Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Cell Prolif ; 55(2): e13189, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35060660

RESUMO

OBJECTIVES: Human pluripotent stem cells (hPSCs) are of great importance in both scientific research and regenerative medicine. The most classic and widely used culture method for hPSCs is co-culture with feeder cells, usually mouse embryonic fibroblasts. However, whether these feeder cell residues can affect the transcriptomic data analysis of hPSCs, especially gene or miRNA expression quantification, is still largely unknown. METHODS AND RESULTS: In this study, reanalysis of published mRNA-Seq and miRNA-Seq data sets revealed the existence of feeder cell-derived reads in the hPSC transcriptomic samples. We identified potentially influenced human genes and miRNAs due to misalignment of sequencing fragments affected by mouse feeder cells. Furthermore, we developed an optimized miRNA analysis pipeline to avoid quantification bias from different miRNA isoforms in the same family. Finally, by comparing the levels of feeder cell residues in hPSC samples isolated by different methods, we found that fluorescence-activated cell sorting and adhesion methods were more effective in feeder cell removal than the gradient centrifugation method. CONCLUSIONS: Collectively, our results demonstrate that feeder cell residues affect the transcriptomic data analysis of hPSCs. To minimize the impact of feeder cell contamination in hPSC samples, we provide solutions for both data analysis and sample preparation.


Assuntos
Diferenciação Celular/fisiologia , Células Alimentadoras/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Técnicas de Cocultura , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos
2.
Stem Cell Reports ; 16(7): 1686-1696, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34214484

RESUMO

Human extended pluripotent stem cells (EPSCs), with bidirectional chimeric ability to contribute to both embryonic and extraembryonic lineages, can be obtained and maintained by converting conventional pluripotent stem cells using chemicals. However, the transition system is based on inactivated mouse fibroblasts, and the underlying mechanism is not clear. Here we report a Matrigel-based feeder-free method to convert human embryonic stem cells and induced pluripotent stem cells into EPSCs and demonstrate the extended pluripotency in terms of molecular features, chimeric ability, and transcriptome. We further identify chemicals targeting glycolysis and histone methyltransferase to facilitate the conversion to and maintenance of feeder-free EPSCs. Altogether, our data not only establish a feeder-free system to generate human EPSCs, which should facilitate the mechanistic studies of extended pluripotency and further applications, but also provide additional insights into the transitions among different pluripotent states.


Assuntos
Células Alimentadoras/citologia , Células-Tronco Pluripotentes/citologia , Linhagem Celular , Quimera/fisiologia , Células Alimentadoras/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Indóis/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Piridonas/farmacologia
3.
Sci Rep ; 11(1): 12392, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34117337

RESUMO

Engineered epithelial cell sheets for clinical replacement of non-functional upper aerodigestive tract mucosa are regulated as medicinal products and should be manufactured to the standards of good manufacturing practice (GMP). The current gold standard for growth of epithelial cells for research utilises growth arrested murine 3T3 J2 feeder layers, which are not available for use as a GMP compliant raw material. Using porcine mucosal tissue, we demonstrate a new method for obtaining and growing non-keratinised squamous epithelial cells and fibroblast cells from a single biopsy, replacing the 3T3 J2 with a growth arrested primary fibroblast feeder layer and using pooled Human Platelet lysate (HPL) as the media serum supplement to replace foetal bovine serum (FBS). The initial isolation of the cells was semi-automated using an Octodissociator and the resultant cell suspension cryopreservation for future use. When compared to the gold standard of 3T3 J2 and FBS containing medium there was no reduction in growth, viability, stem cell population or ability to differentiate to mature epithelial cells. Furthermore, this method was replicated with Human buccal tissue, providing cells of sufficient quality and number to create a tissue engineered sheet.


Assuntos
Células Epiteliais/citologia , Fibroblastos/citologia , Mucosa Bucal/citologia , Engenharia Tecidual/métodos , Células 3T3 , Animais , Automação Laboratorial/instrumentação , Automação Laboratorial/métodos , Células Cultivadas , Criopreservação/métodos , Criopreservação/normas , Meios de Cultura/química , Células Epiteliais/metabolismo , Células Alimentadoras/citologia , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Humanos , Camundongos , Guias de Prática Clínica como Assunto , Engenharia Tecidual/normas
4.
Cells ; 10(4)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33917333

RESUMO

The endoderm, differentiated from human induced pluripotent stem cells (iPSCs), can differentiate into the small intestine and liver, which are vital for drug absorption and metabolism. The development of human iPSC-derived enterocytes (HiEnts) and hepatocytes (HiHeps) has been reported. However, pharmacokinetic function-deficiency of these cells remains to be elucidated. Here, we aimed to develop an efficient differentiation method to induce endoderm formation from human iPSCs. Cells treated with activin A for 168 h expressed higher levels of endodermal genes than those treated for 72 h. Using activin A (days 0-7), CHIR99021 and PI-103 (days 0-2), and FGF2 (days 3-7), the hiPSC-derived endoderm (HiEnd) showed 97.97% CD-117 and CD-184 double-positive cells. Moreover, HiEnts derived from the human iPSC line Windy had similar or higher expression of small intestine-specific genes than adult human small intestine. Activities of the drug transporter P-glycoprotein and drug-metabolizing enzyme cytochrome P450 (CYP) 3A4/5 were confirmed. Additionally, Windy-derived HiHeps expressed higher levels of hepatocyte- and pharmacokinetics-related genes and proteins and showed higher CYP3A4/5 activity than those derived through the conventional differentiation method. Thus, using this novel method, the differentiated HiEnts and HiHeps with pharmacokinetic functions could be used for drug development.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Endoderma/citologia , Enterócitos/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Ativinas/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Dimetil Sulfóxido/farmacologia , Enterócitos/efeitos dos fármacos , Células Alimentadoras/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Furanos/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Intestino Delgado/citologia , Linha Primitiva/citologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Reprodutibilidade dos Testes
5.
Cells ; 10(3)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33668931

RESUMO

Human adipose-derived stromal cells (ADSCs) are receiving unprecedented attention as a potential cellular source for regenerative medicine-based therapies against various diseases and conditions. However, there still have significant issues concerning the translational development of ADSC-based therapies, such as its heterogeneity and being prone to aging. We developed a new simple and economical 3D semi-suspended expansion method in which 3D spheroids reside on an ADSC-derived self-feeder cell layer, producing cells with increased population homogeneity and strong stemness and ensuring that the proliferation and differentiation potency of the cells does not become notably reduced after at least ten passages in culture. To check the potential application of the 3D ADSC spheroids, we discovered that the combination of siEID3, which is a small interfering RNA of EP300 inhibitor of differentiation 3 (EID3), and laminin/poly-d-lysine matrix can rapidly result in trans-differentiation of the 3D spheroid cells to neural progenitor-like cells (NPLCs) in approximately 9 days in vitro. This approach provides a multidisciplinary tool for stem cell research and production in mesenchymal stem cell-related fields.


Assuntos
Proteínas de Transporte/metabolismo , Técnicas de Cultura de Células , Transdiferenciação Celular , Células Alimentadoras/citologia , Células-Tronco Neurais/citologia , Polilisina/farmacologia , Esferoides Celulares/citologia , Células-Tronco/citologia , Tecido Adiposo/citologia , Proliferação de Células , Senescência Celular , Regulação da Expressão Gênica , Humanos , Imunofenotipagem , Laminina/farmacologia , Modelos Biológicos , RNA Interferente Pequeno/metabolismo
6.
Sci Rep ; 11(1): 6217, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33737638

RESUMO

Organotypic skin tissue models have decades of use for basic research applications, the treatment of burns, and for efficacy/safety evaluation studies. The complex and heterogeneous nature of native human skin however creates difficulties for the construction of physiologically comparable organotypic models. Within the present study, we utilized bioprinting technology for the controlled deposition of separate keratinocyte subpopulations to create a reconstructed epidermis with two distinct halves in a single insert, each comprised of a different keratinocyte sub-population, in order to better model heterogonous skin and reduce inter-sample variability. As an initial proof-of-concept, we created a patterned epidermal skin model using GPF positive and negative keratinocyte subpopulations, both printed into 2 halves of a reconstructed skin insert, demonstrating the feasibility of this approach. We then demonstrated the physiological relevance of this bioprinting technique by generating a heterogeneous model comprised of dual keratinocyte population with either normal or low filaggrin expression. The resultant model exhibited a well-organized epidermal structure with each half possessing the phenotypic characteristics of its constituent cells, indicative of a successful and stable tissue reconstruction. This patterned skin model aims to mimic the edge of lesions as seen in atopic dermatitis or ichthyosis vulgaris, while the use of two populations within a single insert allows for paired statistics in evaluation studies, likely increasing study statistical power and reducing the number of models required per study. This is the first report of human patterned epidermal model using a predefined bioprinted designs, and demonstrates the relevance of bioprinting to faithfully reproduce human skin microanatomy.


Assuntos
Bioimpressão/métodos , Queratinócitos/citologia , Modelos Biológicos , Pele/citologia , Engenharia Tecidual/métodos , Animais , Bioimpressão/instrumentação , Células Alimentadoras/citologia , Células Alimentadoras/fisiologia , Proteínas Filagrinas , Imunofluorescência , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Queratinócitos/fisiologia , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Cultura Primária de Células , Impressão Tridimensional/instrumentação , Proteínas S100/genética , Proteínas S100/metabolismo , Pele/anatomia & histologia , Engenharia Tecidual/instrumentação
7.
J Vis Exp ; (168)2021 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-33720120

RESUMO

The early interactions between the nasal epithelial layer and the innate immune cells during viral infections remains an under-explored area. The significance of innate immunity signaling in viral infections has increased substantially as patients with respiratory infections who exhibit high innate T cell activation show a better disease outcome. Hence, dissecting these early innate immune interactions allows the elucidation of the processes that govern them and may facilitate the development of potential therapeutic targets and strategies for dampening or even preventing early progression of viral infections. This protocol details a versatile model that can be used to study early crosstalk, interactions, and activation of innate immune cells from factors secreted by virally infected airway epithelial cells. Using an H3N2 influenza virus (A/Aichi/2/1968) as the representative virus model, innate cell activation of co-cultured peripheral blood mononuclear cells (PBMCs) has been analyzed using flow cytometry to investigate the subsets of cells that are activated by the soluble factors released from the epithelium in response to the viral infection. The results demonstrate the gating strategy for differentiating the subsets of cells and reveal the clear differences between the activated populations of PBMCs and their crosstalk with the control and infected epithelium. The activated subsets can then be further analyzed to determine their functions as well as molecular changes specific to the cells. Findings from such a crosstalk investigation may uncover factors that are important for the activation of vital innate cell populations, which are beneficial in controlling and suppressing the progression of viral infection. Furthermore, these factors can be universally applied to different viral diseases, especially to newly emerging viruses, to dampen the impact of such viruses when they first circulate in naïve human populations.


Assuntos
Imunidade Inata , Vírus da Influenza A Subtipo H3N2/imunologia , Influenza Humana/imunologia , Influenza Humana/virologia , Modelos Biológicos , Células 3T3 , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Impedância Elétrica , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Alimentadoras/citologia , Humanos , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Camundongos , Mitomicina/farmacologia , Mucina-5AC/metabolismo , Mucosa Nasal/patologia , Tubulina (Proteína)/metabolismo
8.
J Vis Exp ; (168)2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33616094

RESUMO

Calcific aortic valve disease (CAVD), an active disease process ranging from mild thickening of the valve to severe calcification, is associated with high mortality, despite new therapeutic options such as transcatheter aortic valve replacement (TAVR). The complete pathways that start with valve calcification and lead to severe aortic stenosis remain only partly understood. By providing a close representation of the aortic valve cells in vivo, the assaying of T lymphocytes from stenotic valve tissue could be an efficient way to clarify their role in the development of calcification. After surgical excision, the fresh aortic valve sample is dissected in small pieces and the T lymphocytes are cultured, cloned then analyzed using fluorescence activated cell sorting (FACS). The staining procedure is simple and the stained tubes can also be fixed using 0.5% of paraformaldehyde and analyzed up to 15 days later. The results generated from the staining panel can be used to track changes in T cell concentrations over time in relation to intervention and could easily be further developed to assess activation states of specific T cell subtypes of interest. In this study, we show the isolation of T cells, performed on fresh calcified aortic valve samples and the steps of analyzing T cell clones using flow cytometry to further understand the role of adaptive immunity in CAVD pathophysiology.


Assuntos
Estenose da Valva Aórtica/patologia , Valva Aórtica/citologia , Valva Aórtica/patologia , Buffy Coat/efeitos da radiação , Calcinose/patologia , Separação Celular/métodos , Células Alimentadoras/citologia , Citometria de Fluxo/métodos , Linfócitos T/citologia , Valva Aórtica/metabolismo , Células Cultivadas , Células Alimentadoras/metabolismo , Humanos , Linfócitos T/metabolismo
9.
J Exp Med ; 218(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33433623

RESUMO

Genome editing is a powerful technique for delineating complex signaling circuitry and enhancing the functionality of immune cells for immunotherapy. Natural killer (NK) cells are potent immune effectors against cell malignancy, but they are challenging to modify genetically by conventional methods due to the toxicity of DNA when introduced into cells coupled with limited transfection and transduction efficiency. Here, we describe an integrated platform that streamlines feeder-free ex vivo expansion of cryopreserved primary human NK cells and nonviral genome editing by the nucleofection of CRISPR-Cas9 ribonucleoproteins (Cas9 RNPs). The optimized Cas9 nucleofection protocol allows efficient and multiplex gene knockout in NK cells while preserving high cell viability and negligible off-target effects. Cointroduction of a DNA template also enables in-frame gene knock-in of an HA affinity tag and a gfp reporter across multiple loci. This work demonstrates the advantages and flexibility of working with cryopreserved NK cells as potential off-the-shelf engineered therapeutic agents.


Assuntos
Edição de Genes , Células Matadoras Naturais/metabolismo , Sequência de Aminoácidos , Biomarcadores/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Cromossomos Humanos/genética , Criopreservação , Citotoxicidade Imunológica , DNA/metabolismo , Células Alimentadoras/citologia , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Plasmídeos/genética , RNA Guia de Cinetoplastídeos/genética , Ribonucleoproteínas/metabolismo , Translocação Genética
10.
J Biosci Bioeng ; 131(3): 305-313, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33262019

RESUMO

During culture with feeder cells, deviation from the undifferentiated state of human induced pluripotent stem cells (hiPSCs) occurs at a very low frequency. Anomalous cell migration in central and peripheral regions of hiPSC colonies has been suggested to be the trigger for this phenomenon. To confirm this hypothesis, sequential cell migration prior to deviation must be demonstrated. This has been difficult using in vitro methods. We therefore developed a kinetic model with a proposed definition of anomalous cell migration as continuous relatively fast or slow cell migration. The developed model was validated via in silico reproduction of deviation phenomenon observed in vitro, such as the positions of deviated cells in a colony and the frequency of deviation in culture. This model suggests that anomalous cell migration-driven hiPSC deviation can be explained by two factors: a mechanical stimulus, represented by cell migration, and duration of the mechanical stimulus. The factor "duration of mechanical stimulus" sets our model apart from others, and helps to realize the ultra-rare trigger (approximately 10-5) of deviation from the undifferentiated state in hiPSC culture.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Alimentadoras/citologia , Humanos , Cinética
11.
Methods Mol Biol ; 2239: 213-234, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33226622

RESUMO

Mouse embryonic fibroblasts (MEFs) can be used in co-culture to support generation of induced pluripotent stem cells (iPSCs) and the normal growth and proliferation of human pluripotent stem cells (hPSCs). Here, we describe the necessary steps to derive, expand, harvest, inactivate, plate, and use MEFs as feeders for iPSC generation and maintenance.


Assuntos
Técnicas de Cultura de Células/métodos , Embrião de Mamíferos/citologia , Células Alimentadoras/efeitos da radiação , Fibroblastos/efeitos da radiação , Animais , Células Cultivadas , Técnicas de Cocultura/métodos , Criopreservação , Células Alimentadoras/citologia , Células Alimentadoras/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos
12.
In Vitro Cell Dev Biol Anim ; 56(10): 888-895, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33140329

RESUMO

Noonan syndrome is an autosomal dominant developmental disorder. Although it is relatively common, and its phenotypical variability is well documented, its pathophysiology is not fully understood. Previously, with the aim of revealing the pathogenesis of genetic disorders, we reported the induction of cleidocranial dysplasia-specific human-induced pluripotent stem cells (hiPSCs) from patient's dental pulp cells (DPCs) under serum-free, feeder-free, and integration-free conditions. Notably, these cells showed potential for application to genetic disorder disease models. Furthermore, using similar procedures, we reported the induction of hiPSCs derived from peripheral blood mononuclear cells (PBMCs) of healthy volunteers. These methods are beneficial, because they are carried out without invasive and painful biopsies. Using those procedures, we reprogrammed DPCs and PBMCs that were derived from a patient with Noonan syndrome (NS) to establish NS-specific hiPSCs (NS-DPC-hiPSCs and NS-PBMC-hiPSCs, respectively). The induction efficiency of NS-hiPSCs was higher than that of WT-hiPSCs. We hypothesize that this was caused by high NANOG expression. Here, we describe the experimental results and findings related to NS-hiPSCs. This is the first report on the establishment of NS-hiPSCs and their disease modeling.


Assuntos
Células Alimentadoras/citologia , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome de Noonan/patologia , Fosfatase Alcalina/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Reprogramação Celular/genética , Meios de Cultura Livres de Soro , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Mutação de Sentido Incorreto/genética , Proteína Homeobox Nanog/química , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
13.
Cell Chem Biol ; 27(12): 1561-1572.e7, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33125912

RESUMO

The proliferation of human pancreatic progenitor cells (PPCs) is critical for developing cell therapies for diabetes. Here, using transcriptome analysis combined with small interfering RNA (siRNA) screening, we revealed that WNT7B is a downstream growth factor of AT7867, a compound known to promote the proliferation of PPCs generated from human pluripotent stem cells. Feeder cell lines stably expressing mouse Wnt7a or Wnt7b, but not other Wnts, enhanced PPC proliferation in the absence of AT7867. Importantly, Wnt7a/b ligands did not activate the canonical Wnt pathway, and PPC proliferation depended on the non-canonical Wnt/PKC pathway. A comparison of the phosphoproteome in response to AT7867 or a newly synthesized AT7867 derivative uncovered the function of YY1 as a transcriptional regulator of WNT7B. Overall, our data highlight unknown roles of non-canonical WNT7B/PKC signaling and YY1 in human PPC proliferation and will contribute to the stable supply of a cell source for pancreatic disease modeling and therapeutic applications.


Assuntos
Pâncreas/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais , Proteínas Wnt/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Células Alimentadoras/citologia , Humanos , Camundongos , Proteína Quinase C/metabolismo
14.
In Vitro Cell Dev Biol Anim ; 56(9): 808-815, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33029688

RESUMO

Homologous feeder culture system can efficiently promote the proliferation of embryonic germ (EG) cells or embryonic stem (ES) cells while avoiding contamination by exogenous proteins and pathogens. In this study, we compared the potency of using homologous porcine embryonic fibroblasts (PEFs), gonadal stromal cells (GSCs), porcine adipose-derived stem cells (PASCs), or porcine amniotic fluid stem (PAFS) cells as feeder cells for porcine EG growth, with the commonly used mouse embryonic fibroblasts (MEFs). We compared the feeder cell growth rates; secretion of growth factors including stem cell factor (SCF), basic fibroblast growth factor (bFGF), and leukemia inhibitory factor (LIF); the effects of growth factors on porcine PGC growth; and EG growth rates when individual cells were used as feeders. Our results showed that feeder cells secreted limited amounts of growth factors, and supplementation of growth factors can significantly improve the formation of EG colonies and number of passages (P < 0.05). GSC and PEF were more suitable for EG growth because of their faster growth rate and their support on EG growth. In conclusion, this study identified novel homologous cells that can be used for EG production.


Assuntos
Técnicas de Cultura de Células/métodos , Células Germinativas Embrionárias/citologia , Animais , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/citologia , Células Alimentadoras/citologia , Feminino , Fibroblastos/citologia , Camundongos , Suínos
15.
J Biosci Bioeng ; 130(5): 545-552, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32782195

RESUMO

Chemically induced liver progenitor (CLiP) cells, converted in vitro from mature hepatocytes, possess the bipotentiality to differentiate into both hepatocytes and cholangiocytes. Here, we aimed to investigate the optimal conditions for bile duct (BD) induction from rat CLiPs. A two-step induction protocol was used for the differentiation of cholangiocytes. We investigated the effects of passage number, preincubation times, Matrigel, and mouse embryonic fibroblast (MEF) feeder cells on the induction of cholangiocytes. Earlier passages of CLiPs were better for BD induction compared with stable CLiPs. Extending the preincubation time of CLiPs before induction delayed the formation of the BD. Matrigel provided cells with space to form three-dimensional (3D) structures, but the long-term use of Matrigel from the induction step did not benefit the differentiation of CLiPs to cholangiocytes. MEF feeder cells, through the Jag/Notch pathway, affected BD formation and function, as well as gene and protein expression. CLiPs were a good cell source for cholangiocyte differentiation under appropriate conditions and may offer a key vehicle for the study of cholangiopathies in vitro.


Assuntos
Ductos Biliares/citologia , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/citologia , Fígado/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Animais , Células Epiteliais/efeitos dos fármacos , Células Alimentadoras/citologia , Hepatócitos/citologia , Camundongos , Ratos
16.
Cells ; 9(6)2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32485910

RESUMO

Non-human primates (NHP) are important surrogate models for late preclinical development of advanced therapy medicinal products (ATMPs), including induced pluripotent stem cell (iPSC)-based therapies, which are also under development for heart failure repair. For effective heart repair by remuscularization, large numbers of cardiomyocytes are required, which can be obtained by efficient differentiation of iPSCs. However, NHP-iPSC generation and long-term culture in an undifferentiated state under feeder cell-free conditions turned out to be problematic. Here we describe the reproducible development of rhesus macaque (Macaca mulatta) iPSC lines. Postnatal rhesus skin fibroblasts were reprogrammed under chemically defined conditions using non-integrating vectors. The robustness of the protocol was confirmed using another NHP species, the olive baboon (Papio anubis). Feeder-free maintenance of NHP-iPSCs was essentially dependent on concurrent Wnt-activation by GSK-inhibition (Gi) and Wnt-inhibition (Wi). Generated NHP-iPSCs were successfully differentiated into cardiomyocytes using a combined growth factor/GiWi protocol. The capacity of the iPSC-derived cardiomyocytes to self-organize into contractile engineered heart muscle (EHM) was demonstrated. Collectively, this study establishes a reproducible protocol for the robust generation and culture of NHP-iPSCs, which are useful for preclinical testing of strategies for cell replacement therapies in NHP.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Animais , Forma Celular , Células Cultivadas , Reprogramação Celular , Células Alimentadoras/citologia , Fibroblastos/citologia , Humanos , Macaca mulatta , Miócitos Cardíacos/citologia , Papio , Fatores de Tempo , Engenharia Tecidual , Transgenes
17.
Methods Cell Biol ; 159: 115-141, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32586440

RESUMO

The lung consists of branched structures that are anatomically, developmentally and functionally divided into airway and alveolar regions. Each region contributes to lung-specific functions involving a defense system and gas exchange, and their dysfunction can cause fatal lung diseases. In the alveolar region, the cuboidal alveolar type 2 (AT2) cells account for 90% of the alveolar epithelial cells and serve as the tissue stem cells secreting pulmonary surfactant, and flattened alveolar type I (AT1) cells cover most of the alveolar surface directly contributing to gas exchange adjacent to capillary vessels. It has been difficult to culture alveolar epithelial cells in vitro, as the lineage-specific features of those cells are rapidly lost in a conventional two-dimensional culture setting. The culture of alveolar organoids (AOs) is an emerging technique that can help maintain the features of alveolar epithelial cells in vitro, and their application to human disease modeling is eagerly awaited. We herein describe our method of generating and culturing alveolar epithelial cells and AOs derived from human induced pluripotent stem cells (iPSCs). iPSCs derived from lung disease patients, including those with rare genetic diseases, will make help elucidate the disease mechanism and hopefully identify therapeutic targets.


Assuntos
Células Epiteliais Alveolares/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Células Epiteliais Alveolares/ultraestrutura , Diferenciação Celular , Células Cultivadas , Células Alimentadoras/citologia , Feto/citologia , Fibroblastos/citologia , Imunofluorescência , Humanos
18.
Exp Cell Res ; 394(2): 112119, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32485182

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a significant obstacle for immunotherapy of cancer. It is of great clinical relevance to study the mechanism of MDSCs accumulation in mouse spleens and establish a stable method to obtain high-purity MDSCs in vitro for further research. Here, we established a new method for amplifying a large number of highly pure MDSCs in vitro. To mimic the microenvironment of MDSCs development in vivo, mouse splenic stroma feeder cells and serum-free medium containing granulocyte-macrophage colony stimulating factor (GM-CSF) were used to induce myeloid precursors in mouse bone marrow cells, which differentiate into MDSCs. Development and immunological functions of the cells were monitored both in vivo and in vitro. A total of 4 × 108 MDSCs could be obtained from the bone marrow from one mouse, the ratio of CD11b+Gr-1+ MDSCs could reach 93.8% ± 3.3% after nine days of culture in vitro. Cultured MDSCs maintained a similar immunophenotype with MDSCs found in tumor-bearing mice. Colony forming assay in vitro and in vivo demonstrated that these were myeloid precursor cells. These cells generated high levels of reactive oxygen species and arginase 1 to prevent proliferation of CD8+ T cells in vitro. These also increased regulatory T (Treg) cells in blood while promoting the growth of lymphoma in vivo. In addition, cultured MDSCs effectively inhibited acute graft-versus-host disease (aGVHD). Our findings suggest that mouse splenic stroma plays an important role in the generation of MDSCs and represent a preliminary mechanism for the accumulation of MDSCs in spleens, and thereby lay the foundation for basic research and the clinical application of MDSCs.


Assuntos
Técnicas de Cultura de Células/métodos , Células Alimentadoras/citologia , Células Supressoras Mieloides/citologia , Baço/citologia , Animais , Arginase/metabolismo , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Feminino , Doença Enxerto-Hospedeiro/imunologia , Imunofenotipagem , Linfoma/patologia , Masculino , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/citologia , Análise de Sobrevida , Linfócitos T Reguladores/citologia
19.
Cytotherapy ; 22(7): 354-368, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32451262

RESUMO

BACKGROUND AIMS: Natural killer (NK) cells are promising cells for immunotherapy of cancer, and there are ongoing efforts to improve their ex vivo expansion to clinically relevant numbers. This study focused on the development of a C1-, C2-, Bw4 killer cell immunoglobulin-like receptor (KIR) ligand and NKG2A ligand-containing feeder cell line for autonomous expansion of functional NK cells. METHODS: PC3PSCA-derived feeder cells expressing IL-2, 4-1BBL and membrane-bound IL-15-mutDAP12 (mIL-15d) fusion protein in combinations or alone were generated and used for expansion. Expanded NK cells were analyzed with respect to subpopulations, expression of NK cell receptors and immune checkpoint molecules as well as their cytotoxicity against K562 cells, cetuximab-marked tumor cells and autologous B cells. RESULTS: Only combinatorial expression of IL-2 plus 4-1BBL or IL-2, 4-1BBL plus mIL-15d in feeder cells efficiently expanded NK cells and supported selective outgrowth of NK cells from peripheral blood mononuclear cell samples. Best expansion of NK cells was achieved using PC3PSCA-IL-2-4-1BBL-mIL-15d feeder cells. Such expanded NK cells exhibited upregulation of natural cytotoxicity receptors, DNAM-1 and NKG2C and induced expression of high affinity IL-2 receptor, which were paralleled by attenuated KIR and increased expression of NKG2A and ILT2. In addition, elevated TIM-3 levels were noted and PD-1 and T cell immunoreceptor with Ig and ITIM domain (TIGIT) levels remained low. Expanded NK cells were highly cytolytic when encountering K562 cells and cetuximab-marked target cells but remained unresponsive to autologous B cells and target cells with protective levels of human leukocyte antigen. CONCLUSIONS: Collectively, the results demonstrate the feasibility of PC3PSCA-IL-2-4-1BBL-mIL-15d feeder cells for robust expansion of NK cells, which remain tolerant to self and could be used in the future for adoptive cell therapy of cancer.


Assuntos
Autoantígenos/imunologia , Células Alimentadoras/citologia , Tolerância Imunológica , Células Matadoras Naturais/citologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos CD/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cetuximab/farmacologia , Células Alimentadoras/efeitos dos fármacos , Células HEK293 , Humanos , Tolerância Imunológica/efeitos dos fármacos , Interleucina-2/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Ligantes
20.
In Vitro Cell Dev Biol Anim ; 56(4): 313-321, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32307668

RESUMO

Bladder dysfunction is one of the most common diseases that occur for a number of reasons and the current treatment modalities do not improve much in its recovery process. Tissue engineering in the last two decades has given great hope for the treatment of these disorders. In this study, a composite nanofibrous scaffold was fabricated from chitosan, collagen, and polyvinyl-alcohol polymer blend while curcumin incorporated in scaffold fibers. The scaffold supportive functions from smooth muscle cell differentiation were studied when human-induced pluripotent stem cells were cultured on the scaffolds under differentiation medium. Biocompatibility of the fabricated scaffold increased significantly by incorporating curcumin in the scaffold fibers, where protein adsorption, cell attachment, and viability were increased in the nanofiber/curcumin group compared with the other groups. In addition, the expression level of smooth muscle cell-related genes, including alpha-smooth muscle actin (αSMA), smooth muscle 22 alpha (SM-22a), Caldesmon1, and Calponin1in the stem cells upregulated while cultured in the presence of curcumin, but this increase was significantly improved while cells cultured on the nanofibers/curcumin. In addition, αSMA protein in the cells cultured on the nanofibers/curcumin expressed significantly higher than those cells cultured on the nanofibers without curcumin. It can be concluded that smooth muscle cell differentiation of the induced pluripotent stem cells promoted by curcumin and this promotion was synergistically improved while curcumin incorporated in the nanofibers. Graphical abstract.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Quitosana/química , Colágeno/química , Curcumina/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos de Músculo Liso/citologia , Nanofibras/química , Álcool de Polivinil/química , Animais , Diferenciação Celular/genética , Linhagem Celular , Células Alimentadoras/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Nanofibras/ultraestrutura , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA