Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Neurochem Res ; 42(8): 2191-2207, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397068

ABSTRACT

Transplantation of human amniotic mesenchymal stem cells (hAM-MSCs) seems to be a promising strategy for the treatment of neurodegenerative disorders, including Alzheimer's disease (AD). However, the clinical therapeutic effects of hAM-MSCs and their mechanisms of action in AD remain to be determined. Here, we used amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic mice to evaluate the effects of hAM-MSC transplantation on AD-related neuropathology and cognitive dysfunction. We found that hAM-MSC transplantation into the hippocampus dramatically reduced amyloid-ß peptide (Aß) deposition and rescued spatial learning and memory deficits in APP/PS1 mice. Interestingly, these effects were associated with increasing in Aß-degrading factors, elevations in activated microglia, and the modulation of neuroinflammation. Furthermore, enhanced hippocampal neurogenesis in the subgranular zone (SGZ) of the dentate gyrus (DG) and enhanced synaptic plasticity following hAM-MSC treatment could be another important factor in reversing the cognitive decline in APP/PS1 mice. Instead, the mechanism underlying the improved cognition apparently involves a robust increase in hippocampal synaptic density and neurogenesis that is mediated by brain-derived neurotrophic factor (BDNF). In conclusion, our data suggest that hAM-MSCs may be a new and effective therapy for the treatment of AD.


Subject(s)
Amniotic Fluid/physiology , Amyloid beta-Peptides/metabolism , Memory Disorders/metabolism , Memory Disorders/therapy , Memory/physiology , Mesenchymal Stem Cell Transplantation/trends , Amniotic Fluid/cytology , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Male , Maze Learning/physiology , Memory Disorders/genetics , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics
2.
Neurochem Res ; 41(10): 2708-2718, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27351200

ABSTRACT

Human amniotic membrane mesenchymal stem cells (hAMSCs) are considered ideal candidate stem cells for cell-based therapy. In this study, we assessed whether hAMSCs transplantation promotes neurological functional recovery in rats after traumatic spinal cord injury (SCI). In addition, the potential mechanisms underlying the possible benefits of this therapy were investigated. Female Sprague-Dawley rats were subjected to SCI using a weight drop device and then hAMSCs, or phosphate-buffered saline (PBS) were immediately injected into the contused dorsal spinal cord at 2 mm rostral and 2 mm caudal to the injury site. Our results indicated that transplanted hAMSCs migrated in the host spinal cord without differentiating into neuronal or glial cells. Compared with the control group, hAMSCs transplantation significantly decreased the numbers of ED1+ macrophages/microglia and caspase-3+ cells. In addition, hAMSCs transplantation significantly increased the levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) in the injured spinal cord, and promoted both angiogenesis and axonal regeneration. These effects were associated with significantly improved neurobehavioral recovery in the hAMSCs transplantation group. These results show that transplantation of hAMSCs provides neuroprotective effects in rats after SCI, and could be candidate stem cells for the treatment of SCI.


Subject(s)
Cell Movement/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Microglia/metabolism , Neuroglia/metabolism , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/physiopathology , Vascular Endothelial Growth Factor A/metabolism
3.
Cell Mol Neurobiol ; 33(4): 465-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23478940

ABSTRACT

Human mesenchymal stem cells (MSCs) are considered a promising tool for cell-based therapies of nervous system diseases. Bone marrow (BM) has been the traditional source of MSCs (BM-MSCs). However, there are some limitations for their clinical use, such as the decline in cell number and differentiation potential with age. Recently, amniotic fluid (AF)-derived MSCs (AF-MSCs) have been shown to express embryonic and adult stem cell markers, and can differentiate into cells of all three germ layers. In this study, we isolated AF-MSCs from second-trimester AF by limiting dilution and compared their proliferative capacity, multipotency, neural differentiation ability, and secretion of neurotrophins to those of BM-MSCs. AF-MSCs showed a higher proliferative capacity and more rapidly formed and expanded neurospheres compared to those of BM-MSCs. Both immunocytochemical and quantitative real-time PCR analyses demonstrated that AF-MSCs showed higher expression of neural stemness markers than those of BM-MSCs following neural stem cell (NSC) differentiation. Furthermore, the levels of brain-derived growth factor and nerve growth factor secreted by AF-MSCs in the culture medium were higher than those of BM-MSCs. In addition, AF-MSCs maintained a normal karyotype in long-term cultures after NSC differentiation and were not tumorigenic in vivo. Our findings suggest that AF-MSCs are a promising and safe alternative to BM-MSCs for therapy of nervous system diseases.


Subject(s)
Amniotic Fluid/cytology , Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Neurogenesis , Neurons/cytology , Adult , Animals , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Proliferation , Cell Separation , Cell Shape , Cell Transformation, Neoplastic/pathology , Chromosomal Instability , Chromosomes, Mammalian/metabolism , Humans , Immunophenotyping , Karyotyping , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nerve Growth Factors/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Young Adult
4.
Neurochem Res ; 38(5): 1022-33, 2013 May.
Article in English | MEDLINE | ID: mdl-23475428

ABSTRACT

Although human amnion derived mesenchymal stem cells (AMSC) are a promising source of stem cells, their therapeutic potential for traumatic brain injury (TBI) has not been widely investigated. In this study, we evaluated the therapeutic potential of AMSC using a rat TBI model. AMSC were isolated from human amniotic membrane and characterized by flow cytometry. After induction, AMSC differentiated in vitro into neural stem-like cells (AM-NSC) that expressed higher levels of the neural stem cell markers, nestin, sox2 and musashi, in comparison to undifferentiated AMSC. Interestingly, the neurotrophic factors, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin 3 (NT-3), glial cell derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) were markedly upregulated after neural stem cell induction. Following transplantation in a rat TBI model, significant improvements in neurological function, brain tissue morphology, and higher levels of BDNF, NGF, NT-3, GDNF and CNTF, were observed in the AM-NSC group compared with the AMSC and Matrigel groups. However, few grafted cells survived with minimal differentiation into neural-like cells. Together, our results suggest that transplantation of AM-NSC promotes functional rehabilitation of rats with TBI, with enhanced expression of neurotrophic factors a likely mechanistic pathway.


Subject(s)
Amnion/cytology , Brain Injuries/therapy , Neural Stem Cells/cytology , Animals , Base Sequence , Cell Differentiation , DNA Primers , Female , Humans , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
5.
Neurochem Res ; 35(10): 1522-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20658188

ABSTRACT

Mesenchymal stem cells are capable of differentiating into dopaminergic-like cells, but currently no report has been available to describe the induction of human umbilical vein mesenchymal stem cells (HUVMSCs) into dopaminergic-like cells. In this study, we induced HUVMSCs in vitro into neurospheres constituted by neural stem-like cells, and further into cells bearing strong morphological, phenotypic and functional resemblances with dopaminergic-like cells. These HUVMSC-derived dopaminergic-like cells, after grafting into the brain of a rat model of Parkinson's disease (PD), showed a partial therapeutic effect in terms of the behavioral improvement. Nerve growth factor was reported to improve the local microenvironment of the grafted cells, and we therefore further tested the effect of dopaminergic-like cell grafting combined with nerve growth factor (NGF) administration at the site of cell transplantation. The results showed that NGF administration significantly promoted the survival of the grafted cells in the host brain and enhanced the content of dopaminergic in the local brain tissue. Behavioral test demonstrated a significant improvement of the motor function of the PD rats after dopaminergic-like cell grafting with NGF administration as compared with that of rats receiving the cell grafting only. These results suggest that transplantation of the dopaminergic-like cells combined with NGF administration may represent a new strategy of stem cell therapy for PD.


Subject(s)
Dopamine/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Motor Activity , Nerve Growth Factor/therapeutic use , Parkinson Disease/therapy , Umbilical Veins/cytology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Differentiation , Humans , Mesenchymal Stem Cells/cytology , Parkinson Disease/metabolism , Parkinson Disease/psychology , Rats , Rats, Sprague-Dawley
6.
Cell Mol Neurobiol ; 29(8): 1283-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19533335

ABSTRACT

Controversies exist concerning the need for mesenchymal stromal cells (MSCs) to be transdifferentiated prior to their transplantation. In the present study, we compared the results of grafting into the rat contused spinal cord undifferentiated, adipose tissue-derived stromal cells (uADSCs) versus ADSCs induced by two different protocols to form differentiated nervous tissue. Using Basso, Beattie, and Bresnahan scores and grid tests, we found that three cell-treated groups, including uADSCs-treated, dADSCs induced by Protocol 1 (dADSC-P1)-treated, and dADSCs induced by Protocol 2 (dADSC-P2)-treated groups, significantly improved locomotor functional recovery in SCI rats, compared with the saline-treated group. Furthermore, functional recovery was better in the uADSC-treated and dADSC-P2-treated groups than in the dADSC-P1-treated group at week 12 postinjury (P < 0.05 for dADSC-P1 group vs. uADSCs or dADSC-P2 groups). Although both protocols could induce high percentages of cells expressing neural markers in vitro, few BrdU-labeled cells survived at the injury sites in the three cell-treated groups, and only a small percentage of BrdU-positive cells expressed neural markers. On the other hand, the number of NF200-positive axons in the uADSC-treated and dADSC-P2-treated groups was significantly larger than those in the dADSC-P1-treated and saline-treated control groups. Our results indicate that ADSCs are able to differentiate into neural-like cells in vitro and in vivo. However, neural differentiated ADSCs did not result in better functional recovery than undifferentiated ones, following SCI. In vitro neural transdifferentiation of ADSCs might therefore not be a necessary pretransplantation step. Furthermore, cellular replacement or integration might not contribute to the functional recovery of the injured spinal cord.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Stromal Cells/cytology , Stromal Cells/transplantation , Animals , Axons/physiology , Behavior, Animal , Biomarkers/metabolism , Cell Shape , Cells, Cultured , Intermediate Filament Proteins/metabolism , Locomotion , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Neurons/metabolism , Rats , Regeneration/physiology , Tubulin/metabolism
7.
Cancer Biol Ther ; 11(11): 974-80, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21502808

ABSTRACT

Cancer stem cells are defined as a subpopulation of cancer cells with the capacity to self-renew and differentiate, which may play critical roles in tumor initiation, progress and resistance to current treatments. It has been reported that Dendritic cells (DCs) transfected with total tumor RNA could induce strong antitumor T-cell responses both in vivo and in vitro. In the study, we investigated the characteristics of 9L tumor spheres, and evaluated the antitumor effects of DCs transfected with 9L tumor spheres RNA in vivo. The results showed that 9L tumor spheres have the properties of cancer stem cells, and the majority of 9L cells were positive for CD133 and nestin. DCs transfected with 9L tumor spheres RNA can significantly inhibit glioma growth and prolong the survival of 9L glioma-bearing rats. These results demonstrated that 9L cancer stem like cells were enriched in tumor spheres, and they were a part of CD133+ cells, DCs transfected with cancer stem cells RNA may be an effective therapy for glioma.


Subject(s)
Brain Neoplasms/pathology , Cancer Vaccines/genetics , Dendritic Cells/transplantation , Gliosarcoma/pathology , Neoplastic Stem Cells/pathology , RNA/immunology , AC133 Antigen , Animals , Antigens, CD/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Cell Culture Techniques , Cell Line, Tumor , Culture Media , Dendritic Cells/metabolism , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gliosarcoma/genetics , Gliosarcoma/therapy , Glycoproteins/metabolism , Interferon-gamma/blood , Intermediate Filament Proteins/metabolism , Kaplan-Meier Estimate , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Nerve Tissue Proteins/metabolism , Nestin , Peptides/metabolism , Rats , Transfection , Xenograft Model Antitumor Assays
8.
Cancer Lett ; 312(2): 168-77, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-21924825

ABSTRACT

Immunotoxins have shown great promise as an alternative treatment for brain malignancies such as gliomas, but their failure to penetrate into the tumor mass remains a major problem. Mesenchymal stem cells exhibit tropism to tumor tissue and may serve as a cellular vehicle for the delivery and local production of antitumor agents. In this study, we used human bone marrow-derived mesenchymal stem cells (hMSCs) as a vehicle for the targeted delivery of EphrinA1-PE38, a very specific immunotoxin against the EphA2 receptor that is overexpressed in gliomas. hMSCs were transduced with adenovirus to express secretable EphrinA1-PE38. Our invitro assays confirmed the expression, release and selective killing effect of the immunotoxin produced by hMSCs. Furthermore, the intratumoral injection of engineered hMSCs was effective at inhibiting tumor growth in a malignant glioma tumor model. These results indicate that gene therapy utilizing EphrinA1-PE38-secreting hMSCs may provide a novel approach for the local treatment of malignant gliomas.


Subject(s)
Bone Marrow Cells/pathology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunotoxins/therapeutic use , Mesenchymal Stem Cells/pathology , Receptor, EphA2/immunology , Animals , Base Sequence , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Primers , Female , Glioma/pathology , Humans , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction
9.
Neurosci Lett ; 458(3): 116-21, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19394407

ABSTRACT

This study is designed to evaluate the therapeutic effects of three types of neurospheres (NSs) derived from brain, bone marrow and adipose tissue in a rat model of spinal contusive injury. As shown by BBB locomotor rating scale and grid test, the optimal therapeutic responses generated by subventricular zone-derived NSs (SVZ-NSs), and followed by adipose-derived (AD-NSs) and bone marrow-derived NSs (BM-NSs) after being grafted into the injured spinal cord. In three cell-treated groups, very few (<1%) grafted cells survived and these survived cells mainly differentiated into oligodendrocytes at week 12 after injury. Additionally, all the cell-treated groups, especially in the SVZ-treated group showed an increase in host oligodendrocytes than control group. Moreover, the level of selective neurotrophins (NTs) in the SVZ-NSs group were significantly higher than those in the BM-NSs and AD-NSs groups, and the level of NTs in the saline group was also significantly higher than sham group. Therefore, not cell replacement or infusion but neuroprotective action associated with endogenous oligodendrocytes and NTs that active by the grafted NSs may contribute to the functional recovery.


Subject(s)
Bone Marrow Transplantation , Neurons/transplantation , Spinal Cord Injuries/surgery , Stem Cell Transplantation , Adipose Tissue/transplantation , Animals , Male , Nerve Growth Factors/metabolism , Nerve Regeneration , Oligodendroglia/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL