Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Stem Cells Int ; 2020: 8878412, 2020.
Article in English | MEDLINE | ID: mdl-32733573

ABSTRACT

The formation of neurofibrillary tangles has been implicated as an important pathological marker for Alzheimer's disease (AD). Studies have revealed that the inhibition of abnormal hyperphosphorylation and aggregation of tau in the AD brain might serve as an important drug target. Using in vitro and in vivo experimental models, such as the AD mouse model (5xFAD mice), we investigated the inhibition of hyperphosphorylation of tau using the human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs). Administration of hUCB-MSCs not only ameliorated the spatial learning and memory impairments but also mitigated the hyperphosphorylation of tau in 5xFAD mice. Furthermore, in vivo experiments in mice and in vitro ThT fluorescence assay validated galectin-3 (GAL-3) as an essential factor of hUCB-MSC. Moreover, GAL-3 was observed to be involved in the removal of aberrant forms of tau, by reducing hyperphosphorylation through decrements in the glycogen synthase kinase 3 beta (GSK-3ß). Our results confirm that GAL-3, secreted by hUCB-MSC, regulates the abnormal accumulation of tau by protein-protein interactions. This study suggests that hUCB-MSCs mitigate hyperphosphorylation of tau through GAL-3 secretion. These findings highlight the potential role of hUCB-MSCs as a therapeutic agent for aberrant tau in AD.

2.
Biochem Biophys Res Commun ; 504(4): 933-940, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30224067

ABSTRACT

Alzheimer's disease (AD), which is the most common progressive neurodegenerative disease, causes learning and memory impairment. The pathological progress of AD can derive from imbalanced homeostasis of amyloid beta (Aß) in the brain. In such cases, microglia play important roles in regulating the brain Aß levels. In the present study, we found that human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) can increase, through paracrine action, the ability of microglial cells to clear Aß. In order to identify the associated paracrine factors, a secretome of hUCB-MSCs co-cultured with Aß-treated BV2 microglial cells was analyzed using a human cytokine protein array. As a result, growth differentiation factor-15 (GDF-15) was identified as a predominant candidate, and its association with Aß clearance by microglial cells was investigated in vitro and in a 5XFAD mouse model. When Aß-treated BV2 cells were treated with exogenous recombinant GDF-15, the Aß levels in the culture medium decreased. Moreover, GDF-15 injection in the brain parenchyma of 5XFAD mice also led to decrease in Aß plaques. In contrast, co-culture of BV2 cells and hUCB-MSCs treated with GDF-15-specific siRNA did not influence the Aß levels in the culture medium. To elucidate how these phenomena are related, we confirmed that GDF-15 specifically increases insulin-degrading enzyme (IDE) expression in microglial cells through TGFß receptor type II (TGFßRII), both in vitro and in vivo. These findings suggest that hUCB-MSCs promote the Aß clearance ability of microglial cells through regulation of GDF-15 secretion, thus elucidating a therapeutic mechanism for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Growth Differentiation Factor 15/metabolism , Mesenchymal Stem Cells/metabolism , Alzheimer Disease/pathology , Animals , Coculture Techniques , Disease Models, Animal , Fetal Blood/cytology , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/pharmacology , Humans , Insulysin/metabolism , Mesenchymal Stem Cells/cytology , Mice, Mutant Strains , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Paracrine Communication , Peptide Fragments/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
3.
Sci Rep ; 8(1): 354, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321508

ABSTRACT

Alzheimer's disease (AD) is an incurable neurodegenerative disease characterised clinically by learning and memory impairments. Amyloid beta (Aß) peptide-induced synaptic dysfunction is a pathological process associated with early-stage AD. Here, we show that paracrine action of human umbilical cord blood-derived-mesenchymal stem cells (hUCB-MSCs) protects the hippocampus from synaptic-density loss in in vitro and in vivo AD models. To identify paracrine factors underlying this rescue effect, we analysed hUCB-MSCs' secretome co-cultured with Aß42-treated mouse hippocampal neurons. Thrombospondin-1 (TSP-1), a protein secreted by hUCB-MSCs in in vitro and 5XFAD AD mouse models, was selected for study. Treatment with exogenous recombinant TSP-1 or co-cultures with hUCB-MSCs significantly increased expression of synaptic-density markers, such as synaptophysin (SYP) and post-synaptic density protein-95 (PSD-95) in Aß42-treated mouse hippocampal neurons. Knockdown of TSP-1 expression in hUCB-MSCs through small interfering RNA (siRNA) abolished the reversal of Aß42-induced hippocampal synaptic-density loss. We demonstrate that the rescue effect of hUCB-MSC-secreted TSP-1 was mediated by neuroligin-1 (NLGN1) or α2δ-1 receptors. Interestingly, NLGN1 and α2δ-1 expression, which was reduced in Aß42-treated hippocampal neurons, increased in co-cultures with hUCB-MSCs or exogenous TSP-1. Together, these findings suggest that hUCB-MSCs can attenuate Aß42-induced synaptic dysfunction by regulating TSP-1 release, thus providing a potential alternative therapeutic option for early-stage AD.


Subject(s)
Alzheimer Disease/metabolism , Fetal Blood/cytology , Mesenchymal Stem Cells/metabolism , Neurons/metabolism , Synapses/metabolism , Thrombospondin 1/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mice , Neurons/drug effects , Paracrine Communication , Pyramidal Cells/metabolism , Thrombospondin 1/cerebrospinal fluid , Thrombospondin 1/pharmacology
4.
Stem Cells Dev ; 24(20): 2378-90, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26154268

ABSTRACT

Our previous studies demonstrated that transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) into the hippocampus of a transgenic mouse model of Alzheimer's disease (AD) reduced amyloid-ß (Aß) plaques and enhanced cognitive function through paracrine action. Due to the limited life span of hUCB-MSCs after their transplantation, the extension of hUCB-MSC efficacy was essential for AD treatment. In this study, we show that repeated cisterna magna injections of hUCB-MSCs activated endogenous hippocampal neurogenesis and significantly reduced Aß42 levels. To identify the paracrine factors released from the hUCB-MSCs that stimulated endogenous hippocampal neurogenesis in the dentate gyrus, we cocultured adult mouse neural stem cells (NSCs) with hUCB-MSCs and analyzed the cocultured media with cytokine arrays. Growth differentiation factor-15 (GDF-15) levels were significantly increased in the media. GDF-15 suppression in hUCB-MSCs with GDF-15 small interfering RNA reduced the proliferation of NSCs in cocultures. Conversely, recombinant GDF-15 treatment in both in vitro and in vivo enhanced hippocampal NSC proliferation and neuronal differentiation. Repeated administration of hUBC-MSCs markedly promoted the expression of synaptic vesicle markers, including synaptophysin, which are downregulated in patients with AD. In addition, in vitro synaptic activity through GDF-15 was promoted. Taken together, these results indicated that repeated cisterna magna administration of hUCB-MSCs enhanced endogenous adult hippocampal neurogenesis and synaptic activity through a paracrine factor of GDF-15, suggesting a possible role of hUCB-MSCs in future treatment strategies for AD.


Subject(s)
Alzheimer Disease/metabolism , Cerebrospinal Fluid/metabolism , Chromosome Pairing/physiology , Growth Differentiation Factor 15/metabolism , Hippocampus/metabolism , Mesenchymal Stem Cells/cytology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cells, Cultured , Disease Models, Animal , Fetal Blood , Hippocampus/cytology , Humans , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Transgenic , Neurogenesis/genetics , Neurogenesis/physiology
5.
Neurotox Res ; 15(4): 332-47, 2009 May.
Article in English | MEDLINE | ID: mdl-19384567

ABSTRACT

Ghrelin is an endogenous ligand for growth hormone (GH) secretagogue receptor 1a (GHS-R1a) and is produced and released mainly from the stomach. It was recently demonstrated that ghrelin can function as a neuroprotective factor by inhibiting apoptotic pathways. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) causes nigrostriatal dopaminergic neurotoxicity in rodents; previous studies suggest that activated microglia actively participate in the pathogenesis of Parkinson's disease (PD) neurodegeneration. However, the role of microglia in the neuroprotective properties of ghrelin is still unknown. Here we show that, in the mouse MPTP PD model generated by an acute regimen of MPTP administration, systemic administration of ghrelin significantly attenuates the loss of substantia nigra pars compacta (SNpc) neurons and the striatal dopaminergic fibers through the activation of GHS-R1a. We also found that ghrelin reduced nitrotyrosine levels and improved the impairment of rota-rod performance. Ghrelin prevents MPTP-induced microglial activation in the SNpc and striatum, the expression of pro-inflammatory molecules tumor necrosis factor alpha (TNF-alpha) and interleukin-1beta (IL-1beta), and the activation of inducible nitric oxide synthase. The inhibitory effect of ghrelin on the activation of microglia appears to be indirect by suppressing matrix metalloproteinase-3 (MMP-3) expression in stressed dopaminergic neurons because GHS-R1a is not expressed in SNpc microglial cells. Finally, in vitro administration of ghrelin prevented 1-methyl-4-phenylpyridinium-induced dopaminergic cell loss, MMP-3 expression, microglial activation, and the subsequent release of TNF-alpha, IL-1beta, and nitrite in mesencephalic cultures. Our data indicate that ghrelin may act as a survival factor for dopaminergic neurons by functioning as a microglia-deactivating factor and suggest that ghrelin may be a valuable therapeutic agent for neurodegenerative diseases such as PD.


Subject(s)
Ghrelin/pharmacology , MPTP Poisoning/physiopathology , Microglia/drug effects , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gliosis/chemically induced , Gliosis/prevention & control , Interleukin-1beta/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Matrix Metalloproteinase 3/metabolism , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurons/drug effects , Nitrites/metabolism , Rats , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Tandem Mass Spectrometry/methods , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
6.
Endocr J ; 55(5): 867-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18506089

ABSTRACT

Intracerebroventricular (icv) administration of glucagon-like peptide-1 (GLP-1) inhibits food intake and induces c-fos expression in the hypothalamus. However, the effects of GLP-1 on hypothalamic neuronal activity or neuropeptide mRNA expression are unknown. In this study, we examined the effects of GLP-1 on fasting-induced changes in the expression of hypothalamic orexigenic and anorexigenic neuropeptide. Food intake was significantly inhibited after icv injection of GLP-1 in 48 h fasted rats. Hypothalamic neuropeptide Y (NPY) and agouti-related peptide (AgRP) mRNAs were significantly increased by fasting, whereas icv GLP-1 treatment significantly attenuated these fasting-induced increases. Both proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) mRNA levels were decreased by fasting, while GLP-1 treatment attenuated fasting-induced decreases in POMC and CART expression. We also determined the mRNA levels of AMP-activated kinase (AMPK) and found that fasting resulted in a significant stimulation of hypothalamic AMPKalpha2 mRNA. Fasting-induced increase in AMPKalpha2 mRNA was almost completely prevented by GLP-1 treatment. Analysis of phosphorylated AMPKalpha and acetyl CoA carboxylase showed similar results. Taken together, our observation suggests that the decreased food intake by GLP-1 is caused by preventing the fasting-induced increase in hypothalamic NPY and AgRP and the fasting-induced decrease in hypothalamic POMC and CART. Our results also suggest that the food intake lowering effect of GLP-1 is caused by reversing the fasting-induced increase in hypothalamic AMPK activity. Therefore we conclude that the anorectic effect of GLP-1 seems to be mediated by, at least in part, by the hypothalamus.


Subject(s)
AMP-Activated Protein Kinases/genetics , Gene Expression/drug effects , Glucagon-Like Peptide 1/administration & dosage , Hypothalamus/drug effects , Neuropeptides/genetics , Acetyl-CoA Carboxylase/genetics , Animals , Eating/drug effects , Fasting , Hypothalamus/chemistry , Hypothalamus/metabolism , Injections, Intraventricular , Male , Proto-Oncogene Proteins c-fos/analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
7.
Endocrinology ; 148(1): 148-59, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17053024

ABSTRACT

Ghrelin is an endogenous ligand for the GH secretagogue receptor, produced and secreted mainly from the stomach. Ghrelin stimulates GH release and induces positive energy balances. Previous studies have reported that ghrelin inhibits apoptosis in several cell types, but its antiapoptotic effect in neuronal cells is unknown. Therefore, we investigated the role of ghrelin in ischemic neuronal injury using primary hypothalamic neurons exposed to oxygen-glucose deprivation (OGD). Here we report that treatment of hypothalamic neurons with ghrelin inhibited OGD-induced cell death and apoptosis. Exposure of neurons to ghrelin caused rapid activation of ERK1/2. Ghrelin-induced activation of ERK1/2 and the antiapoptotic effect of ghrelin were blocked by chemical inhibition of MAPK, phosphatidylinositol 3 kinase, protein kinase C, and protein kinase A. Ghrelin attenuated OGD-induced activation of c-Jun NH2-terminal kinase and p-38 but not ERK1/2. We also investigated ghrelin regulation of apoptosis at the mitochondrial level. Ghrelin protected cells from OGD insult by inhibiting reactive oxygen species generation and stabilizing mitochondrial transmembrane potential. In addition, ghrelin-treated cells showed an increased Bcl-2/Bax ratio, prevention of cytochrome c release, and inhibition of caspase-3 activation. Finally, in vivo administration of ghrelin significantly reduced infarct volume in an animal model of ischemia. Our data indicate that ghrelin may act as a survival factor that preserves mitochondrial integrity and inhibits apoptotic pathways.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/drug therapy , Hypothalamus/pathology , Neurons/drug effects , Peptide Hormones/pharmacology , Animals , Apoptosis/physiology , Brain Ischemia/pathology , Caspase 3/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Ghrelin , Glucose/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/pathology , Oxygen/pharmacology , Peptide Hormones/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL