CPE in the hippocampus in vivo. Overexpression of CPE in hippocampal neurons, or CRS in mice, resulted in elevated prosurvival BCL2 protein/mRNA and p-AKT levels in the hippocampus; however, CPE(-/-) mice showed a decrease. Thus, during mild CRS, CPE expression is up-regulated, possibly contributed by glucocorticoids, to mediate neuroprotection of the hippocampus by enhancing BCL2 expression through AKT signaling, and thereby maintaining allostasis.


Subject(s)
Allostasis , Carboxypeptidase H/biosynthesis , Hippocampus/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Stress, Psychological/metabolism , Up-Regulation , Animals , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Carboxypeptidase H/genetics , Carboxypeptidase H/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Genes, Reporter/drug effects , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Restraint, Physical , Severity of Illness Index , Stress, Physiological , Stress, Psychological/physiopathology , Up-Regulation/drug effects
4.
Gen Comp Endocrinol ; 150(2): 233-45, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17070810

ABSTRACT

Identification of orthologs of vertebrate neuropeptides and hypothalamic hormones in the neural complex of ascidians suggests integral roles of the ascidian neural complex in the endocrine system. In the present study, we investigated endocrine-related genes expressed in the neural complex of Ciona intestinalis. Comprehensive analyses of 3'-end sequences of the neural complex cDNAs placed 10,029 clones into 4051 independent clusters or genes, 1524 of them being expressed preferentially in this organ. Comparison of the 1524 genes with the human proteome databank demonstrated that 476 matched previously identified human proteins with distinct functions. Further analyses of sequence similarity of the 476 genes demonstrated that 21 genes are candidates for those involved in the endocrine system. Although we cannot detect hormone or peptide candidates, we found 21 genes such as receptors for peptide ligands, receptor-modulating proteins, and processing enzymes. We then characterized the Ciona prohormone convertase 2 (Ci-PC2) and carboxypeptidase E (Ci-CPE), which are associated with endoproteolytic processing of peptide hormone precursors. Furthermore, genes encoding these transcripts are expressed specifically in the neural complex of young adult ascidians. These data provide the molecular basis for further functional studies of the endocrine role of the neural complex of ascidians.


Subject(s)
Central Nervous System/physiology , Ciona intestinalis/genetics , Endocrine System/physiology , Expressed Sequence Tags , Amino Acid Sequence , Animals , Base Sequence , Carboxypeptidase H/biosynthesis , Carboxypeptidase H/genetics , Ciona intestinalis/enzymology , Ciona intestinalis/metabolism , Cluster Analysis , Gene Expression , Gene Library , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Proprotein Convertase 2/biosynthesis , Proprotein Convertase 2/genetics , Sequence Alignment
5.
J Cereb Blood Flow Metab ; 24(6): 612-22, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15181368

ABSTRACT

In this study, using both in vivo and in vitro ischemia models, the authors investigated the impact of brain ischemia on the biosynthesis of a key neuropeptide-processing enzyme, carboxypeptidase E (CPE). The response to brain ischemia of animals that lacked an active CPE was also examined. Combined in situ hybridization and immunocytochemical analyses for CPE showed reciprocal changes of CPE mRNA and protein, respectively, in the same cortical cells in rat brains after focal cerebral ischemia. Western blot analysis revealed an accumulation of the precursor protein of CPE in the ischemic cortex in vivo and in ischemic cortical neurons in vitro. Detailed metabolic labeling experiments on ischemic cortical neurons showed that ischemic stress caused a blockade in the proteolytic processing of CPE. When mice lacking an active CPE protease were subjected to a sublethal episode of focal cerebral ischemia, abundant TUNEL-positive cells were seen in the ischemic cortex whereas only a few were seen in the cortex of wild-type animals. These findings suggest that ischemia has an adverse impact on the neuropeptide-processing system in the brain and that the lack of an active neuropeptide-processing enzyme exacerbates ischemic brain injury.


Subject(s)
Brain Ischemia/metabolism , Carboxypeptidase H/biosynthesis , Neuropeptides/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Carboxypeptidase H/genetics , Cells, Cultured , In Situ Hybridization , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...