Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Methods Mol Biol ; 2549: 137-151, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33772463

RESUMO

Inherited retinal diseases (IRDs) are a diverse group of rare eye disorders, resulting in vision loss or blindness. The underlying reason is mutation in one or more than 250 different genes associated with the development and normal physiology of retina largely comprising of rod/cone photoreceptors and retinal pigment epithelium. Interestingly, the sub retinal region of an eye has been shown to be immune privileged, broadening the scope of cell-replacement therapies for patients suffering from retinal degeneration. Several groups around the globe, including ours, have demonstrated safety and efficacy in preclinical studies by employing various approaches of retinal cell therapy. This had largely been possible with the advent of induced pluripotent stem cells (iPSC)-reprogrammed from adult somatic cells, that serves as a starting material for generating retinal cells de novo. Here, we describe a detailed procedure for reprogramming peripheral blood mononuclear cells (PBMC) into iPSC using episomal vectors without any physical disruption in the host genome. The lines thus created were tested for sterility, cytogenetic stability, identity, absence of episomal plasmids and further authenticated for pluripotency and tri-lineage differentiation capacity by embryoid body formation and immunocytochemistry. We believe that this feeder-cell free, animal-product free and gene-insertion free protocol would help people to develop and bank patient-specific cell lines for autologous cell therapies for incurable rare diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular/genética , Reprogramação Celular , Humanos , Leucócitos Mononucleares , Plasmídeos/genética
3.
Toxicol Appl Pharmacol ; 433: 115792, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34742744

RESUMO

Concurrent with the '3R' principle, the embryonic stem cell test (EST) using mouse embryonic stem cells, developed in 2000, remains the solely accepted in vitro method for embryotoxicity testing. However, the scope and implementation of EST for embryotoxicity screening, compliant with regulatory requirements, are limited. This is due to its technical complexity, long testing period, labor-intensive methodology, and limited endpoint data, leading to misclassification of embryotoxic potential. In this study, we used human induced pluripotent stem cell (hiPSC)-derived embryoid bodies (EB) as an in vitro model to investigate the embryotoxic effects of a carefully selected set of pharmacological compounds. Morphology, viability, and differentiation potential were investigated after exposing EBs to folic acid, all-trans-retinoic acid, dexamethasone, and valproic acid for 15 days. The results showed that the compounds differentially repressed cell growth, compromised morphology, and triggered apoptosis in the EBs. Further, transcriptomics was employed to compare subtle temporal changes between treated and untreated cultures. Gene ontology and pathway analysis revealed that dysregulation of a large number of genes strongly correlated with impaired neuroectoderm and cardiac mesoderm formation. This aberrant gene expression pattern was associated with several disorders of the brain like mental retardation, multiple sclerosis, stroke and of the heart like dilated cardiomyopathy, ventricular tachycardia, and ventricular arrhythmia. Lastly, these in vitro findings were validated using in ovo chick embryo model. Taken together, pharmacological compound or drug-induced defective EB development from hiPSCs could potentially be used as a suitable in vitro platform for embryotoxicity screening.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Corpos Embrioides/efeitos dos fármacos , Perfilação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Teratogênicos/toxicidade , Testes de Toxicidade , Transcriptoma/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula , Embrião de Galinha , Dexametasona/toxicidade , Relação Dose-Resposta a Droga , Corpos Embrioides/metabolismo , Corpos Embrioides/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Concentração Inibidora 50 , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurogênese/efeitos dos fármacos , Medição de Risco , Tretinoína/toxicidade , Ácido Valproico/toxicidade
4.
ACS Chem Neurosci ; 12(20): 3785-3794, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34628850

RESUMO

Neural precursor cells (NPCs), derived from pluripotent stem cells (PSCs), with their unique ability to generate multiple neuronal and glial cell types are extremely useful for understanding biological mechanisms in normal and diseased states. However, generation of specific neuronal subtypes (mature) from NPCs in large numbers adequate for cell therapy is challenging due to lack of a thorough understanding of the cues that govern their differentiation. Interestingly, neural stem cells (NSCs) themselves are in consideration for therapy given their potency to form different neural cell types, release of trophic factors, and immunomodulatory effects that confer neuroprotection. With the recent COVID-19 outbreak and its accompanying neurological indications, the immunomodulatory role of NSCs may gain additional significance in the prevention of disease progression in vulnerable populations. In this regard, small-molecule mediated NPC generation from PSCs via NSC formation has become an important strategy that ensures consistency and robustness of the process. The development of the mammalian brain occurs along the rostro-caudal axis, and the establishment of anterior identity is an early event. Wnt signaling, along with fibroblast growth factor and retinoic acid, acts as a caudalization signal. Further, the increasing amount of epigenetic data available from human fetal brain development has enhanced both our understanding of and ability to experimentally manipulate these developmental regulatory programs in vitro. However, the impact on homing and engraftment after transplantation and subsequently on therapeutic efficacy of NPCs based on their derivation strategy is not yet clear. Another formidable challenge in cell replacement therapy for neurodegenerative disorders is the mode of delivery. In this Perspective, we discuss these core ideas with insights from our preliminary studies exploring the role of PSC-derived NPCs in rat models of MPTP-induced Parkinson's disease following intranasal injections.


Assuntos
COVID-19 , Células-Tronco Neurais , Doença de Parkinson , Animais , Humanos , Neurônios , Doença de Parkinson/terapia , Ratos , SARS-CoV-2
5.
Stem Cell Res Ther ; 12(1): 70, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33468244

RESUMO

BACKGROUND: Age-related macular degeneration (AMD) is a result of degeneration/damage of the retinal pigment epithelium (RPE) while retinitis pigmentosa (RP), an inherited early-onset disease, results from premature loss of photoreceptors. A promising therapeutic approach for both is the replacement of lost/damaged cells with human induced pluripotent stem cell (hiPSC)-derived retinal cells. METHODS: The aim of this study was to investigate the in vivo functionality of RPE and photoreceptor progenitor (PRP) cells derived from a clinical-grade hiPSC line through a unified protocol. De novo-generated RPE and PRP were characterized extensively to validate their identity, purity, and potency. RESULTS: RPE expressed tight junction proteins, showed pigmentation and ciliation, and secreted polarization-related factors vascular endothelial growth factor (VEGF) and pigment epithelium-derived factor (PEDF). PRP expressed neural retina proteins and cone and rod markers, and responded to KCl-induced polarization. Transcriptomic analysis demonstrated an increase in the expression of mature retinal tissue-specific genes coupled with concomitant downregulation of genes from undesired lineages. RPE transplantation rescued visual function in RCS rats shown via optokinetic tracking and photoreceptor rescue. PRP transplantation improved light perception in NOD.SCID-rd1 mice, and positive electroretinography signals indicated functional photoreceptor activity in the host's outer nuclear layer. Graft survival and integration were confirmed using immunohistochemistry, and no animals showed teratoma formation or any kind of ectopic growth in the eye. CONCLUSIONS: To our knowledge, this is the first demonstration of a unified, scalable, and GMP-adaptable protocol indicating strong animal efficacy and safety data with hiPSC-derived RPE and PRP cells. These findings provide robust proof-of-principle results for IND-enabling studies to test these potential regenerative cell therapies in patients.


Assuntos
Células-Tronco Pluripotentes Induzidas , Degeneração Retiniana , Animais , Diferenciação Celular , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Epitélio Pigmentado da Retina , Roedores , Fator A de Crescimento do Endotélio Vascular
6.
Stem Cells Dev ; 29(21): 1365-1369, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32867617

RESUMO

The lung is the most vulnerable target for the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, and respiratory failure causing acute respiratory distress syndrome is its foremost outcome. However, the current primary in vitro models in use for SARS-CoV-2 display apparent limitations for modeling such complex human respiratory disease. Although patient cells can directly model the effects of a drug, their availability and capacity for expansion are limited compared with transformed/immortalized cells or tumor-derived cell lines. An additional caveat is that the latter may harbor genetic and metabolic abnormalities making them unsuitable for drug screening. Therefore, it is important to create physiologically relevant human-cell models that can replicate the pathophysiology of SARS-CoV-2, thus facilitating drug testing. In this study, we show preliminary data on how human induced pluripotent stem cells-derived lung epithelial cell system could emerge as a relevant and sensitive platform for modeling SARS-CoV-2 infection and drug screening.


Assuntos
Tratamento Farmacológico da COVID-19 , Avaliação Pré-Clínica de Medicamentos/métodos , Reposicionamento de Medicamentos , Células-Tronco Pluripotentes Induzidas/virologia , SARS-CoV-2/efeitos dos fármacos , COVID-19/patologia , Linhagem Celular , Humanos , Pulmão/citologia , Modelos Biológicos , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/prevenção & controle
7.
Stem Cell Res ; 45: 101757, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32278301

RESUMO

Inherited retinal diseases (IRDs) are clinically and genetically heterogenous diseases affecting the neural retina and retinal pigment epithelium resulting in irreversible blindness. Owing to advantages like ease of access for treatment, one eye being a perfect natural control for the other, and immune privileged environment, research exploring treatment for these retinal diseases has advanced remarkably. We describe the generation of induced pluripotent stem cell (iPSC) lines from peripheral blood mononuclear cells (PBMC) of three patients with IRDs. These well-characterized iPSC lines provide an ideal platform to investigate normal and pathological retinogenesis for drug screening and personalized cell therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Retinianas , Diferenciação Celular , Humanos , Leucócitos Mononucleares , Mutação , Retina , Doenças Retinianas/genética , Doenças Retinianas/terapia
8.
Cell Biol Toxicol ; 35(2): 147-159, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30673990

RESUMO

Manganese exposure is among the many environmental risk factors linked to the progression of neurodegenerative diseases, such as manganese-induced parkinsonism. In animal models, chronic exposure to manganese causes loss of cell viability, neurodegeneration, and functional deficits. Polyamines, such as spermine, have been shown to rescue animals from age-induced neurodegeneration in an autophagy-dependent manner; nonetheless, it is not understood whether polyamines can prevent manganese-induced toxicity. In this study, we used two model systems, the Caenorhabditis elegans UA44 strain and SK-MEL-28 cells, both expressing the protein alpha-synuclein (α-syn) to determine whether spermine could ameliorate manganese-induced toxicity. Manganese caused a substantial reduction in the viability of SK-MEL-28 cells and hastened neurodegeneration in the UA44 strain. Spermine protected both the SK-MEL-28 cells and the UA44 strain from manganese-induced toxicity. Spermine also reduced the age-associated neurodegeneration observed in the UA44 strain compared with a control strain without α-syn expression and led to improved avoidance behavior in a functional assay. Treatment with berenil, an inhibitor of polyamine catabolism, which leads to increased intracellular polyamine levels, also showed similar cellular protection against manganese toxicity. While both translation blocker cycloheximide and autophagy blocker chloroquine caused a reduction in the cytoprotective effect of spermine, transcription blocker actinomycin D had no effect. This study provides new insights on the effect of spermine in preventing manganese-induced toxicity, which is most likely via translational regulation of several candidate genes, including those of autophagy. Thus, our results indicate that polyamines positively influence neuronal health, even when exposed to high levels of manganese and α-syn, and supplementing polyamines through diet might delay the onset of diseases involving degeneration of dopaminergic neurons.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Manganês/toxicidade , Fármacos Neuroprotetores/farmacologia , Espermina/farmacologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Diminazena/análogos & derivados , Diminazena/farmacologia , Humanos , Degeneração Neural/prevenção & controle , alfa-Sinucleína/metabolismo
9.
J Cell Commun Signal ; 11(4): 381-394, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28748508

RESUMO

Increased cellular concentration of α-synuclein (α-syn) predisposes it to misfolding and aggregation that in turn impair the degradation pathways. This poses a limitation to the use of overexpression models for studies on α-syn clearance by autophagy, which is widely investigated for its therapeutic potential. This limitation can be overcome with the use of endogenous models. In this study, SK-MEL-28, a melanoma cell model with endogenous α-syn expression, was employed to study α-syn clearance through autophagy. We demonstrated the dual localization of α-syn to nucleus and cytoplasm that varied in response to changes in cellular environment. Autophagy inhibition and exposure to dopamine favored cytoplasmic localization of α-syn, while autophagy induction favored increased localization to the nucleus. The inhibitory effect of dopamine on autophagy was heightened in presence of α-syn. Additionally, because α-syn had a regulatory effect on autophagy, cells showed an increased resistance to autophagy induction in presence of α-syn. This resistance prevented effective induction of autophagy even under conditions of prolonged autophagy inhibition. These results highlight alternate physiological roles of α-syn, particularly in non-neuronal cells. Because autophagy enhancement could reverse neither the increase in α-syn levels nor the autophagy inhibition, there arises a need to evaluate the efficacy of autophagy-based therapeutic strategies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...