Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników

Znaleziono wyników: 39

Liczba wyników na stronie
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 2 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 / 2 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników
1
Artykuł dostępny w postaci pełnego tekstu - kliknij by otworzyć plik
Content available

Stem cells and their outstanding concerns

100%
Stem cells have captured considerable scientific and clinic interest because of their potential to renew themselves and to differentiate into one or more adult cell types. Thus stem cells have been recognized as a potential tool for the development of innovative therapeutic strategies in different disease disorders. Stem cells can be discriminating based on their differentiated potential as totipotent, pluripotent, multipotent or unipotent cells. There are in general three types of stem cells: embryonic, fetal and adult stem cells. While embryonic stem cell therapy has a lot of ethical concerns due to their obtaining but also unlimited proliferation and uncontrolled differentiation, fetal and adult stem cells have been used in the treatment of different diseases. The bone marrow, peripheral blood and umbilical cord blood are ideal sources of adult stem cells because there are easily accessible and contain two types of stem cells: hematopoietic stem cells giving rise to all blood cell types and mesenchymal stem cells differentiating into cells of mesodermal lineage. This review describes the general characteristics of these stem cell populations and their current applications in regenerative medicine. Additionally induced pluripotent stem cells generated through the reprogramming of differentiated adult cells are described.
Ischemic stroke remains a major health problem associated with high mortality and severe morbidity. In spite of the extensive research in the field of stroke biology, there is little effective treatment for a completed disease onset. Numerous neuroprotective strategies have failed in clinical trials because of lack of efficacy or unacceptable side-effects. The challenge of clinical trial design is now to understand the process leading to ischemic brain injury and thus identify the targets for intervention in stroke. Therefore, uncovering cellular and molecular processes involved in ischemic brain injury is of critical importance. The review discusses the current understanding of these processes engaged in pathogenesis of stroke including excitotoxicity and inflammation. In addition recruitment of endogenous progenitors engaged in neurogenesis and vascular regeneration has been implicated. All of the aforementioned changes opted for therapeutic intervention to protect neurons in the region adjacent to the ischemic cerebral tissue and enhance cell recovery. Stem cell transplantation seems to offer a major promise of this therapy in stroke disorders.
Astrocytes are the main class of neuroglia, serving a wide range of adaptive functions in the mammalian nervous system. They interact with neurons, providing structural, metabolic and trophic support for them. In pathological circumstances, astrocytes have the potential to induce neuronal dysfunction, but they can also play a neuroprotective role, releasing neuronal growth factors. Here we review recent findings regarding the role of astrocytes in the biology of the brain in physiological conditions, as well as their reaction following the onset of neurodegenerative disorders.
Brain infl ammation contributes to the propagation of neuropathological events that involves activation of astrocytes and microglia. It remains obscure how activated glial cells affect the survival and differentiation of neural stem cells (NSC). The aim of the study was to analyze neuronal commitment of Human Umbilical Cord Blood derived Neural Stem Cells (HUCB-NSC) cultured in the presence of normal and LPS- or TMT-activated glial cells. Methods: HUCB-NSC (5 × 104/cm2 ) were co-cultured with normal or LPS (0.1 μg/ ml) and TMT (1μM)-stimulated astrocytes and microglial cells isolated from neonatal rat brain for proliferation and cell phenotype assessment. Pro-infl ammatory cytokines were estimated (ELISA). Results: Normal rat astrocytes induce HUCB-NSC to differentiate mostly into neurones but microglia stimulate HUCB-NSC to differentiate into neurons as well as into astrocytes. LPS- and TMTinduced astrocytes diminish neurogenesis of HUCB-NSC and increase astrocyte differentiation in comparison to non-stimulated astrocytes. Microglia activation by LPS and TMT decreases HUCBNSC differentiation into neurons but enhances oligodendrogenesis compared to normal microglia. Stimulation of astrocytes and microglia by LPS and TMT declines HUCB-NSC proliferation cocultured with astrocytes or with microglia. The presence of IL-1β, IL-6, TNF-α and NO was observed in glia cell culture supernatants after LPS and TMT implementation. Conclusion: Activation of astrocytes and microglia induced by LPS and TMT attenuate pro-neural effect of non-stimulated (resting) glia and suppress proliferation of HUCB-NSC in vitro. The release of pro-infl ammatory cytokines and NO might be partly responsible for this effect. Supported by MSHE grant No 142/P01/2008/35
Cerebrovascular diseases are the leading cause of severe disability worldwide, with an enormous financial burden for society. There is growing evidence that stem cell-based therapy may positively influence recovery from stroke. Cord blood is an attractive source of ontogenetically young, yet safe, stem cells. Conceptually, preclinical studies in which donor cells were of human origin have been the most valuable, and thus, it is likely that these cells will be used in clinical trials. Unfortunately, immunological barriers impede discordant xenotransplantations. We have previously observed acute rejection of cord blood- derived neural stem cells (HUCB-NSC) after transplantation to the brains of intact animals. Since it was reported recently that a brain lesion may actually improve the chances of graft survival, in this study, we used infarcted animals as graft recipients. In ongoing studies, we tested three immunosuppressive regimes: group I received cyclosporine A (CsA: 10 mg/ kg i.p.); group II received a triple-drug therapy (CsA: 10 mg/kg i.p., azathioprine: 5 mg/kg i.p., and methylprednisolone: 1.5 mg/kg i.m.); group III included rats that were formerly desensitized with HUCB, group IV had not undergone immunosuppression.. Animals were sacrificed at five time-points: 1, 3, 7, 14, and 21 days post-transplantation to evaluate graft survival and the time-course of immunological response. We observed a gradual decrease in the number of transplanted cells, with complete disappearance by day 14, surprisingly, with no difference among the experimental groups. The involvement of the innate immune system in the process of graft rejection dominated over an adaptive immunoresponse, with the highest activity on day 3, and subsequent fading of immune cell infiltration. In this work, we have shown that none of our immunosuppressive strategies proved adequate to prevent rejection of human stem cell grafts after transplantation into immunocompetent animals.
Potentially therapeutic neural stem cell line from human cord blood (HUCB-NSC) has been established in our laboratory. Reaching appropriate target by transplanted cells is a prerequisite for success of cell therapy for stroke. The question arises what is the migration potential of HUCB-NSC towards infarcted brain tissue. The migration of HUCB-NSC towards rat tissue homogenates from healthy brain (THHB) and ouabain-induced focal brain injury (THIB) obtained 6 h, 48 h and 7 days after insult was studied in vitro using transwells. Additionally the migratory activities of HUCB-NSC was checked in the presence of migration inducing proteins IGF-1 (200 ng/ml) and SDF-1 (10 ng/ml) dissolved in culture medium. Immunocytochemical analysis of migration-related receptors (CXCR-4, IGF-1R) on HUCB-NSC was performed. Results: Immunohistochemistry of HUCB-NSC unveiled expression of CXCR-4, IGF-1R. HUCB-NSC revealed robust migration toward THIB in comparison to THHB, which was most pronounced in the presence of 48 h postinfarct brain tissue (900 vs. 300 cells/well, P<0.05). The presence of IGF-1 and SDF-1 in medium increased signifi cantly HUCB-NSC migration but the effect was much weaker in comparison to injured brain tissue. The ability of robust in vitro migration of HUCB-NSC towards infarcted rat brain tissue has been confi rmed. Neither IGF-1 nor SDF-1 seems to play a pivotal role in this lesion-induced migration of HUCB-NSC. Supported by MSHE grant: N N401 332636.
Many types of neural progenitors from various sources have been evaluated for therapy of CNS disorders. Prerequisite for success in cell therapy is the ability for transplanted cells to reach appropriate target such as stroke lesion. We have established neural stem cell line from human umbilical cord blood neural stem (HUCB-NSC). In the present study we evaluated migratory capabilities of cells (HUCB-NSC) and the presence of various migration-related receptors. Immunocytochemical analysis revealed abundant expression of CXCR4, PDGFRa, PDGFRp, c-Met, VEGFR, IGF-1R and PSA-NCAM receptors in non-adherent population of HUCB-NSC cultured in serum free (SF) conditions (SF cells). Biological activity of selected receptors was confirmed by HUCB-NSC in vitro migration towards SDF-1 and IGF-1 ligands. Additionally, rat brain-derived homogenates have been assessed for their chemoattractive activity of HUCB-NSC. Our experiments unveiled that brain tissue was more attracted for HUCB-NSC than single ligands with higher potency of injured than intact brain. Moreover, adherent HUCB-NSC cultured in low serum (LS) conditions (LS cells) were employed to investigate an impact of different extracellular matrix (ECM) proteins on cell motility. It turned out that laminin provided most permissive microenvironment for cell migration, followed by fibronectin and gelatin. Unexpected nuclear localization of CXCR4 in SF cells prompted us to characterize intracellular pattern of this expression in relation to developmental stage of cells cultured in different conditions. Continuous culture of LS cells revealed cytoplasmatic pattern of CXCR4 expression while HUCB-NSC cultured in high serum conditions (HS cells) resulted in gradual translocation of CXCR4 from nucleus to cytoplasm and then to arising processes. Terminal differentiation of HUCB-NSC was followed by CXCR4 expression decline.
Focal brain damage following stroke leads to severe functional impairments. The aim of the study was to compare therapeutic effectiveness of intra-arterial infusion of HUCB-MNs at different stages of their neural conversion in vitro. Methods: Focal brain damage of dorsolateral striatum was induced in Wistar rats by stereotactic injection of previously established low dose of ouabain (1 μl, 5 mmol). Three days later 107 HUCB-MNs cells were infused (during 3 min) into carotid artery. Thirty days following surgery groups of 7ñ8 rats were housed in large enriched environment cages with various toys. Rats were behaviorally tested for 30 days after lesion. Results: Freshly isolated cells were much more effective in enhancing recovery from motor defi cits measured in walking beam task. Rats treated with HUCB-MNs cells presented also tendency to reduce turning bias and apomorphine induced rotations affected by unilateral lesion. This therapy enhances also recovery from impairments visible in object recognition task. However, rats treated with neurally directed HUCB-MNs also showed a signifi cant improvement in this task. The observed effects were much more prominent in T-maze habit learning task where cell treatment attenuated substantially lesion-induced learning defi cits. What interesting, the mechanism underlying this improvement seems to be different from this observed spontaneously in non-injured animals. Conclusions: Freshly isolated and neurally directed HUCB-MNs differently enhance recovery from distinct functional defi cits induced by focal brain damage. Non-cultured HUCB-MNs seems to be more effective in reducing motor defi cits. Neurally directed HUCB-MNs may be more potent in restoration of impaired habit learning processes. Supported by MSHE grant no 2PO5A05430
Stem cell therapy is a promising strategy for overcoming the limitations of current treatment methods. The modification of stem cell properties may be necessary to fully exploit their potential. Genetic engineering, with an abundance of methodology to induce gene expression in a precise and well-controllable manner, is particularly attractive for this purpose. There are virus-based and non-viral methods of genetic manipulation. Genome-integrating viral vectors are usually characterized by highly efficient and long-term transgene expression, at a cost of safety. Non-integrating viruses are also highly efficient in transduction, and, while safer, offer only a limited duration of transgene expression. There is a great diversity of transfectable forms of nucleic acids; however, for efficient shuttling across cell membranes, additional manipulation is required. Both physical and chemical methods have been employed for this purpose. Stem cell engineering for clinical applications is still in its infancy and requires further research. There are two main strategies for inducing transgene expression in therapeutic cells: transient and permanent expression. In many cases, including stem cell trafficking and using cell therapy for the treatment of rapid-onset disease with a short healing process, transient transgene expression may be a sufficient and optimal approach. For that purpose, mRNA-based methods seem ideally suited, as they are characterized by a rapid, highly efficient transfection, with outstanding safety. Permanent transgene expression is primarily based on the application of viral vectors, and, due to safety concerns, these methods are more challenging. There is active, ongoing research toward the development of non-viral methods that would induce permanent expression, such as transposons and mammalian artificial chromosomes.
The oligodendrocyte precursors exhibit many features of neural stem cells and constitute the abundant population of dividing progenitors in the young and adult brain. A question arises if their commitment and development could be modulated by either local tissue-specific or neuropathological signals. The aim of our study was to evaluate the effect of distinct microenvironments (provided by either the spinal cord or the hippocampal slices) on the differentiation of rat neonatal NG2 cells. The hippocampal slice cultures subjected to an ischemic injury (OGD) were used to mimic the traumatized tissue microenvironment. Both the hippocampal and spinal cord slice cultures were established from the same 7-day old rats. The model of an indirect contact (i.e. exclusively by the culture media) in co-culture system was chosen to eliminate the influence of cell-cell contact. The NG2 cells were obtained from 10-day old mixed primary culture of neonatal rat hemispheres. After 7 days in co-culture, the cells were either stained with neural markers or collected for the RNA isolation and real-time PCR. The medium conditioned by hippocampal slices effectively promoted neurogenesis: ~30% of NG2 cells differentiated into TUJ 1-positive neurons. The remaining fraction mostly formed premyelinating and mature oligodendrocytes. The exposition of hippocampal slices to the OGD injury abolished the effect of pro-neuronal induction in cocultures. In media conditioned by spinal cord slices, neurogenesis was less pronounced (20% neurons) and the oligodendrocyte differentiation was significantly slowed-down. The NG2 precursors have the intrinsic potency for neurogenesis. Heterogeneity of local microenviroment might modify the fate of endogenous or transplanted NG2 cells what should be taken into consideration in potential neurorepair strategies. Supported by grant 0345/B/P01/2010/38.
Searching for a reliable source of alternative neural stem cells for experimental treatment of neurological disorders we have established neural stem cell line derived from human umbilical cord blood (HUCB-NSC) (Buzanska et al. 2006). These cells have been shown to differentiate along neuronal and glial lineages in the promoting in vitro conditions. In the current study we transplanted HUCB-NSC into rat brain to determine whether the neural progenitors would be able to survive, migrate and eventually adopt neural phenotypes after exposure to central nervous system (CNS) microenvironment. Our experiments revealed that HUCB-NSC grafting into the brain of adult rats limited their survival up-to two weeks probably due to their elimination by severe immunological host reaction evoked by xenotransplantation. HUCB-NSC graft in neonates survived longer time in rat brain, migrated, proliferated and differentiated into neuronal cells however their presence in the host tissue did not exceed more than five weeks after transplantation.
The NG2-positive cells are the oligodendrocyte precursors, which, when terminally differentiated, are capable of myelinating the central nervous system. There is however an ever-growing list of evidences that NG2 cells actually possess an intrinsic neurogenic potential and they are capable of neuronal differentiation in response to environmental stimuli. To address the question, we have established a model of an indirect co-culture system of the freshly isolated rat neonatal NG2 cells and organotypic slices derived from two distinct CNS regions (hippocampus and spinal cord) to mimic the nervous tissue microenviroment. The cell differentiation in microenvironment of OGD-injured hippocampal slices has been studied as well. The molecular analysis of selected trophic factors has been performed to determine the patterns of their expression. Indeed, the comparison of the cell commitment and development in various microenvironments has pointed to significant dissimilarities. First of all, the medium being continuously conditioned by the hippocampal slices efficiently promoted neurogenesis. The effect has been significantly abolished in co-cultures with the injured tissue. The less pronounced susceptibility to adopting neuronal phenotype and the considerable slowdown of oligodendroglial development was observed in the co-cultures with the spinal cord slices. The role of BDNF in oligodendroglial progenitor commitment and development has been investigated proving that it is one of the key players in the examined processes. The specificity of the instructive clues cocktail might module the fate choice of mobilized endogenous or transplanted cells, which should be taken into consideration while planning neurorepair strategies.
Studies in experimental stroke demonstrate that cerebral ischemic injury promotes neurogenesis in the subventricular zone (SVZ) and subgranular zone (SGZ) of the dentate gyrus. Spontaneously occurring injury-induced neurogenesis is insufficient to fully reverse disease pathophysiology. Exogenous neural progenitors transplanted into damaged brain might be useful for facilitating the repair of damaged tissue by instructing several endogenous processes. However, the molecular and cellular mechanisms stimulating cell proliferation and mediating the migration of arising neuroblasts towards the ischemic boundary still remain to be characterized. Building evidence suggests that matrix metalloproteinases (MMPs) seem to play a role in neurogenesis-associated processes, providing an environment which may be instructive or permissive to stem cells activation. The overall goal of our present studies was to examine whether HUCB-NSC transplantation modulates migration of endogenous progenitor cells and MMPs activity in adult rat brain after focal ischemia. Methods: 2x104 neural stem cells from human cord blood (HUCB-NSC) were transplanted into corpus callosum of naive or focally injured (induced by 1µl/50nmol OUA injection) rat brain. At 1, 3, 7 and 14 days rat brains were removed. Then immunocytochemical analysis of doublecortin (DCX) (marker expressed by immature migratory neuroblasts) and in situ zymography of MMPs activity was performed. Results: OUA-induced brain lesion resulted in increase of DCX+ cells in SVZ and SGZ in comparison to intact rats. This response has been potentiated by HUCBNSC transplantation. Moreover, the activation of MMPs in cells was visible in SVZ. At 7th day after HUCB-NSC transplantation the intense migration of DCX+ cells from SVZ towards ischemic boundary regions of the striatum was observed. 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. Conclusions: 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 localization of MMPs in cell nuclei implies the involvement of these proteases in proteolytical activation of pro-neural gene transcription. Supported by MSHE grant no N401 014235.
Adult bone marrow-derived mesenchymal stem cells (hMSCs) display a spectrum of functional properties. Transplantation of these cells improves the clinical outcome in experimental models of cerebral ischemia and spinal cord injury. Therapeutic effects have been reported in stroke after the systemic delivery of MSCs. A minimally invasive, intraarterial route is an attractive method for stem cell transplantation to the injured brain. However, MSCs lack the intrinsic mechanisms that enable homing of the cells to the area of infarction. Recent studies suggest that genetic manipulation can promote the forced expression of certain molecules responsible for adhesion and transendothelial migration of systemically delivered cells. It is anticipated that, for cell homing to the brain after intra-arterial delivery, the transient expression of integrins should be sufficient for diapedesis to occur. Since the capacity of MSCs to undergo functional transfection using pDNA is very low, we investigated an mRNA transfection method for the expression of transgenes in MSCs in order to overcome the limitations of the pDNA approach. Methods: Human mesenchymal stem cells (hMSC, PT-2501, Lonza) were thawed and cultured in medium MSCBM (PT-3238, Lonza) supplemented with 10% MCGS (PT-4106E, Lonza), L-glutamine (PT4107E, Lonza), and gentamicin sulfate (GA-1000, PT-4504E, Lonza). Cells were maintained in a humidified atmosphere at 37°C and 5% CO2 using 75 cm2 flasks. For transfection experiments, hMSCs were transferred to 24-well plates and seeded at a density of 15 000 cells/well. For transgene induction experiments, pDNA-eGFP (BD Biosciences) at a dose of 0.5 and 1.0 µg/well, and mRNA-eGFP (StemGent) at doses of 0.12, 0.25, and 0.5 µg/well were used. The Lipofectamine® 2000 (Invitrogen), TransIT-2020 (Mirus), and StemfectTM RNA Transfection Kit (StemGent) were used as transfection agents. After transfection, cells were maintained in culture conditions up to 21 days. Transfection efficiency was assessed by confocal microscopy using GFP fluorescent signal detection. Results: MSC pDNA-eGFP transfection results in a dramatically low efficiency, less than 1% of the cell population in each of the tested conditions. In contrast, mRNA-eGFP transfection resulted in an efficiency exceeding 95% in each of the tested conditions. This difference was highly statistically significant (P<0.001). Furthermore, cellular GFP level, and the persistence of transfection was dependent on the mRNA dose and the type of transfection agent. It was found that the dose of mRNA-eGFP 0.5 µg/well and the use of Lipofectamine was the most effective method with transgene expression up to three weeks. Conclusions: The mRNA transfection is a robust, clinically applicable tool for inducing the transient expression of transgenes in hMSCs, which are otherwise difficult to transfect by vectors that do not incorporate into the host genome. Using this method, application of engineered MSC could revolutionize regenerative medicine. Supported by a National Centre for Research and Development grant No 101 in ERA-NET NEURON project: “MEMS-IRBI”
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 2 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.