Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 282
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 36(10): 1552-1566, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30004605

RESUMO

Although autologous induced pluripotent stem cells (iPSCs) can potentially be useful for treating patients without immune rejection, in reality it will be extremely expensive and labor-intensive to make iPSCs to realize personalized medicine. An alternative approach is to make use of human leukocyte antigen (HLA) haplotype homozygous donors to provide HLA matched iPSC products to significant numbers of patients. To establish a haplobank of iPSCs, we repurposed the cord blood bank by screening ∼4,200 high resolution HLA typed cord blood samples, and selected those homozygous for the 10 most frequent HLA-A,-B,-DRB1 haplotypes in the Korean population. Following the generation of 10 iPSC lines, we conducted a comprehensive characterization, including morphology, expression of pluripotent markers and cell surface antigens, three-germ layer formation, vector clearance, mycoplasma/microbiological/viral contamination, endotoxin, and short tandem repeat (STR) assays. Various genomic analyses using microarray and comparative genomic hybridization (aCGH)-based single nucleotide polymorphism (SNP) and copy number variation (CNV) were also conducted. These 10 HLA-homozygous iPSC lines match 41.07% of the Korean population. Comparative analysis of HLA population data shows that they are also of use in other Asian populations, such as Japan, with some limited utility in ethnically diverse populations, such as the UK. Taken together, the generation of the 10 most frequent Korean HLA-homozygous iPSC lines serves as a useful pointer for the development of optimal methods for iPSC generation and quality control and indicates the benefits and limitations of collaborative HLA driven selection of donors for future stocking of worldwide iPSC haplobanks. Stem Cells 2018;36:1552-1566.


Assuntos
Armazenamento de Sangue/métodos , Instabilidade Genômica/genética , Antígenos HLA/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Haplótipos , Antígenos de Histocompatibilidade Classe II , Humanos
2.
Psychiatry Clin Neurosci ; 73(1): 11-19, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30367527

RESUMO

AIM: Severe mental illnesses (SMI), such as bipolar disorder and schizophrenia, are highly heritable, and have a complex pattern of inheritance. Genome-wide association studies detect a part of the heritability, which can be attributed to common genetic variation. Examination of rare variants with next-generation sequencing may add to the understanding of the genetic architecture of SMI. METHODS: We analyzed 32 ill subjects from eight multiplex families and 33 healthy individuals using whole-exome sequencing. Prioritized variants were selected by a three-step filtering process, which included: deleteriousness by five in silico algorithms; sharing within families by affected individuals; rarity in South Asian sample estimated using the Exome Aggregation Consortium data; and complete absence of these variants in control individuals from the same gene pool. RESULTS: We identified 42 rare, non-synonymous deleterious variants (~5 per pedigree) in this study. None of the variants were shared across families, indicating a 'private' mutational profile. Twenty (47.6%) of the variant harboring genes were previously reported to contribute to the risk of diverse neuropsychiatric syndromes, nine (21.4%) of which were of Mendelian inheritance. These included genes carrying novel deleterious variants, such as the GRM1 gene implicated in spinocerebellar ataxia 44 and the NIPBL gene implicated in Cornelia de Lange syndrome. CONCLUSION: Next-generation sequencing approaches in family-based studies are useful to identify novel and rare variants in genes for complex disorders like SMI. The findings of the study suggest a potential phenotypic burden of rare variants in Mendelian disease genes, indicating pleiotropic effects in the etiology of SMI.


Assuntos
Transtorno Bipolar/genética , Exoma , Predisposição Genética para Doença , Esquizofrenia/genética , Feminino , Variação Genética , Estudo de Associação Genômica Ampla , Humanos , Masculino , Linhagem , Fenótipo
3.
Cytotherapy ; 20(6): 861-872, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29793831

RESUMO

BACKGROUND AIMS: We have previously reported the generation of a current Good Manufacture Practice (cGMP)-compliant induced pluripotent stem cell (iPSC) line for clinical applications. Here we show that multiple cellular products currently being considered for therapy can be generated from a single master cell bank of this or any other clinically compliant iPSC line METHODS: Using a stock at passage 20 prepared from the cGMP-compliant working cell bank (WCB), we tested differentiation into therapeutically relevant cell types of the three germ layers using standardized but generic protocols. Cells that we generated include (i) neural stem cells, dopaminergic neurons and astrocytes; (ii) retinal cells (retinal pigment epithelium and photoreceptors); and (iii) hepatocyte, endothelial and mesenchymal cells. To confirm that these generic protocols can also be used for other iPSC lines, we tested the reproducibility of our methodology with a second clinically compliant line RESULTS: Our results confirmed that well-characterized iPSC lines have broad potency, and, despite allelic variability, the same protocols could be used with minimal modifications with multiple qualified lines. In addition, we introduced a constitutively expressed GFP cassette in Chr13 safe harbor site using a standardized previously described method and observed no significant difference in growth and differentiation between the engineered line and the control line indicating that engineered products can be made using a standardized methodology CONCLUSIONS: We believe that our demonstration that multiple products can be made from the same WCB and that the same protocols can be used with multiple lines offers a path to a cost-effective strategy for developing cellular products from iPSC lines.


Assuntos
Engenharia Celular/métodos , Engenharia Celular/normas , Linhagem da Célula , Fidelidade a Diretrizes , Células-Tronco Pluripotentes Induzidas/citologia , Astrócitos/citologia , Astrócitos/fisiologia , Diferenciação Celular , Linhagem Celular , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Fidelidade a Diretrizes/normas , Hepatócitos/citologia , Hepatócitos/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Mesoderma/citologia , Mesoderma/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Guias de Prática Clínica como Assunto/normas , Padrões de Referência , Reprodutibilidade dos Testes , Retina/citologia , Bancos de Tecidos/normas
4.
BMC Psychiatry ; 18(1): 106, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29669557

RESUMO

BACKGROUND: There is emerging evidence that there are shared genetic, environmental and developmental risk factors in psychiatry, that cut across traditional diagnostic boundaries. With this background, the Discovery biology of neuropsychiatric syndromes (DBNS) proposes to recruit patients from five different syndromes (schizophrenia, bipolar disorder, obsessive compulsive disorder, Alzheimer's dementia and substance use disorders), identify those with multiple affected relatives, and invite these families to participate in this study. The families will be assessed: 1) To compare neuro-endophenotype measures between patients, first degree relatives (FDR) and healthy controls., 2) To identify cellular phenotypes which differentiate the groups., 3) To examine the longitudinal course of neuro-endophenotype measures., 4) To identify measures which correlate with outcome, and 5) To create a unified digital database and biorepository. METHODS: The identification of the index participants will occur at well-established specialty clinics. The selected individuals will have a strong family history (with at least another affected FDR) of mental illness. We will also recruit healthy controls without family history of such illness. All recruited individuals (N = 4500) will undergo brief clinical assessments and a blood sample will be drawn for isolation of DNA and peripheral blood mononuclear cells (PBMCs). From among this set, a subset of 1500 individuals (300 families and 300 controls) will be assessed on several additional assessments [detailed clinical assessments, endophenotype measures (neuroimaging- structural and functional, neuropsychology, psychophysics-electroencephalography, functional near infrared spectroscopy, eye movement tracking)], with the intention of conducting repeated measurements every alternate year. PBMCs from this set will be used to generate lymphoblastoid cell lines, and a subset of these would be converted to induced pluripotent stem cell lines and also undergo whole exome sequencing. DISCUSSION: We hope to identify unique and overlapping brain endophenotypes for major psychiatric syndromes. In a proportion of subjects, we expect these neuro-endophenotypes to progress over time and to predict treatment outcome. Similarly, cellular assays could differentiate cell lines derived from such groups. The repository of biomaterials as well as digital datasets of clinical parameters, will serve as a valuable resource for the broader scientific community who wish to address research questions in the area.


Assuntos
Predisposição Genética para Doença , Testes Genéticos/métodos , Leucócitos Mononucleares , Adulto , Transtorno Bipolar/diagnóstico , Eletroencefalografia , Feminino , Variação Genética/genética , Humanos , Masculino , Esquizofrenia/diagnóstico , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia
5.
Trends Immunol ; 35(4): 178-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24661777

RESUMO

Regenerative medicine holds great promise in replacing tissues and organs lost to degenerative disease and injury. Application of the principles of cellular reprogramming for the treatment of cancer, however, is not well established. Here, we present an overview of cellular reprogramming techniques used in regenerative medicine, and within this context, envision how the scope of regenerative medicine may be expanded to treat metastatic cancer by revitalizing an exhausted and senescent immune system.


Assuntos
Reprogramação Celular/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Animais , Humanos
6.
Stem Cells ; 33(12): 3504-18, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26235913

RESUMO

The derivation of three-dimensional (3D) stratified neural retina from pluripotent stem cells has permitted investigations of human photoreceptors. We have generated a H9 human embryonic stem cell subclone that carries a green fluorescent protein (GFP) reporter under the control of the promoter of cone-rod homeobox (CRX), an established marker of postmitotic photoreceptor precursors. The CRXp-GFP reporter replicates endogenous CRX expression in vitro when the H9 subclone is induced to form self-organizing 3D retina-like tissue. At day 37, CRX+ photoreceptors appear in the basal or middle part of neural retina and migrate to apical side by day 67. Temporal and spatial patterns of retinal cell type markers recapitulate the predicted sequence of development. Cone gene expression is concomitant with CRX, whereas rod differentiation factor neural retina leucine zipper protein (NRL) is first observed at day 67. At day 90, robust expression of NRL and its target nuclear receptor NR2E3 is evident in many CRX+ cells, while minimal S-opsin and no rhodopsin or L/M-opsin is present. The transcriptome profile, by RNA-seq, of developing human photoreceptors is remarkably concordant with mRNA and immunohistochemistry data available for human fetal retina although many targets of CRX, including phototransduction genes, exhibit a significant delay in expression. We report on temporal changes in gene signatures, including expression of cell surface markers and transcription factors; these expression changes should assist in isolation of photoreceptors at distinct stages of differentiation and in delineating coexpression networks. Our studies establish the first global expression database of developing human photoreceptors, providing a reference map for functional studies in retinal cultures.


Assuntos
Antígenos de Diferenciação/biossíntese , Diferenciação Celular , Proteínas do Olho/biossíntese , Células-Tronco Embrionárias Humanas/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Transcriptoma , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Bastonetes/citologia
7.
J Biol Chem ; 289(8): 4553-4, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24362035

RESUMO

With the advent of human induced pluripotent stem cell (hiPSC) technology, it is now possible to derive patient-specific cell lines that are of great potential in both basic research and the development of new therapeutics for human diseases. Not only do hiPSCs offer unprecedented opportunities to study cellular differentiation and model human diseases, but the differentiated cell types obtained from iPSCs may become therapeutics themselves. These cells can also be used in the screening of therapeutics and in toxicology assays for potential liabilities of therapeutic agents. The remarkable achievement of transcription factor reprogramming to generate iPSCs was recognized by the award of the Nobel Prize in Medicine to Shinya Yamanaka in 2012, just 6 years after the first publication of reprogramming methods to generate hiPSCs (Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., and Yamanaka, S. (2007) Cell 131, 861-872). This minireview series highlights both the promises and challenges of using iPSC technology for disease modeling, drug screening, and the development of stem cell therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco , Avaliação Pré-Clínica de Medicamentos , Humanos
8.
Cytotherapy ; 17(6): 756-764, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25770678

RESUMO

The discovery of induced pluripotent stem cells (iPSCs) and the rapid evolution of clinically compliant protocols to generate such lines from a variety of tissue sources has raised the possibility that personalized medicine may be achievable in the near future. Several strategies to deliver iPSCs for iPSC-derived cell-based therapy have been proposed: one such model has been the cell-banking model, using processes developed by the cord blood industry. The cord blood industry has evolved primarily as a banking model in which units of cord blood harvested from discarded placenta are stored either in a public or a private cord blood bank for future use. The consideration of a cord blood--like banking model has been further spurred by the realization that this population of cells is an ideal starting sample to generate pluripotent cells. Spurred by these technological advances, major efforts are underway to develop a current Good Manufacturing Practice--compliant protocol to generate iPSCs from cord blood and to develop a haplobanking strategy. In this article, we discuss the issues that may affect such an effort.


Assuntos
Bancos de Sangue , Sangue Fetal/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Transplante de Células-Tronco
9.
Cytotherapy ; 17(9): 1169-77, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26276001

RESUMO

Mesenchymal stem/stromal cells (MSCs) have been extensively investigated for their regenerative, immune-modulatory, and wound healing properties. While the laboratory studies have suggested that MSC's have a unique potential for modulating the etiopathology of multiple diseases, the results from clinical trials have not been encouraging or reproducible. One of the explanations for such variability is explained by the "art" of isolating and propagating MSCs. Therefore, establishing more than minimal criteria to define MSC would help understand best protocols to isolate, propagate and deliver MSCs. Developing a calibration standard, a database and a set of functional tests would be a better quality metric for MSCs. In this review, we discuss the importance of selecting a standard, issues associated with coming up with such a standard and how these issues can be mitigated.


Assuntos
Separação Celular/normas , Células-Tronco Mesenquimais/citologia , Técnicas de Cultura de Células , Células Cultivadas , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Padrões de Referência
10.
Toxicol Pathol ; 43(1): 115-25, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25476796

RESUMO

Cell-based therapies have the potential to treat a diversity of disease conditions, many representing significant and long-standing unmet medical needs. Certain properties of cell-based therapies, such as differentiation potential and proliferative potential, present safety concerns uniquely distinct from those of small molecule drugs and other macromolecule biologics. These cellular products carry risks associated with localized host tissue response, long-term persistence, ectopic tissue formation, differentiation to undesirable cell and tissue types, uncontrollable biodistribution, tumorigenicity, and immunogenicity. Such risks are generally evaluated in preclinical animal model studies as part of a comprehensive safety program prior to administration in humans. However, safety assessment for these products can be challenging because of inconsistent approaches to product characterization, inadequately defined product parameters that anticipate adverse events, and the lack of standardized approaches in evaluating in vivo host responses. In this symposium, we introduced cell-based therapies as an emerging product class to the Society of Toxicologic Pathology (STP) and highlighted key challenges for consideration during product biosafety evaluation.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Medicina Regenerativa/métodos , Medicina Regenerativa/normas , Engenharia Tecidual , Animais , Terapia Baseada em Transplante de Células e Tecidos/normas , Rejeição de Enxerto/prevenção & controle , Humanos , Células-Tronco Mesenquimais/citologia , Segurança
11.
Biologicals ; 43(5): 283-97, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26315651

RESUMO

The regulation of human cell therapy products is a key factor in their development and use to treat human diseases. In that regard, there is a recognized need for a global effort to develop a set of common principles that may serve to facilitate a convergence of regulatory approaches to ensure the smooth and efficient evaluation of products. This conference, with experts from regulatory agencies, industry, and academia, contributed to the process of developing such a document. Elements that could form a minimum consensus package of requirements for evaluating human cell therapy products were the overall focus of the conference. The important regulatory considerations that are unique to human cell therapy products were highlighted. Sessions addressed specific points that are different from those of traditional biological/biotechnological protein products. Panel discussions complemented the presentations. The conference concluded that most of the current regulatory framework is appropriate for cell therapy, but there are some areas where the application of the requirements for traditional biologicals is inappropriate. In addition, it was agreed that there is a need for international consensus on core regulatory elements, and that one of the major international organizations should take the lead in formulating such a consensus document.


Assuntos
Biotecnologia/legislação & jurisprudência , Terapia Baseada em Transplante de Células e Tecidos , Produtos Biológicos , Humanos
13.
Stem Cells ; 31(5): 941-52, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23341249

RESUMO

Astrocytes can be generated from various tissue sources including human pluripotent stem cells (PSC). In this manuscript, we describe a chemically defined xeno-free medium culture system for rapidly generating astrocytes from neural stem cells derived from PSC. We show that astrocyte development in vitro, mimics normal development in vivo, and also passes through a CD44(+) astrocyte precursor stage. Astrocytes generated by our method display similar gene expression patterns, morphological characteristics and functional properties to primary astrocytes, and they survive and integrate after xenotransplantation. Whole genome expression profiling of astrocyte differentiation was performed at several time points of differentiation, and the results indicate the importance of known regulators and identify potential novel regulators and stage-specific lineage markers.


Assuntos
Astrócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Animais , Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Criopreservação , Técnicas Citológicas/métodos , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transplante Heterólogo
14.
Cytotherapy ; 16(9): 1305-12, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25065637

RESUMO

BACKGROUND AIMS: We have previously reported a Good Manufacturing Practice (GMP)-compatible process for generating authentic dopaminergic neurons in defined media from human pluripotent stem cells and determined the time point at which dopaminergic precursors/neurons (day 14 after neuronal stem cell [NSC] stage) can be frozen, shipped and thawed without compromising their viability and ability to mature in vitro. One important issue we wished to address is whether dopaminergic precursors/neurons manufactured by our GMP-compatible process can be cryopreserved and engrafted in animal Parkinson disease (PD) models. METHODS: In this study, we evaluated the efficacy of freshly prepared and cryopreserved dopaminergic neurons in the 6-hydroxydopamine-lesioned rat PD model. RESULTS: We showed functional recovery up to 6 months post-transplantation in rats transplanted with our cells, whether freshly prepared or cryopreserved. In contrast, no motor improvement was observed in two control groups receiving either medium or cells at a slightly earlier stage (day 10 after NSC stage). Histologic analysis at the end point of the study (6 months post-transplantation) showed robust long-term survival of donor-derived tyrosine hydroxylase (TH)(+) dopaminergic neurons in rats transplanted with day 14 dopaminergic neurons. Moreover, TH(+) fibers emanated from the graft core into the surrounding host striatum. Consistent with the behavioral analysis, no or few TH(+) neurons were detected in animals receiving day 10 cells, although human cells were present in the graft. Importantly, no tumors were detected in any grafted rats, but long-term tumorigenic studies will need to determine the safety of our products. CONCLUSIONS: Dopaminergic neurons manufactured by a GMP-compatible process from human ESC survived and engrafted efficiently in the 6-OHDA PD rat model.


Assuntos
Criopreservação/métodos , Neurônios Dopaminérgicos/fisiologia , Células-Tronco Neurais/fisiologia , Doença de Parkinson/terapia , Transplante de Células-Tronco , Animais , Biotecnologia , Linhagem Celular , Sobrevivência Celular , Corpo Estriado/patologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/patologia , Feminino , Humanos , Indústria Manufatureira , Fibras Nervosas/patologia , Células-Tronco Neurais/patologia , Neurogênese , Oxidopamina/administração & dosagem , Oxidopamina/efeitos adversos , Doença de Parkinson/etiologia , Guias de Prática Clínica como Assunto , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Transplante Heterólogo
15.
Nature ; 455(7211): 401-5, 2008 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-18724358

RESUMO

Stem cells are defined as self-renewing cell populations that can differentiate into multiple distinct cell types. However, hundreds of different human cell lines from embryonic, fetal and adult sources have been called stem cells, even though they range from pluripotent cells-typified by embryonic stem cells, which are capable of virtually unlimited proliferation and differentiation-to adult stem cell lines, which can generate a far more limited repertoire of differentiated cell types. The rapid increase in reports of new sources of stem cells and their anticipated value to regenerative medicine has highlighted the need for a general, reproducible method for classification of these cells. We report here the creation and analysis of a database of global gene expression profiles (which we call the 'stem cell matrix') that enables the classification of cultured human stem cells in the context of a wide variety of pluripotent, multipotent and differentiated cell types. Using an unsupervised clustering method to categorize a collection of approximately 150 cell samples, we discovered that pluripotent stem cell lines group together, whereas other cell types, including brain-derived neural stem cell lines, are very diverse. Using further bioinformatic analysis we uncovered a protein-protein network (PluriNet) that is shared by the pluripotent cells (embryonic stem cells, embryonal carcinomas and induced pluripotent cells). Analysis of published data showed that the PluriNet seems to be a common characteristic of pluripotent cells, including mouse embryonic stem and induced pluripotent cells and human oocytes. Our results offer a new strategy for classifying stem cells and support the idea that pluripotency and self-renewal are under tight control by specific molecular networks.


Assuntos
Perfilação da Expressão Gênica , Células-Tronco/classificação , Células-Tronco/metabolismo , Algoritmos , Animais , Inteligência Artificial , Diferenciação Celular , Linhagem Celular , Biologia Computacional , Bases de Dados Factuais , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Células-Tronco Multipotentes/classificação , Células-Tronco Multipotentes/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oócitos/classificação , Oócitos/metabolismo , Fenótipo , Células-Tronco Pluripotentes/classificação , Células-Tronco Pluripotentes/metabolismo , Ligação Proteica
16.
Nat Genet ; 37(10): 1099-103, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16142235

RESUMO

Cultured human embryonic stem cell (hESC) lines are an invaluable resource because they provide a uniform and stable genetic system for functional analyses and therapeutic applications. Nevertheless, these dividing cells, like other cells, probably undergo spontaneous mutation at a rate of 10(-9) per nucleotide. Because each mutant has only a few progeny, the overall biological properties of the cell culture are not altered unless a mutation provides a survival or growth advantage. Clonal evolution that leads to emergence of a dominant mutant genotype may potentially affect cellular phenotype as well. We assessed the genomic fidelity of paired early- and late-passage hESC lines in the course of tissue culture. Relative to early-passage lines, eight of nine late-passage hESC lines had one or more genomic alterations commonly observed in human cancers, including aberrations in copy number (45%), mitochondrial DNA sequence (22%) and gene promoter methylation (90%), although the latter was essentially restricted to 2 of 14 promoters examined. The observation that hESC lines maintained in vitro develop genetic and epigenetic alterations implies that periodic monitoring of these lines will be required before they are used in in vivo applications and that some late-passage hESC lines may be unusable for therapeutic purposes.


Assuntos
Embrião de Mamíferos/citologia , Genoma Humano/genética , Mutação , Células-Tronco/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , DNA/genética , DNA/metabolismo , Metilação de DNA , DNA Mitocondrial/química , Humanos , Regiões Promotoras Genéticas
17.
Cells ; 13(7)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38607035

RESUMO

Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofias Musculares , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Músculo Esquelético/fisiologia , Distrofias Musculares/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Diferenciação Celular
18.
Stem Cells ; 30(1): 55-60, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22069231

RESUMO

Hematopoietic cell transplantation (HCT) has become a standard practice to treat a number of malignant and nonmalignant hematologic diseases. Bone marrow, mobilized peripheral blood, and umbilical cord blood can all serve as primary sources of cells for HCT. The number of cord blood units currently stored is large, although it represents only a fraction of potential collections. With much of the collection being sequestered in private banks for possible autologous use, there is a reason to expect that public banks may not be able to provide for the demand in coming years as use of cord blood for treatment of patients with diseases such as leukemia and lymphoma continues to increase. We suggest that a possible solution to encourage private banks to share their valuable units is to apply recent methodologies to generate induced pluripotent stem cells from cord cells and to optimize techniques to generate hematopoietic lineages from them. This strategy would allow us to take advantage of the units already collected under appropriate regulatory guidelines, to access a pristine cell that can be converted to a pluripotent cell at a much higher efficiency and in a shorter time period than other cells. The ability to potentially replenish a used cord unit with new cells, as well as extend the potential utility of cord blood for additional therapeutic applications, should allow banks to develop an appropriate business model for both private and public cord blood banks to flourish.


Assuntos
Bancos de Sangue , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Bancos de Sangue/ética , Medula Óssea/fisiologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Setor Privado , Setor Público , Transplante Autólogo
19.
Cytotherapy ; 15(8): 999-1010, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23664011

RESUMO

BACKGROUND AIMS: We have previously described a xeno-free scalable system to generate transplantable dopaminergic neurons from human pluripotent stem cells. However, several important questions remain to be answered about our cell therapy efforts. These include determining the exact time at which cells should be transplanted and whether cells at this stage can be frozen, shipped, thawed and injected without compromising their ability to mature and survive the transplantation procedure. We also needed to determine whether further optimization of the culture process could shorten the development time and reduce variability and whether a current Good Manufacture Practice (CGMP) facility could manufacture cells with fidelity. METHODS: We developed an optimized protocol that included modulating the sonic hedgehog homolog gradient with bone morphogenetic proteins (BMP2) and addition of activin to the culture medium, which shortened the time to generate Lmx1A and FoxA2 immunoreactive cells by 4-6 days. RESULTS: We showed that cells at this stage could be safely frozen and thawed while retaining an excellent ability to continue to mature in vitro and survive transplant in vivo. Importantly, we successfully adapted this process to a CGMP facility and manufactured two lots of transplant-ready dopaminergic neurons (>250 vials) under CGMP-compatible conditions. In vitro characterization, including viability/recovery on thawing, whole genome expression as well as expression of midbrain/dopaminergic markers, showed that the cells manufactured under GMP-compatible conditions were similar to cells produced at lab scale. CONCLUSIONS: Our results suggest that this optimized protocol can be used to generate dopaminergic neurons for Investigational New Drug enabling studies.


Assuntos
Técnicas de Cultura de Células , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/transplante , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurogênese , Células-Tronco Pluripotentes/citologia , Ativinas , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proliferação de Células , Sobrevivência Celular , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Criopreservação/métodos , Dopamina/análise , Dopamina/biossíntese , Descoberta de Drogas/métodos , Proteínas Hedgehog/metabolismo , Fator 3-beta Nuclear de Hepatócito/imunologia , Humanos , Proteínas com Homeodomínio LIM/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/imunologia
20.
J Cell Biochem ; 113(10): 3061-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22573568

RESUMO

The discovery of the ability to induce somatic cells to a pluripotent state through the overexpression of specific transcription factors has the potential to transform the ways in which pharmaceutical agents and cellular transplantation therapies are developed. Proper utilization of the technology to generate induced pluripotent stem cells (iPSCs) requires that researchers select the appropriate reprogramming method for generating iPSCs so that the resulting iPSCs can be transitioned towards clinical applications effectively. This article reviews all of the currently available reprogramming techniques with a focus on critiquing them on the basis of their utility in translational medicine.


Assuntos
Diferenciação Celular , Engenharia Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Pesquisa Translacional Biomédica/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Medicina Regenerativa , Reprodutibilidade dos Testes , Transfecção , Transgenes
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa