Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35055039

RESUMO

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.


Assuntos
Regeneração Tecidual Guiada , Miocárdio , Regeneração Nervosa , Medicina Regenerativa , Animais , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Encefalopatias/diagnóstico , Encefalopatias/etiologia , Encefalopatias/terapia , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Gerenciamento Clínico , Vesículas Extracelulares/metabolismo , Regeneração Tecidual Guiada/métodos , Cardiopatias/diagnóstico , Cardiopatias/etiologia , Cardiopatias/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Organoides , Medicina Regenerativa/métodos , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo
2.
J Appl Toxicol ; 39(12): 1610-1622, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31168795

RESUMO

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).


Assuntos
Biotecnologia/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Testes de Toxicidade/métodos , Animais , Bovinos , Técnicas de Cultura de Células , Galinhas , Cães , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Modelos Biológicos , Especificidade de Órgãos , Organoides/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Especificidade da Espécie , Suínos
3.
Folia Neuropathol ; 62(1): 32-46, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38741435

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Vírus Sendai , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Linhagem Celular , Fibroblastos , Diferenciação Celular/fisiologia , Transdução Genética/métodos , Fatores Sexuais
4.
Expert Opin Biol Ther ; 24(9): 933-954, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39162129

RESUMO

INTRODUCTION: Cell therapy development represents a critical challenge in amyotrophic lateral sclerosis (ALS) research. Despite more than 20 years of basic and clinical research, no definitive safety and efficacy results of cell-based therapies for ALS have been published. AREAS COVERED: This review summarizes advances using stem cells (SCs) in pre-clinical studies to promote clinical translation and in clinical trials to treat ALS. New technologies have been developed and new experimental in vitro and animal models are now available to facilitate pre-clinical research in this field and to determine the most promising approaches to pursue in patients. New clinical trial designs aimed at developing personalized SC-based treatment with biological endpoints are being defined. EXPERT OPINION: Knowledge of the basic biology of ALS and on the use of SCs to study and potentially treat ALS continues to grow. However, a consensus has yet to emerge on how best to translate these results into therapeutic applications. The selection and follow-up of patients should be based on clinical, biological, and molecular criteria. Planning of SC-based clinical trials should be coordinated with patient profiling genetically and molecularly to achieve personalized treatment. Much work within basic and clinical research is still needed to successfully transition SC therapy in ALS.


Assuntos
Esclerose Lateral Amiotrófica , Transplante de Células-Tronco , Esclerose Lateral Amiotrófica/terapia , Esclerose Lateral Amiotrófica/genética , Humanos , Animais , Modelos Animais de Doenças , Ensaios Clínicos como Assunto
5.
Biomed Microdevices ; 15(3): 495-507, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23404262

RESUMO

Two methods for protein patterning on antifouling surfaces have been applied to analyze the density and bioactivity of the proteins after deposition. Microcontact printing has been used as a technique to transfer fibronectin through conformal contact, while piezoelectric deposition has been employed as a non-contact technique for producing arrays of fibronectin (FN). Plasma deposited polyethylene oxide-like (PEO-like) films have been used as non-fouling background to achieve the bioadhesive/biorepellent surface contrast. Both patterning methods allow the direct fabrication of protein arrays on a non-fouling substrate, and the subsequent formation of a pattern of stem cells by cell attachment on the arrayed substrates. Microcontact printing produced fully packed homogeneous fibronectin patterns, much denser than microspotted patterns. Both printing and spotting technologies generated functional protein arrays, their bioactivity being primarily modulated by the density of the deposited protein layer. Optimization of the FN parameters used for deposition has lead to the achievement of high-quality microarrays with large population of neural stem cells immobilized in the patterns in serum-free conditions, where cells exhibit a more homogeneous starting population and factors influencing fate decisions can be more easily tracked. The immunorecognition of fibronectin targeted antibodies, as well as the cell density, increase with the protein density up to a saturation point. Over 100 ng/cm² of fibronectin on the surface leads to a decrease in the number of attached cells and a raise of cell spreading.


Assuntos
Fibronectinas/química , Microtecnologia/métodos , Gases em Plasma/química , Polietilenoglicóis/química , Impressão/métodos , Células-Tronco/citologia , Adesivos/química , Fibronectinas/metabolismo , Humanos , Polimerização , Propriedades de Superfície
6.
Postepy Biochem ; 59(2): 175-86, 2013.
Artigo em Polonês | MEDLINE | ID: mdl-24044282

RESUMO

Maintenance of developmental and regenerative capability of the tissue highly depends upon mutual interaction of the stem cells with the components of their microenvironment (niche). The nature of this interaction is determined by the biochemical and biophysical properties of the niche constituencies. Although knowledge about the components of the stem cell microenvironment and their architecture is growing quickly, we still need to unravel the mechanisms underlying the control of the niche functioning, enabling stem cells differentiation and homeostasis of the tissue. Advancement in biotechnology provides tools to build up in vitro "biomimetic" microenvironments resembling a natural stem cell niche, where the cell is provided with diverse extracellular signals exerted by soluble and structural cues, mimicking those found in vivo. To obtain such microenvironment in vitro emerging nano/biotechnology methods were applied, using biomaterials of new generation, which enable controlling of the stem cell differentiation by time and special related release of the active factors. This article is providing an overview of the new research strategies for the bioengineering of the stem cell niche and gives the examples of the cell/biomaterial 2D and 3D complex systems used for basic and preclinical research as well as entering clinical applications for the therapy of the nervous system.


Assuntos
Bioengenharia/métodos , Células-Tronco Neurais/citologia , Nicho de Células-Tronco , Materiais Biocompatíveis , Biomimética/métodos , Diferenciação Celular , Homeostase
7.
Cells ; 12(3)2023 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-36766732

RESUMO

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.


Assuntos
Corpos Embrioides , Mitofagia , Humanos , Mitofagia/genética , Reparo do DNA , Oxigênio/farmacologia , Oxigênio/metabolismo
8.
Cells ; 12(2)2023 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-36672274

RESUMO

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.


Assuntos
Epilepsias Mioclônicas , Células-Tronco Pluripotentes Induzidas , Humanos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Epilepsias Mioclônicas/genética , Neurônios/metabolismo , Prosencéfalo/metabolismo
9.
Cells ; 11(20)2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36291101

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais , Doenças Neurodegenerativas , Animais , Adulto , Humanos , Doenças Neurodegenerativas/metabolismo , Qualidade de Vida , Neurogênese/fisiologia , Hipocampo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Antidepressivos/farmacologia
10.
Cells ; 11(1)2021 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-35011673

RESUMO

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.


Assuntos
Sistema Nervoso Central/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Humanos , Biogênese de Organelas
11.
Dev Neurobiol ; 81(5): 591-607, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33725382

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Animais , Feminino , Humanos , Mitocôndrias , Doenças Neurodegenerativas/metabolismo , Neurogênese , Organoides/metabolismo , Gravidez
12.
Cells ; 10(4)2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33804841

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais/citologia , Neuroproteção , Oxigênio/farmacologia , Geleia de Wharton/citologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Células Clonais , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores
13.
Stem Cells ; 27(10): 2591-601, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19609937

RESUMO

The aim of our study was to investigate whether a human neural stem cell line derived from umbilical cord blood (HUCB-NSC) can serve as a reliable test model for developmental neurotoxicity (DNT). We assessed the sensitivity of HUCB-NSCs at different developmental stages to a panel of neurotoxic (sodium tellurite, methylmercury chloride, cadmium chloride, chlorpyrifos, and L-glutamate) and non-neurotoxic (acetaminophen, theophylline, and D-glutamate) compounds. In addition, we investigated the effect of some compounds on key neurodevelopmental processes like cell proliferation, apoptotic cell death, and neuronal and glial differentiation. Less differentiated HUCB-NSCs were generally more sensitive to neurotoxicants, with the notable exception of L-glutamate, which showed a higher toxicity to later stages. The relative potencies of the compounds were: cadmium chloride > methylmercury chloride >> chlorpyrifos >> L-glutamate. Fifty nanomolar methylmercury chloride (MeHgCl) inhibited proliferation and induced apoptosis in early-stage cells. At the differentiated stage, 1 muM MeHgCl induced selective loss of S100 beta-expressing astrocytic cells. One millimolar L-glutamate did not influence the early stages of HUCB-NSC development, but it affected late stages of neuronal differentiation. A valuable system for in vitro DNT assessment should be able to discriminate between neurotoxic and non-neurotoxic compounds and show different susceptibilities to chemicals according to developmental stage and cell lineage. Although not exhaustive, this work shows that the HUCB-NSC model fulfils these criteria and may serve as a human in vitro model for DNT priority setting.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Sangue Fetal/citologia , Sistema Nervoso/efeitos dos fármacos , Neurotoxinas/toxicidade , Testes de Toxicidade/métodos , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Biomarcadores/metabolismo , Cloreto de Cádmio/toxicidade , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Clorpirifos/toxicidade , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Ácido Glutâmico/toxicidade , Humanos , Compostos de Metilmercúrio/toxicidade , Fatores de Crescimento Neural/análise , Fatores de Crescimento Neural/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Valor Preditivo dos Testes , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/análise , Proteínas S100/metabolismo , Sensibilidade e Especificidade
14.
Mech Ageing Dev ; 190: 111296, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32621937

RESUMO

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.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia , Autofagia/fisiologia , Senescência Celular/genética , Neurônios/fisiologia , Ataxia Telangiectasia/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Dano ao DNA , Descoberta de Drogas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Mitofagia , Mutação , Estresse Oxidativo/fisiologia , Transdução de Sinais , beta-Galactosidase/metabolismo
15.
Sci Rep ; 10(1): 16946, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-33037314

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais/citologia , Neuroproteção/fisiologia , Nicho de Células-Tronco/fisiologia , Geleia de Wharton/citologia , Animais , Antígenos de Diferenciação/metabolismo , Biomimética/métodos , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Cocultura/métodos , Citocinas/metabolismo , Hipocampo/fisiologia , Humanos , Hidrogéis/farmacologia , Ratos , Ratos Wistar , Cordão Umbilical/metabolismo
16.
Acta Neurobiol Exp (Wars) ; 69(1): 24-36, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19325638

RESUMO

Bio-functionalized surfaces were prepared to study the adherence and differentiation capacity of neural stem cells derived from human umbilical cord blood (HUCB-NSC). Cell growth platforms containing arranged arrays of adhesive molecules were created by microcontact printing on a biologically inert surface. Biomolecules used to prepare microarray platforms included the extracellular matrix protein fibronectin and the polyaminoacid poly-L-lysine. HUCB-NSC plated on microplatforms at various serum conditions showed serum and molecule type dependent capacity for adhesion and differentiation. Poly-L-lysine allowed the maintenance of stem-like non differentiated cells attached to the surface, whereas fibronectin promoted spreading and neural commitment. Serum deprivation did not influence the attachment of HUCB-NSC to fibronectin, but significantly enhanced the attachment to poly-L-lysine and promoted dBcAMP induced neuronal differentiation. A bio-pattern of squares with interconnecting lines was used to guide neuronal differentiation by directing cell protrusion outgrowth. Tailoring the geometry of the bio-pattern enabled directing and monitoring of the neural stem cells. development in the large scale multiparameter biotests.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Proliferação de Células , Sangue Fetal/citologia , Neurônios/citologia , Células-Tronco/fisiologia , Astrócitos/citologia , Bucladesina/farmacologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Fibronectinas/fisiologia , Humanos , Análise em Microsséries/métodos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Polilisina/fisiologia , Impressão , Células-Tronco/efeitos dos fármacos , Propriedades de Superfície
17.
Int J Dev Biol ; 52(2-3): 237-48, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18311714

RESUMO

From the time of discovery that among the cord blood mononuclear cell population there are cells capable of changing their fate towards the neural lineage and producing functional neurons and macroglial cells, our attempts have been focused on the understanding of the underlying mechanism of this transition. We have deciphered the first steps of neural stem/progenitor gene induction in aggregating culture of cord blood mononuclear cells, their rapid phenotypic conversion under the influence of neuromorphogenic signals due to mitogen activation and their ability to expand and develop a prototypic, long-living line with neural stem cell properties. Evidence has accumulated that human umbilical cord-derived and neurally committed cells, due to their capacity for self-renewal, multilineage differentiation, plasticity and ability for long-lasting growth in vitro, provide unique material for the cell therapy of a wide spectrum of neurological diseases. The putative regenerating potential of these cord blood-derived neural stem/progenitor cells was evaluated after transplantation in experimental models of brain injury. In spite of initial promising data, the results indicate an urgent need to improve available animal model protocols in order to increase immuno-tolerance toward transplanted human cells.


Assuntos
Sangue Fetal/citologia , Neurônios/citologia , Células-Tronco/citologia , Cordão Umbilical , Animais , Linhagem da Célula , Sistema Hematopoético/citologia , Humanos
18.
Mol Neurobiol ; 56(10): 6820-6832, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30927132

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase em Tempo Real/normas , Algoritmos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Humanos , Padrões de Referência
19.
Exp Neurol ; 319: 112813, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30171864

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Transdiferenciação Celular/fisiologia , Regeneração Nervosa/fisiologia , Neuroglia/fisiologia , Animais , Humanos , Tecido Nervoso/citologia , Tecido Nervoso/crescimento & desenvolvimento , Traumatismos dos Nervos Periféricos/terapia
20.
Front Biosci (Schol Ed) ; 11(1): 105-121, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30844739

RESUMO

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.


Assuntos
Diferenciação Celular , Colágeno/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Alicerces Teciduais , Animais , Materiais Biocompatíveis , Varredura Diferencial de Calorimetria , Técnicas de Cocultura , Humanos , Microscopia Confocal , Neurônios/citologia , Porosidade , Espectroscopia de Infravermelho com Transformada de Fourier , Suínos , Tendões/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA