Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Folia Neuropathol ; 62(1): 32-46, 2024.
Article in English | MEDLINE | ID: mdl-38741435

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a potential source of somatic cells for cell therapies due to their ability to self-renew and differentiate into various cells of the body. To date, the clinical application of hiPSCs has been limited due to safety issues. The present study aims to standardize the safety procedure of the derivation of GMP-compliant induced pluripotent stem cell (iPSC) lines from human fibroblasts. The hiPSC lines were generated using the nonintegrative Sendai virus method to incorporate Yamanaka reprogramming factors (OCT3/4, SOX2, KLF4 and c-MYC) into cells. A constant temperature was maintained during the cell culture, including all stages of the culture after transduction with Sendai virus. Pluripotency was proved in six independently generated hiPSC lines from adult female (47 years old) and male (57 years old) donors' derived fibroblasts via alkaline phosphatase live (ALP) staining, qPCR, and immunocytochemistry. The hiPSC lines showed a gradual decrease in the presence of the virus with each subsequent passage, and this reduction was specific to the hiPSC line. The frequency and probability of chromosomal aberrations in hiPSCs were dependent on both the iPSC clone identity and sex of the donor. In summary, the generation of hiPSC for clinical applications requires safety standards application (biosafety protocol, quality control of hiPSC lines, viral and genetic integrity screening) from the first stages of the clonal selection of hiPSC from the same donor.


Subject(s)
Induced Pluripotent Stem Cells , Kruppel-Like Factor 4 , Sendai virus , Humans , Female , Male , Middle Aged , Cell Line , Fibroblasts , Cell Differentiation/physiology , Transduction, Genetic/methods , Sex Factors
2.
Cells ; 12(3)2023 01 21.
Article in English | MEDLINE | ID: mdl-36766732

ABSTRACT

The formation of embryoid bodies (EBs) from human pluripotent stem cells resembles the early stages of human embryo development, mimicking the organization of three germ layers. In our study, EBs were tested for their vulnerability to chronic exposure to low doses of MeHgCl (1 nM) under atmospheric (21%O2) and physioxia (5%O2) conditions. Significant differences were observed in the relative expression of genes associated with DNA repair and mitophagy between the tested oxygen conditions in nontreated EBs. When compared to physioxia conditions, the significant differences recorded in EBs cultured at 21% O2 included: (1) lower expression of genes associated with DNA repair (ATM, OGG1, PARP1, POLG1) and mitophagy (PARK2); (2) higher level of mtDNA copy number; and (3) higher expression of the neuroectodermal gene (NES). Chronic exposure to a low dose of MeHgCl (1 nM) disrupted the development of EBs under both oxygen conditions. However, only EBs exposed to MeHgCl at 21% O2 revealed downregulation of mtDNA copy number, increased oxidative DNA damage and DNA fragmentation, as well as disturbances in SOX17 (endoderm) and TBXT (mesoderm) genes expression. Our data revealed that physioxia conditions protected EBs genome integrity and their further differentiation.


Subject(s)
Embryoid Bodies , Mitophagy , Humans , Mitophagy/genetics , DNA Repair , Oxygen/pharmacology , Oxygen/metabolism
3.
Cells ; 12(2)2023 01 16.
Article in English | MEDLINE | ID: mdl-36672274

ABSTRACT

Dravet syndrome (DRVT) is a rare form of neurodevelopmental disorder with a high risk of sudden unexpected death in epilepsy (SUDEP), caused mainly (>80% cases) by mutations in the SCN1A gene, coding the Nav1.1 protein (alfa-subunit of voltage-sensitive sodium channel). Mutations in SCN1A are linked to heterogenous epileptic phenotypes of various types, severity, and patient prognosis. Here we generated iPSC lines from fibroblasts obtained from three individuals affected with DRVT carrying distinct mutations in the SCN1A gene (nonsense mutation p.Ser1516*, missense mutation p.Arg1596His, and splicing mutation c.2589+2dupT). The iPSC lines, generated with the non-integrative approach, retained the distinct SCN1A gene mutation of the donor fibroblasts and were characterized by confirming the expression of the pluripotency markers, the three-germ layer differentiation potential, the absence of exogenous vector expression, and a normal karyotype. The generated iPSC lines were used to establish ventral forebrain organoids, the most affected type of neurons in the pathology of DRVT. The DRVT organoid model will provide an additional resource for deciphering the pathology behind Nav1.1 haploinsufficiency and drug screening to remediate the functional deficits associated with the disease.


Subject(s)
Epilepsies, Myoclonic , Induced Pluripotent Stem Cells , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Induced Pluripotent Stem Cells/metabolism , Epilepsies, Myoclonic/genetics , Neurons/metabolism , Prosencephalon/metabolism
5.
Cells ; 11(20)2022 10 14.
Article in English | MEDLINE | ID: mdl-36291101

ABSTRACT

The hippocampus is one of the few privileged regions (neural stem cell niche) of the brain, where neural stem cells differentiate into new neurons throughout adulthood. However, dysregulation of hippocampal neurogenesis with aging, injury, depression and neurodegenerative disease leads to debilitating cognitive impacts. These debilitating symptoms deteriorate the quality of life in the afflicted individuals. Impaired hippocampal neurogenesis is especially difficult to rescue with increasing age and neurodegeneration. However, the potential to boost endogenous Wnt signaling by influencing pathway modulators such as receptors, agonists, and antagonists through drug and cell therapy-based interventions offers hope. Restoration and augmentation of hampered Wnt signaling to facilitate increased hippocampal neurogenesis would serve as an endogenous repair mechanism and contribute to hippocampal structural and functional plasticity. This review focuses on the possible interaction between neurogenesis and Wnt signaling under the control of antidepressants and mesenchymal stem cells (MSCs) to overcome debilitating symptoms caused by age, diseases, or environmental factors such as stress. It will also address some current limitations hindering the direct extrapolation of research from animal models to human application, and the technical challenges associated with the MSCs and their cellular products as potential therapeutic solutions.


Subject(s)
Mesenchymal Stem Cells , Neurodegenerative Diseases , Animals , Adult , Humans , Neurodegenerative Diseases/metabolism , Quality of Life , Neurogenesis/physiology , Hippocampus/metabolism , Mesenchymal Stem Cells/metabolism , Antidepressive Agents/pharmacology
6.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055039

ABSTRACT

From the first success in cultivation of cells in vitro, it became clear that developing cell and/or tissue specific cultures would open a myriad of new opportunities for medical research. Expertise in various in vitro models has been developing over decades, so nowadays we benefit from highly specific in vitro systems imitating every organ of the human body. Moreover, obtaining sufficient number of standardized cells allows for cell transplantation approach with the goal of improving the regeneration of injured/disease affected tissue. However, different cell types bring different needs and place various types of hurdles on the path of regenerative neurology and regenerative cardiology. In this review, written by European experts gathered in Cost European action dedicated to neurology and cardiology-Bioneca, we present the experience acquired by working on two rather different organs: the brain and the heart. When taken into account that diseases of these two organs, mostly ischemic in their nature (stroke and heart infarction), bring by far the largest burden of the medical systems around Europe, it is not surprising that in vitro models of nervous and heart muscle tissue were in the focus of biomedical research in the last decades. In this review we describe and discuss hurdles which still impair further progress of regenerative neurology and cardiology and we detect those ones which are common to both fields and some, which are field-specific. With the goal to elucidate strategies which might be shared between regenerative neurology and cardiology we discuss methodological solutions which can help each of the fields to accelerate their development.


Subject(s)
Guided Tissue Regeneration , Myocardium , Nerve Regeneration , Regenerative Medicine , Animals , Brain/anatomy & histology , Brain/metabolism , Brain Diseases/diagnosis , Brain Diseases/etiology , Brain Diseases/therapy , Cell Differentiation , Cell- and Tissue-Based Therapy , Disease Management , Extracellular Vesicles/metabolism , Guided Tissue Regeneration/methods , Heart Diseases/diagnosis , Heart Diseases/etiology , Heart Diseases/therapy , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Organoids , Regenerative Medicine/methods , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Stem Cells/cytology , Stem Cells/metabolism
7.
Cells ; 10(4)2021 03 24.
Article in English | MEDLINE | ID: mdl-33804841

ABSTRACT

To optimise the culture conditions for human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) intended for clinical use, we investigated ten different properties of these cells cultured under 21% (atmospheric) and 5% (physiological normoxia) oxygen concentrations. The obtained results indicate that 5% O2 has beneficial effects on the proliferation rate, clonogenicity, and slowdown of senescence of hWJ-MSCs; however, the oxygen level did not have an influence on the cell morphology, immunophenotype, or neuroprotective effect of the hWJ-MSCs. Nonetheless, the potential to differentiate into adipocytes, osteocytes, and chondrocytes was comparable under both oxygen conditions. However, spontaneous differentiation of hWJ-MSCs into neuronal lineages was observed and enhanced under atmospheric oxygen conditions. The cells relied more on mitochondrial respiration than glycolysis, regardless of the oxygen conditions. Based on these results, we can conclude that hWJ-MSCs could be effectively cultured and prepared under both oxygen conditions for cell-based therapy. However, the 5% oxygen level seemed to create a more balanced and appropriate environment for hWJ-MSCs.


Subject(s)
Mesenchymal Stem Cells/cytology , Neuroprotection , Oxygen/pharmacology , Wharton Jelly/cytology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Clone Cells , Humans , Immunophenotyping , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents
8.
Dev Neurobiol ; 81(5): 591-607, 2021 07.
Article in English | MEDLINE | ID: mdl-33725382

ABSTRACT

Mitochondria are cellular organelles involved in generating energy to power various processes in the cell. Although the pivotal role of mitochondria in neurogenesis was demonstrated (first in animal models), very little is known about their role in human embryonic neurodevelopment and its pathology. In this respect human-induced pluripotent stem cells (hiPSC)-derived cerebral organoids provide a tractable, alternative model system of the early neural development and disease that is responsive to pharmacological and genetic manipulations, not possible to apply in humans. Although the involvement of mitochondria in the pathogenesis and progression of neurodegenerative diseases and brain dysfunction has been demonstrated, the precise role they play in cell life and death remains unknown, compromising the development of new mitochondria-targeted approaches to treat human diseases. The cerebral organoid model of neurogenesis and disease in vitro provides an unprecedented opportunity to answer some of the most fundamental questions about mitochondrial function in early human neurodevelopment and neural pathology. Largely an unexplored territory due to the lack of tools and approaches, this review focuses on recent technological advancements in fluorescent and molecular tools, imaging systems, and computational approaches for quantitative and qualitative analyses of mitochondrial structure and function in three-dimensional cellular assemblies-cerebral organoids. Future developments in this direction will further facilitate our understanding of the important role or mitochondrial dynamics and energy requirements during early embryonic development. This in turn will provide a further understanding of how dysfunctional mitochondria contribute to disease processes.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Animals , Female , Humans , Mitochondria , Neurodegenerative Diseases/metabolism , Neurogenesis , Organoids/metabolism , Pregnancy
9.
Cells ; 11(1)2021 12 30.
Article in English | MEDLINE | ID: mdl-35011673

ABSTRACT

The peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a well-known transcriptional coactivator involved in mitochondrial biogenesis. PGC-1α is implicated in the pathophysiology of many neurodegenerative disorders; therefore, a deep understanding of its functioning in the nervous system may lead to the development of new therapeutic strategies. The central nervous system (CNS)-specific isoforms of PGC-1α have been recently identified, and many functions of PGC-1α are assigned to the particular cell types of the central nervous system. In the mice CNS, deficiency of PGC-1α disturbed viability and functioning of interneurons and dopaminergic neurons, followed by alterations in inhibitory signaling and behavioral dysfunction. Furthermore, in the ALS rodent model, PGC-1α protects upper motoneurons from neurodegeneration. PGC-1α is engaged in the generation of neuromuscular junctions by lower motoneurons, protection of photoreceptors, and reduction in oxidative stress in sensory neurons. Furthermore, in the glial cells, PGC-1α is essential for the maturation and proliferation of astrocytes, myelination by oligodendrocytes, and mitophagy and autophagy of microglia. PGC-1α is also necessary for synaptogenesis in the developing brain and the generation and maintenance of synapses in postnatal life. This review provides an outlook of recent studies on the role of PGC-1α in various cells in the central nervous system.


Subject(s)
Central Nervous System/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Humans , Organelle Biogenesis
10.
Sci Rep ; 10(1): 16946, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037314

ABSTRACT

Tuning stem cells microenvironment in vitro may influence their regenerative properties. In this study Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) were encapsulated in 3D hydrogels derived from human fibrin (FB) or platelet lysate (PL) and the oxygen level was adjusted to physiological normoxia (5% O2). The influence of the type of the scaffold and physiological normoxia conditions was tested on the WJ-MSCs' survivability, proliferation, migratory potential, the level of expression of selected trophic factors, cytokines, and neural markers. Encapsulated WJ-MSCs revealed high survivability, stable proliferation rate, and ability to migrate out of the hydrogel and the up-regulated expression of all tested factors, as well as the increased expression of neural differentiation markers. Physiological normoxia stimulated proliferation of encapsulated WJ-MSCs and significantly enhanced their neuronal, but not glial, differentiation. Ex vivo studies with indirect co-culture of organotypic hippocampal slices and cell-hydrogel bio-constructs revealed strong neuroprotective effect of WJ-MSCs against neuronal death in the CA1 region of the rat hippocampus. This effect was potentiated further by FB scaffolds under 5% O2 conditions. Our results indicating significant effect of oxygen and 3D cytoarchitecture suggest the urgent need for further optimization of the microenvironmental conditions to improve therapeutical competence of the WJ-MSCs population.


Subject(s)
Mesenchymal Stem Cells/cytology , Neuroprotection/physiology , Stem Cell Niche/physiology , Wharton Jelly/cytology , Animals , Antigens, Differentiation/metabolism , Biomimetics/methods , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Coculture Techniques/methods , Cytokines/metabolism , Hippocampus/physiology , Humans , Hydrogels/pharmacology , Rats , Rats, Wistar , Umbilical Cord/metabolism
11.
Mech Ageing Dev ; 190: 111296, 2020 09.
Article in English | MEDLINE | ID: mdl-32621937

ABSTRACT

ATM is a kinase involved in DNA damage response (DDR), regulation of response to oxidative stress, autophagy and mitophagy. Mutations in the ATM gene in humans result in ataxi A-Telangiectasia disease (A-T) characterized by a variety of symptoms with neurodegeneration and premature ageing among them. Since brain is one of the most affected organs in A-T, we have focused on senescence of neural progenitor cells (NPCs) derived from A-T reprogrammed fibroblasts. Accordingly, A-T NPCs obtained through neural differentiation of iPSCs in 5% oxygen possessed some features of senescence including increased activity of SA-ß-gal and secretion of IL6 and IL8 in comparison to control NPCs. This phenotype of A-T NPC was accompanied by elevated oxidative stress. A-T NPCs exhibited symptoms of impaired autophagy and mitophagy with lack of response to chloroquine treatment. Additional sources of oxidative stress like increased oxygen concentration (20 %) and H2O2 respectively aggravated the phenotype of senescence and additionally disturbed the process of mitophagy. In both cases only A-T NPCs reacted to the treatment. We conclude that oxidative stress may be responsible for the phenotype of senescence and impairment of autophagy in A-T NPCs. Our results point to senescent A-T cells as a potential therapeutic target in this disease.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Autophagy/physiology , Cellular Senescence/genetics , Neurons/physiology , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , Drug Discovery , Humans , Induced Pluripotent Stem Cells/physiology , Interleukin-6/metabolism , Interleukin-8/metabolism , Mitophagy , Mutation , Oxidative Stress/physiology , Signal Transduction , beta-Galactosidase/metabolism
12.
J Appl Toxicol ; 39(12): 1610-1622, 2019 12.
Article in English | MEDLINE | ID: mdl-31168795

ABSTRACT

Organoids are three-dimensional self-aggregating structures generated from stem cells (SCs) or progenitor cells in a process that recapitulates molecular and cellular stages of early organ development. The differentiation process leads to the appearance of specialized mature cells and is connected with changes in the organoid internal structure rearrangement and self-organization. The formation of organ-specific structures in vitro with highly ordered architecture is also strongly influenced by the extracellular matrix. These features make organoids as a powerful model for in vitro toxicology. Nowadays this technology is developing very quickly. In this review we present, from a toxicological and species-specific point of view, the state of the art of organoid generation from adult SCs and pluripotent SCs: embryonic SCs or induced pluripotent SCs. The current culture organoid techniques are discussed for their main advantages, disadvantages and limitations. In the second part of the review, we concentrated on the characterization of species-specific organoids generated from tissue-specific SCs of different sources: mammary (bovine), epidermis (canine), intestinal (porcine, bovine, canine, chicken) and liver (feline, canine).


Subject(s)
Biotechnology/methods , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Toxicity Tests/methods , Animals , Cattle , Cell Culture Techniques , Chickens , Dogs , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/drug effects , Models, Biological , Organ Specificity , Organoids/drug effects , Pluripotent Stem Cells/drug effects , Species Specificity , Swine
13.
Mol Neurobiol ; 56(10): 6820-6832, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30927132

ABSTRACT

Correct selection of the reference gene(s) is the most important step in gene expression analysis. The aims of this study were to identify and evaluate the panel of possible reference genes in neural stem cells (NSC), early neural progenitors (eNP) and neural progenitors (NP) obtained from human-induced pluripotent stem cells (hiPSC). The stability of expression of genes commonly used as the reference in cells during neural differentiation is variable and does not meet the criteria for reference genes. In the present work, we evaluated the stability of expression of 16 candidate reference genes using the four most popular algorithms: the ΔCt method, BestKeeper, geNorm and NormFinder. All data were analysed using the online tool RefFinder to obtain a comprehensive ranking. Our results indicate that NormFinder is the best tool for reference gene selection in early stages of hiPSC neural differentiation. None of the 16 tested genes is suitable as reference gene for all three stages of development. We recommend using different genes (panel of genes) to normalise RT-qPCR data for each of the neural differentiation stages.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/metabolism , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Algorithms , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Humans , Reference Standards
14.
Front Biosci (Schol Ed) ; 11(1): 105-121, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30844739

ABSTRACT

The human induced pluripotent stem cells (hiPSC) are one of the promising candidates as patient specific cell source for autologous transplantation or modeling of diseases. The collagen (Col) scaffolds have been shown suitable to create in vitro biomimetic microenvironment for human neural stem cells, but their ability to accommodate stem cells at different stages of neural differentiation has not been verified yet. In this paper we compare lineage related hiPSC during neural differentiation for their ability to colonize Col scaffold. We have also focused on modification of collagen physicochemical properties with improved mechanical and thermal stability, without loss of its biological activity. The hiPSC expressing markers of pluripotency (OCT4, SOX2, NANOG) after neural commitment are NESTIN, GFAP, PDGFR alpha, beta- TUBULIN III, MAP-2, DCX, GalC positive. We have shown, that Col scaffold was not preferable for hiPSC culture, while the neurally committed population after seeding on Col scaffolds revealed good adhesion, viability, proliferation, along with sustaining markers of neuronal and glial differentiation. The Col scaffold-based 3D culture of hiPSC-NSCs may serve as a research tool for further translational studies.


Subject(s)
Cell Differentiation , Collagen/chemistry , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Tissue Scaffolds , Animals , Biocompatible Materials , Calorimetry, Differential Scanning , Coculture Techniques , Humans , Microscopy, Confocal , Neurons/cytology , Porosity , Spectroscopy, Fourier Transform Infrared , Swine , Tendons/pathology
15.
Mol Neurobiol ; 56(6): 4346-4363, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30315479

ABSTRACT

Bezafibrate (BZ) regulates mitochondrial biogenesis by activation of PPAR's receptors and enhancing the level of PGC-1α coactivator. In this report, we investigated the effect of BZ on the expression of genes (1) that are linked to different pathways involved in mitochondrial biogenesis, e.g., regulated by PPAR's receptors or PGC-1α coactivator, and (2) involved in neuronal or astroglial fate, during neural differentiation of hiPSC. The tested cell populations included hiPSC-derived neural stem cells (NSC), early neural progenitors (eNP), and neural progenitors (NP). RNA-seq analysis showed the expression of PPARA, PPARD receptors and excluded PPARG in all tested populations. The expression of PPARGC1A encoding PGC-1α was dependent on the stage of differentiation: NSC, eNP, and NP differed significantly as compared to hiPSC. In addition, BZ-evoked upregulation of PPARGC1A, GFAP, S100B, and DCX genes coexist with downregulation of MAP2 gene only at the eNP stage of differentiation. In the second task, we investigated the cell sensitivity and mitochondrial biogenesis upon BZ treatment. BZ influenced the cell viability, ROS level, mitochondrial membrane potential, and total cell number in concentration- and stage of differentiation-dependent manner. Induction of mitochondrial biogenesis evoked by BZ determined by the changes in the level of SDHA and COX-1 protein, and mtDNA copy number, as well as the expression of NRF1, PPARGC1A, and TFAM genes, was detected only at NP stage for all tested markers. Thus, developmental stage-specific sensitivity to BZ of neurally differentiating hiPSC can be linked to mitochondrial biogenesis, while fate commitment decisions to PGC-1α (encoded by PPARGC1A) pathway.


Subject(s)
Bezafibrate/pharmacology , Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Organelle Biogenesis , Up-Regulation/drug effects , Cell Line , Cell Survival/drug effects , Computer Simulation , Cyclooxygenase 1/metabolism , DNA, Mitochondrial/genetics , Electron Transport Complex II/metabolism , Gene Dosage , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Reactive Oxygen Species/metabolism , Reference Standards , Reproducibility of Results
16.
Exp Neurol ; 319: 112813, 2019 09.
Article in English | MEDLINE | ID: mdl-30171864

ABSTRACT

Glial cells which are indispensable for the central nervous system development and functioning, are proven to be vulnerable to a harmful influence of pathological cues and tissue misbalance. However, they are also highly sensitive to both in vitro and in vivo modulation of their commitment, differentiation, activity and even the fate-switch by different types of bioactive molecules. Since glial cells (comprising macroglia and microglia) are an abundant and heterogeneous population of neural cells, which are almost uniformly distributed in the brain and the spinal cord parenchyma, they all create a natural endogenous reservoir of cells for potential neurogenerative processes required to be initiated in response to pathophysiological cues present in the local tissue microenvironment. The past decade of intensive investigation on a spontaneous and enforced conversion of glial fate into either alternative glial (for instance from oligodendrocytes to astrocytes) or neuronal phenotypes, has considerably extended our appreciation of glial involvement in restoring the nervous tissue cytoarchitecture and its proper functions. The most effective modulators of reprogramming processes have been identified and tested in a series of pre-clinical experiments. A list of bioactive compounds which are potent in guiding in vivo cell fate conversion and driving cell differentiation includes a selection of transcription factors, microRNAs, small molecules, exosomes, morphogens and trophic factors, which are helpful in boosting the enforced neuro-or gliogenesis and promoting the subsequent cell maturation into desired phenotypes. Herein, an issue of their utility for a directed glial differentiation and transdifferentiation is discussed in the context of elaborating future therapeutic options aimed at restoring the diseased nervous tissue.


Subject(s)
Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Nerve Regeneration/physiology , Neuroglia/physiology , Animals , Humans , Nerve Tissue/cytology , Nerve Tissue/growth & development , Peripheral Nerve Injuries/therapy
17.
Results Probl Cell Differ ; 66: 21-87, 2018.
Article in English | MEDLINE | ID: mdl-30209654

ABSTRACT

Human somatic stem cells can be identified and isolated from different types of tissues and are grouped here based on their developmental maturation and ability to undergo neural differentiation. The first group will represent afterbirth somatic tissues, which are perinatal stem cells including placental blood and tissue, amniotic fluid and tissue, and umbilical cord blood- and umbilical cord tissue-derived cells. The second group of cells discussed in this chapter is the adult stem cells, generally those in a transient period of development, thus placing them in the special position of transitioning from the perinatal to young somatic tissue, and they include the menstrual blood-, the peripheral blood-, and the bone marrow-derived stem cells.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation , Neural Stem Cells/cytology , Adult , Bone Marrow Cells/cytology , Female , Fetal Blood/cytology , Humans , Placenta/cytology , Pregnancy , Umbilical Cord/cytology
18.
Results Probl Cell Differ ; 66: 207-230, 2018.
Article in English | MEDLINE | ID: mdl-30209661

ABSTRACT

Human neural stem/progenitor cells of the developing and adult organisms are surrounded by the microenvironment, so-called neurogenic niche. The developmental processes of stem cells, such as survival, proliferation, differentiation, and fate decisions, are controlled by the mutual interactions between cells and the niche components. Such interactions are tissue specific and determined by the biochemical and biophysical properties of the niche constituencies and the presence of other cell types. This dynamic approach of the stem cell niche, when translated into in vitro settings, requires building up "biomimetic" microenvironments resembling natural conditions, where the stem/progenitor cell is provided with diverse extracellular signals exerted by soluble and structural cues, mimicking those found in vivo. The neural stem cell niche is characterized by a unique composition of soluble components including neurotransmitters and trophic factors as well as insoluble extracellular matrix proteins and proteoglycans. Biotechnological innovations provide tools such as a new generation of tunable biomaterials capable of releasing specific signals in a spatially and temporally controlled manner, thus creating in vitro nature-like conditions and, when combined with stem cell-derived tissue specific progenitors, producing differentiated neuronal tissue structures. In addition, substantial progress has been made on the protocols to obtain stem cell-derived cell aggregates such as neurospheres and self-assembled organoids.In this chapter, we have assessed the application of bioengineered human neural stem cell microenvironments to produce in vitro models of different levels of biological complexity for the efficient control of stem cell fate. Examples of biomaterial-supported two-dimensional and three-dimensional (2D and 3D) complex culture systems that provide artificial neural stem cell niches are discussed in the context of their application for basic research and neurotoxicity testing.


Subject(s)
Bioengineering , Cell Differentiation , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurotoxins/pharmacology , Stem Cell Niche/drug effects , Biocompatible Materials , Cell Lineage , Humans
19.
Expert Opin Biol Ther ; 18(8): 865-881, 2018 08.
Article in English | MEDLINE | ID: mdl-30025485

ABSTRACT

INTRODUCTION: Amyotrophic Lateral Sclerosis (ALS) is a progressive, incurable neurodegenerative disease that targets motoneurons. Cell-based therapies have generated widespread interest as a potential therapeutic approach but no conclusive results have yet been reported either from pre-clinical or clinical studies. AREAS COVERED: This is an integrated review of pre-clinical and clinical studies focused on the development of cell-based therapies for ALS. We analyze the biology of stem cell treatments and results obtained from pre-clinical models of ALS and examine the methods and the results obtained to date from clinical trials. We discuss scientific, clinical, and ethical issues and propose some directions for future studies. EXPERT OPINION: While data from individual studies are encouraging, stem-cell-based therapies do not yet represent a satisfactory, reliable clinical option. The field will critically benefit from the introduction of well-designed, randomized and reproducible, powered clinical trials. Comparative studies addressing key issues such as the nature, properties, and number of donor cells, the delivery mode and the selection of proper patient populations that may benefit the most from cell-based therapies are now of the essence. Multidisciplinary networks of experts should be established to empower effective translation of research into the clinic.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Stem Cell Transplantation/trends , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Humans , Randomized Controlled Trials as Topic/methods , Stem Cell Transplantation/methods
20.
Folia Neuropathol ; 55(2): 110-123, 2017.
Article in English | MEDLINE | ID: mdl-28677368

ABSTRACT

The biomimetic, standardized conditions for in vitro cultures of human neural progenitors derived from induced pluripotent stem cells (hiPSC-NPs) should meet the requirements to serve as the template and protective environment for therapeutically competent cell population. In this study, two different collagen scaffolds: bi-component consisting of collagen and chondroitin sulphate (Col-CS), and collagen modified by crosslinking agent 2,3-dialdehyde cellulose (Col-DAC) have been used for the first time to encapsulate hiPSC-NPs and compared for the ability to create permissive microenvironment enabling cell survival, growth and differentiation. In our previous report, physicochemical comparison of the scaffolds revealed different elasticity, and diverse size and distribution of the pores within the 3D structure. Binary systems of Col-CS and Col-DAC tested in the current study have the correct balance of properties to serve as a biomimetic niche: they accommodate hiPSC-NPs sustaining their ability to proliferate and differentiate into neural lineages. However, a dense, network structure and rounded in shape pores of the Col-DAC microenvironment resulted in differential cell distributions within the scaffolds, with a tendency for augmented formation of highly proliferating cell aggregates as compared to Col-CS scaffolds. In contrast, Col-CS, which exhibited formation of the network of ellipsoidal and inner interconnected parallel pore channels, promoted enhanced cell viability and neuronal differentiation.


Subject(s)
Collagen Type I , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biomimetic Materials/chemistry , Cell Differentiation , Humans , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...