Free access supported by contributions and sponsoring — share your knowledge or support us financially
Search / Compare / Validate Lab equipment & Methods

β mercaptoethanol

Manufactured by Thermo Fisher Scientific
Sourced in United States, Germany, United Kingdom, Switzerland, Canada, France, China, Australia, Italy, Belgium, Japan, Austria, Netherlands, Denmark, Israel, Ireland, Sweden, Norway, Macao, Brazil, Spain, Colombia, New Zealand
About the product

β-mercaptoethanol is a reducing agent commonly used in biochemical applications. It functions by breaking disulfide bonds and maintaining proteins in a reduced state.

Automatically generated - may contain errors

Market Availability & Pricing

β-Mercaptoethanol is actively commercialized by Thermo Fisher Scientific under various product lines, including Thermo Scientific™ Pierce™, Gibco™, and Thermo Scientific Chemicals. It is available through authorized distributors such as Fisher Scientific. Prices vary depending on the product line and packaging, ranging from $12.57 for a 50 mM, 20 mL Gibco™ product to $618.87 for a 2.5 L Thermo Scientific Chemicals product.

Need Operating Instructions, SDS, or distributor details? Just ask our AI Agent.

Is this product still available?

Get pricing insights and sourcing options

Product FAQ

4 095 protocols using «β mercaptoethanol»

1

Isolation and Culture of CD8+ T Cells

2025
CD8 + T cells were obtained using a CD8a + T cell isolation kit (Miltenyi, #130-104-075, Germany). The average purity assessed by flow cytometry was more than 90% (Fig. 2E). Cells were centrifuged and resuspended in a complete medium (CM) containing RPMI 1640, 10% heat-inactivated FBS (Gibco, USA), 1% L-glutamine (Life Technologies #35050–061) with penicillin-streptomycin, 0.1% of 50 mM β-mercaptoethanol (Gibco, #21985-023), 10ng/mL IL-2 (PeproTech Inc., #212 − 12, USA) and 2% of 100 mM sodium pyruvate solution.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
2

Derivation of Induced Pluripotent Stem Cells from Tail Tip Fibroblasts

2025
TTFs were isolated from 8‐week‐old C57BL/6 mice. Briefly, mice were euthanized by carbon dioxide asphyxiation, and their entire tail was excised and washed with 70% ethanol. A surgical cut was made along the lateral axis of the tail, and the superficial dermis layer was removed. The tail was further rinsed with phosphate buffer saline (PBS; Corning), cut into small 2–3‐mm sections, and submerged in several wells of a six‐well plate coated with 0.1% gelatin (Millipore Sigma) in Dulbecco's modified Eagle medium (DMEM; Corning) supplemented with 10% fetal bovine serum (FBS; Gibco) and 1% penicillin‐streptomycin (Gibco). The TTFs were allowed to naturally migrate out of the tail at 37 °C, and the plate was also shaken daily to facilitate even distribution of the cells. Tail pieces and media were discarded after 5 days, and the wells were replenished with fresh medium. Once confluent, the TTFs were used immediately for iPSC reprogramming.
Reprogramming was carried out by using the CytoTune‐iPS 2.0 Sendai reprogramming kit (Invitrogen) following the manufacturer's instructions. Briefly, TTFs were seeded into six‐well plates at 1 × 105 per well and TTFs were infected 2 days later with the KOS, c‐Myc, and Klf4 Sendai viruses at a multiplicity of infection of 5. The media was replaced daily for 1 week, after which the reprogrammed TTFs were plated onto 0.1% gelatin‐coated six‐well plates with 5 × 105 mouse embryonic fibroblasts (MEFs; Gibco) as feeders. The following day, the medium was discarded and replaced with KnockOut DMEM/F‐12 (Gibco) supplemented with 20% KnockOut serum replacement (Gibco), 1% non‐essential amino acids (Gibco), 1% GlutaMAX (Gibco), 0.1 mm β‐mercaptoethanol (Gibco), and 10 ng mL−1 leukemia inhibitory factor (StemCell Technologies). The medium was changed daily, and emerging iPSC colonies exhibiting compact embryonic stem cell‐like morphology were manually picked and transferred onto fresh MEF feeder layers. Established iPSC clones were expanded by passaging every 4 days. The reprogramming process was performed in the absence of penicillin‐streptomycin, and the iPSCs were subsequently cultured without any antibiotics.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
3

Feeder-free mESC Maintenance Protocol

2025
M. musculus embryonic stem cells (mESC E14tg2A) were cultured under feeder-free conditions using 0.1% Gelatine (Millipore, #ES-006-B) coated plates (ThermoFisher, #140675) and grown in mESC medium prepared as follows: KnockOut™ DMEM (ThermoFisher, 10829018) supplemented with 10% FBS (in-house tested for ES competence) MEM Non-Essential Amino Acids Solution 1X (ThermoFisher, #11140050), GlutaMAX™ 2 mM (ThermoFisher, #35050061), Pen/Strep 1X (ThermoFisher, #15140122), β-mercaptoethanol 50 µM (Gibco, #31350010), HEPES 30 mM (Gibco, #15630080), 0,22 µm filtered and then supplemented with LIF conditioned (may 2021- in house tested). Cells were passed every 2-3 days in a 1:6-1:8 dilution.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
4

Expansion and Transduction of Human T Cells

2025
PBMCs were stimulated with ImmunoCult Human CD3/CD28/CD2 T cell Activator (StemCell Technologies) at 25 μL/mL in RPMI 1640 (Gibco) containing 10% FBS (Gibco), 2 mM L-glutamine (BioShop), 10 mM HEPES (Roche Diagnostics), 1 mM sodium pyruvate (Sigma-Aldrich), 1 mM non-essential amino acids (Gibco), 55 μM β-mercaptoethanol (Gibco), 100 U/mL penicillin (Gibco), 100 μg/mL streptomycin (Gibco), 100 I.U./mL rhL-2 and 10 ng/mL rhIL-7 (PeproTech). 16-24 hours later, cells were transduced with lentivirus at an MOI of 0.5-2. For in vitro experiments, cells were sorted with EasySep Human CD271 Positive Selection Kit (StemCell Technologies) on day 7 of culture and expanded for a total culture period of 14 days prior to use. T cells were cryopreserved in Cryostor CS10 (StemCell Technologies) according to manufacturer’s instructions.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
5

BMDCS Stimulation and DMR Analysis

2025
For DMR experiments, stable hCCR7-HEK293
cells were seeded at a density of 18,000 cells per well into fibronectin-coated
384-well biosensor plates. Murine BMDCs were plated on 100 mm Petri
dishes (nonadhesive plastic; Fisher; cat.nr: 10470613) at a density
of 2 × 106 cells/plate in complete medium (RPMI 1640
supplemented with 10% fetal calf serum, 2 mM l-glutamine,
100 U/mL penicillin, 100 μg/mL streptomycin, 50 μM β-mercaptoethanol;
all purchased from Thermo Fisher Scientific) containing 10% GM-CSF
(supernatant from hybridoma culture). DC differentiation was induced
with 200 ng/mL LPS from E. coli 0127:B8
(Sigma-Aldrich) for 24–48 h at 37 °C and 5% CO2. The supernatant was then collected to harvest the mature nonadherent
BMDCs, cells were washed with assay buffer (HBSS with 20 mM HEPES)
and seeded at a density of 80,000 cells per well on 384-well fibronectin-coated
biosensor plates. Cells were incubated for 60 min at 37 °C in
the EPIC reader (EnSight Multimode Plate Reader, PerkinElmer, MA,
US), followed by a 3 min baseline read. Thereafter, SLW131 (10) and SLW132 (21m) were added using the Selma
semiautomatic liquid handling system (Analytik Jena AG, Jena, DE)
for another 60 min. After a second 3 min baseline read, CCL19 was
added and wavelength shifts over time were recorded for 3600 s. Real-time
DMR recordings were buffer-corrected and are presented as wavelength
shift over time; concentration-effect-curves were derived from the
peak wavelength shifts, concentration-inhibition-relationships are
expressed as percentage of the indicated concentration of CCL19.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols

Top 5 protocols citing «β mercaptoethanol»

1

Primed and Naive Human Stem Cell Culture

Conventional (primed) human iPSC line C1 (Whitehead Institute Center for Human Stem Cell Research, Cambridge, MA) (Hockemeyer et al., 2008 (link)) and human ESC lines WIBR2 and WIBR3 (Whitehead Institute Center for Human Stem Cell Research, Cambridge, MA) (Lengner et al., 2010 (link)) were maintained on mitomycin C inactivated MEF feeder layers and passaged mechanically using a drawn Pasteur pipette or enzymatically by treatment for 20 min with 1 mg/ml Collagenase type IV (GIBCO) followed by sequential sedimentation steps in human ESC medium (hESM) to remove single cells. Primed human ESCs and human iPSCs were cultured in hESM—DMEM/F12 (Invitrogen) supplemented with 15% FBS (Hyclone), 5% KSR (Invitrogen), 1 mM glutamine (Invitrogen), 1% nonessential amino acids (Invitrogen), penicillin-streptomycin (Invitrogen), 0.1 mM β-mercaptoethanol (Sigma), and 4 ng/ml FGF2 (R&D systems). Naive human ESCs/hiPSCs were cultured on mitomycin C-inactivated MEF feeder cells and were passaged every 5–7 days by a brief PBS wash followed by single-cell dissociation using 3–5 min treatment with Accutase (GIBCO) and centrifugation in fibroblast medium (DMEM [Invitrogen] supplemented with 10% FBS [Hyclone], 1 mM glutamine [Invitrogen], 1% nonessential amino acids [Invitrogen], penicillin-streptomycin [Invitrogen], and 0.1 mM β-mercaptoethanol). For conversion of preexisting primed human ESC lines, we seeded 2 × 105 trypsinized single cells on an MEF feeder layer in hESM supplemented with ROCK inhibitor Y-27632 (Stemgent, 10 μM). One or two days later, medium was switched to 5i/L/A-containing naive hESM. Following an initial wave of widespread cell death, dome-shaped naive colonies appeared within 10 days and could be picked or expanded polyclonally using 3–5 min treatment with Accutase (GIBCO) on an MEF feeder layer. Naive human pluripotent cells were derived and maintained in serum-free N2B27-based media supplemented with 5i/L/A. Medium (500 ml) was generated by inclusion of the following: 240 ml DMEM/F12 (Invitrogen; 11320), 240 ml Neurobasal (Invitrogen; 21103), 5 ml N2 supplement (Invitrogen; 17502048), 10 ml B27 supplement (Invitrogen; 17504044), 10 μg recombinant human LIF (made in-house), 1 mM glutamine (Invitrogen), 1% nonessential amino acids (Invitrogen), 0.1 mM β-mercaptoethanol (Sigma), penicillin-streptomycin (Invitrogen), 50 μg/ml BSA (Sigma), and the following small molecules and cytokines: PD0325901 (Stemgent, 1 μM), IM-12 (Enzo, 1 μM), SB590885 (R&D systems, 0.5 μM), WH-4-023 (A Chemtek, 1 μM), Y-27632 (Stemgent, 10 μM), and Activin A (Peprotech, 20 ng/ml). 0.5% KSR (GIBCO) can be included to enhance conversion efficiency. FGF2 (R&D systems, 8 ng/ml) enhanced the generation of OCT4-ΔPE-GFP+ cells from the primed state, but it was dispensable for maintenance of naive human ESCs. Additional chemicals described in this work include: CHIR99021 (Stemgent, 0.3–3 μM as indicated), SP600125 (R&D systems, 10 μM), PD173074 (Stemgent, 0.1 μM), SB431542 (Tocris, 5 μM), BIRB796 (Axon Medchem, 2 μM), and doxycycline (Sigma-Aldrich, 2 μg/ml). Tissue culture media were filtered using a low protein-binding binding 0.22 μM filter (Corning). Alternative formulations for naive human ESC culture were followed as described elsewhere (Chan et al., 2013; Gafni et al., 2013; Valamehr et al., 2014; Ware et al., 2014 ). All experiments in this paper were performed under physiological oxygen conditions (5% O2, 3% CO2) in the presence of a MEF feeder layer unless stated otherwise.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
2

Culturing Murine Dendritic Cells

The cell lines were derived and kept in culture at 37°C in a humidified incubator with 5% CO2. The complete medium composition was IMDM-glutamax (GIBCO 31980) adjusted with NaHCO3 to 310 mOsm if required and supplemented with 8–10% heat inactivated FCS (tested for endotoxin toxicity toward DC cultures), 10 mM Hepes (GIBCO 15630), 50 μM β-mercaptoethanol (GIBCO 31350), and 50 U/mL of penicillin and 50 μg/mL streptomycin (GIBCO 15070). The medium was not supplemented with additional growth factors. MutuDC line cells were harvested by incubation in non-enzymatic, 5 mM EDTA-based cell dissociation buffer (5 mM EDTA in 20 mM Hepes-PBS).
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
3

Maintenance and Manipulation of Mouse ESCs

Mouse feeder free E14Tg2A ES cells were maintained on gelatin-coated dishes in Glasgow Minimum Essential medium (GMEM; GIBCO), supplemented with 15% heat-inactivated fetal bovine serum, 55 µM β-mercaptoethanol (GIBCO), 2 mM L-glutamine, 0.1 mM MEM nonessential amino acid, 5,000 units/ml penicillin/streptomycin and 1,000 units/ml of ESGRO (Chemicon) under feeder-free conditions. J1 ES cells and Dnmt1−/−Dnmt3a−/−Dnmt3b−/− (DNMT TKO) ES cells were maintained on gelatin-coated dishes with mitotically inactivated mouse embryonic fibroblasts. Alkaline phosphatase staining was performed with Alkaline Phosphatase Detection Kit (Chemicon). For growth curve analysis, control and knockdown ESCs were plated at 1×103 cells/cm2 and counted for 6 consecutive days. For self-renewal analysis cells were plated on 96 well plates at single cell density and the number of colonies on each plate was counted 6 days after plating.
To knockdown Tet proteins, lentiviral transduction was performed in mouse ES cells as described previously 19 (link). Short-hairpin RNA (shRNA) sequences (Supplementary Table 2) were cloned into pTY vetctor under the U6 promoter. To rescue Tet1 knockdown with Nanog, cDNA of Nanog was placed downstream of puromycin-resistant gene and foot-and-mouth disease virus 2A segment (Fig. S12), which enables multicistronic expression of transgenes in ES cells using a single promoter 21 (link).
Total RNA from mouse tissues was isolated using Trizol reagent (Invitrogen) and total RNA from cultured cells was isolated using RNeasy Mini Kit (Qiagen), and cDNA was generated with Improm-IITM Reverse Transcription System (Promega). Real-time quantitative PCR reactions were performed on an ABI PRISM 7700 Sequence Detection System (Applied Biosystems) using SYBR Green reagent (Invitrogen). cDNA levels of target genes were analyzed using comparative Ct methods, where Ct is the cycle threshold number and normalized to GAPDH. RT-qPCR primers are listed in Supplementary table 3.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
4

Derivation and Characterization of iPSCs

Skin biopsy samples were obtained from four individuals in a previously characterized American family, pedigree H12 (Fig. 1a). C1 fibroblasts were from ATCC (CRL-2097). All studies followed institutional IRB, ISCRO and animal protocols approved by Johns Hopkins University School of Medicine. Informed consents were obtained from individuals from pedigree H. Mouse embryonic fibroblasts (MEFs) were derived from E13.5 CF-1 mouse embryos as previously described16 (link). Fibroblasts were cultured in Dulbecco’s modified Eagle’s medium (DMEM, Mediatech) supplemented with 10% fetal bovine serum (FBS, HyClone) and 2 mM L-glutamine (Invitrogen).
iPS cells were generated with the EBV-based vectors as previously described16 (link). Briefly, plasmids pEP4 EO2S ET2K (Addgene Plasmid 20927), pEP4 EO2S EN2L (Addgene Plasmid 20922), and pEP4 EO2S EM2K (Addgene Plasmid 20923) were transfected intohuman fibroblasts by Amaxa Nucleofector (Lonza; program U-023) at a concentration of 2 μg per 100 μl electroporation solution per 2 × 106 cells. Colonies of iPS cells were manually picked after 3–6 weeks for further expansion and characterization. Lack of vector integration was confirmed by qRT–PCR analysis as previously described16 (link). Two lines from each individual that passed stringent criteria were used for the current study (Supplementary Table 1a). iPS cells (passage ≤ 35) were cultured on irradiated MEFs in human iPS cell medium consisting of D-MEM/F12 (Invitrogen), 20% Knockout Serum Replacement (KSR, Invitrogen), 2 mM L-glutamine (Invitrogen), 100 μM MEM NEAA (Invitrogen), 100 μM β-mercaptoethanol (Invitrogen), and 10 ng ml−1 human basic FGF (bFGF, PeproTech) as described16 (link),31 (link). For feeder-free culture of iPS cells, cells were cultured on Matrigel (BD Biosciences) with mTeSR1 media (Stem Cell Technologies). Media were changed daily and iPS cell lines were passaged by collagenase (Invitrogen, 1 mg ml−1 in D-MEM/F12 for 30 min at 37°C).
Karyotyping analysis by standard G-banding technique was carried out by the Cytogenetics Core Facility at the Johns Hopkins Hospital or Cell Line Genetics. Results were interpreted by clinical laboratory specialists of the Cytogenetics Core or Cell Line Genetics. Genotyping analysis was performed as described previously16 (link). Genomic DNA of fibroblasts and derived iPS cells was extracted by DNeasy Blood & Tissue Kit (Qiagen) following the manufacturer’s recommended protocol. Apair of specific primers was used to amplify the region around the 4-bp deletion (Supplementary Table 1c). PCR products were cloned by TA cloning and sequenced. Bisulphite genomic sequencing was carried out with the EZ DNA Methylation-Direct Kit (Zymo Research) as previously described32 (link). After bisulphite conversion of genomic DNA from iPS cells, primers specific to human OCT4 and NANOG promoters (Supplementary Table 1c) were used to amplify genomic DNA sequences with Platinum Taq DNA Polymerase High Fidelity (Invitrogen) for sequencing.
To assess the in vivo pluripotency of iPS cell lines, teratoma formation assays were performed16 (link). iPS cells were injected subcutaneously into the dorsal flank of SCID mice. Animals were monitored and teratomas were dissected at 8 to 10 weeks post-injection. Tissues were fixed in 10% neutralized formalin solution (Sigma). Embedding, sectioning and haematoxylin and eosin staining were carried out by the Pathology Core Facility at the Johns Hopkins University Hospital.
TALEN designs and constructions were based on a Golden Gate Assembly protocol with modifications to the vector backbone33 (link). Donor DNA vectors with a loxP-flanked PGK-hygromycin cassette were cloned between 5′ and 3′ homology arms (Fig. 3a), which were amplified from genomic DNA of a healthy subject and a patient with the DISC14-bp mutation. For targeting, TALENs (4 μg DNA of each plasmid) and linearized donor vectors (10 μg DNA) were electroporated into individual iPS cells (1 × 106 to 2 × 106 cells pretreated with 5 μM ROCK inhibitor, Y-27632, Cellagentech) using Nucleofector 2b (Lonza; program A-023). Transfected cells were transferred onto a 6-well dish pre-plated with inactivated MEFs and supplemented with Y-27632 in standard iPS cell medium. Positive colonies were selected by 10mg ml−1 hygromycin B (Invitrogen) after 5 days of culture or until small colonies appeared. Resistant colonies were sub-cloned and expanded in 48-well plates. Over 200 clonal lines were screened. The loxP-flanked PGK-hygromycin cassette was removed by electroporation of a Cre recombinase expression vector (4 μg DNA). Specific integration, correct genetic editing and efficient removal of PGK-hygromycin cassette at each stage were verified by Sanger sequencing.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols
5

hESC Maintenance and Propagation

The human ESC line HUES6 was used in this study and was purchased from the hESC Collection (Harvard University). The derivation of this line has been reported previously [32] (link). HUES6 and iPSCs were cultured on a layer of mitotically inactivated (with mitomycin C (Tocris)) mouse embryo fibroblasts (MEFs) in hESC medium. The mouse embryonic fibroblasts (MEFs) used in this study were derived in the laboratory of Prof. Dr. Hans Schöler and have been reported previously [31] (link). hESC medium consisted of Knockout DMEM (Invitrogen) with 20% Knockout Serum Replacement (Invitrogen), 1 mM beta-mercaptoethanol (Invitrogen), 1% nonessential amino acids (NEAA, Invitrogen), 1% penicillin/streptomycin/glutamine (PAA), freshly supplemented with 5 ng/mL FGF2 (Peprotech). Pluripotent stem cells were split 1∶5 to 1∶8 every 5–7 days. Colonies were mechanically disaggregated with 1 mg/mL collagenase IV (Invitrogen). 10 µM ROCK Inhibitor (Ascent Scientific) was added for 24 hours after splitting.
+ Open protocol
+ Expand Check if the same lab product or an alternative is used in the 5 most similar protocols

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!

🧪 Need help with an experiment or choosing lab equipment?
I search the PubCompare platform for you—tapping into 40+ million protocols to bring you relevant answers from scientific literature and vendor data.
1. Find protocols
2. Find best products for an experiment
3. Validate product use from papers
4. Check Product Compatibility
5. Ask a technical question
Want to copy this response? Upgrade to Premium to unlock copy/paste and export options.