Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(1)2019 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-31877913

RESUMO

The clinical effectiveness of human induced pluripotent stem cells (iPSCs) is highly dependent on a few key quality characteristics including the generation of high quality cell bank, long-term genomic stability, post-thaw viability, plating efficiency, retention of pluripotency, directed differentiation, purity, potency, and sterility. We have already reported the establishment of iPSC master cell banks (MCBs) and working cell banks (WCBs) under current good manufacturing procedure (cGMP)-compliant conditions. In this study, we assessed the cellular and genomic stability of the iPSC lines generated and cryopreserved five years ago under cGMP-compliant conditions. iPSC lines were thawed, characterized, and directly differentiated into cells from three germ layers including cardiomyocytes (CMs), neural stem cells (NSCs), and definitive endoderm (DE). The cells were also expanded in 2D and 3D spinner flasks to evaluate their long-term expansion potential in matrix-dependent and feeder-free culture environment. All three lines successfully thawed and attached to the L7TM matrix, and formed typical iPSC colonies that expressed pluripotency markers over 15 passages. iPSCs maintained their differentiation potential as demonstrated with spontaneous and directed differentiation to the three germ layers and corresponding expression of specific markers, respectfully. Furthermore, post-thaw cells showed normal karyotype, negative mycoplasma, and sterility testing. These cells maintained both their 2D and 3D proliferation potential after five years of cryopreservation without acquiring karyotype abnormality, loss of pluripotency, and telomerase activity. These results illustrate the long-term stability of cGMP iPSC lines, which is an important step in establishing a reliable, long-term source of starting materials for clinical and commercial manufacturing of iPSC-derived cell therapy products.


Assuntos
Criopreservação , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fatores de Tempo
2.
PLoS One ; 11(9): e0161229, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27606941

RESUMO

Human pluripotent stem cells (hPSCs) present an unprecedented opportunity to advance human health by offering an alternative and renewable cell resource for cellular therapeutics and regenerative medicine. The present demand for high quality hPSCs for use in both research and clinical studies underscores the need to develop technologies that will simplify the cultivation process and control variability. Here we describe the development of a robust, defined and xeno-free hPSC medium that supports reliable propagation of hPSCs and generation of human induced pluripotent stem cells (hiPSCs) from multiple somatic cell types; long-term serial subculturing of hPSCs with every-other-day (EOD) medium replacement; and banking fully characterized hPSCs. The hPSCs cultured in this medium for over 40 passages are genetically stable, retain high expression levels of the pluripotency markers TRA-1-60, TRA-1-81, Oct-3/4 and SSEA-4, and readily differentiate into ectoderm, mesoderm and endoderm. Importantly, the medium plays an integral role in establishing a cGMP-compliant process for the manufacturing of hiPSCs that can be used for generation of clinically relevant cell types for cell replacement therapy applications.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/farmacologia , Células-Tronco Pluripotentes/citologia , Animais , Biomarcadores/metabolismo , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos SCID , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Fatores de Tempo
3.
Stem Cell Reports ; 5(4): 647-59, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26411904

RESUMO

The discovery of induced pluripotent stem cells (iPSCs) and the concurrent development of protocols for their cell-type-specific differentiation have revolutionized our approach to cell therapy. It has now become critical to address the challenges related to the generation of iPSCs under current good manufacturing practice (cGMP) compliant conditions, including tissue sourcing, manufacturing, testing, and storage. Furthermore, regarding the technical challenges, it is very important to keep the costs of manufacturing and testing reasonable and solve logistic hurdles that permit the global distribution of these products. Here we describe our efforts to develop a process for the manufacturing of iPSC master cell banks (MCBs) under cGMPs and announce the availability of such banks.


Assuntos
Biotecnologia/métodos , Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos , Marcação de Genes/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Transplante de Células-Tronco , Bancos de Tecidos
4.
Methods Mol Biol ; 767: 433-47, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21822894

RESUMO

Efficient derivation and isolation of hematopoietic stem cells (HSCs) from human pluripotent stem cell (hPSC) populations remains a major goal in the field of developmental hematopoiesis. These enticing pluripotent stem cells (comprising both human embryonic stem cells and induced pluripotent stem cells) have been successfully used to generate a wide array of hematopoietic cells in vitro, from primitive hematoendothelial precursors to mature myeloid, erythroid, and lymphoid lineage cells. However, to date, PSC-derived cells have demonstrated only limited potential for long-term multilineage hematopoietic engraftment in vivo - the test by which putative HSCs are defined. Successful generation and characterization of HSCs from hPSCs not only requires an efficient in vitro differentiation system that provides insight into the developmental fate of hPSC-derived cells, but also necessitates an in vivo engraftment model that allows identification of specific mechanisms that hinder or promote hematopoietic engraftment. In this chapter, we will describe a method that utilizes firefly luciferase-expressing hPSCs and bioluminescent imaging to noninvasively track the survival, proliferation, and migration of transplanted hPSC-derived cells. Combined with lineage and functional analyses of engrafted cells, this system is a useful tool to gain insight into the in vivo potential of hematopoietic cells generated from hPSCs.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Células Sanguíneas/citologia , Células da Medula Óssea/citologia , Diferenciação Celular , Separação Celular , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imageamento Tridimensional , Fígado/citologia , Fígado/metabolismo , Luciferases/metabolismo , Medições Luminescentes , Camundongos , Células-Tronco Pluripotentes/metabolismo , Baço/citologia
5.
Stem Cells ; 27(11): 2675-85, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19711457

RESUMO

Human embryonic stem cells (hESCs) provide an important resource for novel regenerative medicine therapies and have been used to derive diverse cell populations, including hematopoietic and endothelial cells. However, it remains a challenge to achieve significant engraftment of hESC-derived blood cells when transplanted into animal models. To better understand mechanisms that enhance or limit the in vivo developmental potential of hESC-derived cells, we utilized hESCs that express firefly luciferase (luc) to allow noninvasive, real-time bioluminescent imaging of hESC-derived CD34(+) cells transplanted into the liver of neonatal immunodeficient mice. Serial imaging demonstrated stable engraftment and expansion of the luc(+) hESC-derived cells in vivo over several months. While we found that these hESC-derived CD34(+) cells have bipotential ability to generate both hematopoietic and endothelial lineages in vitro, these studies demonstrate preferential differentiation into endothelial cells in vivo, with only low levels of hematopoietic cell engraftment. Therefore, these studies reveal key differences in the developmental potential of hESC-derived cells using in vitro and in vivo analyses. Although transplanted hESC-derived CD34(+) cells are well-suited for revascularization therapies, additional measures are needed to provide higher levels of long-term hematopoietic engraftment.


Assuntos
Antígenos CD34/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hematopoese/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Células Cultivadas , Células-Tronco Embrionárias/transplante , Sangue Fetal/citologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Fígado/metabolismo , Medições Luminescentes , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
Blood ; 113(24): 6094-101, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19365083

RESUMO

Natural killer (NK) cells serve as important effectors for antitumor immunity, and CD56+CD45+ NK cells can be routinely derived from human embryonic stem cells (hESCs). However, little is know about the ability of hESC-derived NK cells to mediate an effective in vivo antitumor response. Using bioluminescent imaging, we now demonstrate that H9 line hESC-derived NK cells mediate effective clearance of human tumor cells in vivo. In addition to increased in vitro killing of diverse tumor targets, the in vivo tumor clearance by H9 hESC-derived NK cells was more effective compared with NK cells derived from umbilical cord blood (UCB). Phenotypic analysis demonstrates the hESC-derived NK cells are uniformly CD94+CD117(low/-), an NK-cell population characterized by potent cytolytic activity and thus more competent to mediate tumor clearance. These studies demonstrate that hESCs provide an important model to study human lymphocyte development and may serve as a novel source for antitumor immunotherapy.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células Matadoras Naturais/imunologia , Leucemia Experimental/prevenção & controle , Animais , Medula Óssea/imunologia , Células Cultivadas , Sangue Fetal/citologia , Citometria de Fluxo , Humanos , Neoplasias Renais/imunologia , Neoplasias Renais/prevenção & controle , Neoplasias Renais/secundário , Leucemia Experimental/imunologia , Leucemia Experimental/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Estromais/imunologia
7.
Curr Opin Hematol ; 15(4): 312-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18536568

RESUMO

PURPOSE OF REVIEW: Hematopoietic development from embryonic stem cells has been one of the most productive areas of stem cell biology. Recent studies have progressed from work with mouse to human embryonic stem cells. Strategies to produce defined blood cell populations can be used to better understand normal and abnormal hematopoiesis, as well as potentially improve the generation of hematopoietic cells with therapeutic potential. RECENT FINDINGS: Molecular profiling, phenotypic and functional analyses have all been utilized to demonstrate that hematopoietic cells derived from embryonic stem cells most closely represent a stage of hematopoiesis that occurs at embryonic/fetal developmental stages. Generation of hematopoietic stem/progenitor cells comparable to hematopoietic stem cells found in the adult sources, such as bone marrow and cord blood, still remains challenging. However, genetic manipulation of intrinsic factors during hematopoietic differentiation has proven a suitable approach to induce adult definitive hematopoiesis from embryonic stem cells. SUMMARY: Concrete evidence has shown that embryonic stem cells provide a powerful approach to study the early stage of hematopoiesis. Multiple hematopoietic lineages can be generated from embryonic stem cells, although most of the evidence suggests that hematopoietic development from embryonic stem cells mimics an embryonic/fetal stage of hematopoiesis.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Linhagem da Célula , Hematopoese
8.
Methods Mol Biol ; 430: 119-33, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370295

RESUMO

The successful isolation and characterization of human embryonic stem cells (hESCs) provides a powerful tool to study the cellular and genetic mechanisms that mediate cell-fate decisions toward distinct developmental lineages. hESC-derived cells may also be suitable for novel cellular therapies. Significant progress in hematopoietic development of hESCs has demonstrated production of many types of blood cells from hESCs including myeloid, erythroid and lymphoid lineage cells, and possibly hematopoietic stem cells. Current established approaches to generate specific hematopoietic lineages are based on the initial pre-differentiation of hESCs into a heterogeneous mixture of cell populations. In this chapter, we describe two methods that have been successfully used in our laboratory: (1) co-culture with stromal cells derived from hematopoietic microenvironments and (2) x embryoid body (EB) formation. Subsequent to this early differentiation step, distinct progenitor cell populations can be derived, sorted, and utilized for further lineage-specific developmental studies.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Embrionárias/citologia , Linhagem da Célula , Células Cultivadas , Citometria de Fluxo , Humanos
9.
Stem Cells ; 25(11): 2919-27, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17673526

RESUMO

Efficient and stable genetic modification of human embryonic stem (ES) cells is required to realize the full scientific and potential therapeutic use of these cells. Currently, only limited success toward this goal has been achieved without using a viral vector. The Sleeping Beauty (SB) transposon system mediates nonviral gene insertion and stable expression in target cells and tissues. Here, we demonstrate use of the nonviral SB transposon system to effectively mediate stable gene transfer in human ES cells. Transposons encoding (a) green fluorescent protein coupled to the zeocin gene or (b) the firefly luciferase (luc) gene were effectively delivered to undifferentiated human ES cells with either a DNA or RNA source of transposase. Only human ES cells cotransfected with transposon- and transposase-encoding sequences exhibited transgene expression after 1 week in culture. Molecular analysis of transposon integrants indicated that 98% of stable gene transfer resulted from transposition. Stable luc expression was observed up to 5 months in human ES cells cotransfected with a transposon along with either DNA or RNA encoding SB transposase. Genetically engineered human ES cells demonstrated the ability to differentiate into teratomas in vivo and mature hematopoietic cells in vitro while maintaining stable transgene expression. We conclude that the SB transposon system provides an effective approach with several advantages for genetic manipulation and durable gene expression in human ES cells.


Assuntos
Elementos de DNA Transponíveis/fisiologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Transferência de Genes , Transgenes , Sequência de Bases , Células-Tronco Embrionárias/metabolismo , Humanos , Dados de Sequência Molecular , Transposases/biossíntese , Transposases/genética , Transposases/fisiologia
10.
Stem Cells ; 24(5): 1370-80, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16456127

RESUMO

Human embryonic stem cells (hESCs) provide an important means to characterize early stages of hematopoietic development. However, the in vivo potential of hESC-derived hematopoietic cells has not been well defined. We demonstrate that hESC-derived cells are capable of long-term hematopoietic engraftment when transplanted into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Human CD45(+) and CD34(+) cells are identified in the mouse bone marrow (BM) more than 3 months after injection of hESCs that were allowed to differentiate on S17 stromal cells for 7-24 days. Secondary engraftment studies further confirm long-term repopulating cells derived from hESCs. We also evaluated two mechanisms that may inhibit engraftment: host immunity and requirement for homing to BM. Treatment with anti-ASGM1 antiserum that primarily acts by depletion of natural killer cells in transplanted mice leads to improved engraftment, likely due to low levels of HLA class I expressed on hESCs and CD34(+) cells derived from hESCs. Intra-BM injection also provided stable engraftment, with hematopoietic cells identified in both the injected and contra-lateral femur. Importantly, no teratomas are evident in animals injected with differentiated hESCs. These results demonstrate that SCID-repopulating cells, a close surrogate for hematopoietic stem cells, can be derived from hESCs. Moreover, both adaptive and innate immune effector cells may be barriers to engraftment of these cells.


Assuntos
Células-Tronco Embrionárias/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Soros Imunes/farmacologia , Imunidade Inata , Células Matadoras Naturais/imunologia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
11.
Biochemistry (Mosc) ; 71 Suppl 1: S60-4, 4-5, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16487070

RESUMO

Killer cell immunoglobulin-like receptor (KIR) 2DL4 is the only KIR member reported to be expressed by all human natural killer (NK) cells. It differs from other KIR members in both structure and function. Its specific interaction with HLA-G, a non-classical MHC class I molecule, has been suggested to play an important role in regulating NK cell-mediated cytotoxicity. However, this interaction is still in doubt. In addition, the soluble KIR2DL4 extracellular domain used in many studies was produced by eukaryotic expression, which is less efficient than prokaryotic expression. In this study, we describe a method of rapid production a large amount of soluble KIR2DL4 extracellular domain based on a prokaryotic expression system. With this soluble KIR2DL4, we verified the interaction between KIR2DL4 and HLA-G1.


Assuntos
Antígenos HLA/química , Antígenos de Histocompatibilidade Classe I/química , Receptores Imunológicos/química , Expressão Gênica , Antígenos HLA/genética , Antígenos HLA/imunologia , Antígenos HLA-G , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células K562 , Células Matadoras Naturais/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores KIR , Receptores KIR2DL4 , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Relação Estrutura-Atividade
12.
Blood ; 107(5): 2180-3, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16278307

RESUMO

The human/sheep xenograft model has proven valuable in assessing the in vivo hematopoietic activity of stem cells from a variety of fetal and postnatal human sources. CD34+/lineage- or CD34+/CD38- cells isolated from human embryonic stem cells (hESCs) differentiated on S17 feeder layer were transplanted by intraperitoneal injections into fetal sheep. Chimerism in primary transplants was established with polymerase chain reaction (PCR) and flow cytometry of bone marrow and peripheral blood samples. Whole bone marrow cells harvested from a primary recipient were transplanted into a secondary recipient. Chimerism was established as described before. This animal was stimulated with human GM-CSF, and an increase in human hematopoietic activity was noted by flow cytometry. Bone marrow aspirations cultured in methylcellulose generated colonies identified by PCR to be of human origin. We therefore conclude that hESCs are capable of generating hematopoietic cells that engraft primary recipients. These cells also fulfill the criteria for long-term engrafting hematopoietic stem cells as demonstrated by engraftment and differentiation in the secondary recipient.


Assuntos
Embrião de Mamíferos/fisiologia , Feto/fisiologia , Sobrevivência de Enxerto/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Ovinos/fisiologia , ADP-Ribosil Ciclase 1/metabolismo , Animais , Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Ensaio de Unidades Formadoras de Colônias/métodos , Embrião de Mamíferos/citologia , Citometria de Fluxo/métodos , Células-Tronco Hematopoéticas/citologia , Humanos , Reação em Cadeia da Polimerase/métodos , Quimeras de Transplante/fisiologia , Transplante Heterólogo
13.
Methods Mol Biol ; 290: 149-62, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15361661

RESUMO

The isolation of embryonic stem (ES) cells from human preimplantation blastocysts creates an exciting new starting point to analyze the earliest stages of human blood development. This chapter describes two methods to promote hematopoietic differentiation of human ES cells: stromal cell coculture and embryoid body formation. Better understanding of basic human hematopoiesis through the study of human ES cells will likely have future therapeutic benefits.


Assuntos
Células da Medula Óssea/citologia , Embrião de Mamíferos/citologia , Células-Tronco/citologia , Técnicas de Cultura de Células , Técnicas de Cocultura , Citometria de Fluxo , Humanos
14.
Methods Mol Med ; 105: 425-36, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15492412

RESUMO

The isolation of embryonic stem (ES) cells from human preimplantation blastocytes creates an exciting new starting point to analyze the earliest stages of human blood development. This chapter describes two methods for the promotion of hematopoietic differentiation of human ES cells: stromal cell co-culture and embryoid body formation. Better understanding of basic human hematopoiesis through the study of human ES cells will likely have future therapeutic benefits.


Assuntos
Diferenciação Celular/fisiologia , Hematopoese/fisiologia , Células-Tronco Totipotentes/fisiologia , Animais , Células Cultivadas , Técnicas de Cocultura/métodos , Humanos , Camundongos , Células Estromais/fisiologia
15.
Exp Hematol ; 32(10): 1000-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15504555

RESUMO

OBJECTIVE: Human embryonic stem (ES) cells can be induced to differentiate into hematopoietic lineages either by stromal cell coculture or by formation of embryoid bodies (EBs). Here, we better characterize cell-bound and secreted factors that support this hematopoietic development. METHODS: Human ES cells either cocultured on the mouse bone marrow cell line S17, or allowed to form EBs, were induced to differentiate in the presence of serum, serum-free conditions, and serum-free media supplemented with defined cytokines. To better characterize the requirement for stromal cell-bound or secreted proteins, S17 conditioned media and transwell cultures were also utilized. RESULTS: In both models, CD34(+), CD45(+), and hematopoietic colony-forming cells (CFCs) were routinely derived. While hematopoietic development was diminished without serum, here we demonstrate with the stromal cell coculture model that addition of the growth factors stem cell factor (SCF), thrombopoietin (TPO), and Flt-3 ligand (Flt3L) to serum-free media does allow isolation of hematopoietic progenitors. However, these same three growth factors added to serum-free media do not support significant hematopoiesis in the EB system. However, addition of the mesoderm-inducing factors bone morphogenic protein-4 and vascular endothelial growth factor to EBs grown in serum-free media plus SCF, TPO, and Flt-3L does improve hematopoietic development. CONCLUSION: These results demonstrate the utility of human ES cell to evaluate specific stimuli that regulate cell fate decisions and the survival of specific lineages. Moreover, the method used to promote differentiation of ES cells may alter the cytokines or growth factors required to isolate specific cell types.


Assuntos
Comunicação Celular , Citocinas/farmacologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Células Estromais/citologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Técnicas de Cocultura , Meios de Cultura/farmacologia , Embrião de Mamíferos/citologia , Citometria de Fluxo , Substâncias de Crescimento/farmacologia , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA