Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników

Znaleziono wyników: 9

Liczba wyników na stronie
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników

Wyniki wyszukiwania

help Sortuj według:

help Ogranicz wyniki do:
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników
Here we provide a comprehensive data on the unique features of mesenchymal stem cells (MSCs) which makes them feasible and preferred candidate for cell-based therapy in neurological clinic. From this point of view the most important features of these cells are: (1) availability from autologous sources independently from age of patient; (2) extensive expansion in vitro; (3) immunomodulatory "bystander" function after transplantation in vivo; (4) potentiality to protect, repair or eventually replace impaired or dysfunctional host cells. For complete these last task of functional regeneration of central nervous system, we have to take advantages of MSCs capability for transient, time-locked proliferation, migration to site of injury and their commitment to neuronal differentiation. However, if we are to make progress in the use of MSCs for therapy in the clinic it will be necessary to establish more unified, advanced standards for cells processing in vitro as well as safer and improved procedures for their delivery in vivo.
Mesenchymal stem cells (MSC) are of clinical interest because of their potential use in autologous transplantation. The ability of MSC to differentiate into multiple different cells of mesodermal origin has offered therapeutic tool for the treatment of hematopoietic malignancies and graft versus host disease. Recently, MSC have been shown to ameliorate a variety of neurological dysfunction. This effect is believed to be mediated to their paracrine functions since it is known that MSC produce bioactive substances that promote endogenous neurogenesis. However, the critical question that remains unanswered is whether MSC can transdifferentiate into neural cells. To be “primed” toward a neuronal fate, MSC have to express neural antigens. In an attempt to clarify this issue we explored the constitutive expression of different markers by mesenchymal cells isolated from human umbilical cord Wharton jelly (HUC-MSC) and compare their expression with neural stem cell line derived from human umbilical cord blood (HUCB-NSC) established in our laboratory. Materials and methods: Gene expression pattern in HUC-MSC (passage 5 of cells cultured in MSCGM hMSC Lonza medium) and HUCB-NSC (cultured in DMEM/F12 medium+2% FBS) was performed by real time PCR and RT-PCR reactions. Total RNA was extracted using TRIzol (Invitrogen). Then cDNA was synthesized from total RNA, using High Capacity RNA-to-cDNA kit (Applied Biosystems). PCR reactions were carried out using template cDNA in the presence of specific primers. Concomitantly immunocytochemical analysis of gene-related proteins was employed. The results of our studies have demonstrated that HUC-MSC, in addition to pluripotent (Oct3/4, Nanog1), mesenchymal (CD73, CD90, CD105, CD166) and extracellular matrix (Fibronectin, Vimentin, Collagen1) genes, spontaneously express neural genes i.e. Nestin, NF200, βIIITubulin, MAP2 and GFAP. The initially expressed neuroectodermal genes were comparable with mRNA level of the same neural genes in HUCB-NSC. In addition to neural genes noninduced expression of neural proteins was found. Subsets of HUCMSC were positive for several neural markers, including: Nestin, NF200, βIIITubulin and GFAP. Summary and conclusion: We have demonstrated that MSC derived from human umbilical cord Wharton jelly acquire neural progenitor-like properties by expressing neuronal and astrocytic specific markers. However, it is of clinical interest whether transplanted MSC respond with an appropriate neural pattern of differentiation when exposed to the environment of the host brain. Supported by MSHE grant no N401 014235 and Foundation Jerome Lejeune grant as part of the Novus Sanguis research consortium.
There is a great interest in the possibility of repairing the nervous system by transplantation new cells that can replace those lost through damage in neurological disorders. Key functions such as the replacement of neural cells have been recently challenged by intrinsic bystander capacities of undifferentiated donor cells to restore these cells. A comprehensive knowledge how transplanted stem cells exert their therapeutic achievements is still lacking. Here we investigated the effects of HUCB-NSC infused into the damaged rat brain at 72 h post ischemia on endogenous neurogenesis. The goal of our studies was to examine the proliferation and migration of host progenitor cells, analyze the substantial matrix remodeling of tissue and the presence of neurotrophic factors in rat brain after focal ischemia followed by HUCB-NSC transplantation. Methods: 2×104 HUCB-NSC were transplanted into corpus callosum of naive or focally injured rat brain 3 days after ischemic insult. At 1, 3, 7 and 14 days rat brains were removed. Endogenous cell proliferation was determined by BrdU incorporation. Then immunocytochemical analysis of doublecortin (DCX) and PSA-NCAM (markers expressed by immature migratory neuroblasts), and in situ zymography of MMPs activity was performed. Additionally, total RNA was isolated from rat brain tissue and RTPCR was performed using sets of primers of each of human and rat neurotrophic factor genes. Results: OUA-induced brain lesion resulted in increase of proliferating (BrdU+) and migrating (DCX+ and PSA-NCAM+) cells in subventricular zone (SVZ) and subgranular zone (SGZ) regions in comparison to intact rats. This response has been potentiated by HUCB-NSC transplantation. At 7th day after HUCB-NSC infusion the intense migration of DCX+cells from SVZ towards ischemic boundary regions of the striatum was observed. Moreover, the activation of MMPs in cells was visible in SVZ. Double-labeling showed co-localization of DCX marker with MMPs activity. The presence of MMPs appeared to be associated with cell nuclei and cytoplasm but interestingly it was also seen outside the cell bodies and in the neuronal protrusions. In OUAinduced lesion rat brain tissue, the expression pattern of rat-origin neurotrophic factors mRNA was higher than in intact rats. HUCBNSC transplantation into focal brain ischemic tissue significantly increased mRNA expression of several rat-origin growth factors, such as GDNF, CNTF responsible for regulation of proliferation and maturation of stem cells as well as IGF-1, HGF and presaposin functioning as anti-apoptotic mediators. The significant increment was observed 7 days after HUCB-NSC infusion. Using Real Time PCR method we were able to detect the presence of mRNA of BDNF, GDNF, NT3, IGF-1, HGF, semaphorin and presaposin of human-origin factors in the rat brain recipients of HUCB-NSC grafts. Conclusions: Transplantation of HUCB-NSC triggers early expansion of endogenous progenitor pool increasing fraction of proliferating cells in SVZ and SGZ of brain ischemic rats. Proteolytic activity of MMPs in extracellular compartment suggests its ability to remodel extracellular matrix and facilitate migration of neuroblasts to the damaged brain areas. The mechanism promoting recovery from ischemic injury remains to be clarified, although it is likely that it might be due to HUCB-NSC graft-induced release of neurotrophic factors by the host cells as well as the presence of human neural stem cells derived factors. Supported by MMRC statutory fund.
Cerebral ischemia causes severe functional deficits due to the death of neuronal and glial cells in the cortex and sub-cortical regions. Stem cell-based therapy could be used to restore lost cells and thus may enhance functional recovery. The aim of the study was to compare therapeutic effectiveness of intra-arterial infusion of human umbilical cord-blood derived mononuclear cells (HUCB-MNC) at different stages of their neural conversion in vitro. Materials and methods. Freshly isolated HUCB-MNC (D-0) neurally directed progenitors (D-3) obtained during 3 days culture of HUCB-MNC and neural-like stem cells (HUCB-NSC) line derived from human cord blood cells were assessed. Focal brain damage was induced in Wistar rats by stereotactic injection of previously established low dose of ouabain into dorsolateral striatum Three days later 107 HUCB cells were infused into internal carotid artery. Following surgery rats were housed in large enriched environment cages, in groups of 7-8 animals per cage, for 30 days observation period. Behavioral assessment consisted of tests for sensorimotor deficits (walking beam task, rotarod, vibrissae elicited forelimb placing), cognitive impairments (habit learning task and object recognition test), exploratory behavior (open field test) and apomorphine induced rotations. At the end of 30 days observation the lesion volume was measured and the presence of donor cells visualized by the expression of mRNA of human reference gene β-2-microglobulin. Results. Functional effects of different subsets of HUCB-MNC treatment shared substantial diversity in various behavioral tests. In walking beam test the most effective in recovery the impaired sensomotor functions in focal brain injured rats were freshly isolated HUCB-MNC (D-0). Also, in rotarod task and in apomorphine induced rotations the tendency to improve scores was observed 30 days following HUCB-MNC (D-0) treatment. In parameters describing open field exploratory behavior the positive effects of HUCB-MNC (D-0) as well as HUCB-NSC cells treatment were observed. However, in cognitive tasks none of tested cell subsets reduced the functional deficits induced by ouabain injection. Thirty days after HUCB cell transplantation we did not observed any mRNA expression of human reference gene in the rat brain samples. Conclusions. Our observation reveals that freshly isolated D-0 HUCB-MNC are the most effective in functional recovery of injured rats. These cells are also the most potent in reducing the ouabain-induced brain lesion volume. The best functional outcome observed after transplantation of HUCB-MNC (D-0) is probably due to the positive effect of therapeutic molecules secreted by these cells than the persistence of donor per se in the host since we did not detect systemically infused human cells in rat brains. Supported by MSHE grant no. 0394/B/P01/2010/38.
Mesenchymal stem cells (MSC) exert unique ability to differentiate into various cells of mesodermal origin. These properties place MSC as a very promising source of cells for regenerative medicine and tissue engineering. Recently, it has been shown that experimental transplantation of MSC improves a variety of neurological dysfunctions. Bone marrow (BM) represents the mostly exploited source of human therapeutic stem cells but similar populations have been recently identified in many other tissues and organs. Among them umbilical cord Wharton jelly (WJ) has been recognized for its safety, accessibility and differentiation potential. This study compares human Wharton jelly-derived MSC (WJ-MSC) and human bone marrow-derived MSC (BM-MSC) in terms of cell phenotype, optimal growth and multilineage differentiation characteristics with special attention to neurogenic potential demonstrated by both type of cells. Materials and Methods: MSC were isolated from human Wharton jelly and human bone marrow then cultured in vitro in Lonza medium in defined conditions. Then both cell types were subjected to the specific induction media (Gibco) to analyze their potential to differentiate into osteo-, chondro- adipo- and myogenic lineages. Transcriptional activity of genes characteristic for early and late stages of cell differentiation has been examined using RT-PCR. Concomitantly immunochemical analysis of certain gene-related proteins has been performed by immunocytochemical methods. Results: We have demonstrated that both isolated WJ-MSC and BM-MSC exhibited characteristic, mesenchymal cell specific phenotypes by expressing the panels of surface antigens (CD73, CD90, CD105, CD166) as well as typical for MSC multilineage differentiation markers. However, efficiency of these processes differs markedly between the cells derived from each of examined tissues. Thus, WJ-MSC appeared to be much less prone to adipogenic differentiation in comparison to BM-MSC. In contrast, WJMSC revealed higher proliferation and neural differentiations potential than BM-MSC. Consistently, only WJ-MSC-derived cells unveiled neural progenitor characteristics expressing panel of cellular markers typical for neural lineage differentiation, i.e. Nestin, NF200, GFAP. All together, these data allow us to hypothesize that the fetal origin of WJ tissue determines its distinguished neuro-mesenchymal characteristic. This is consisted with the data of Takashima et al. (2007) showing that neonatal MSC cultures contain substantially high number of cells being descendants of the earliest wave of developmentally discern neuroepithelial MSC lineage derived from cranial part of neural crest and clearly partitioned from MSC residing in adult bon marrow niche. Conclusions: The study demonstrated that WJ-MSC, similarly to BM-MSC, can be effectively expanded in culture up to 6–8 passages when maintaining cells in undifferentiated state expressing common MSC markers. In contrast, the both MSC lines differ markedly in their ability to lineage differentiation. The most striking difference was that only WJ-MSC can be induced to neural phenotypes. Consisted with this observation WJMSC seems to be more favorable than BM-MSC to cell replacement therapy of neurodegenerative diseases. Supported by NSC grant No 2011/01/B/NZ3/0540
Mesenchymal stem cells (MSC) emerged as promising candidates for therapeutic applications in regenerative medicine and tissue engineering. The ability of MSC to differentiate into multiple different cells of mesodermal origin has offered therapeutic tool for the treatment leukaemias and other malignant diseases. Recently, MSC have been shown to ameliorate a variety of neurological dysfunction. The critical question that remains unanswered is whether MSC can trans-differentiate into neural cells. In an attempt to clarify this issue we explored the expression profile of different markers by mesenchymal cells isolated from human umbilical cord Wharton jelly (HUC-MSC) and compare their expression with neural stem cell line derived from human umbilical cord blood (HUCB-NSC) established in our laboratory. Materials and methods: Gene expression pattern in HUC-MSC (hMSC Lonza medium) and HUCB-NSC (DMEM/F12 medium+2% FBS) was performed by RT-PCR and quantitative RT-PCR reactions. Total RNA was isolated from cultured cells and RT-PCR was performed by using gene-specific primers. The target gene value of each sample was normalized by the GAPDH value. Concomitantly immunocytochemical analysis of gene-related proteins was employed. Results: Direct comparison of the expression profiles demonstrated that HUC-MSC, in addition to pluripotent (Oct-4, Nanog) genes, spontaneously express neural genes: Nestin, NF-200, βIIITubulin, and GFAP. Concomitantly non-induced expression of neural proteins was found. The subsets of HUC-MSC were positive for several markers including: SSEA-4, Nestin, NF-200, βIIITubulin, GFAP and A2B5. Summary and conclusions: We have demonstrated that MSC derived from human umbilical cord Wharton jelly cultured in vitro acquire neural progenitor-like properties by expressing neuronal and astrocytic specific markers. Supported by MSHE grant No. N401 014235 and Foundation Jerome Lejeune grant as part of the Novus Sanguis research consortium.
The oxygen tension is an important factor modulating cell fate and developmental decisions. There are evidences that HIFs (Hypoxia Inducible Factors) family is implicated in the regulation of pluripotency and differentiation genes. The goal of this study is to compare the influence of close to physiological oxygen conditions (5%) to atmospheric oxygen tension on differentiation process and pluripotent activity in HUCB-NSC. The expression of Hypoxia Inducible Factors, stemness and neural differentiation markers in NSC, cultured under 5% and 21% oxygen were checked on the transcriptional and translational level. We were looking at the interaction between HIFs (HIF1 alpha, HIF 2 alpha) and activity of neural differentiations genes (MAP2, GFAP, β-tubulin) as well as expression of pluripotency genes (Oct4, Sox2, Rex1 and Nanog). In order to demonstrate the impact of increased HIF1α and/ or HIF2α level on cell differentiation we used DMOG (Sigma) which is of prolyl-4-hydroksylase inhibitor to increase HIF alpha levels. Our data show, that low oxygen conditions promote proliferation of HUCBNSC at early stage of development and can activate Oct4 and Nanog genes in HUCB-NSC. The time of cultivation of the cells in low oxygen conditions and the developmental stage of the cells are the important factors for the induction of the expression of “pluripotency” genes.Hypoxia Inducible Factors HIF 1α and HIF 2α, but not HIF3α are expressed in HUCB-NSC at all stages of development. During neuronal differentiation of HUCB-NSC by using dBcAMP, 5% oxygen level act synergistically, promoting further differentiation (enhanced MAP2 expression). Application of prolyl hydroxylase inhibitor – DMOG resulted in increased expression of HIF1α but not HIF2α and increased the expression of MAP2 (only in 21% oxygen conditions) referring to variants without DMOG. Sponsored by grant from Polish Ministry of Scientific Research and Higher Education No N N302 597838 and by NSC grant No 2011/01/B/NZ3/05401
The aim of the study was to evaluate therapeutic effectiveness of intra-arterial infusion of human umbilical cord blood (HUCB) derived cells at different stages of their neural conversion. Freshly isolated mononuclear cells (D-0), neurally directed progenitors (D-3) and neural-like stem cells derived from umbilical cord blood (NSC) were compared. Focal brain damage was induced in rats by stereotactic injection of ouabain into dorsolateral striatum Three days later 107 of different subsets of HUCB cells were infused into the right internal carotid artery. Following surgery rats were housed in enriched environment for 30 days. Behavioral assessment consisted of tests for sensorimotor deficits (walking beam, rotarod, vibrissae elicited forelimb placing, apomorphine induced rotations), cognitive impairments (habit learning and object recognition) and exploratory behavior (open field). Thirty days after surgery the lesion volume was measured and the presence of donor cells was detected in the brain at mRNA level. At the same time immunohistochemical analysis of brain tissue was performed to estimate the local tissue response of ouabain injured rats and its modulation after HUCB cells systemic treatment. Functional effects of different subsets of cord blood cells shared substantial diversity in various behavioral tests. An additional analysis showed that D-0 HUCB cells were the most effective in functional restoration and reduction of brain lesion volume. None of transplanted cord blood derived cell fractions were detected in rat's brains at 30th day after treatment. This may suggest that the mechanism(s) underlying positive effects of HUCB derived cell may concern the other than direct neural cell supplementation. In addition increased immunoreactivity of markers indicating local cells proliferation and migration suggests stimulation of endogenous reparative processes by HUCB D-0 cell interarterial infusion.
Mesenchymal stem cell (MSC) transplantation offers new therapeutic avenue for neurological diseases, however limitted survival of exogenous cells in the host brain is a major setback. The aim of this study was to evaluate the efficacy of using biodegradable scaffolds or Wharton jelly implants contained MSC after their transplantation into rat brain.Materials and methods: Adult Wistar rats were transplanted with MSC derived from human umbilical cord (hUC-MSC) (i), hUC-MSC localized inside biodegradable gelatin/laminin scaffolds (hUC-MSC/GL) (ii) or Wharton jelly implants (hUC-MSC/WJ) (iii). Results: hUC-MSC cultured in vitro expressed CD73, CD90, CD166 and Oct3/4, Nanog1, Nestin markers. 7 days after hUC-MSC transplantation only few viable donor cells were observed in the rat brain surrounded by heavy infiltration of macrophages/microglia (ED1+) and activating astrocytes (GFAP+). Hence transplantation of hUC-MSC/GL resulted with better cell survival compared with hUCMSC grafted in suspension. There was no donor cell migration out of the scaffolds however hUC-MSC lodged inside G/L scaffolds attained early neural markers. The inflammatory cell influx observed around the scaffolds was less intense with few ED1+ cells present in the core of scaffold. Similarly, all transplanted hUC-MSC/WJ remained in 3D tissue implants. Some of these cells adopted NF200, A2B5 or GFAP phenotypes during 7 days of observation. Concomitantly only scarce infiltration of immunoreactive cells was seen. Conclusions: Transplantation of hUC-MSC in 3D G/L scaffolds or hUC-MSC/WJ implants into adult rat brain improves survival of donor cells and induces their spontaneous transition into cell of neural lineage. It seems that 3D structures protect cells localized inside them from the host immune cells and may allow the diffusion of nutrients and other factors to propagate cell survival and differentiation into neuronal lineage. Supported by MSHE grant no N401 014235 and Fondation Jerome Lejeune grant.
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników
JavaScript jest wyłączony w Twojej przeglądarce internetowej. Włącz go, a następnie odśwież stronę, aby móc w pełni z niej korzystać.