Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Stem Cells ; 34(3): 653-67, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26661057

ABSTRACT

Degenerative disc disease (DDD) primarily affects the central part of the intervertebral disc namely the nucleus pulposus (NP). DDD explains about 40% of low back pain and is characterized by massive cellular alterations that ultimately result in the disappearance of resident NP cells. Thus, repopulating the NP with regenerative cells is a promising therapeutic approach and remains a great challenge. The objectives of this study were to evaluate the potential of growth factor-driven protocols to commit human adipose stromal cells (hASCs) toward NP-like cell phenotype and the involvement of Smad proteins in this differentiation process. Here, we demonstrate that the transforming growth factor-ß1 and the growth differentiation factor 5 synergistically drive the nucleopulpogenic differentiation process. The commitment of the hASCs was robust and highly specific as attested by the expression of NP-related genes characteristic of young healthy human NP cells. In addition, the engineered NP-like cells secreted an abundant aggrecan and type II collagen rich extracellular matrix comparable with that of native NP. Furthermore, we demonstrate that these in vitro engineered cells survived, maintained their specialized phenotype and secretory activity after in vivo transplantation in nude mice subcutis. Finally, we provide evidence suggesting that the Smad 2/3 pathway mainly governed the acquisition of the NP cell molecular identity while the Smad1/5/8 pathway controlled the NP cell morphology. This study offers valuable insights for the development of biologically-inspired treatments for DDD by generating adapted and exhaustively characterized autologous regenerative cells.


Subject(s)
Cell Differentiation/genetics , Growth Differentiation Factor 5/genetics , Intervertebral Disc Degeneration/therapy , Mesenchymal Stem Cell Transplantation , Transforming Growth Factor beta1/genetics , Adipocytes/cytology , Adipocytes/transplantation , Animals , Cell Engineering/methods , Extracellular Matrix , Growth Differentiation Factor 5/therapeutic use , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Low Back Pain , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Nucleus Pulposus/cytology , Nucleus Pulposus/transplantation , Smad Proteins/genetics , Transforming Growth Factor beta1/therapeutic use
2.
Stem Cell Res Ther ; 6: 9, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25971780

ABSTRACT

INTRODUCTION: Huntington's disease (HD) is an autosomal dominant disorder caused by an expanded CAG repeat (greater than 38) on the short arm of chromosome 4, resulting in loss and dysfunction of neurons in the neostriatum and cortex, leading to cognitive decline, motor dysfunction, and death, typically occurring 15 to 20 years after the onset of motor symptoms. Although an effective treatment for HD has remained elusive, current studies using transplants of bone-marrow-derived mesenchymal stem cells provides considerable promise. This study further investigates the efficacy of these transplants with a focus on comparing how passage number of these cells may affect subsequent efficacy following transplantation. METHODS: In this study, mesenchymal stem cells isolated from the bone-marrow of mice (BM MSCs), were labeled with Hoechst after low (3 to 8) or high (40 to 50) numbers of passages and then transplanted intrastriatally into 5-week-old R6/2 mice, which carries the N-terminal fragment of the human HD gene (145 to 155 repeats) and rapidly develops symptoms analogous to the human form of the disease. RESULTS: It was observed that the transplanted cells survived and the R6/2 mice displayed significant behavioral and morphological sparing compared to untreated R6/2 mice, with R6/2 mice receiving high passage BM MSCs displaying fewer deficits than those receiving low-passage BM MSCs. These beneficial effects are likely due to trophic support, as an increase in brain derived neurotrophic factor mRNA expression was observed in the striatum following transplantation of BM MSCs. CONCLUSION: The results from this study demonstrate that BM MSCs hold significant therapeutic value for HD, and that the amount of time the cells are exposed to in vitro culture conditions can alter their efficacy.


Subject(s)
Bone Marrow Cells/cytology , Huntington Disease/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Disease Models, Animal , Female , Humans , Huntingtin Protein , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Motor Activity , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Tissue Proteins/genetics , Receptor, trkB/genetics , Receptor, trkB/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
J Cell Mol Med ; 19(1): 124-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25310920

ABSTRACT

Foetal pig neuroblasts are interesting candidates as a cell source for transplantation, but xenotransplantation in the brain requires the development of adapted immunosuppressive treatments. As systemic administration of high doses of cyclosporine A has side effects and does not protect xenotransplants forever, we focused our work on local control of the host immune responses. We studied the advantage of cotransplanting syngenic mesenchymal stem cells (MSC) with porcine neuroblasts (pNb) in immunocompetent rat striata. Two groups of animals were transplanted, either with pNb alone or with both MSC and pNb. At day 63, no porcine neurons were detected in the striata that received only pNb, while four of six rats transplanted with both pNb and MSC exhibited healthy porcine neurons. Interestingly, 50% of the cotransplanted rats displayed healthy grafts with pNF70+ and TH+ neurons at 120 days post-transplantation. qPCR analyses revealed a general dwindling of pro- and anti-inflammatory cytokines in the striata that received the cotransplants. Motor recovery was also observed following the transplantation of pNb and MSC in a rat model of Parkinson's disease. Taken together, the present data indicate that the immunosuppressive properties of MSC are of great interest for the long-term survival of xenogeneic neurons in the brain.


Subject(s)
Brain/immunology , Immunity , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Transplantation, Heterologous , Animals , CD11b Antigen/metabolism , Cell Survival , Chemokines/genetics , Chemokines/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Graft Survival/immunology , Immunity, Cellular , Immunocompetence , Male , Mesencephalon/cytology , Molecular Sequence Data , Motor Activity , Neurons/cytology , Neurons/metabolism , Neurons/transplantation , Oxidopamine , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred Lew , Recovery of Function , Sus scrofa
4.
Med Sci (Paris) ; 30(12): 1091-100, 2014 Dec.
Article in French | MEDLINE | ID: mdl-25537039

ABSTRACT

A large proportion of low back pain may be explained by intervertebral disc (IVD) degeneration. Currently, the process leading to IVD degeneration highlights the pivotal role of IVD cells. The number of these cells drastically decreases and does not support a spontaneous repair of the tissue. In order to counteract IVD degeneration, regenerative medicine, based on a cell supplementation of the damaged tissue is considered as a promising approach. After a description of IVD physiopathology, we will develop the different strategies based on cell therapy and tissue engineering and currently under investigation to improve altered IVD degeneration. Finally, results from the current pre-clinical and clinical studies will be discussed.


Subject(s)
Intervertebral Disc Degeneration/therapy , Intervertebral Disc/physiology , Regeneration , Biocompatible Materials , Cell- and Tissue-Based Therapy , Humans , Tissue Engineering , Treatment Outcome
6.
Stem Cells Transl Med ; 3(5): 620-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24657963

ABSTRACT

Induced pluripotent stem cells (iPSCs) show considerable promise for cell replacement therapies for Huntington's disease (HD). Our laboratory has demonstrated that tail-tip fibroblasts, reprogrammed into iPSCs via two adenoviruses, can survive and differentiate into neuronal lineages following transplantation into healthy adult rats. However, the ability of these cells to survive, differentiate, and restore function in a damaged brain is unknown. To this end, adult rats received a regimen of 3-nitropropionic acid (3-NP) to induce behavioral and neuropathological deficits that resemble HD. At 7, 21, and 42 days after the initiation of 3-NP or vehicle, the rats received intrastriatal bilateral transplantation of iPSCs. All rats that received 3-NP and vehicle treatment displayed significant motor impairment, whereas those that received iPSC transplantation after 3-NP treatment had preserved motor function. Histological analysis of the brains of these rats revealed significant decreases in optical densitometric measures in the striatum, lateral ventricle enlargement, as well as an increase in striosome size in all rats receiving 3-NP when compared with sham rats. The 3-NP-treated rats given transplants of iPSCs in the 7- or 21-day groups did not exhibit these deficits. Transplantation of iPSCs at the late-stage (42-day) time point did not protect against the 3-NP-induced neuropathology, despite preserving motor function. Transplanted iPSCs were found to survive and differentiate into region-specific neurons in the striatum of 3-NP rats, at all transplantation time points. Taken together, these results suggest that transplantation of adenovirus-generated iPSCs may provide a potential avenue for therapeutic treatment of HD.


Subject(s)
Adenoviridae , Corpus Striatum , Huntington Disease , Induced Pluripotent Stem Cells , Stem Cell Transplantation , Transduction, Genetic , Animals , Behavior, Animal , Convulsants/adverse effects , Convulsants/pharmacology , Disease Models, Animal , Female , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/therapy , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Male , Nitro Compounds/adverse effects , Nitro Compounds/pharmacology , Propionates/adverse effects , Propionates/pharmacology , Rats , Rats, Sprague-Dawley
7.
Cell Transplant ; 23(11): 1407-23, 2014.
Article in English | MEDLINE | ID: mdl-23879897

ABSTRACT

Induced pluripotent stem cells (iPSCs) offer certain advantages over embryonic stem cells in cell replacement therapy for a variety of neurological disorders. However, reliable procedures, whereby transplanted iPSCs can survive and differentiate into functional neurons, without forming tumors, have yet to be devised. Currently, retroviral or lentiviral reprogramming methods are often used to reprogram somatic cells. Although the use of these viruses has proven to be effective, formation of tumors often results following in vivo transplantation, possibly due to the integration of the reprogramming genes. The goal of the current study was to develop a new approach, using an adenovirus for reprogramming cells, characterize the iPSCs in vitro, and test their safety, survivability, and ability to differentiate into region-appropriate neurons following transplantation into the rat brain. To this end, iPSCs were derived from bone marrow-derived mesenchymal stem cells and tail-tip fibroblasts using a single cassette lentivirus or a combination of adenoviruses. The reprogramming efficiency and levels of pluripotency were compared using immunocytochemistry, flow cytometry, and real-time polymerase chain reaction. Our data indicate that adenovirus-generated iPSCs from tail-tip fibroblasts are as efficient as the method we used for lentiviral reprogramming. All generated iPSCs were also capable of differentiating into neuronal-like cells in vitro. To test the in vivo survivability and the ability to differentiate into region-specific neurons in the absence of tumor formation, 400,000 of the iPSCs derived from tail-tip fibroblasts that were transfected with the adenovirus pair were transplanted into the striatum of adult, immune-competent rats. We observed that these iPSCs produced region-specific neuronal phenotypes, in the absence of tumor formation, at 90 days posttransplantation. These results suggest that adenovirus-generated iPSCs may provide a safe and viable means for neuronal replacement therapies.


Subject(s)
Adenoviridae/physiology , Cellular Reprogramming/physiology , Corpus Striatum/surgery , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/virology , Stem Cell Transplantation/methods , Animals , Cell Differentiation/physiology , Cell Survival/physiology , Corpus Striatum/cytology , Male , Rats , Rats, Sprague-Dawley , Transfection
8.
Joint Bone Spine ; 81(2): 125-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23932724

ABSTRACT

Lumbar intervertebral discs (IVDs) are prone to degeneration upon skeletal maturity. In fact, this process could explain approximately 40% of the cases of low back pain in humans. Despite the efficiency of pain-relieving treatments, the scientific community seeks to develop innovative therapeutic approaches that might limit the use of invasive surgical procedures (e.g., spine fusion and arthroplasty). As a prerequisite to the development of these strategies, we must improve our fundamental knowledge regarding IVD pathophysiology. Recently, several studies have demonstrated that there is a singular phenotype associated with Nucleus pulposus (NP) cells, which is distinct from that of articular chondrocytes. In parallel, recent studies concerning the origin and development of NP cells, as well as their role in intervertebral tissue homeostasis, have yielded new insights into the complex mechanisms involved in disc degeneration. This review summarizes our current understanding of IVD physiology and the complex cell-mediated processes that contribute to IVD degeneration. Collectively, these recent advances could inspire the scientific community to explore new biotherapeutic strategies.


Subject(s)
Intervertebral Disc Degeneration/physiopathology , Intervertebral Disc/physiology , Lumbar Vertebrae , Aging/physiology , Humans , Intervertebral Disc/embryology , Intervertebral Disc/growth & development , Intervertebral Disc Degeneration/complications , Low Back Pain/etiology
9.
Stem Cells ; 32(2): 500-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23939879

ABSTRACT

Stem cells have gained significant interest as a potential treatment of neurodegenerative diseases, including Huntington's disease (HD). One source of these cells is adult neural stem cells (aNSCs), which differentiate easily into neuronal lineages. However, these cells are vulnerable to immune responses following transplantation. Another source is bone-marrow-derived mesenchymal stem cells (MSCs), which release neurotrophic factors and anti-inflammatory cytokines following transplantation, and are less vulnerable to rejection. The goal of this study was to compare the efficacy of transplants of MSCs, aNSCs, or cotransplants of MSCs and aNSCs for reducing deficits in a transgenic rat model of HD. HD rats received intrastriatal transplantations of 400,000 MSCs, aNSCs, or a combination of MSCs/aNSCs, while wild-type and HD controls were given vehicle. Rats were tested on the rotarod over the course of 20 weeks. The results indicated that transplants of: (a) aNSCs produced a strong immune response and conferred short-term behavioral benefits; (b) MSCs elicited a relatively weak immune response, and provided a longer term behavioral benefit; and (c) combined MSCs and aNSCs conferred long-term behavioral benefits and increased survival of the transplanted aNSCs. The finding that cotransplanting MSCs with aNSCs can prolong aNSC survival and provide greater behavioral sparing than when the transplants contains only aNSCs suggests that MSCs are capable of creating a more suitable microenvironment for aNSC survival. This cotransplantation strategy may be useful as a future therapeutic option for treating HD, especially if long-term survival of differentiated cells proves to be critically important for preserving lasting functional outcomes.


Subject(s)
Cell- and Tissue-Based Therapy , Huntington Disease/therapy , Mesenchymal Stem Cell Transplantation , Neural Stem Cells/transplantation , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Disease Models, Animal , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Rats , Rats, Transgenic
10.
J Neurosci ; 33(47): 18672-85, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259588

ABSTRACT

The immunoreceptor-associated protein CD3ζ is known for its role in immunity and has also been implicated in neuronal development and synaptic plasticity. However, the mechanism by which CD3ζ regulates synaptic transmission remains unclear. In this study, we showed that mice lacking CD3ζ exhibited defects in spatial learning and memory as examined by the Barnes maze and object location memory tasks. Given that peripheral T cells have been shown to support cognitive functions and neural plasticity, we generated CD3ζ(-/-) mice in which the peripheral T cells were repopulated to a normal level by syngeneic bone marrow transplantation. Using this approach, we showed that T-cell replenishment in CD3ζ(-/-) mice did not restore spatial memory defects, suggesting that the cognitive deficits in CD3ζ(-/-) mice were most likely mediated through a T-cell-independent mechanism. In support of this idea, we showed that CD3ζ proteins were localized to glutamatergic postsynaptic sites, where they interacted with the NMDAR subunit GluN2A. Loss of CD3ζ in brain decreased GluN2A-PSD95 association and GluN2A synaptic localization. This effect was accompanied by a reduced interaction of GluN2A with the key NMDAR downstream signaling protein calcium/calmodulin-dependent protein kinase II (CaMKII). Using the glycine-induced, NMDA-dependent form of chemical long-term potentiation (LTP) in cultured cortical neurons, we showed that CD3ζ was required for activity-dependent CaMKII autophosphorylation and for the synaptic recruitment of the AMPAR subunit GluA1. Together, these results support the model that the procognitive function of CD3ζ may be mediated through its involvement in the NMDAR downstream signaling pathway leading to CaMKII-dependent LTP induction.


Subject(s)
CD3 Complex/metabolism , Memory Disorders/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , T-Lymphocytes/pathology , Animals , Bone Marrow Transplantation , CD3 Complex/genetics , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation/genetics , Glycine/pharmacology , Leukocyte Common Antigens/genetics , Maze Learning , Memory Disorders/physiopathology , Memory Disorders/surgery , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Recognition, Psychology/physiology
11.
Stem Cell Res Ther ; 4(5): 130, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24456799

ABSTRACT

INTRODUCTION: Huntington's disease (HD) is an autosomal dominant disorder caused by an expanded CAG repeat on the short arm of chromosome 4 resulting in cognitive decline, motor dysfunction, and death, typically occurring 15 to 20 years after the onset of motor symptoms. Neuropathologically, HD is characterized by a specific loss of medium spiny neurons in the caudate and the putamen, as well as subsequent neuronal loss in the cerebral cortex. The transgenic R6/2 mouse model of HD carries the N-terminal fragment of the human HD gene (145 to 155 repeats) and rapidly develops some of the behavioral characteristics that are analogous to the human form of the disease. Mesenchymal stem cells (MSCs) have shown the ability to slow the onset of behavioral and neuropathological deficits following intrastriatal transplantation in rodent models of HD. Use of MSCs derived from umbilical cord (UC) offers an attractive strategy for transplantation as these cells are isolated from a noncontroversial and inexhaustible source and can be harvested at a low cost. Because UC MSCs represent an intermediate link between adult and embryonic tissue, they may hold more pluripotent properties than adult stem cells derived from other sources. METHODS: Mesenchymal stem cells, isolated from the UC of day 15 gestation pups, were transplanted intrastriatally into 5-week-old R6/2 mice at either a low-passage (3 to 8) or high-passage (40 to 50). Mice were tested behaviorally for 6 weeks using the rotarod task, the Morris water maze, and the limb-clasping response. Following behavioral testing, tissue sections were analyzed for UC MSC survival, the immune response to the transplanted cells, and neuropathological changes. RESULTS: Following transplantation of UC MSCs, R6/2 mice did not display a reduction in motor deficits but there appeared to be transient sparing in a spatial memory task when compared to untreated R6/2 mice. However, R6/2 mice receiving either low- or high-passage UC MSCs displayed significantly less neuropathological deficits, relative to untreated R6/2 mice. CONCLUSIONS: The results from this study demonstrate that UC MSCs hold promise for reducing the neuropathological deficits observed in the R6/2 rodent model of HD.


Subject(s)
Behavior, Animal , Huntington Disease/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Motor Activity , Umbilical Cord/cytology , Animals , Behavior, Animal/physiology , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Huntington Disease/pathology , Male , Memory , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology
12.
Behav Neurosci ; 126(3): 479-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22642889

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder in humans caused by an expansion of a CAG trinucleotide repeat that produces choreic movements, which are preceded by cognitive deficits. The HD transgenic rat (tgHD), which contains the human HD mutation with a 51 CAG repeat allele, exhibits motor deficits that begin when these rats are 12 months of age. However, there are no reports of cognitive dysfunction occurring prior to this. To assess whether cognitive dysfunction might precede motor deficits in tgHD rats, one group of 9-month-old male rats with homozygotic mutated genes and one group of wild-type (WT) rats underwent three testing phases in a unique Spatial Operant Reversal Test (SORT) paradigm, as well as assessment of spontaneous motor activity. After testing, morphological and histological examination of the brains were made. Results indicated that tgHD rats acquired the cued-response (Phase 1) portion of the SORT, but made significantly more errors during the reversal (Phase 2) and during the pseudorandomized reversals (Phase 3) portion of the study, when compared to WT rats. Analysis of the data using mathematical principles of reinforcement revealed no memory, motor, or motivational deficits. These results indicate that early cognitive dysfunction, as measured by the SORT, occur prior to motor deficits, gross anatomical changes, or cell loss in the tgHD rat with 51 CAG repeats, and suggest that this protocol could provide a useful screen for therapeutic studies.


Subject(s)
Cognition Disorders/genetics , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Trinucleotide Repeats/genetics , Animals , Cell Death/genetics , Cognition Disorders/psychology , Conditioning, Operant/physiology , Disease Models, Animal , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/psychology , Male , Random Allocation , Rats , Rats, Transgenic
13.
Methods Mol Biol ; 879: 147-64, 2012.
Article in English | MEDLINE | ID: mdl-22610559

ABSTRACT

Treatments for neurodegenerative diseases have little impact on the long-term patient health. However, cellular transplants of neuroblasts derived from the aborted embryonic brain tissue in animal models of neurodegenerative disorders and in patients have demonstrated survival and functionality in the brain. However, ethical and functional problems due to the use of this fetal tissue stopped most of the clinical trials. Therefore, new cell sources were needed, and scientists focused on neural (NSCs) and mesenchymal stem cells (MSCs). When transplanted in the brain of animals with Parkinson's or Huntington's disease, NSCs and MSCs were able to induce partial functional recovery by promoting neuroprotection and immunomodulation. MSCs are more readily accessible than NSCs due to sources such as the bone marrow. However, MSCs are not capable of differentiating into neurons in vivo where NSCs are. Thus, transplantation of NSCs and MSCs is interesting for brain regenerative medicine. In this chapter, we detail the methods for NSCs and MSCs isolation as well as the transplantation procedures used to treat rodent models of neurodegenerative damage.


Subject(s)
Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Neurodegenerative Diseases/therapy , Stem Cell Transplantation/methods , Animals , Cell Separation/methods , Cells, Cultured , Rats , Rats, Sprague-Dawley
14.
Parkinsons Dis ; 2011: 987084, 2011.
Article in English | MEDLINE | ID: mdl-21766003

ABSTRACT

A transgenic Sprague Dawley rat bearing the A30P and A53T α-synuclein (α-syn) human mutations under the control of the tyrosine hydroxylase promoter was generated in order to get a better understanding of the role of the human α-syn mutations on the neuropathological events involved in the progression of the Parkinson's disease (PD). This rat displayed olfactory deficits in the absence of motor impairments as observed in most early PD cases. In order to investigate the role of the mutated α-syn on cell proliferation, we focused on the subventricular zone (SVZ) and the olfactory bulbs (OB) as a change of the proliferation could affect OB function. The effect on OB dopaminergic innervation was investigated. The human α-syn co-localized in TH-positive OB neurons. No human α-syn was visualized in the SVZ. A significant increase in resident cell proliferation in the glomerular but not in the granular layers of the OB and in the SVZ was observed. TH innervation was significantly increased within the glomerular layer without an increase in the size of the glomeruli. Our rat could be a good model to investigate the role of human mutated α-syn on the development of olfactory deficits.

15.
PLoS One ; 6(5): e19820, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21589877

ABSTRACT

BACKGROUND: Adult mesenchymal stem cells (MSCs) can be maintained over extended periods of time before activation and differentiation. Little is known about the programs that sustain the survival of these cells. PRINCIPAL FINDINGS: Undifferentiated adult human MSCs (hMSCs) did not undergo apoptosis in response to different cell death inducers. Conversely, the same inducers can readily induce apoptosis when hMSCs are engaged in the early stages of differentiation. The survival of undifferentiated cells is linked to the expression of Bcl-Xl and Bcl-2 in completely opposite ways. Bcl-Xl is expressed at similar levels in undifferentiated and differentiated hMSCs while Bcl-2 is expressed only in differentiated cells. In undifferentiated hMSCs, the down-regulation of Bcl-Xl is associated with an increased sensitivity to apoptosis while the ectopic expression of Bcl-2 induced apoptosis. This apoptosis is linked to the presence of cytoplasmic Nur 77 in undifferentiated hMSCs. SIGNIFICANCE: In hMSCs, the expression of Bcl-2 depends on cellular differentiation and can be either pro- or anti-apoptotic. Bcl-Xl, on the other hand, exhibits an anti-apoptotic activity under all conditions.


Subject(s)
Apoptosis/physiology , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Proto-Oncogene Proteins c-bcl-2/physiology , bcl-X Protein/physiology , Humans
16.
Behav Brain Res ; 217(2): 369-78, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21070819

ABSTRACT

Transplantation of mesenchymal stem cells (MSCs) may offer a viable treatment for Huntington's disease (HD). We tested the efficacy of MSC transplants to reduce deficits in a 3-nitropropionic acid (3NP) rat model of HD. Five groups of rats (Sham, 3NP, 3NP+vehicle, 3NP+TP(low), 3NP+TP(high)), were given PBS or 3NP intraperitoneally, twice daily for 42 days. On day 28, rats in all groups except Sham and 3NP, received intrastriatal injections of either 200,000 MSCs (TP(low)), 400,000 (TP(high)) MSCs or DMEM (VH, the vehicle for transplantation). MSCs survived 72 days without inducing a strong inflammatory response from the striatum. Behavioral sparing was observed on tests of supported-hindlimb-retraction, unsupported-hindlimb-retraction, visual paw placement and stepping ability for 3NP+TP(low) rats and on the unsupported-hindlimb-retraction and rotarod tasks for 3NP+VH rats. Relative to 3NP controls, all treated groups were protected from 3NP-induced enlargement of the lateral ventricles. In vitro, MSCs expressed transcripts for numerous neurotrophic factors. In vivo, increased striatal labeling in BDNF, collagen type-I and fibronectin (but not GDNF or CNTF) was observed in the brains of MSC-transplanted rats but not in DMEM-treated rats. In addition, none of the transplanted MSCs expressed neural phenotypes. These findings suggest that factors other than neuronal replacement underlie the behavioral sparing observed in 3NP rats after MSC transplantation.


Subject(s)
Convulsants/toxicity , Huntington Disease/chemically induced , Huntington Disease/surgery , Mesenchymal Stem Cell Transplantation/methods , Nitro Compounds/toxicity , Propionates/toxicity , Animals , Behavior, Animal , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation/drug effects , Hindlimb/drug effects , Hindlimb/physiopathology , Huntington Disease/pathology , Huntington Disease/physiopathology , Male , Motor Activity/drug effects , Motor Activity/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Osteoblasts/drug effects , Osteoblasts/physiology , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
17.
Behav Brain Res ; 214(2): 193-200, 2010 Dec 25.
Article in English | MEDLINE | ID: mdl-20493905

ABSTRACT

The purpose of this study was to evaluate the therapeutic effects of the transplantation of bone-marrow mesenchymal stem cells (MSCs), genetically engineered to over-express brain-derived neurotrophic factor (BDNF) or nerve growth factor (NGF) on motor deficits and neurodegeneration in YAC 128 transgenic mice. MSCs, harvested from mouse femurs, were genetically engineered to over-express BDNF and/or NGF and these cells, or the vehicle solution, were injected into the striata of four-month old YAC 128 transgenic and wild-type mice. Assessments of motor ability on the rotarod and the severity of clasping were made one day prior to transplantation and once monthly, thereafter, to determine the effects of the transplanted cells on motor function. The mice were sacrificed at 13-months of age for immunohistological examination. All YAC 128 mice receiving transplants had reduced clasping, relative to vehicle-treated YAC 128 mice, while YAC 128 mice that were transplanted with MSCs which were genetically engineered to over-express BDNF, had the longest latencies on the rotarod and the least amount of neuronal loss within the striatum of the YAC 128 mice. These results indicate that intrastriatal transplantation of MSCs that over-express BDNF may create an environment within the striatum that slows neurodegenerative processes and provides behavioral sparing in the YAC 128 mouse model of HD. Further research on the long-term safety and efficacy of this approach is needed before its potential clinical utility can be comprehensively assessed.


Subject(s)
Chromosomes, Artificial, Yeast/genetics , Genetic Engineering/methods , Huntington Disease/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Motor Skills/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/pathology , Corpus Striatum/surgery , Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Nerve Growth Factor/metabolism
18.
J Cell Mol Med ; 13(8B): 2547-58, 2009 Aug.
Article in English | MEDLINE | ID: mdl-20141619

ABSTRACT

Mesenchymal stem cells (MSCs) have attracted attention for their potential use in regenerative medicine such as brain transplantation. As MSCs are considered to be hypoimmunogenic, transplanted MSCs should not trigger a strong host inflammatory response. To verify this hypothesis, we studied the brain immune response after transplantation of human or rat MSCs into the rat striatum and MSC fate at days 5, 14, 21 and 63 after transplantation. Flow cytometry analysis indicated that both MSCs express CD90 and human leucocyte antigen (MHC) class I, but no MHC class II molecules. They do not express CD45 or CD34 antigens. However, MSC phenotype varies with passage number. Human MSCs have mRNAs for interleukin (IL)-6, IL-8, IL-12, tumour necrosis factor (TNF)-alpha and TGF-beta(1), whereas rat MSCs express IL-6-, IL-10-, IL-12- and TGF-beta(1)-mRNAs. The quantification shows higher levels of mRNAs for the anti-inflammatory molecules IL-6 and TGF-beta(1) than for pro-inflammatory cytokines IL-8 and IL-12; ELISA analysis showed no IL-12 whereas TGF-beta(1) and IL-6 were detected. Transplant size did not significantly vary between 14 and 63 days after transplantation, indicating an absence of immune rejection of the grafts. Very few mast cells and moderate macrophage and microglial infiltrations, observed at day 5 remained stable until day 63 after transplantation in both rat and human MSC grafts. The observations of very few dendritic cells, T alphabeta-cells, and no T gammadelta-lymphocytes, all three being associated with Tp rejection in the brain, support the contention that MSCs are hypoimmunogenic. Our results suggest that MSCs are of great interest in regenerative medicine in a (xeno)transplantation setting.


Subject(s)
Corpus Striatum/immunology , Mesenchymal Stem Cells/cytology , Transplantation, Heterologous , Transplantation, Homologous , Animals , Cells, Cultured , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic
19.
Mov Disord ; 22(4): 533-9, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17230470

ABSTRACT

Parkinson's disease (PD) is characterized by the bilateral degeneration of the midbrain dopamine-containing neurons with the most severe lesion in the posterolateral part of the substantia nigra pars compacta (SNpc). In humans, such lesions lead to specific motor abnormalities (i.e., akinesia, rigidity, and tremor) that are greatly improved by levodopa treatment. After a few years, the beneficial effect of the treatment is frequently offset by the development of dyskinesias. To improve treatment strategies, an animal model showing most of the histological and clinical characteristics of the human disease is mandatory. Ten rats received a bilateral injection of small doses of 6-OHDA in the medial forebrain bundle (MFB) and were compared with five sham-lesioned rats. The 6-OHDA-lesioned rats progressively developed abnormal motor behavior (assessed by the stepping test) compared with the sham-lesioned rats. The lesioned rats greatly improved under levodopa treatment, but developed concomitant dyskinesias. All 6-OHDA-lesioned animals had bilateral partial lesions of the SNpc, with the most severe lesion being in its posterolateral part. There was a significant correlation between the severity of the dopaminergic cell loss and the severity of the levodopa-induced dyskinesias. These rats constitute an interesting model of PD, sharing some of the main characteristics of the human disease.


Subject(s)
Antiparkinson Agents/therapeutic use , Disease Models, Animal , Dyskinesias/drug therapy , Dyskinesias/physiopathology , Extremities/physiopathology , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Animals , Disease Progression , Dopamine/metabolism , Dyskinesias/diagnosis , Male , Mesencephalon/metabolism , Mesencephalon/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Parkinson Disease/pathology , Prosencephalon/metabolism , Prosencephalon/pathology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
20.
Restor Neurol Neurosci ; 25(5-6): 485-92, 2007.
Article in English | MEDLINE | ID: mdl-18334766

ABSTRACT

PURPOSE: The purpose of this study was to test the potential therapeutic effects of the substituted pyrimidine, KP544, which has been shown to amplify the effects of nerve growth factor in vitro, on motor deficits in the R6/2 transgenic mouse model of Huntington's disease (HD). METHODS: Young, female R6/2 mice were given daily oral intubation of either 10 mg/kg KP544 or vehicle (0.5% methylcellulose) at 6 weeks of age and tested from postnatal weeks 8 through 12 on a battery of motor tasks, including assessments of clasping (drawing of the limbs to the torso when suspended by the tail), motor coordination on the rotarod, and spontaneous motor activity in the open-field. Following testing, the mice were sacrificed and the brains were sectioned and stained with cresyl violet for histological examination. RESULTS: KP544 treatment decreased balance deficits on the rotarod task, reduced clasping, delayed the onset of hypoactivity, and reduced enlargement of the lateral ventricles in R6/2 mice. CONCLUSION: These results suggest that KP544 can reduce motor deficits and anatomical alterations in R6/2 mice. Further research into the use of KP544 as a potential pharmacotherapy HD is warranted.


Subject(s)
Cyclohexanols/therapeutic use , Huntington Disease/complications , Huntington Disease/genetics , Movement Disorders/drug therapy , Movement Disorders/etiology , Pyrimidines/therapeutic use , Age Factors , Analysis of Variance , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Disease Models, Animal , Exploratory Behavior/drug effects , Female , Huntington Disease/pathology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Skills/drug effects , Movement Disorders/genetics , Mutation , Reaction Time
SELECTION OF CITATIONS
SEARCH DETAIL
...