Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
PeerJ ; 11: e16358, 2023.
Article En | MEDLINE | ID: mdl-38025691

Background: Cell therapy using neural progenitor cells (NPCs) is a promising approach for ischemic stroke treatment according to the results of multiple preclinical studies in animal stroke models. In the vast majority of conducted animal studies, the therapeutic efficacy of NPCs was estimated after intracerebral transplantation, while the information of the effectiveness of systemic administration is limited. Nowadays, several clinical trials aimed to estimate the safety and efficacy of NPCs transplantation in stroke patients were also conducted. In these studies, NPCs were transplanted intracerebrally in the subacute/chronic phase of stroke. The results of clinical trials confirmed the safety of the approach, however, the degree of functional improvement (the primary efficacy endpoint) was not sufficient in the majority of the studies. Therefore, more studies are needed in order to investigate the optimal transplantation parameters, especially the timing of cell transplantation after the stroke onset. This study aimed to evaluate the therapeutic effects of intra-arterial (IA) and intravenous (IV) administration of NPCs derived from induced pluripotent stem cells (iNPCs) in the acute phase of experimental stroke in rats. Induced pluripotent stem cells were chosen as the source of NPCs as this technology is perspective, has no ethical concerns and provides the access to personalized medicine. Methods: Human iNPCs were transplanted IA or IV into male Wistar rats 24 h after the middle cerebral artery occlusion stroke modeling. Therapeutic efficacy was monitored for 14 days and evaluated in comparison with the cell transplantation-free control group. Additionally, cell distribution in the brain was assessed. Results: The obtained results show that both routes of systemic transplantation (IV and IA) significantly reduced the mortality and improved the neurological deficit of experimental animals compared to the control group. At the same time, according to the MRI data, only IA administration led to faster and prominent reduction of the stroke volume. After IA administration, iNPCs transiently trapped in the brain and were not detected on day 7 after the transplantation. In case of IV injection, transplanted cells were not visualized in the brain. The obtained data demonstrated that the systemic transplantation of human iNPCs in the acute phase of ischemic stroke can be a promising therapeutic strategy.


Induced Pluripotent Stem Cells , Ischemic Stroke , Neural Stem Cells , Stroke , Humans , Rats , Male , Animals , Rats, Wistar , Stroke/therapy , Neural Stem Cells/transplantation , Infarction, Middle Cerebral Artery/therapy
2.
Life (Basel) ; 13(2)2023 Jan 20.
Article En | MEDLINE | ID: mdl-36836645

Systemic transplantation of mesenchymal stem cells (MSCs) is a promising approach for the treatment of ischemia-associated disorders, including stroke. However, exact mechanisms underlying its beneficial effects are still debated. In this respect, studies of the transplanted cells distribution and homing are indispensable. We proposed an MRI protocol which allowed us to estimate the dynamic distribution of single superparamagnetic iron oxide labeled MSCs in live ischemic rat brain during intravenous transplantation after the transient middle cerebral artery occlusion. Additionally, we evaluated therapeutic efficacy of cell therapy in this rat stroke model. According to the dynamic MRI data, limited numbers of MSCs accumulated diffusely in the brain vessels starting at the 7th minute from the onset of infusion, reached its maximum by 29 min, and gradually eliminated from cerebral circulation during 24 h. Despite low numbers of cells entering brain blood flow and their short-term engraftment, MSCs transplantation induced long lasting improvement of the neurological deficit, but without acceleration of the stroke volume reduction compared to the control animals during 14 post-transplantation days. Taken together, these findings indicate that MSCs convey their positive action by triggering certain paracrine mechanisms or cell-cell interactions or invoking direct long-lasting effects on brain vessels.

3.
Polymers (Basel) ; 14(19)2022 Sep 26.
Article En | MEDLINE | ID: mdl-36235965

Restoring the anatomical and functional characteristics of the cornea using various biomaterials is especially relevant in the context of a global shortage of donor tissue. Such biomaterials must be biocompatible, strong, and transparent. Here, we report a Viscoll collagen membrane with mechanical and optical properties suitable for replacing damaged stromal tissue. After removing a portion of the stroma, a Viscoll collagen membrane was implanted into the corneas of rabbits. After 6 months, the active migration of host cells into Viscoll collagen membranes was noted, with the preservation of corneal transparency in all experimental animals. Effective integration of the Viscoll collagen membrane with corneal tissue promoted nerve regeneration in vivo, as confirmed by in vivo confocal microscopy. We also demonstrated the safety and efficacy of the Viscoll collagen membrane for corneal stroma regeneration. Thus, in combination with the proposed packaging format that provides long-term storage of up to 10 months, this material has great potential for replacing and regenerating damaged stromal tissues.

4.
Biomedicines ; 10(2)2022 Feb 01.
Article En | MEDLINE | ID: mdl-35203560

Intra-arterial (IA) mesenchymal stem cells (MSCs) transplantation providing targeted cell delivery to brain tissue is a promising approach to the treatment of neurological disorders, including stroke. Factors determining cell distribution after IA administration have not been fully elucidated. Their decoding may contribute to the improvement of a transplantation technique and facilitate translation of stroke cell therapy into clinical practice. The goal of this work was to quantitatively assess the impact of brain tissue perfusion on the distribution of IA transplanted MSCs in rat brains. We performed a selective MR-perfusion study with bolus IA injection of gadolinium-based contrast agent and subsequent IA transplantation of MSCs in intact rats and rats with experimental stroke and evaluated the correlation between different perfusion parameters and cell distribution estimated by susceptibility weighted imaging (SWI) immediately after cell transplantation. The obtained results revealed a certain correlation between the distribution of IA transplanted MSCs and brain perfusion in both intact rats and rats with experimental stroke with the coefficient of determination up to 30%. It can be concluded that the distribution of MSCs after IA injection can be partially predicted based on cerebral perfusion data, but other factors requiring further investigation also have a significant impact on the fate of transplanted cells.

5.
Cells ; 10(11)2021 11 03.
Article En | MEDLINE | ID: mdl-34831220

Animal model studies and first clinical trials have demonstrated the safety and efficacy of the mesenchymal stem cells' (MSCs) transplantation in stroke. Intra-arterial (IA) administration looks especially promising, since it provides targeted cell delivery to the ischemic brain, is highly effective, and can be safe as long as the infusion is conducted appropriately. However, wider clinical application of the IA MSCs transplantation will only be possible after a better understanding of the mechanism of their therapeutic action is achieved. On the way to achieve this goal, the study of transplanted cells' fate and their interactions with the blood-brain barrier (BBB) structures could be one of the key factors. In this review, we analyze the available data concerning one of the most important aspects of the transplanted MSCs' action-the ability of cells to cross the blood-brain barrier (BBB) in vitro and in vivo after IA administration into animals with experimental stroke. The collected data show that some of the transplanted MSCs temporarily attach to the walls of the cerebral vessels and then return to the bloodstream or penetrate the BBB and either undergo homing in the perivascular space or penetrate deeper into the parenchyma. Transmigration across the BBB is not necessary for the induction of therapeutic effects, which can be incited through a paracrine mechanism even by cells located inside the blood vessels.


Blood-Brain Barrier/pathology , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Stroke/therapy , Animals , Clinical Trials as Topic , Humans , Injections, Intra-Arterial
6.
Front Neurosci ; 15: 641970, 2021.
Article En | MEDLINE | ID: mdl-33737862

Cell therapy is an emerging approach to stroke treatment with a potential to limit brain damage and enhance its restoration after the acute phase of the disease. In this study we tested directly reprogrammed neural precursor cells (drNPC) derived from adult human bone marrow cells in the rat middle cerebral artery occlusion (MCAO) model of acute ischemic stroke using human placenta mesenchymal stem cells (pMSC) as a positive control with previously confirmed efficacy. Cells were infused into the ipsilateral (right) internal carotid artery of male Wistar rats 24 h after MCAO. The main goal of this work was to evaluate real-time distribution and subsequent homing of transplanted cells in the brain. This was achieved by performing intra-arterial infusion directly inside the MRI scanner and allowed transplanted cells tracing starting from their first pass through the brain vessels. Immediately after transplantation, cells were observed in the periphery of the infarct zone and in the brain stem, 15 min later small numbers of cells could be discovered deep in the infarct core and in the contralateral hemisphere, where drNPC were seen earlier and in greater numbers than pMSC. Transplanted cells in both groups could no longer be detected in the rat brain 48-72 h after infusion. Histological and histochemical analysis demonstrated that both the drNPC and pMSC were localized inside blood vessels in close contact with the vascular wall. No passage of labeled cells through the blood brain barrier was observed. Additionally, the therapeutic effects of drNPC and pMSC were compared. Both drNPC and pMSC induced substantial attenuation of neurological deficits evaluated at the 7th and 14th day after transplantation using the modified neurological severity score (mNSS). Some of the effects of drNPC and pMSC, such as the influence on the infarct volume and the survival rate of animals, differed. The results suggest a paracrine mechanism of the positive therapeutic effects of IA drNPC and pMSC infusion, potentially enhanced by the cell-cell interactions. Our data also indicate that the long-term homing of transplanted cells in the brain is not necessary for the brain's functional recovery.

7.
Biomedicines ; 8(12)2020 Dec 15.
Article En | MEDLINE | ID: mdl-33333803

Cell therapy of neurological diseases is gaining momentum. Various types of stem/progenitor cells and their derivatives have shown positive therapeutic results in animal models of neurological disorders and in clinical trials. Each tested cell type proved to have its advantages and flaws and unique cellular and molecular mechanism of action, prompting the idea to test combined transplantation of two or more types of cells (combined cell therapy). This review summarizes the results of combined cell therapy of neurological pathologies reported up to this point. The number of papers describing experimental studies or clinical trials addressing this subject is still limited. However, its successful application to the treatment of neurological pathologies including stroke, spinal cord injury, neurodegenerative diseases, Duchenne muscular dystrophy, and retinal degeneration has been reported in both experimental and clinical studies. The advantages of combined cell therapy can be realized by simple summation of beneficial effects of different cells. Alternatively, one kind of cells can support the survival and functioning of the other by enhancing the formation of optimum environment or immunomodulation. No significant adverse events were reported. Combined cell therapy is a promising approach for the treatment of neurological disorders, but further research needs to be conducted.

8.
Plast Reconstr Surg Glob Open ; 8(2): e2610, 2020 Feb.
Article En | MEDLINE | ID: mdl-32309071

BACKGROUND: The regeneration of the peripheral nerves after injuries is still a challenging fundamental and clinical problem. The cell therapy and nerve guide conduit construction are promising modern approaches. Nowadays, different sources of cells for transplantation are available. But it is little known about the interaction between fetal central nervous system cells and peripheral nerve tissue. In this study, we analyzed the development of the fetal neocortex and spinal cord solid grafts injected into the gelatin hydrogel conduits and their effects on sciatic nerve regeneration after cut injury. METHODS: Frontal neocortex tissue was obtained from E19.5 and spinal cord tissue was obtained from E14.5 fetuses harvested from transgenic EGFP mice. The grafts were injected into the hydrogel conduits which were connected to the nerve stumps after cut injury. The recovery of motor function was estimated with walking track analysis at 2, 5, and 8 weeks after surgery. Then immunohistochemical study was performed. RESULTS: The histological examination showed that only fetal neocortex solid graft cells had survived after implantation. Immunostaining revealed that some of the transplanted cells expressed neural markers such as neurofilament protein and NeuN. But the cells mostly differentiated in glial lineage, which was confirmed with immunostaining for GFAP and S100ß. The walking-track analysis has shown that 8 weeks after surgery bioengineered conduit differed significantly from the control. CONCLUSIONS: We revealed that the hydrogel conduit is suitable for nerve re-growth and that the fetal neocortex grafted cells can survive and differentiate. Bioengineered conduit can stimulate functional recovery after the nerve injury.

9.
Tissue Cell ; 54: 94-104, 2018 Oct.
Article En | MEDLINE | ID: mdl-30309515

Neural crest stem cells that located in the postnatal hair follicle (HF-NCSC) are considered a promising tool for treatment of nervous system diseases and injuries. It is well known that HF-NCSC can be used in the spinal cord and sciatic nerve reparation but their ability to restore brain structures is poorly studied. In this article we are investigating the interaction between HF-NCSC and a nerve tissue (embryonic and adult). We have found out that HF-NCSC isolated from adult mice grow and differentiate in accordance with the mouse embryo developmental stage when co-cultured with the embryonic nerve tissue. The HF-NCSC migration is slower in the late embryonic tissue co-culture system compared to the early one. This phenomenon is related to the motor function of the cells but not to their proliferation level. We have demonstrated that the embryonic nerve tissue maintains HF-NCSC an undifferentiated status, while an adult brain tissue inhibits the cell proliferation and activates the differentiation processes. Besides, HF-NCSC pre-differentiated into the neuronal direction shows a higher survival and migration rate after the transplantation into the adult brain tissue compared to the undifferentiated HF-NCSC. Thus, we have investigated the postnatal HF-NCSC response to the nerve tissue microenvironment to analyze their possible application to the brain repair processes.


Cell Differentiation/physiology , Hair Follicle/cytology , Neural Stem Cells/cytology , Stem Cell Transplantation/methods , Animals , Brain/cytology , Cell Proliferation/physiology , Coculture Techniques , Embryo, Mammalian , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/transplantation , Neural Crest/cytology , Neural Stem Cells/transplantation , Organ Culture Techniques , Spinal Cord/cytology
10.
PLoS One ; 12(10): e0186717, 2017.
Article En | MEDLINE | ID: mdl-29049361

In vivo tracking of transplanted mesenchymal stem cells (MSCs) migration and homing is vital for understanding the mechanisms of beneficial effects of MSCs transplantation in animal models of diseases and in clinical trials. Transplanted cells can be labeled with superparamagnetic iron oxide (SPIO) particles and visualized in vivo using a number of iron sensitive MRI techniques. However, the applicability of those techniques for SPIO-labeled MSCs tracking in live brain has not been sufficiently investigated. The goal of this study was to estimate the efficiency of various MRI techniques of SPIO-labeled cell tracing in the brain. To achieve that goal, the precision and specificity of T2WI, T2*WI and SWI (Susceptibility-Weighted Imaging) techniques of SPIO-labeled MSCs tracing in vitro and in live rat brain were for the first time compared in the same experiment. We have shown that SWI presents the most sensitive pulse sequence for SPIO-labeled MSCs MR visualization. After intracerebral administration due to limitations caused by local micro-hemorrhages the visualization threshold was 102 cells, while after intra-arterial transplantation SWI permitted detection of several cells or even single cells. There is just one publication claiming detection of individual SPIO-labeled MSCs in live brain, while the other state much lower sensitivity, describe detection of different cell types or high resolution tracing of MSCs in other tissues. This study confirms the possibility of single cell tracing in live brain and outlines the necessary conditions. SWI is a method convenient for the detection of single SPIO labeled MSCs and small groups of SPIO labeled MSCs in brain tissue and can be appropriate for monitoring migration and homing of transplanted cells in basic and translational neuroscience.


Cell Transplantation , Corpus Striatum , Ferric Compounds/chemistry , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Proliferation , Cell Survival , Humans , Phantoms, Imaging , Rats
...