The largest database of trusted experimental protocols

19 protocols using g csf

1

Monocyte Differentiation into Macrophages

Check if the same lab product or an alternative is used in the 5 most similar protocols
Peripheral blood mononuclear cells were isolated from buffy coats from healthy donors (Blutspende Zürich, Zurich, Switzerland) by density gradient centrifugation using Ficoll-paque PLUS (Cat: 17–1440–02; GE Healthcare). The monocyte population was magnetically enriched by negative selection using the Human Monocyte Isolation Kit II (Cat: 130–091–153; MACS, Miltenyi Biotech). The monocyte purity was >95% as confirmed by FACS analysis with an anti-CD14 antibody (BD Biosciences).
Freshly isolated human monocytes were differentiated into macrophages by incubation with either 50 ng/mL of human M-CSF (Cat: 14–8789) or G-CSF (Cat: 578602) (both from BioLegend) in RPMI medium (Cat: 61870–010; Gibco) supplemented with 1% heat-inactivated human AB serum (Cat: 34005100; Invitrogen), for up to 8 d. 20 ng/mL of recombinant human IL-4 (Cat: 204-IL; R&D Systems) was used as a control to induce M2-type macrophage differentiation and anti-human G-CSF (1 μg/mL, clone BVD13–3A5, Cat: 502101; BioLegend) or isotype control (rat IgG1 Cat: 400401) was used to neutralize G-CSF in MDA-MB-231 conditioned medium.
+ Open protocol
+ Expand
2

Neutrophil Progenitor Differentiation and Autophagy

Check if the same lab product or an alternative is used in the 5 most similar protocols
Neutrophil progenitors were maintained in growth media supplemented with 2% conditioned CHO media containing Stem Cell Factor (SCF) and 0.5 μM β-estradiol (Sigma-Aldrich, E2758, Saint Louis, MO, USA). The CRISPRed neutrophil cell lines were authenticated with WB.
HL-60 (ATCC, ccl240) were propagated and differentiated per manufacturer’s instructions without authentication.
To differentiate into granulocytes, cells were cultured in RPMI-1640 media supplemented with 10% FBS, 100 U/ml penicillin/100 μg/mL streptomycin, 2 mM L-Glutamine, 2% SCF in the presence or absence of 20 ng/mL GCSF (BioLegend, 574602, San Diego, CA, USA) or GCSF and 5 ng/ml GM-CSF (BioLegend, 576302, San Diego, CA, USA), and maintained for either 3 or 5 days before performing experiments. Cells were seeded into 6-well plates at a density of 2 × 106 cells/4 mL.
To examine changes in autophagy, neutrophils were treated with 50 μM Chloroquine phosphate (Sigma-Aldrich, 50-63-5, Saint Louis, MO, USA) or vehicle for 4 h before preparing protein lysates. To examine impact of blocking prenylation, neutrophils were treated with 10 μM Cysmethynil (Cayman chemicals, 14745, Ann Arbor, Michigan, USA) for three days.
+ Open protocol
+ Expand
3

Generation of Tamoxifen-Induced Neutrophil Progenitors

Check if the same lab product or an alternative is used in the 5 most similar protocols
HoxB8 conditionally immortalized neutrophil progenitors were generated as previously described (20 (link)–22 (link)). Briefly, C57BL/6 mouse bone marrow was isolated, cultured, and transduced with a tamoxifen-inducible Hoxb8 transgene. All cells were grown in Opti-Mem media supplemented with GlutaMax (Gibco), 30uM beta-mercaptoethanol (Sigma-Aldrich), 10% fetal bovine serum (Gemini Bio-Products, West Sacramento, CA), 1x penicillin/streptomycin (Gibco), 1x non-essential amino acids (Gibco) at a density of ~106 cells/mL. The progenitor cell culture was supplemented with stem cell factor (SCF) and 100nM Z-4-hydroxytamoxifen (Tocris Bioscience, Bristol, UK) to induce Hoxb8 expression. SCF was derived from the supernatant of CHO cells that secrete recombinant murine SCF (a gift from Patrice Dubreuil, Centre de Recherche en Cancérologie de Marseille). For differentiation, cells were washed 3x in PBS to remove Z-4-hydroxytamoxifen induction. Cells were pelleted by centrifugation at 400xg for 3 minutes. The differentiation culture was supplemented with SCF and 20 ng/mL G-CSF (BioLegend) for 2-3 days, and then just G-CSF until use on days 5-7.
+ Open protocol
+ Expand
4

Culturing Primary Cells and CML Cell Lines

Check if the same lab product or an alternative is used in the 5 most similar protocols
Primary cells were cultured in SFM supplemented with stem cell factor (SCF; 0.2 ng/ml; BioLegend, #573902), granulocyte colony-stimulating factor (G-CSF; 1 ng/ml; BioLegend, #578602), granulocyte-macrophage CSF (GM-CSF; 0.2 ng/ml; BioLegend, #572902), interleukin-6 (IL6; 1 ng/ml; BioLegend, #570802), macrophage inflammatory protein α (MIPα; 0.2 ng/ml; PeproTech, #300-08), leukemia inhibitory factor (LIF; 0.05 ng/ml; PeproTech, #300-05), [bovine serum albumin (BSA), insulin, and transferrin (BIT)] (20%; STEMCELL Technologies, #09500), low-density lipoprotein (40 μg/ml; Sigma-Aldrich, #L4646), 2-mercaptoethanol (0.1 mM; Invitrogen, #31350-010), and 1% penicillin/streptomycin (LifeTech, #15140-122) and resuspended in Iscove’s modified Dulbecco’s medium (LifeTech, #12440-053). The CML cell lines K562 and KCL22 (DSMZ) were cultured in RPMI 1640 medium (LifeTech, #11875-093) supplemented with 1% penicillin/streptomycin, 1% l-glutamine, and 10% (v/v) fetal calf serum. Both primary cells and cell lines were passaged every 2 to 3 days, maintained at a concentration of 2 × 105 cells/ml at 37°C with 5% CO2, and routinely tested for mycoplasma. ULK1 KO single-cell cloning was performed with serial dilution in a 96-well plate (100 μl per well). Puromycin selection (3 μg/ml) was performed after cell expansion, and Western blotting was performed to validate ULK1 KO.
+ Open protocol
+ Expand
5

Cytokine-Induced Proliferation of MeT-5A Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
MeT-5A cells were plated at a density of 3×105 cells per 10-cm dish and cultured 72 h prior to the following experiments. The cells were then cultured in 96-well tissue culture plates (6×103 cells per well) in 100 µl of medium containing 12.5 µl of the supernatant collected from the PBMC culture. After 48 h of culture, the growth properties were analyzed using a water-soluble tetrazolium salt (WST-1) assay, as described previously (21 (link)). MeT-5A cells were also treated with the following recombinant cytokines: G-CSF (1.0 ng/ml; catalog no., 561701; BioLegend, San Diego, CA, USA), GM-CSF (10 ng/ml; catalog no., 572902), IL-1α (2.5 ng/ml; catalog no., 570002), IL-1β (2.5 ng/ml; catalog no., 579402), IL-3 (1.0 ng/ml; catalog no., 560503), IL-5 (1.0 ng/ml; catalog no., 560701), IL-13 (5.0 ng/ml; catalog no., 571102) and IL-17A (50 ng/ml; catalog no., 570502) for 48 h; the concentrations of cytokines were determined according to the supernatant of PBMCs. The WST-1 assay was then performed using the same protocol. Optical density at 450 nm (OD450 nm) was measured and normalized to a standard condition, which was the OD450 nm without supernatants or cytokines.
+ Open protocol
+ Expand
6

Flow Cytometry with Recombinant Cytokines

Check if the same lab product or an alternative is used in the 5 most similar protocols
Commercial antibodies for use in flow cytometry were purchased from BD Biosciences, Biolegend or eBioscience. Recombinant mouse IL-2 and α-IL-2 monoclonal antibody, clone JES6-5H4 (eBioscience). IL-2/α-IL-2 complex was generated by incubating 1.5 μg recombinant mouse IL-2 with 8 μg JES6-5H4 for 15 minutes at RT. TL1A-Ig was generated as described previously28 (link). The A20luc/YFP cell line (derived from BALB/c mice) was a generous gift of Dr. Robert Negrin (Stanford University) and used for GVL experiments30 (link). Luciferin was purchased from Perkin Elmer; G-CSF (Biolegend) and AMD3100 (Sigma Aldrich).
+ Open protocol
+ Expand
7

Modulation of Neutrophil Function by AMs

Check if the same lab product or an alternative is used in the 5 most similar protocols
AMs co-cultured with BAL–neutrophils at a 1:1 ratio were treated with or without different inhibitors in medium, including G-CSF (10 ng/mL, BioLegend), Q-VD-Oph (50 μM, Selleckchem, Houston, TX, USA), NAC (1 mM, Selleckchem), and DFO (1 μM, Selleckchem), the control was co-culture treated without inhibitors. After 24 h, the cells were cultured in complete medium containing 10 ng/mL recombinant murine GM-CSF, and cells were analyzed by flow cytometry.
+ Open protocol
+ Expand
8

Myeloid Cell Growth and Differentiation Analysis

Check if the same lab product or an alternative is used in the 5 most similar protocols
To assess myeloid cell growth and differentiation in bulk liquid culture, human HSPC were maintained in Iscove’s Modified Dulbecco’s Medium (IMDM; Fisher Scientific, Loughborough, UK) supplemented with IL-3, SCF, G-CSF and GM-CSF (BioLegend, London, UK) at 5 ng/mL for 13 days. Transduced cultures were analyzed by flow cytometry for cell surface markers expression using a BD FACSCanto™II (Supplemental Materials and Methods) as previously described [12 , 14 (link)]. The gating strategy employed in these studies is shown in Supplemental Fig. S1. Granulocytic cells were defined as CD13−/+CD36, monocytic cells as CD13+CD36+, and erythroid cells as CD13CD36+. Measurement of cell motility using the Transwell® cell migration assay was performed on day 6 of culture using stromal cell-derived factor 1 (SDF-1) as a chemoattractant (Supplemental Materials and Methods). Morphology was assessed on day 17 of culture (Supplemental Materials and Methods) [13 (link)].
+ Open protocol
+ Expand
9

Inducing Myeloid-Derived Suppressor Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
The in vitro differentiation of bone marrow (BM) cells into MDSCs was performed as described previously (Xie et al., 2018 (link)). Briefly, BM cells from C57BL/6J mice were stimulated with 40 ng/ml recombinant Granulocyte-macrophage CSF (GM-CSF) (Peprotech, NJ, United States) for 4 days in the absence or presence of G-CSF (5 ng/ml, BioLegend, CA, United States) and/or GGsTop (50 mΜ, FUJIFILM Wako) to examine their effects on MDSCs.
+ Open protocol
+ Expand
10

Quantifying Pulmonary S. aureus Burden

Check if the same lab product or an alternative is used in the 5 most similar protocols
Twenty-four hours after oropharyngeal S. aureus or saline inhalation, mice were euthanized by intraperitoneal injection of Fatal Plus euthanasia solution. Immediately following euthanasia, mouse whole blood was collected by cardiac puncture and serum was separated from the cellular components of blood by centrifugation. All lobes of the lungs were dissected free and dissociated in gentleMACS C tubes on a gentleMACS dissociator (Miltenyi, Auburn, CA) in a solution of in 2 mg/mL collagenase V (Sigma-Aldrich, St. Louis, MO) in Hanks Buffered Saline Solution and digested at 37°C for 40 min. The S. aureus burden was calculated by plating 10 μL of unfiltered serial dilutions of digested lungs in triplicate on tryptic soy agar plates. The digested lung was then filtered through a 70-μm cell strainer (Falcon, Corning, NY) and centrifuged to remove the cellular components. For determining the cytokine profile of serum 24 h after hemorrhage, blood was collected by saphenous vein puncture and separated by centrifugation. The lung and serum cytokine and chemokine profile were determined by cytometric bead array assay for multiplexed assessment of murine IL-6, CXCL1, TNFα, IL-1β, IL-10, G-CSF, GM-CSF, and CXCL5 per the manufacturer's protocol (BioLegend).
+ Open protocol
+ Expand

About PubCompare

Our mission is to provide scientists with the largest repository of trustworthy protocols and intelligent analytical tools, thereby offering them extensive information to design robust protocols aimed at minimizing the risk of failures.

We believe that the most crucial aspect is to grant scientists access to a wide range of reliable sources and new useful tools that surpass human capabilities.

However, we trust in allowing scientists to determine how to construct their own protocols based on this information, as they are the experts in their field.

Ready to get started?

Sign up for free.
Registration takes 20 seconds.
Available from any computer
No download required

Sign up now

Revolutionizing how scientists
search and build protocols!