Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Stem Cell Res Ther ; 15(1): 245, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39113095

RESUMO

BACKGROUND: The establishment of stable porcine embryonic stem cells (pESCs) can contribute to basic and biomedical research, including comparative developmental biology, as well as assessing the safety of stem cell-based therapies. Despite these advantages, most pESCs obtained from in vitro blastocysts require complex media and feeder layers, making routine use, genetic modification, and differentiation into specific cell types difficult. We aimed to establish pESCs with a single cell-passage ability, high proliferative potency, and stable in long-term culture from in vitro-derived blastocysts using a simplified serum-free medium. METHODS: We evaluated the establishment efficiency of pESCs from in vitro blastocysts using various basal media (DMEM/F10 (1:1), DMEM/F12, and a-MEM) and factors (FGF2, IWR-1, CHIR99021, and WH-4-023). The pluripotency and self-renewal capacity of the established pESCs were analyzed under feeder or feeder-free conditions. Ultimately, we developed a simplified culture medium (FIW) composed of FGF2, IWR-1, and WH-4-023 under serum-free conditions. RESULTS: The pESC-FIW lines were capable of single-cell passaging with short cell doubling times and expressed the pluripotency markers POU5F1, SOX2, and NANOG, as well as cell surface markers SSEA1, SSEA4, and TRA-1-60. pESC-FIW showed a stable proliferation rate and normal karyotype, even after 50 passages. Transcriptome analysis revealed that pESC-FIW were similar to reported pESC maintained in complex media and showed gastrulating epiblast cell characteristics. pESC-FIW were maintained for multiple passages under feeder-free conditions on fibronectin-coated plates using mTeSR™, a commercial medium used for feeder-free culture, exhibiting characteristics similar to those observed under feeder conditions. CONCLUSIONS: These results indicated that inhibition of WNT and SRC was sufficient to establish pESCs capable of single-cell passaging and feeder-free expansion under serum-free conditions. The easy maintenance of pESCs facilitates their application in gene editing technology for agriculture and biomedicine, as well as lineage commitment studies.


Assuntos
Células-Tronco Embrionárias , Animais , Meios de Cultura Livres de Soro/farmacologia , Suínos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/citologia , Diferenciação Celular , Células Alimentadoras/citologia , Células Alimentadoras/metabolismo , Técnicas de Cultura de Células/métodos , Proliferação de Células , Blastocisto/citologia , Blastocisto/metabolismo , Células Cultivadas
2.
Stem Cell Rev Rep ; 20(2): 484-494, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38079087

RESUMO

Pluripotent stem cells (PSCs) are widely recognized as one of the most promising types of stem cells for applications in regenerative medicine, tissue engineering, disease modeling, and drug screening. This is due to their unique ability to differentiate into cells from all three germ layers and their capacity for indefinite self-renewal. Initially, PSCs were cultured using animal feeder cells, but these systems presented several limitations, particularly in terms of Good Manufacturing Practices (GMP) regulations. As a result, feeder-free systems were introduced as a safer alternative. However, the precise mechanisms by which feeder cells support pluripotency are not fully understood. More importantly, it has been observed that some aspects of the need for feeder cells like the optimal density and cell type can vary depending on conditions such as the developmental stage of the PSCs, phases of the culture protocol, the method used in culture for induction of pluripotency, and intrinsic variability of PSCs. Thus, gaining a better understanding of the divergent roles and necessity of feeder cells in various conditions would lead to the development of condition-specific defined feeder-free systems that resolve the failure of current feeder-free systems in some conditions. Therefore, this review aims to explore considerable feeder-related issues that can lead to the development of condition-specific feeder-free systems.


Assuntos
Células-Tronco Pluripotentes , Animais , Células Alimentadoras/metabolismo , Medicina Regenerativa , Engenharia Tecidual
3.
Adv Healthc Mater ; 12(8): e2201708, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36455286

RESUMO

The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting  HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Fibroblastos , Células HeLa , Células Alimentadoras/metabolismo , Diferenciação Celular
4.
FEBS Open Bio ; 13(2): 279-292, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36537760

RESUMO

Feeder cells play an important role in the culture of human pluripotent stem cells (hPSCs) in vitro. Previously, we used methanol as a fixative to prepare feeder cells for the cultivation of pluripotent stem cells (PSCs), and this method could maintain the self-renewal and pluripotency of PSCs. However, methanol is toxic, and so here we examined whether ethanol could be used to prepare feeder cells as a fixative for hPSC culturing. Primed, naïve, and extended human embryonic stem cells and induced pluripotent stem cells can maintain self-renewal and undifferentiated potential on feeder cells treated with ethanol for an extended period. RNA sequencing analysis showed that the expression of collagen-related genes in hPSCs cultured on feeder cells treated with ethanol was significantly lower as compared with hPSCs cultured on feeder cells treated with mitomycin C. Therefore, we speculate that the signaling pathway mediated by collagen-related genes may, at least in part, contribute to the maintenance of self-renewal and pluripotency of PSCs induced by feeder cells treated with chemicals.


Assuntos
Etanol , Células-Tronco Pluripotentes , Humanos , Células Alimentadoras/metabolismo , Etanol/farmacologia , Etanol/metabolismo , Metanol , Fixadores/metabolismo , Células-Tronco Pluripotentes/metabolismo , Colágeno/metabolismo
5.
Sci Rep ; 12(1): 9177, 2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35654935

RESUMO

The pluripotency maintenance of pluripotent stem cells (PSCs) requires the suitable microenvironment, which commonly provided by feeder layers. However, the preparation of feeder layers is time consuming and labor exhaustive, and the feeder cells treated with mitomycin C or γ-ray irradiation bring heterologous contamination. In this study, mouse embryonic fibroblasts (MEFs) were treated by methanol to generate chemical fixed feeder cells, and bovine embryonic stem cells F7 (bESC-F7) cultured on this feeder layer. Then the pluripotency and metabolism of bESC-F7 cultured on methanol-fixed MEFs (MT-MEFs) named MT-F7 was compared with mitomycin C treated MEFs (MC-MEFs). The results showed that bESC-F7 formed alkaline phosphatase positive colonies on MT-MEFs, the relative expression of pluripotent markers of these cells was different from the bESCs cultured on the MC-MEFs (MC-F7). The long-term cultured MT-F7 formed embryoid bodies, showed the ability to differentiate into three germ layers similar to MC-F7. The analyses of RNA-seq data showed that MT-MEFs lead bESCs to novel steady expression patterns of genes regulating pluripotency and metabolism. Furthermore, the bovine expanded pluripotent stem cells (bEPSCs) cultured on MT-MEFs formed classical colonies, maintained pluripotency, and elevated metabolism. In conclusion, MT-MEFs were efficient feeder layer that maintain the distinctive pluripotency and metabolism of PSCs.


Assuntos
Metanol , Células-Tronco Pluripotentes , Animais , Bovinos , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Alimentadoras/metabolismo , Fibroblastos , Metanol/metabolismo , Camundongos , Mitomicina/metabolismo , Células-Tronco Pluripotentes/metabolismo
6.
Zygote ; 30(4): 550-560, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35485762

RESUMO

Mouse embryonic fibroblast (MEF) cells are commonly used as feeder cells to maintain the pluripotent state of stem cells. MEFs produce growth factors and provide adhesion molecules and extracellular matrix (ECM) compounds for cellular binding. In the present study, we compared the expression levels of Fgf2, Bmp4, ActivinA, Lif and Tgfb1 genes at the mRNA level and the level of Fgf2 protein secretion and Lif cytokine secretion at passages one, three and five of MEFs isolated from 13.5-day-old and 15.5-day-old embryos of NMRI and C57BL/6 mice using real-time PCR and enzyme-linked immunosorbent assay. We observed differences in the expression levels of the studied genes and secretion of the two growth factors in the three passages of MEFs isolated from 13.5-day-old and 15.5-day-old embryos, respectively. These differences were also observed between the NMRI and C57BL/6 strains. The results of this study suggested that researchers should use mice embryos that have different genetic backgrounds and ages, in addition to different MEF passages, when producing MEFs based on the application and type of their study.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Fibroblastos , Animais , Diferenciação Celular , Células Cultivadas , Células Alimentadoras/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Patrimônio Genético , Camundongos , Camundongos Endogâmicos C57BL
7.
Sci Rep ; 12(1): 1082, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058512

RESUMO

Contractile activity is a fundamental property of skeletal muscles. We describe the establishment of a "feeder-supported in vitro exercise model" using human-origin primary satellite cells, allowing highly-developed contractile myotubes to readily be generated by applying electrical pulse stimulation (EPS). The use of murine fibroblasts as the feeder cells allows biological responses to EPS in contractile human myotubes to be selectively evaluated with species-specific analyses such as RT-PCR. We successfully applied this feeder-supported co-culture system to myotubes derived from primary satellite cells obtained from sporadic inclusion body myositis (sIBM) patients who are incapable of strenuous exercise testing. Our results demonstrated that sIBM myotubes possess essentially normal muscle functions, including contractility development, de novo sarcomere formation, and contraction-dependent myokine upregulation, upon EPS treatment. However, we found that some of sIBM myotubes, but not healthy control myotubes, often exhibit abnormal cytoplasmic TDP-43 accumulation upon EPS-evoked contraction, suggesting potential pathogenic involvement of the contraction-inducible TDP-43 distribution peculiar to sIBM. Thus, our "feeder-supported in vitro exercise model" enables us to obtain contractile human-origin myotubes, potentially utilizable for evaluating exercise-dependent intrinsic and pathogenic properties of patient muscle cells. Our approach, using feeder layers, further expands the usefulness of the "in vitro exercise model".


Assuntos
Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Estimulação Elétrica/métodos , Células Alimentadoras/metabolismo , Humanos , Camundongos , Modelos Biológicos , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Miosite de Corpos de Inclusão/fisiopatologia , Sarcômeros/fisiologia , Células Satélites de Músculo Esquelético/metabolismo
8.
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
9.
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
10.
Cell Cycle ; 19(24): 3639-3649, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33323026

RESUMO

The present study aimed to assess the role of miR-1275 in cardiac ischemia reperfusion injury. H9 human embryonic stem cell (hESC)-derived cardiomyocytes stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were used to simulate myocardial injury in vitro. miR-1275 expression levels in cells were measured by RT-qPCR. The release of lactate dehydrogenase (LDH) and creatine kinase (CK) was examined through LDH and CK ELISA kits. Cell apoptosis was detected through flow cytometry. A Fura-2 Calcium Flux Assay Kit and a Fluo-4 assay kit were used to determine the Ca2+ concentration. Expression levels of proteins were tested by Western blotting. The binding effect of miR-1275 and neuromedin U type 1 receptor (NMUR1) was detected by dual-luciferase activity assay. The results showed that miR-1275 was upregulated in OGD/R-stimulated cardiomyocytes. Inhibition of miR-1275 suppressed the increased activity of LDH and CK, cell apoptosis, reactive oxygen species (ROS) production, intracellular Ca2+ concentration and sarcoplasmic reticulum (SR) Ca2+ leak induced by OGD/R treatment in cardiomyocytes. miR-1275 directly targets 3'UTR of NMUR1 and negatively regulates NMUR1 expression. Silence of NMUR1 abolished the protecting effect of the miR-1275 antagomir on myocardial OGD/R injury. Our study indicated that the miR-1275 antagomir protects cardiomyocytes from OGD/R injury through the promotion of NMUR1.


Assuntos
Sinalização do Cálcio/genética , Técnicas de Silenciamento de Genes/métodos , MicroRNAs/genética , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptores de Neurotransmissores/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Antagomirs/farmacologia , Apoptose/genética , Cálcio/metabolismo , Hipóxia Celular/genética , Linhagem Celular , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Glucose/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Camundongos , Traumatismo por Reperfusão Miocárdica/genética , Estresse Oxidativo/genética , Oxigênio/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Neurotransmissores/genética , Regulação para Cima/genética
11.
Reprod Fertil Dev ; 32(9): 822-834, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32527373

RESUMO

In a feeder-dependent culture system of human pluripotent stem cells (hPSCs), coculture with mouse embryonic fibroblasts may limit the clinical use of hPSCs. The aim of this study was to determine the feasibility of using human Caesarean scar fibroblasts (HSFs) as feeder cells for the culture of hPSCs. HSFs were isolated and characterised and cocultured with hPSCs, and the pluripotency, differentiation ability and karyotypic stability of hPSCs were determined. Inactivated HSFs expressed genes (including inhibin subunit beta A (INHBA), bone morphogenetic protein 4 (BMP4), fibroblast growth factor 2 (FGF2), transforming growth factor-ß1 (TGFB1), collagen alpha-1(I) (COL1A1) and fibronectin-1 (FN1) that have been implicated in the maintenance of hPSC pluripotency. When HSFs were used as feeder cells, the pluripotency and karyotypic stability of hPSC lines did not change after prolonged coculture. Interestingly, exogenous FGF2 could be omitted from the culture medium when HSFs were used as feeder cells for hESCs but not hiPSCs. hESCs cocultured with HSF feeder cells in medium without FGF2 supplementation maintained their pluripotency (as confirmed by the expression of pluripotency markers and genes), differentiated invitro into embryonic germ layers and maintained their normal karyotype. The present study demonstrates that HSFs are a novel feeder cell type for culturing hPSCs and that supplementation of exogenous FGF2 is not necessary for the Chula2.hES line.


Assuntos
Cesárea/efeitos adversos , Cicatriz/metabolismo , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Comunicação Parácrina , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular , Linhagem Celular , Cicatriz/etiologia , Cicatriz/patologia , Técnicas de Cocultura , Estudos de Viabilidade , Células Alimentadoras/patologia , Feminino , Fibroblastos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cariótipo , Fenótipo , Gravidez , Transdução de Sinais
12.
Cells ; 9(4)2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230942

RESUMO

Obtaining sufficient numbers of functional natural killer (NK) cells is crucial for the success of NK-cell-based adoptive immunotherapies. While expansion from peripheral blood (PB) is the current method of choice, ex vivo generation of NK cells from hematopoietic stem and progenitor cells (HSCs) may constitute an attractive alternative. Thereby, HSCs mobilized into peripheral blood (PB-CD34+) represent a valuable starting material, but the rather poor and donor-dependent differentiation of isolated PB-CD34+ cells into NK cells observed in earlier studies still represents a major hurdle. Here, we report a refined approach based on ex vivo culture of PB-CD34+ cells with optimized cytokine cocktails that reliably generates functionally mature NK cells, as assessed by analyzing NK-cell-associated surface markers and cytotoxicity. To further enhance NK cell expansion, we generated K562 feeder cells co-expressing 4-1BB ligand and membrane-anchored IL-15 and IL-21. Co-culture of PB-derived NK cells and NK cells that were ex-vivo-differentiated from HSCs with these feeder cells dramatically improved NK cell expansion, and fully compensated for donor-to-donor variability observed during only cytokine-based propagation. Our findings suggest mobilized PB-CD34+ cells expanded and differentiated according to this two-step protocol as a promising source for the generation of allogeneic NK cells for adoptive cancer immunotherapy.


Assuntos
Antineoplásicos/metabolismo , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Células Matadoras Naturais/citologia , Ligante 4-1BB/metabolismo , Antígenos CD34/metabolismo , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Células Alimentadoras/metabolismo , Células HEK293 , Mobilização de Células-Tronco Hematopoéticas , Humanos , Interleucinas/metabolismo , Células Matadoras Naturais/metabolismo , Fenótipo , Doadores de Tecidos
13.
Biotechniques ; 68(5): 270-274, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31939319

RESUMO

Chemically defined stem cell culture media are often costly, and the use of mitotically arrested mouse embryonic fibroblasts (MEFs) as feeder cells is a popular and cost-efficient way to maintain induced pluripotent stem cells (iPSCs). However, the commonly used mitotic inhibitor mitomycin-C (MMC) is known to cause cellular metabolic stress. Therefore, our aim was to determine whether such stress in feeder cells indirectly affects iPSC growth during coculture. We report that prolonged exposure to MMC causes metabolic stress in MEFs in the form of oxidative dysregulation. Through optimization of MMC exposure time, we show how to effectively arrest MEFs without inducing oxidative stress, thus promoting significantly better colony growth rates (p < 0.05), improved viability and longer periods between passages of iPSCs in coculture.


Assuntos
Técnicas de Cocultura/métodos , Células Alimentadoras , Células-Tronco Pluripotentes Induzidas , Mitomicina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Alimentadoras/citologia , Células Alimentadoras/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Estresse Oxidativo/efeitos dos fármacos
14.
Sci Rep ; 9(1): 18561, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31811191

RESUMO

The long-term expansion of keratinocytes under conditions that avoid xenogeneic components (i.e. animal serum- and feeder cell-free) generally causes diminished proliferation and increased terminal differentiation. Here we present a culture system free of xenogeneic components that retains the self-renewal capacity of primary human keratinocytes. In vivo the extracellular matrix (ECM) of the tissue microenvironment has a major influence on a cell's fate. We used ECM from human dermal fibroblasts, cultured under macromolecular crowding conditions to facilitate matrix deposition and organisation, in a xenogeneic-free keratinocyte expansion protocol. Phospholipase A2 decellularisation produced ECM whose components resembled the core matrix composition of natural dermis by proteome analyses. Keratinocytes proliferated rapidly on these matrices, retained their small size, expressed p63, lacked keratin 10 and rarely expressed keratin 16. The colony forming efficiency of these keratinocytes was enhanced over that of keratinocytes grown on collagen I, indicating that dermal fibroblast-derived matrices maintain the in vitro expansion of keratinocytes in a stem-like state. Keratinocyte sheets formed on such matrices were multi-layered with superior strength and stability compared to the single-layered sheets formed on collagen I. Thus, keratinocytes expanded using our xenogeneic-free protocol retained a stem-like state, but when triggered by confluence and calcium concentration, they stratified to produce epidermal sheets with a potential clinical use.


Assuntos
Técnicas de Cultura de Células/métodos , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Queratinócitos/fisiologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Derme/citologia , Células Alimentadoras/citologia , Células Alimentadoras/metabolismo , Fibroblastos/citologia , Humanos , Queratinócitos/transplante , Transplante de Pele/métodos
15.
Int J Mol Sci ; 20(24)2019 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-31847118

RESUMO

Because of the worldwide shortage of graftable corneas, alternatives to restore visual impairments, such as the production of a functional human cornea by tissue engineering, have emerged. Self-renewal of the corneal epithelium through the maintenance of a sub-population of corneal stem cells is required to maintain the functionality of such a reconstructed cornea. We previously reported an association between stem cell differentiation and the level to which they express the transcription factors Sp1 and NFI. In this study, we investigated the impact of replacing irradiated 3T3 (i3T3) murine fibroblast feeder cells by irradiated human corneal fibroblasts (iHFL) on the expression of Sp1 and NFI and evaluated their contribution to the proliferative properties of human corneal epithelial cells (hCECs) in both monolayer cultures and human tissue engineered corneas (hTECs). hCECs co-cultured with iHFL could be maintained for up to two more passages than when they were grown with i3T3. Western Blot and electrophoretic mobility shift assays (EMSAs) revealed no significant difference in the feeder-layer dependent increase in Sp1 at both the protein and DNA binding level, respectively, between HCECs grown with either i3T3 or iHFL. On the other hand, a significant increase in the expression and DNA binding of NFI was observed at each subsequent passage when hCECs were co-cultured along with i3T3. These changes were found to result from an increased expression of the NFIA and NFIB isoforms in hCECs grown with i3T3. Exposure of hCECs to cycloheximide revealed an increased stability of NFIB that likely resulted from post-translational glycosylation of this protein when these cells were co-cultured with i3T3. In addition, iHFL were as efficient as i3T3 at preserving corneal, slow-cycling, epithelial stem cells in the basal epithelium of the reconstructed hTECs. Furthermore, we observed an increased expression of genes whose encoded products promote hCECs differentiation along several passages in hCECs co-cultured with either type of feeder layer. Therefore, the iHFL feeder layer appears to be the most effective at maintaining the proliferative properties of hCECs in culture most likely by preserving high levels of Sp1 and low levels of NFIB, which is known for its gene repressor and cell differentiation properties.


Assuntos
Células Epiteliais/metabolismo , Epitélio Corneano/metabolismo , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Células-Tronco/metabolismo , Engenharia Tecidual , Células 3T3 , Animais , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Células Epiteliais/citologia , Epitélio Corneano/citologia , Células Alimentadoras/citologia , Fibroblastos/citologia , Humanos , Camundongos , Células-Tronco/citologia
16.
Sci Rep ; 9(1): 14916, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31624330

RESUMO

NK cell adoptive therapy is a promising cancer therapeutic approach, but there are significant challenges that limiting its feasibility and clinical efficacy. One difficulty is the paucity of clinical grade manufacturing platforms to support the large scale expansion of highly active NK cells. We created an NK cell feeder cell line termed 'NKF' through overexpressing membrane bound IL-21 that is capable of inducing robust and sustained proliferation (>10,000-fold expansion at 5 weeks) of highly cytotoxic NK cells. The expanded NK cells exhibit increased cytotoxic function against a panel of blood cancer and solid tumor cells as compared to IL-2-activated non-expanded NK cells. The NKF-expanded NK cells also demonstrate efficacy in mouse models of human sarcoma and T cell leukemia. Mechanistic studies revealed that membrane-bound IL-21 leads to an activation of a STAT3/c-Myc pathway and increased NK cell metabolism with a shift towards aerobic glycolysis. The NKF feeder cell line is a promising new platform that enables the large scale proliferation of highly active NK cells in support of large scale third party NK cell clinical studies that have been recently intiatied. These results also provide mechanistic insights into how membrane-bound IL-21 regulates NK cell expansion.


Assuntos
Células Alimentadoras/metabolismo , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Neoplasias/terapia , Cultura Primária de Células/métodos , Animais , Linhagem Celular Tumoral , Membrana Celular/imunologia , Membrana Celular/metabolismo , Proliferação de Células , Técnicas de Cocultura , Voluntários Saudáveis , Humanos , Interleucinas/imunologia , Interleucinas/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/transplante , Camundongos , Neoplasias/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Regen Med ; 14(6): 535-553, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31115264

RESUMO

Aim: To examine whether AKT-modified stromal cells expand human CD34+ hematopoietic stem cells (HSCs). Methods: Coculture, in vitro functional assays, immuno-fluorescence microscopy, flow cytometry. Results: M2-10B4 stromal cells (M2) modified with AKT1 (M2-AKT) expanded primitive CD34+38- HSCs, but affected their functionality. A chimeric feeder layer comprising naive human bone marrow-derived mesenchymal stromal cells and M2-AKT not only overcame the negative effects of M2-AKT, but, unexpectedly, also gave a synergistic effect on the growth and functionality of the HSCs. Conditioned medium of bone marrow stromal cells worked as effectively, but cell-cell contact between HSCs and M2-AKT cells was necessary for the synergistic effect of M2-AKT and bone marrow-derived mesenchymal stromal cells or their CM. Conclusion: Chimeric feeders expand HSCs.


Assuntos
Proliferação de Células , Células Alimentadoras/metabolismo , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Proto-Oncogênicas c-akt , Animais , Técnicas de Cocultura , Células Alimentadoras/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Células Estromais/citologia , Células Estromais/metabolismo
18.
J Cell Physiol ; 234(11): 20072-20084, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30953369

RESUMO

Human umbilical cord blood (HUCB) is a suitable source of hematopoietic stem cells (HSCs) for therapeutic transplantation. Different approaches have been used to expand the number of HSCs to increase the rate of HSC transplantation success in patients, such as using different cocktails of cytokines, feeder cell layers, and biocompatible scaffolds. microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression posttranscriptionally. They play crucial roles in hematopoiesis including stem cell proliferation, differentiation, stemness, and self-renewal properties. Here, we studied the UCB-derived CD34+ cell expansion and the miRNA signatures of CD34+ cells on two- and three-dimensional (2D and 3D) culture conditions. We successfully expanded the UCB-derived CD34+ cells in both liquid culture (2D) and on aminated polyethersulfone nanofiber scaffolds (3D). Next, we identified the miRNA signature of CD34+ cells and their target genes. We found 58 dysregulated miRNAs in 3D culture condition and 34 dysregulated miRNAs in 2D culture condition when compared to the freshly isolated CD34+ cells. Various types of target genes were also predicted in both conditions using two online databases.


Assuntos
Antígenos CD34/metabolismo , Sangue Fetal/metabolismo , MicroRNAs/metabolismo , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Células Alimentadoras/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Nanofibras/química
19.
Adipocyte ; 8(1): 178-189, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31033380

RESUMO

Adipose stromal/progenitor cells (ASCs) can differentiate into adipocytes in the course of adipogenesis. This process is governed by systemic factors and signals of the adipose stem cell niche. ASCs isolated from fat tissues and amplified in vitro provide an essential and reliable model system to study adipogenesis. However, current cell culture models routinely grow ASCs on plastic surfaces largely missing niche parameters. In the present communication, we employed human foreskin fibroblasts (HFFs) monolayers as feeder cells for ASCs, which were isolated from human subcutaneous white adipose tissue and amplified in vitro. We found that PPARγ2 and several adipocyte markers were significantly higher expressed in differentiated ASCs growing on feeder layers relative to plastic dishes. Moreover, a significant higher number of adipocytes was generated from ASCs cultured on feeder layer and these adipocytes contained larger fat droplets. Insulin strongly stimulated glucose uptake into adipocytes produced on feeder layer suggesting that these cells show characteristic metabolic features of fat cells.  Finally, we show that the HFF feeder layer allows adipogenic differentiation of low-density-seeded ASCs. In conclusion, we demonstrate that the HFF feeder layer increases adipocyte differentiation of ASCs and allows differentiation of low density seeded progenitor cells  into functional adipocytes.


Assuntos
Adipogenia , Tecido Adiposo/citologia , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Prepúcio do Pênis/citologia , Células-Tronco Mesenquimais/citologia , Adulto , Técnicas de Cocultura/métodos , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade
20.
Biochim Biophys Acta Mol Cell Res ; 1866(5): 945-956, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30826331

RESUMO

The maintenance of stem cells often requires the support of feeder cells. Primary mouse embryonic fibroblasts (MEFs) have traditionally been used as feeder cells, and although these MEF-derived feeder cells have exhibited a reasonable performance, they require repeated cell isolation, since MEFs cannot expand indefinitely. To overcome this limitation, immortalized cells, such as STO cells, have been used. However, one major disadvantage is that previously reported immortalized cells can only support stem cell cultures for a relatively short period, typically 4 to 7 days. In this study, we found that our newly established rat-derived fibroblasts immortalized by the expression of mutant cyclin-dependent kinase 4, cyclin D, and telomerase reverse transcriptase, can function as feeder cells for relatively long cell culture periods of approximately 14 days. The rat-derived immortalized cells developed in this study should be a useful source of feeder cells to support stem cell research.


Assuntos
Ciclina D/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Células Alimentadoras/metabolismo , Fibroblastos/metabolismo , Mutação , Células-Tronco/metabolismo , Telomerase/biossíntese , Animais , Linhagem Celular Transformada , Técnicas de Cocultura , Ciclina D/genética , Quinase 4 Dependente de Ciclina/genética , Células Alimentadoras/citologia , Fibroblastos/citologia , Camundongos , Ratos , Células-Tronco/citologia , Telomerase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA