Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 16(7): 1686-1696, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34214484

ABSTRACT

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.


Subject(s)
Feeder Cells/cytology , Pluripotent Stem Cells/cytology , Cell Line , Chimera/physiology , Feeder Cells/drug effects , Glycolysis/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Indoles/pharmacology , Pluripotent Stem Cells/drug effects , Pyridones/pharmacology
3.
Leuk Res ; 108: 106617, 2021 09.
Article in English | MEDLINE | ID: mdl-34022745

ABSTRACT

The present study aimed to investigate the ability of a novel form of the anti-angiogenic molecular antibody drug to induce Myeloma cell death after cultivation upon endothelial feeder cells. Bevacizumab-loaded chitosan (BCS) nanoparticles (NPs) were prepared by the ionic gelation method. Human U266 cell line and human umbilical vein endothelial cells were co-cultured for 72 h and treated with BCS nanoparticles (10µM) to study their impact on inhibition of cell growth and induction of apoptosis. Death assessments, P53 pro-apoptotic marker expression and the VEGF level were investigated by flow-cytometric analyses of the Annexin V, immunocytochemistry and ELISA, respectively. The endothelial monolayer co-culture showed protection of myeloma cells from apoptosis when exposed to NPs or without any treatment. In present of bevacizumab, the VEGF factor was effectively suppressed, and the p53 expression was significantly increased in bevacizumab-treated myeloma cells co-cultured with HUVECs, compared to other groups. BCS was capable of disturbing the effective interconnections of myeloma cells and HUVECs as supportive cells. Disruptions of tumor cells' connections with their microenvironment and blocking their possible supportive pathways might be a potential strategy to eradicate tumor cells and finally cure such types of cancer.


Subject(s)
Apoptosis , Bevacizumab/pharmacology , Feeder Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Multiple Myeloma/pathology , Nanoparticles/administration & dosage , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Cell Proliferation , Coculture Techniques , Humans , Multiple Myeloma/drug therapy , Nanoparticles/chemistry , Neovascularization, Pathologic/pathology
4.
Methods Mol Biol ; 2239: 199-211, 2021.
Article in English | MEDLINE | ID: mdl-33226621

ABSTRACT

The development of porcine expanded potential stem cells (pEPSCs) provides an invaluable tool for investigation of porcine stem cell pluripotency and opens a venue for research in biotechnology, agriculture, and regenerative medicine. Since the derivation of pEPSC from porcine pre-implantation embryos has been demanding in resource supply and technical challenges, it is more feasible and convenient for most laboratories to derive this new type of porcine stem cells by reprogramming somatic cells. In this chapter, we describe the detailed procedures for reprogramming porcine fetal fibroblast cells to EPSCiPSC with the eight reprogramming factors cloned on the piggyBac vectors followed by a selection for pluripotent cells independent of transgene expression using the EPSC media. This technique allows the generation of pEPSCs for stem cell research, genome editing, biotechnology, and agriculture.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Animals , Cryopreservation/methods , Feeder Cells/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mitomycin/pharmacology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Real-Time Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Swine , Transcription Factors/genetics , Transfection
5.
Cytotherapy ; 22(7): 354-368, 2020 07.
Article in English | MEDLINE | ID: mdl-32451262

ABSTRACT

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.


Subject(s)
Autoantigens/immunology , Feeder Cells/cytology , Immune Tolerance , Killer Cells, Natural/cytology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Cetuximab/pharmacology , Feeder Cells/drug effects , HEK293 Cells , Humans , Immune Tolerance/drug effects , Interleukin-2/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Ligands
6.
Cytotherapy ; 22(6): 322-328, 2020 06.
Article in English | MEDLINE | ID: mdl-32278551

ABSTRACT

BACKGROUND: Optimal expansion of therapeutic natural killer (NK) cell products has required media supplementation with human or fetal bovine serum, which raises safety and regulatory concerns for clinical manufacturing. Serum-free media (SFM) have been optimized for T-cell expansion, but few SFM systems have been developed for NK cells. Here, we compare six commercial clinical-grade SFM with our standard fetal bovine serum-containing medium for their ability to support NK cell expansion and function. METHODS: Human peripheral blood NK cells were expanded in selected media by recursive weekly stimulation with K562-based feeder cells expressing membrane-bound interleukin-21 and CD137L. Expansion was the primary readout, and the best-performing SFM was then compared with standard medium for cytotoxicity, phenotype, degranulation and cytokine secretion. Multiple lots were compared for consistency, and media was analyzed throughout for nutrient consumption and metabolic byproducts. RESULTS: TexMACS, OpTmizer, SCGM, ABS-001 and StemXVivo demonstrated equal or inferior NK cell expansion kinetics compared with standard medium, but expansion was markedly superior with AIM V + 5% Immune Cell Serum Replacement (ICSR; mean 5448 vs. 2621-fold expansion in 14 days). Surprisingly, NK cells expanded in AIM V + ICSR also showed increased cytotoxicity, tumor necrosis factor α secretion and DNAM-1, NKG2D, NKp30, FasL, granzyme B and perforin expression. Lot-to-lot variability was minimal. Glucose and glutamine consumption were inversely related to lactate and ammonia production. DISCUSSION: The AIM V + ICSR SFM system supports excellent ex vivo expansion of clinical-grade NK cells with the phenotype and function needed for adoptive immunotherapy.


Subject(s)
Culture Media, Serum-Free/pharmacology , Feeder Cells/drug effects , Killer Cells, Natural/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Culture Techniques/methods , Cell Proliferation/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Culture Media, Serum-Free/chemistry , Cytotoxicity, Immunologic , Fas Ligand Protein/metabolism , Humans , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 3/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Immunol Lett ; 220: 79-87, 2020 04.
Article in English | MEDLINE | ID: mdl-31901377

ABSTRACT

Adoptive cell therapy using ex vivo expanded lymphocytes has shown remarkable efficacy in tumor immunotherapy recently. Among various transfused immune cells, T lymphocytes are the most widely used since they are critical mediators of the immune system and have the capacity to kill tumor cells. However, there are drawbacks in the expanded T cells for transfusion including limited cytotoxicity, limited proliferation and lack of specificity. To improve the quality of these ex vivo expanded T cells, we have designed a new method to expand a group of T cells which are named bispecific antibodies activated T cells. It is the first time that such cells are induced by introducing the bispecific antibody drug (blinatumomab) and feeder cells (normal B cells and irradiated B cell originated lymphoma cells) to the traditional T cells culture system. Culture of freshly isolated human peripheral blood mononuclear cells in this newly designed cell culture system enabled these expanded T cells that (a) displayed a robust proliferation ability; (b) showed fully activated phenotype and enhanced cytokines production; (c) had a low proportion of CD4+CD25+ T regulatory cells and (d) exhibited strengthened cytotoxicity at relatively low effector: target ratios. This work further confirmed the feasibility of rapid induction and expansion of large amounts of human T cells in vitro by using bispecific antibodies and feeder cells. This strategy could also be used for other immune cells rapid expansion and help to improve the quality of these expanded immune cells for adoptive transfusion.


Subject(s)
Antibodies, Bispecific/pharmacology , Cytotoxicity, Immunologic/drug effects , Immunotherapy, Adoptive , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , Antigens, CD19/analysis , Antigens, CD19/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD4 Antigens/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Feeder Cells/drug effects , Feeder Cells/immunology , Humans , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocytes, Regulatory/immunology
8.
Cold Spring Harb Protoc ; 2019(11)2019 11 01.
Article in English | MEDLINE | ID: mdl-31676582

ABSTRACT

Single-cell cloning during hybridoma production ensures that cells that produce the antibody of interest are truly monoclonal and that the secretion of this antibody can be stably maintained. Cloning of hybridoma cells in semisolid medium is one of the most commonly used methods for producing single-cell clones. The technique is easy, but, because it is performed in two stages, it does take longer than other methods. Not all cells will grow in soft agar, and there may be a bias on the type of colony that appears. However, most of the commonly used myeloma fusion partners have relatively good cloning efficiencies in soft agar, and, consequently, so do most hybridomas. Even though every attempt is made to ensure that the cells are in a single-cell suspension before plating, there is no way to guarantee that the colonies do not arise from two cells that were stuck together. Therefore, single-cell cloning in soft agar should be repeated at least twice before the cells are considered clonal.


Subject(s)
Agar/pharmacology , Cloning, Molecular/methods , Hybridomas/cytology , Single-Cell Analysis , Animals , Cell Proliferation/drug effects , Feeder Cells/cytology , Feeder Cells/drug effects , Humans
9.
Int J Mol Med ; 43(5): 2230-2240, 2019 May.
Article in English | MEDLINE | ID: mdl-30864673

ABSTRACT

Hair follicles (HFs) are a well­characterized niche for adult stem cells (SCs), and include epithelial and melanocytic SCs. HF cells are an accessible source of multipotent adult SCs for the generation of the interfollicular epidermis, HF structures and sebaceous glands in addition to the reconstitution of novel HFs in vivo. In the present study, it was demonstrated that HF cells are able to be induced to differentiate into cardiomyocyte­like cells in vitro under specific conditions. It was determined that HF cells cultured on OP9 feeder cells in KnockOut­Dulbecco's modified Eagle's medium/B27 in the presence of vascular endothelial growth factors differentiated into cardiomyocyte­like cells that express markers specific to cardiac lineage, but do not express non­cardiac lineage markers including neural stem/progenitor cell, HF bulge cells or undifferentiated spermatogonia markers. These cardiomyocyte­like cells exhibited a spindle­ and filament­shaped morphology similar to that presented by cardiac muscles and exhibited spontaneous beating that persisted for over 3 months. These results demonstrate that SC reprogramming and differentiation may be induced without resulting in any genetic modification, which is important for the clinical applications of SCs including tissue and organ regeneration.


Subject(s)
Hair Follicle/cytology , Myocytes, Cardiac/cytology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cells, Cultured , Culture Media/pharmacology , Feeder Cells/cytology , Feeder Cells/drug effects , Gene Expression Regulation/drug effects , Mice , Myocytes, Cardiac/drug effects , Phenotype , Vascular Endothelial Growth Factor A/pharmacology
10.
Cell Rep ; 25(3): 598-610.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332641

ABSTRACT

Despite substantial self-renewal capability in vivo, epithelial stem and progenitor cells located in various tissues expand for a few passages in vitro in feeder-free condition before they succumb to growth arrest. Here, we describe the EpiX method, which utilizes small molecules that inhibit PAK1-ROCK-Myosin II and TGF-ß signaling to achieve over one trillion-fold expansion of human epithelial stem and progenitor cells from skin, airway, mammary, and prostate glands in the absence of feeder cells. Transcriptomic and epigenomic studies show that this condition helps epithelial cells to overcome stresses for continuous proliferation. EpiX-expanded basal epithelial cells differentiate into mature epithelial cells consistent with their tissue origins. Whole-genome sequencing reveals that the cells retain remarkable genome integrity after extensive in vitro expansion without acquiring tumorigenicity. EpiX technology provides a solution to exploit the potential of tissue-resident epithelial stem and progenitor cells for regenerative medicine.


Subject(s)
Epithelial Cells/cytology , Myosin Type II/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Stem Cells/cytology , Transforming Growth Factor beta/antagonists & inhibitors , p21-Activated Kinases/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Feeder Cells/cytology , Feeder Cells/drug effects , Feeder Cells/metabolism , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Myosin Type II/genetics , Myosin Type II/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
11.
Int J Mol Sci ; 19(8)2018 Jul 25.
Article in English | MEDLINE | ID: mdl-30044428

ABSTRACT

Human keratinocyte culture has provided the means to treat burns, wounds and skin pathologies. To date, to efficiently culture keratinocytes, cells are cultured on an irradiated feeder layer (iFL), either comprising human (iHFL) or murine (i3T3FL) fibroblasts, and the culture medium is supplemented with a cyclic adenosine monophosphate (cAMP) accumulation inducing agent such as isoproterenol (ISO) or cholera toxin (CT). Previous studies have characterized how the feeder layer type and the cAMP inducer type influence epithelial cells' phenotype independently from one another, but it is still unknown if an optimal combination of feeder layer and cAMP inducer types exists. We used sophisticated statistical models to search for a synergetic effect of feeder layer and cAMP inducer types on human keratinocytes' proliferative potential. Our data suggests that, when culturing human keratinocytes, using iHFL over i3T3FL increases population doublings and colony-forming efficiency through signaling pathways involving Ak mouse strain thymoma (Akt, also known as protein kinase B) isoforms 1 to 3, signal transducer and activator of transcription 5 (STAT5), p53, and adenosine monophosphate activated protein kinase α1 (AMPKα1). Both tested cAMP inducers ISO and CT yielded comparable outcomes. However, no significant synergy between feeder layer and cAMP inducer types was detected. We conclude that, to promote human keratinocyte growth in the early passages of culture, co-culturing them with a human feeder layer is preferable to a murine feeder layer.


Subject(s)
Cholera Toxin/pharmacology , Feeder Cells/drug effects , Fibroblasts/drug effects , Isoproterenol/pharmacology , Keratinocytes/drug effects , 3T3 Cells , Adult , Aged , Animals , Cell Proliferation/drug effects , Coculture Techniques , Culture Media/pharmacology , Cyclic AMP/pharmacology , Female , Fibroblasts/cytology , Humans , Keratinocytes/cytology , Mice , Middle Aged , Models, Statistical
12.
Methods Mol Biol ; 1773: 31-39, 2018.
Article in English | MEDLINE | ID: mdl-29687379

ABSTRACT

In this chapter, we describe a protocol to induce at a high rate the differentiation of brown/brown like adipocyte progenitors (BAPs) derived from human induced pluripotent stem cells (hiPSCs). We also describe culture conditions to maintain hiPSCs and to derive hiPSC-BAPs.This novel culture system provides an unlimited source of human brown adipocytes and a unique means for studying events regulating the generation and recruitment of human BAPs.


Subject(s)
Adipocytes, Brown/cytology , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Induced Pluripotent Stem Cells/cytology , Amides/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Feeder Cells/drug effects , Fibroblasts/drug effects , Humans , Mice , Mitomycin/pharmacology , Pyridines/pharmacology
13.
Clin Cancer Res ; 24(16): 4006-4017, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29666301

ABSTRACT

Purpose: Daratumumab and its use in combination with other agents is becoming a new standard of care for the treatment of multiple myeloma. We mechanistically studied how daratumumab acts on natural killer (NK) cells.Experimental Design: Quantities of NK cells in peripheral blood and/or bone marrow of patients with multiple myeloma or healthy donors were examined by flow cytometry. NK-cell apoptosis and the associated mechanism were assessed by flow cytometry and immunoblotting. Patients' NK cells were expanded in vitro using feeder cells. Combination treatment of daratumumab and expanded NK cells was performed using an MM.1S xenograft animal model.Results: CD38-/low NK cells survived, whereas CD38+ NK cells were almost completely eliminated, in peripheral blood and bone marrow of daratumumab-treated multiple myeloma patients. NK-cell depletion occurred due to daratumumab-induced NK-cell fratricide via antibody-dependent cellular cytotoxicity. Consequently, CD38-/low NK cells were more effective for eradicating multiple myeloma cells than were CD38+ NK cells in the presence of daratumumab. Blockade of CD38 with the F(ab)2 fragments of daratumumab inhibited the antibody-mediated NK-cell fratricide. CD38-/low NK cells displayed a significantly better potential for expansion than CD38+ NK cells, and the expanded NK cells derived from the former population were more cytotoxic than those derived from the latter against multiple myeloma cells. Therefore, infusion of ex vivo-expanded autologous NK cells from daratumumab-treated patients may improve the antibody therapy.Conclusions: We unravel a fratricide mechanism for daratumumab-mediated NK-cell depletion and provide a potential therapeutic strategy to overcome this side effect in daratumumab-treated patients with multiple myeloma. Clin Cancer Res; 24(16); 4006-17. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Killer Cells, Natural/immunology , Multiple Myeloma/drug therapy , ADP-ribosyl Cyclase 1/immunology , Animals , Antibodies, Monoclonal/adverse effects , Antibody-Dependent Cell Cytotoxicity/immunology , Apoptosis/drug effects , Cell Line, Tumor , Feeder Cells/drug effects , Flow Cytometry , Heterografts , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Membrane Glycoproteins/immunology , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology
14.
Biomaterials ; 171: 1-11, 2018 07.
Article in English | MEDLINE | ID: mdl-29677519

ABSTRACT

Graphene oxide (GO), with good hydrophilicity and biocompatibility, is widely explored as a carrier for various factors in the field of stem cell differentiation. However, its function of sustaining the stemness of mouse embryonic stem cells (mESCs) and the underlying mechanisms of this process remains undiscovered. Herein, we explored the biofunction of GO on mESCs and revealed the involved signaling pathways and key gene. The alkaline phosphatase activity detection, pluripotency genes quantification and the teratomas formation in vivo confirmed that GO nanosheets could sustain the self-renewal ability of mESCs instead of influencing its pluripotency. The underlying signaling pathways were uncovered by RNA-seq that integrin signaling pathway was involved in the biofunction of GO on mESCs and Vinculin turned to be a key gene for the effect of GO. Further experiments confirmed that the downregulation of Vinculin influenced the fate of mESCs through decreasing the expression of MEK1. Altogether, the study demonstrated for the first time that GOs hold the potential in sustaining the self-renewal of mESCs and clarified the mechanism of this function, which make it play a new role in stem cell research and regenerative medicine.


Subject(s)
Cell Self Renewal/drug effects , Down-Regulation/drug effects , Graphite/pharmacology , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nanostructures/chemistry , Vinculin/genetics , Alkaline Phosphatase/metabolism , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Embryo, Mammalian/cytology , Feeder Cells/cytology , Feeder Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Integrins/metabolism , Mice , Mouse Embryonic Stem Cells/drug effects , Nanostructures/ultrastructure , Signal Transduction/drug effects , Vinculin/metabolism
15.
PLoS One ; 12(7): e0181985, 2017.
Article in English | MEDLINE | ID: mdl-28750033

ABSTRACT

The developmental progression of conventional DC has been quite well defined, yet the developmental pathway of monocyte-derived, GM-CSF-driven DC is less well understood. We addressed this issue by establishing an isolation strategy that identifies five distinct GM-CSF derived cell types. Expression of Ly6C and CD115 (Csf-1R) was used to identify and isolate four populations. One of the populations could be further separated based on CD11c expression, distinguishing five populations. We further defined these cells based on expression of transcription factors and markers of early and later stages of myeloid development. These discreet developmental stages corresponded well with previously defined populations: Common Myeloid Progenitors (CMP), Granulocyte/Macrophage Progenitors (GMP), Monocytes, as well as Monocyte-derived macrophages (moMac) and Monocyte-derived DC (moDC). Finally, within the moMac population we also identified moDC precursor activity (moDP) that could be distinguished from moMac and moDC based on their level of MHC class II expression and developmental plasticity.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Animals , Antigens, Ly/metabolism , Biomarkers/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Separation , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Feeder Cells/cytology , Feeder Cells/drug effects , Feeder Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class II/metabolism , Inflammation/genetics , Inflammation/pathology , Kinetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Models, Biological , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Myeloid Cells/drug effects , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/metabolism
16.
Wound Repair Regen ; 25(3): 526-531, 2017 05.
Article in English | MEDLINE | ID: mdl-28437853

ABSTRACT

Cultured epidermal autografts have been used worldwide since 1981 for patients with extensive third-degree burn wounds and limited skin donor sites. Despite significant progress in techniques toward improving clinical outcome of skin grafts, the long in vitro preparation time of cultured autografts has remained a major factor limiting its widespread use. Here, we show that pharmacological inhibition of TGF-ß signaling promotes the expansion of human epidermal keratinocytes (HEKs) with high proliferative potential in co-cultures with both murine 3T3-J2 cells and human feeder cells, including dermal fibroblasts and preadipocytes. In contrast, TGF-ß signaling inhibition does not enhance the growth of HEKs in a serum- and feeder-free condition, an alternative approach to propagate HEKs for subsequent autograft production. Our results have important implications for the use of TGF-ß signaling inhibition as a viable therapeutic strategy for improving Green's methodology and for more efficient production of customized skin autografts with human feeder cells.


Subject(s)
Coculture Techniques , Epidermal Cells , Feeder Cells/cytology , Keratinocytes/cytology , Keratinocytes/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , 3T3 Cells , Animals , Disease Models, Animal , Epidermis/drug effects , Feeder Cells/drug effects , Humans , Mice , Skin Transplantation/methods
17.
Nat Protoc ; 12(2): 439-451, 2017 02.
Article in English | MEDLINE | ID: mdl-28125105

ABSTRACT

Historically, it has been difficult to propagate cells in vitro that are derived directly from human tumors or healthy tissue. However, in vitro preclinical models are essential tools for both the study of basic cancer biology and the promotion of translational research, including drug discovery and drug target identification. This protocol describes conditional reprogramming (CR), which involves coculture of irradiated mouse fibroblast feeder cells with normal and tumor human epithelial cells in the presence of a Rho kinase inhibitor (Y-27632). CR cells can be used for various applications, including regenerative medicine, drug sensitivity testing, gene expression profiling and xenograft studies. The method requires a pathologist to differentiate healthy tissue from tumor tissue, and basic tissue culture skills. The protocol can be used with cells derived from both fresh and cryopreserved tissue samples. As approximately 1 million cells can be generated in 7 d, the technique is directly applicable to diagnostic and predictive medicine. Moreover, the epithelial cells can be propagated indefinitely in vitro, yet retain the capacity to become fully differentiated when placed into conditions that mimic their natural environment.


Subject(s)
Cellular Reprogramming , Coculture Techniques/methods , Neoplasms/pathology , Amides/pharmacology , Animals , Cell Transformation, Neoplastic , Feeder Cells/cytology , Feeder Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , rho-Associated Kinases/antagonists & inhibitors
18.
Bull Exp Biol Med ; 162(1): 127-133, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27882459

ABSTRACT

A series of experiments on co-culturing of Mel IL melanoma cells and mesenhymal stem cells showed that these cells do not influence proliferation of each other, but we observed weaker adhesion of stromal stem cells to plastic in cocultures where with melanoma cells were grown on mesenhymal stem cells feeder. Cell proliferation was also considerably influenced by experimental conditions, which should be taken into account for correct interpretation of obtained results. The principles of experiments on co-culturing of cancer and stromal cells are formulated that take into account the most important factors influencing cell behavior and minimize the probability of artifact results. It was concluded that co-culturing conditions cells significantly affect the experimental results and can be the source of conflicting conclusions on mutual influence of stromal and cancer cells in vitro.


Subject(s)
Adipose Tissue/drug effects , Culture Media/pharmacology , Feeder Cells/drug effects , Melanoma/metabolism , Mesenchymal Stem Cells/drug effects , Skin Neoplasms/metabolism , Adipose Tissue/cytology , Animals , Cell Communication/drug effects , Cell Count , Cell Proliferation/drug effects , Coculture Techniques , Data Interpretation, Statistical , Feeder Cells/cytology , Humans , Melanoma/pathology , Mesenchymal Stem Cells/cytology , Primary Cell Culture , Rats , Skin Neoplasms/pathology , Tumor Cells, Cultured
19.
Curr Protoc Stem Cell Biol ; 38: 1G.6.1-1G.6.18, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27532814

ABSTRACT

Hepatocyte-like cells (HLCs) generated in vitro from human pluripotent stem cells (hPSCs) provide an invaluable resource for basic research, regenerative medicine, drug screening, toxicology, and modeling of liver disease and development. This unit describes a small-molecule-driven protocol for in vitro differentiation of hPSCs into HLCs without the use of growth factors. hPSCs are coaxed through a developmentally relevant route via the primitive streak to definitive endoderm (DE) using the small molecule CHIR99021 (a Wnt agonist), replacing the conventional growth factors Wnt3A and activin A. The small-molecule-derived DE is then differentiated to hepatoblast-like cells in the presence of dimethyl sulfoxide. The resulting hepatoblasts are then differentiated to HLCs with N-hexanoic-Tyr, Ile-6 aminohexanoic amide (Dihexa, a hepatocyte growth factor agonist) and dexamethasone. The protocol provides an efficient and reproducible procedure for differentiation of hPSCs into HLCs utilizing small molecules. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Cell Differentiation/drug effects , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Small Molecule Libraries/pharmacology , Tissue Culture Techniques/methods , Endoderm/cytology , Feeder Cells/cytology , Feeder Cells/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
20.
Article in English | MEDLINE | ID: mdl-27178105

ABSTRACT

INTRODUCTION: Growth-arrested feeder cells following Mitomycin C treatment are instrumental in stem cell culture allowing development of regenerative strategies and alternatives to animal testing in drug discovery. The concentration of Mitomycin C and feeder cell type was described to affect feeder performance but the criticality of feeder cell exposure density was not addressed. We hypothesize that the exposure cell density influences the effectiveness of Mitomycin C in an arithmetic manner. METHODS: Three different exposure cell densities of Swiss 3T3 fibroblasts were treated with a range of Mitomycin C concentrations for 2h. The cells were replaced and the viable cells counted on 3, 6, 9, 12 and 20days. The cell extinctions were compared with doses per cell which were derived by dividing the product of concentration and volume of Mitomycin C solution with exposure cell number. RESULTS: The periodic post-treatment feeder cell extinctions were not just dependent on Mitomycin C concentration but also on dose per cell. Analysis of linearity between viable cell number and Mitomycin C dose per cell derived from the concentrations of 3 to 10µg/ml revealed four distinct categories of growth-arrest. Confluent cultures exposed to low concentration showed growth-arrest failure. DISCUSSION: The in vitro cell density titration can facilitate prediction of a compound's operational in vivo dosing. For containing the growth arrest failure, an arithmetic volume derivation strategy is proposed by fixing the exposure density to a safe limit. The feeder extinction characteristics are critical for streamlining the stem cell based pharmacological and toxicological assays.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cell Culture Techniques/methods , Cell Cycle Checkpoints/drug effects , Feeder Cells/drug effects , Mitomycin/toxicity , Stem Cells/drug effects , Animals , Cell Count , Cell Division/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Mice , Stimulation, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...