Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Vis Exp ; (180)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35285824

RESUMO

Cardiac reprogramming has become a potentially promising therapy to repair a damaged heart. By introducing multiple transcription factors, including Mef2c, Gata4, Tbx5 (MGT), fibroblasts can be reprogrammed into induced cardiomyocytes (iCMs). These iCMs, when generated in situ in an infarcted heart, integrate electrically and mechanically with the surrounding myocardium, leading to a reduction in scar size and an improvement in heart function. Because of the relatively low reprogramming efficiency, purity, and quality of the iCMs, characterization of iCMs remains a challenge. The currently used methods in this field, including flow cytometry, immunocytochemistry, and qPCR, mainly focus on cardiac-specific gene and protein expression but not on the functional maturation of iCMs. Triggered by action potentials, the opening of voltage-gated calcium channels in cardiomyocytes leads to a rapid influx of calcium into the cell. Therefore, quantifying the rate of calcium influx is a promising method to evaluate cardiomyocyte function. Here, the protocol introduces a method to evaluate iCM function by calcium (Ca2+) flux. An αMHC-Cre/Rosa26A-Flox-Stop-Flox-GCaMP3 mouse strain was established by crossing Tg(Myh6-cre)1Jmk/J (referred to as Myh6-Cre below) with Gt(ROSA)26Sortm38(CAG-GCaMP3)Hze/J (referred to as Rosa26A-Flox-Stop-Flox-GCaMP3 below) mice. Neonatal cardiac fibroblasts (NCFs) from P0-P2 neonatal mice were isolated and cultured in vitro, and a polycistronic construction of MGT was introduced to NCFs, which led to their reprogramming to iCMs. Because only successfully reprogrammed iCMs will express GCaMP3 reporter, the functional maturation of iCMs can be visually assessed by Ca2+ flux with fluorescence microscopy. Compared with un-reprogrammed NCFs, NCF-iCMs showed significant calcium transient flux and spontaneous contraction, similar to CMs. This protocol describes in detail the mouse strain establishment, isolation and selection of neonatal mice hearts, NCF isolation, production of retrovirus for cardiac reprogramming, iCM induction, the evaluation of iCM Ca2+ flux using our reporter line, and related statistical analysis and data presentation. It is expected that the methods described here will provide a valuable platform to assess the functional maturation of iCMs for cardiac reprogramming studies.


Assuntos
Cálcio , Reprogramação Celular , Animais , Cálcio/metabolismo , Fibroblastos/metabolismo , Camundongos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo
2.
Methods Mol Biol ; 2352: 183-199, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34324188

RESUMO

Direct reprogramming is an emerging research field where you can generate neurons from a somatic cell, such as a skin or glial cell by overexpressing neurogenic transcription factors. This technique allows fast generation of subtype-specific and functional neurons from both human and mouse cells. Despite the fact that neurons have been successfully generated both in vitro and in vivo, a more extensive analysis of the induced neurons including phenotypic functional identity or gradual maturity is still lacking. This is an important step for a further development of induced neurons towards cell therapy or disease modeling of neurological diseases. In this protocol, we describe a method for functional assessment of direct reprogrammed neuronal cells both in vitro and in vivo. Using a synapsin-driven reporter, our protocol allows for a direct identification of the reprogrammed neurons that permits functional assessment using patch-clamp electrophysiology. For in vitro reprogramming we further provide an optimized coating condition that allows a long-term maturation of human induced neurons in vitro.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Reprogramação Celular/genética , Técnicas de Reprogramação Celular , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/biossíntese , Vetores Genéticos/genética , Vetores Genéticos/isolamento & purificação , Humanos , Lentivirus/genética , Camundongos , Técnicas de Patch-Clamp , Fatores de Transcrição/genética , Transdução Genética
4.
PLoS Comput Biol ; 16(1): e1007491, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31923173

RESUMO

Recent high-dimensional single-cell technologies such as mass cytometry are enabling time series experiments to monitor the temporal evolution of cell state distributions and to identify dynamically important cell states, such as fate decision states in differentiation. However, these technologies are destructive, and require analysis approaches that temporally map between cell state distributions across time points. Current approaches to approximate the single-cell time series as a dynamical system suffer from too restrictive assumptions about the type of kinetics, or link together pairs of sequential measurements in a discontinuous fashion. We propose Dynamic Distribution Decomposition (DDD), an operator approximation approach to infer a continuous distribution map between time points. On the basis of single-cell snapshot time series data, DDD approximates the continuous time Perron-Frobenius operator by means of a finite set of basis functions. This procedure can be interpreted as a continuous time Markov chain over a continuum of states. By only assuming a memoryless Markov (autonomous) process, the types of dynamics represented are more general than those represented by other common models, e.g., chemical reaction networks, stochastic differential equations. Furthermore, we can a posteriori check whether the autonomy assumptions are valid by calculation of prediction error-which we show gives a measure of autonomy within the studied system. The continuity and autonomy assumptions ensure that the same dynamical system maps between all time points, not arbitrarily changing at each time point. We demonstrate the ability of DDD to reconstruct dynamically important cell states and their transitions both on synthetic data, as well as on mass cytometry time series of iPSC reprogramming of a fibroblast system. We use DDD to find previously identified subpopulations of cells and to visualise differentiation trajectories. Dynamic Distribution Decomposition allows interpretation of high-dimensional snapshot time series data as a low-dimensional Markov process, thereby enabling an interpretable dynamics analysis for a variety of biological processes by means of identifying their dynamically important cell states.


Assuntos
Reprogramação Celular/fisiologia , Biologia Computacional/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Análise de Célula Única/métodos , Algoritmos , Animais , Linhagem Celular , Cadeias de Markov , Camundongos
5.
Biomaterials ; 189: 23-36, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30384126

RESUMO

Human induced pluripotent stem cells (iPSCs) have unlimited proliferation capability and potential to differentiate into all somatic cells. Their derivatives contain patients' genetic information and can model many diseases. Additionally, derivatives of patient-specific iPSCs induce minimal immune rejection in vivo. With this unique combination of properties, iPSCs open the avenue to personalized medicine including personalized drug screening, toxicity test, cell therapy and tissue engineering. However, the further advance of iPSC-based personalized medicine is currently limited by the difficulty to generate iPSCs for large populations and at affordable cost. We here report a low-cost device to address this challenge. The device allows the entire bioprocess for generating high quality and quantity of iPSCs for one patient to be done automatically within a closed conical tube without cell passaging. Additionally, iPSCs can be further differentiated into somatic cells in the device. Thus, the device also allows integrated iPSCs generation, expansion and differentiation to produce any somatic cell types. This device can be made in large quantities at low cost for manufacturing iPSCs (and their derivatives in necessary) for large populations at affordable cost. It will significantly advance the iPSCs-based personalized medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Engenharia Tecidual/instrumentação , Alginatos/química , Materiais Biocompatíveis/química , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Reprogramação Celular , Desenho de Equipamento , Humanos , Engenharia Tecidual/economia
6.
Int J Cardiol ; 269: 229-234, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30224033

RESUMO

BACKGROUND: Recent developments in cellular reprogramming technology enable the production of virtually unlimited numbers of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Although hiPSC-CM share various characteristic hallmarks with endogenous cardiomyocytes, it remains a question as to what extent metabolic characteristics are equivalent to mature mammalian cardiomyocytes. Here we set out to functionally characterize the metabolic status of hiPSC-CM in vitro by employing a radionuclide tracer uptake assay. MATERIAL AND METHODS: Cardiac differentiation of hiPSC was induced using a combination of well-orchestrated extrinsic stimuli such as WNT activation (by CHIR99021) and BMP signalling followed by WNT inhibition and lactate based cardiomyocyte enrichment. For characterization of metabolic substrates, dual tracer uptake studies were performed with 18F­2­fluoro­2­deoxy­d­glucose (18F-FDG) and 125I­ß­methyl­iodophenyl­pentadecanoic acid (125I-BMIPP) as transport markers of glucose and fatty acids, respectively. RESULTS: After cardiac differentiation of hiPSCs, in vitro tracer uptake assays confirmed metabolic substrate shift from glucose to fatty acids that was comparable to those observed in native isolated human cardiomyocytes. Immunostaining further confirmed expression of fatty acid transport and binding proteins on hiPSC-CM. CONCLUSIONS: During in vitro cardiac maturation, we observed a metabolic shift to fatty acids, which are known as a main energy source of mammalian hearts, suggesting hi-PSC-CM as a potential functional phenotype to investigate alteration of cardiac metabolism in cardiac diseases. Results also highlight the use of available clinical nuclear medicine tracers as functional assays in stem cell research for improved generation of autologous differentiated cells for numerous biomedical applications.


Assuntos
Reprogramação Celular/fisiologia , Ácidos Graxos/metabolismo , Fluordesoxiglucose F18/metabolismo , Glucose/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Radioisótopos do Iodo/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Miócitos Cardíacos/metabolismo
7.
Cell Rep ; 24(13): 3582-3592, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30257217

RESUMO

To assess the utility of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as an in vitro proarrhythmia model, we evaluated the concentration dependence and sources of variability of electrophysiologic responses to 28 drugs linked to low, intermediate, and high torsades de pointes (TdP) risk categories using two commercial cell lines and standardized protocols in a blinded multisite study using multielectrode array or voltage-sensing optical approaches. Logistical and ordinal linear regression models were constructed using drug responses as predictors and TdP risk categories as outcomes. Three of seven predictors (drug-induced arrhythmia-like events and prolongation of repolarization at either maximum tested or maximal clinical exposures) categorized drugs with reasonable accuracy (area under the curve values of receiver operator curves ∼0.8). hiPSC-CM line, test site, and platform had minimal influence on drug categorization. These results demonstrate the utility of hiPSC-CMs to detect drug-induced proarrhythmic effects as part of the evolving Comprehensive In Vitro Proarrhythmia Assay paradigm.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Eletrofisiologia/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/efeitos dos fármacos , Torsades de Pointes/induzido quimicamente , Cardiotoxicidade , Linhagem Celular , Reprogramação Celular , Avaliação Pré-Clínica de Medicamentos/normas , Eletrofisiologia/normas , Humanos , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia
8.
Stem Cell Reports ; 9(2): 499-512, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28757167

RESUMO

Liver or hepatocytes transplantation is limited by the availability of donor organs. Functional hepatocytes independent of the donor sources may have wide applications in regenerative medicine and the drug industry. Recent studies have demonstrated that chemical cocktails may induce reprogramming of fibroblasts into a range of functional somatic cells. Here, we show that mouse fibroblasts can be transdifferentiated into the hepatocyte-like cells (iHeps) using only one transcription factor (TF) (Foxa1, Foxa2, or Foxa3) plus a chemical cocktail. These iHeps show typical epithelial morphology, express multiple hepatocyte-specific genes, and acquire hepatocyte functions. Genetic lineage tracing confirms the fibroblast origin of these iHeps. More interestingly, these iHeps are expandable in vitro and can reconstitute the damaged hepatic tissues of the fumarylacetoacetate hydrolase-deficient (Fah-/-) mice. Our study provides a strategy to generate functional hepatocyte-like cells by using a single TF plus a chemical cocktail and is one step closer to generate the full-chemical iHeps.


Assuntos
Transdiferenciação Celular/genética , Reprogramação Celular/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Fatores de Transcrição/genética , Animais , Biomarcadores , Linhagem da Célula , Transplante de Células , Células Cultivadas , Imunofluorescência , Expressão Gênica , Hidrolases/deficiência , Imunofenotipagem , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Medicina Regenerativa , Fatores de Transcrição/metabolismo
10.
Stem Cell Reports ; 8(4): 1101-1111, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28410643

RESUMO

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) offers the possibility of studying the molecular mechanisms underlying human diseases in cell types difficult to extract from living patients, such as neurons and cardiomyocytes. To date, studies have been published that use small panels of iPSC-derived cell lines to study monogenic diseases. However, to study complex diseases, where the genetic variation underlying the disorder is unknown, a sizable number of patient-specific iPSC lines and controls need to be generated. Currently the methods for deriving and characterizing iPSCs are time consuming, expensive, and, in some cases, descriptive but not quantitative. Here we set out to develop a set of simple methods that reduce cost and increase throughput in the characterization of iPSC lines. Specifically, we outline methods for high-throughput quantification of surface markers, gene expression analysis of in vitro differentiation potential, and evaluation of karyotype with markedly reduced cost.


Assuntos
Variação Genética , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariotipagem/métodos , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Reprogramação Celular/genética , Análise Custo-Benefício , Genótipo , Ensaios de Triagem em Larga Escala/economia , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Cariotipagem/economia , Miócitos Cardíacos/citologia , Neurônios/citologia , Fenótipo
11.
Stem Cell Reports ; 6(6): 873-884, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27161365

RESUMO

We previously reported the generation of integration-free induced pluripotent stem cells from adult peripheral blood (PB) with an improved episomal vector (EV) system, which uses the spleen focus-forming virus U3 promoter and an extra factor BCL-XL (B). Here we show an ∼100-fold increase in efficiency by optimizing the vector combination. The two most critical factors are: (1) equimolar expression of OCT4 (O) and SOX2 (S), by using a 2A linker; (2) a higher and gradual increase in the MYC (M) to KLF4 (K) ratio during the course of reprogramming, by using two individual vectors to express M and K instead of one. The combination of EV plasmids (OS + M + K + B) is comparable with Sendai virus in reprogramming efficiency but at a fraction of the cost. The generated iPSCs are indistinguishable from those from our previous approach in pluripotency and phenotype. This improvement lays the foundation for broad applications of episomal vectors in PB reprogramming.


Assuntos
Reprogramação Celular , Engenharia Genética/métodos , Vetores Genéticos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Plasmídeos/metabolismo , Adulto , Biomarcadores/metabolismo , Diferenciação Celular , Expressão Gênica , Engenharia Genética/economia , Vetores Genéticos/química , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Leucócitos Mononucleares/citologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Plasmídeos/química , Cultura Primária de Células , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
12.
Sci Rep ; 5: 11319, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-26066579

RESUMO

Factors limiting the adoption of iPSC technology include the cost of developing lines and the time period that it takes to characterize and bank them, particularly when integration free, feeder free, and Xeno-free components are used. In this manuscript we describe our optimization procedure that enables a single technician to make 20-40 lines at a time in a 24-96 well format in a reliable and reproducible fashion. Improvements spanned the entire workflow and included using RNA virus, reducing cytotoxicity of reagents, developing improved transfection and freezing efficiencies, modifying the manual colony picking steps, enhancing passaging efficiency and developing early criteria of success. These modifications allowed us to make more than two hundred well-characterized lines per year.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Reprogramação Celular/métodos , Reprogramação Celular , Meios de Cultura/química , Células-Tronco Pluripotentes Induzidas , Técnicas de Cultura de Células/economia , Linhagem Celular , Técnicas de Reprogramação Celular/economia , Humanos
13.
Circ Res ; 116(8): 1378-91, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858064

RESUMO

The discovery of induced pluripotent stem cells changed the field of regenerative medicine and inspired the technological development of direct reprogramming or the process by which one cell type is directly converted into another without reverting a stem cell state by overexpressing lineage-specific factors. Indeed, direct reprogramming has proven sufficient in yielding a diverse range of cell types from fibroblasts, including neurons, cardiomyocytes, endothelial cells, hematopoietic stem/progenitor cells, and hepatocytes. These studies revealed that somatic cells are more plastic than anticipated, and that transcription factors, microRNAs, epigenetic factors, secreted molecules, as well as the cellular microenvironment are all important for cell fate specification. With respect to the field of cardiology, the cardiac reprogramming presents as a novel method to regenerate damaged myocardium by directly converting endogenous cardiac fibroblasts into induced cardiomyocyte-like cells in situ. The first in vivo cardiac reprogramming reports were promising to repair infarcted hearts; however, the low induction efficiency of fully reprogrammed, functional induced cardiomyocyte-like cells has become a major challenge and hampered our understanding of the reprogramming process. Nevertheless, recent studies have identified several critical factors that may affect the efficiency and quality of cardiac induction and have provided new insights into the mechanisms of cardiac reprogramming. Here, we review the progress in direct reprogramming research and discuss the perspectives and challenges of this nascent technology in basic biology and clinical applications.


Assuntos
Reprogramação Celular , Cardiopatias/cirurgia , Células-Tronco Pluripotentes Induzidas/transplante , Miocárdio/patologia , Miócitos Cardíacos/transplante , Regeneração , Medicina Regenerativa/métodos , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Resultado do Tratamento
16.
Vet J ; 198(1): 34-42, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24129109

RESUMO

Induced pluripotent stem cells (iPSCs) can now be derived from a tissue biopsy and represent a promising new platform for disease modelling, drug and toxicity testing, biomarker development and cell-based therapies for regenerative medicine. In regenerative medicine, large animals may represent the best models for man, and thereby provide invaluable systems in which to test the safety and the potential of iPSCs. Hence, testing iPSCs in veterinary species may serve a double function, namely, developing therapeutic products for regenerative medicine in veterinary patients while providing valuable background information for human clinical trials. The production of iPSCs from livestock or wild species is attractive because it could improve efficiency and reduce costs in various fields, such as transgenic animal generation and drug development, preservation of biological diversity, and because it also offers an alternative to xenotransplantation for in vivo generation of organs. Although the technology of cellular reprogramming using the so-called 'Yamanaka factors' is in its peak expectation phase and many concerns still need to be addressed, the rapid technical progress suggests that iPSCs could contribute significantly to novel therapies in veterinary and biomedical practice in the near future. This review provides an overview of the potential applications of iPSCs in veterinary medicine.


Assuntos
Doenças dos Animais/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Medicina Regenerativa/métodos , Medicina Veterinária/métodos , Animais , Diferenciação Celular , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Animais , Medicina Regenerativa/economia , Medicina Veterinária/economia
19.
BMC Syst Biol ; 5 Suppl 2: S8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22784579

RESUMO

BACKGROUND: Somatic cells can be reprogrammed to induced-pluripotent stem cells (iPSCs) by introducing few reprogramming factors, which challenges the long held view that cell differentiation is irreversible. However, the mechanism of induced pluripotency is still unknown. METHODS: Inspired by the phenomenological reprogramming model of Artyomov et al (2010), we proposed a novel Markov model, stepwise reprogramming Markov (SRM) model, with simpler gene regulation rules and explored various properties of the model with Monte Carlo simulation. We calculated the reprogramming rate and showed that it would increase in the condition of knockdown of somatic transcription factors or inhibition of DNA methylation globally, consistent with the real reprogramming experiments. Furthermore, we demonstrated the utility of our model by testing it with the real dynamic gene expression data spanning across different intermediate stages in the iPS reprogramming process. RESULTS: The gene expression data at several stages in reprogramming and the reprogramming rate under several typically experiment conditions coincided with our simulation results. The function of reprogramming factors and gene expression change during reprogramming could be partly explained by our model reasonably well. CONCLUSIONS: This lands further support on our general rules of gene regulation network in iPSC reprogramming. This model may help uncover the basic mechanism of reprogramming and improve the efficiency of converting somatic cells to iPSCs.


Assuntos
Reprogramação Celular/genética , Perfilação da Expressão Gênica/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cadeias de Markov , Ciclo Celular , Diferenciação Celular/genética , Simulação por Computador , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Toxicology ; 270(1): 18-34, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-19948204

RESUMO

Given the reality of the inadequacies of current concepts of the mechanisms of chemical toxicities, of the various assays to predict toxicities from current molecular, biochemical, in vitro and animal bioassays, and of the failure to generate efficacious and safe chemicals for medicines, food supplements, industrial, consumer and agricultural chemicals, the recent NAS Report, "Toxicity Testing in the 21st Century: A Vision and a Strategy", has drawn attention to a renewed examination of what needs to be done to improve our current approach for better assessment of potential risk to human health. This "Commentary" provides a major paradigm challenge to the current concepts of how chemicals induce toxicities and how these various mechanisms of toxicities can contribute to the pathogenesis of some human diseases, such as birth defects and cancer. In concordance with the NAS Report to take "... advantage of the on-going revolution in biology and biotechnology", this "Commentary" supports the use of human embryonic and adult stem cells, grown in vitro under simulated "in vivo niche conditions". The human being should be viewed "as greater than the sum of its parts". Homeostatic control of the "emergent properties" of the human hierarchy, needed to maintain human health, requires complex integration of endogenous and exogenous signaling molecules that control cell proliferation, differentiation, apoptosis and senescence of stem, progenitor and differentiated cells. Currently, in vitro toxicity assays (mutagenesis, cytotoxicity, epigenetic modulation), done on 2-dimensional primary rodent or human cells (which are always mixtures of cells), on immortalized or tumorigenic rodent or human cell lines do not represent normal human cells in vivo [which do not grow on plastic and which are in micro-environments representing 3 dimensions and constantly interacting factors]. In addition, with the known genetic, gender, and developmental state of cells in vivo, any in vitro toxicity assay will need to mimic these conditions in vitro. More specifically, while tissues contain a few stem cells, many progenitor/transit cells and terminally differentiated cells, it should be obvious that both embryonic and adult stem cells would be critical "target" cells for toxicity testing. The ultimate potential for in vitro testing of human stem cells will to try to mimic a 3-D in vitro micro-environment on multiple "organ-specific and multiple genotypic/gender "adult stem cells. The role of stem cells in many chronic diseases, such as cancer, birth defects, and possibly adult diseases after pre-natal and early post-natal exposures (Barker hypothesis), demands toxicity studies of stem cells. While alteration of gene expression ("toxico-epigenomics") is a legitimate endpoint of these toxicity studies, alteration of the quantity of stem cells during development must be serious considered. If the future utility of human stem cells proves to be valid, the elimination of less relevant, expensive and time-consuming rodent and 2-D human in vitro assays will be eliminated.


Assuntos
Descoberta de Drogas/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Células-Tronco/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Indústria Farmacêutica , Células-Tronco Embrionárias , Epigênese Genética , Feminino , Humanos , Testes de Mutagenicidade , Gravidez , Segurança , Toxicologia/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA