The largest database of trusted experimental protocols

14 protocols using s trityl l cysteine stlc

1

Cell Cycle Synchronization Techniques

Check if the same lab product or an alternative is used in the 5 most similar protocols
To synchronize in G1, cells were placed in serum-free media for 24 h. To arrest cells in S phase, cells were incubated for 16 h with 2 mM thymidine, released by washing three times with PBS and placed into fresh medium for 6 h, followed by a second incubation with 2 mM thymidine for 16 h. Cells in the G2 phase of the cell cycle were obtained 4 h after release from double thymidine block. Where nocodazole was used to arrest cells in mitosis, treatment was with 50 μg ml−1 nocodazole (Sigma) for 16 h. Where Eg5 inhibitor was used to arrest cells in mitosis, cells were incubated with S-trityl-L-cysteine (STLC, Sigma, 5 μM) or monastrol (Tocris, 100 μM) for 16 h unless otherwise stated. Release from monastrol was achieved by washing cells three times in PBS and replacing media. Cdk1 inhibitor (RO-3306, Alexis) was used at 10 μM. Treatment with the Pak inhibitor FRAX597 (a kind gift from Jonathan Chernoff, Fox Chase Cancer Center, Philadelphia, USA) was at 2 μM for 6 h, and with NSC 23766 for 24 h at 100 μM. For dox-inducible expression of shRNA or Pak2-CFP or Tiam1-HA constructs, cells were treated with doxycyline (1 μg μl−1) for 3–4 days (shRNA), or 24 h (Pak2-CFP, Tiam1-HA constructs).
+ Open protocol
+ Expand
2

Monopolar Spindle Formation and Visualization

Check if the same lab product or an alternative is used in the 5 most similar protocols
To make monopolar spindles, 5 µM S-trityl-l-cysteine (STLC; Sigma-Aldrich) was added 15 min before imaging (10 mM stock). To rigor CenpE to microtubules, 90 nM GSK-923295 (MedChem Express) was added 15 min before imaging (30 µM stock; Magidson et al., 2015 (link)). To visualize tubulin as a third color, 100 nM SiR-tubulin dye (Cytoskeleton, Inc.) was added 1 h before imaging (1 mM stock) along with 10 µM verapamil (10 mM stock; Cytoskeleton, Inc.) to prevent dye efflux.
+ Open protocol
+ Expand
3

Cell Synchronization and Cytoskeletal Disruption

Check if the same lab product or an alternative is used in the 5 most similar protocols
HeLa Kyoto cells were cultured in DMEM, and Jurkat cells (immortalised human T lymphocytes) as well as MAVER1 CML B cells in RPMI 1640 (Gibco) at 37°C and 5% CO2. All media were supplemented with 10% foetal bovine serum and penicillin/streptomycin (Gibco). Cells were synchronised in prometaphase using S‐trityl‐L‐cysteine (STLC; Sigma) at 5 μM treatment for 18 h (Skoufias et al, 2006). Forced mitotic exit was performed by the addition of 20 μM RO‐3306 (Enzo Life Sciences), an inhibitor of Cdk1/cyclin B1 and Cdk1/cyclin A to STLC‐treated cells. For the Arp2/3 complex inhibition experiments, we used 0.2 mM CK666 (Sigma‐Aldrich). Control experiments were performed using DMSO. For CK666 treatment (Fig 3), cells were plated with 2 mM thymidine for 22 h, were released from thymidine block for 9 h, following which they were treated with DMSO or 0.2 mM CK666 for 15 min. They were then processed for staining. Actin network disruption was performed by adding 10 μg/ml cytochalasin D (Sigma‐Aldrich). Microtubule depolymerisation was performed by adding 1 μM nocodazole (Sigma‐Aldrich) for 1 h at 37°C and 5% CO2 and for 30 min at 4°C.
+ Open protocol
+ Expand
4

Modulating Spindle Dynamics with Eg5 Inhibitors

Check if the same lab product or an alternative is used in the 5 most similar protocols
To increase microtubule crosslinking by rigor binding Eg5, we treated with 20 μM FCPT (gift of T. Mitchison, Harvard Medical School, Boston, MA) for 15–30 min. Under these conditions, flux was slowed to 0.1 +/− 0.1 μm/min (s.e.m., n=5) vs. 0.8 +/− 0.1 μm/min in control cells (s.e.m.; n=10), but chromosomes still oscillated, indicating that spindle crosslinking was increased but that spindles remained dynamic (allowing us to normalize for centromere stretch over an oscillation cycle as described).
To inhibit Eg5 motor activity, we treated with 5 μM S-trityl-L-cysteine (STLC, Sigma) in MEM for 30 min, and chose cells that remained bipolar, indicating that they entered mitosis before drug treatment began [32 (link)]. Under these conditions, we observed many monopolar spindles, which indicated that the drug treatment effectively inhibited Eg5.
For speckle experiments, we used SiR-tubulin (alongside speckle strategy described above) to visualize microtubules [47 (link)]. We treated with 10 μM verapamil and 100 nM SiR-tubulin (Spirochrome) for 2–6 h to allow time for incorporation of SiR-tubulin into spindles. Under these conditions, there was no detected defect in spindle appearance or behavior.
+ Open protocol
+ Expand
5

Synchronizing and Arresting Cell Lines

Check if the same lab product or an alternative is used in the 5 most similar protocols
All cell lines were maintained at 37°C with 5% CO2 (see Supplemental Experimental Procedures for details of origin and media). hTERT-RPE-1 H2B-RFP stable cell lines were generated after transfection with lentiviral construct H2B-RFP (26001; Addgene). RNAi was achieved by transfection of cells for 48 hr with 30 nM small interfering RNA (siControl [D-001210-02] and siWAPL SMART pool [M-026287-01]; Dharmacon) using Lipofectamine RNAiMAX (Invitrogen) and Optimem (Gibco). Drug concentrations used were 10 μM MG132, 100 ng/mL nocodazole, 10 μM S-trityl-L-cysteine (STLC), 100 μM monastrol, and 250 nM reversine (all from Sigma-Aldrich). Release from mitotic arrest was achieved by washing drug out of cells with prewarmed media three to five times, then leaving in incubator for 1 hr (nocodazole), 1.5 hr (STLC and monastrol), or 2.5 hr (MG132).
+ Open protocol
+ Expand
6

Inhibitors of DNA Damage Repair

Check if the same lab product or an alternative is used in the 5 most similar protocols
DNA damage repair inhibitors were dissolved in DMSO and used at the following concentrations: 15 μM ABT-888 (PARP inhibitor, Enzo), 10 μM Olaparib (PARP inhibitor, ApexBio), and 10 μM NU7026 (DNA-PKcs inhibitor, Abcam). Methotrexate (454126, Calbiochem) was dissolved in DMSO and used at indicated concentrations. Doxorubicin (Sigma) was dissolved in water and cells were treated at a final concentration of 0.5 μM for 1 hour. Hydroxyurea (Sigma) was dissolved in PBS and used at a final concentration of 100 μM. S-Trityl-L-cysteine (STLC, Sigma) was used at a final concentration of 10μM, and zeocin (Gibco) was used at a final concentration of 100μg/ml.
+ Open protocol
+ Expand
7

Inducing and Correcting Kinetochore-Microtubule Defects

Check if the same lab product or an alternative is used in the 5 most similar protocols
To analyze the response to defective kinetochore-microtubule attachments, an established assay for error correction in human cells was modified for extracts. The addition of an Eg5 inhibitor, S-Trityl-L-Cysteine (STLC, Sigma Aldrich), results in monopolar spindles with kinetochores in a tension-less, mono-oriented (syntelic) configuration (Houghtaling et al., 2009 (link)). Subsequent removal of Eg5 inhibition allows spindles to bipolarize and chromosomes to align at the metaphase plate through removal of improper kinetochore-microtubules by Aurora B kinase (Lampson et al., 2004 (link)). A range of concentrations of STLC was tested in extracts, and the ratio of monopolar and bipolar spindles generated were recorded in order to determine the IC50 (0.2 µM). In the modified assay for extracts, STLC was added to a final concentration of 0.5 µM at the beginning of metaphase to induce the majority of spindle structures into a monopolar state. After monopolar spindles were generated in metaphase (around 40 minutes), experiments demonstrating error correction could be performed by diluting the STLC-treated extract with ten-fold untreated CSF extract and letting the monopolar spindles re-bipolarize. At the final concentration of 0.05 µM, no monopolar spindles were observed. Samples for Western blot and immunofluorescence were taken 30 minutes after STLC was diluted.
+ Open protocol
+ Expand
8

Targeted protein degradation in mitotic cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
HaloPROTAC-E, SGK3-PROTAC1 and VH298 were synthesised as previously described (Tovell et al., 2019a (link); Tovell et al., 2019b (link); Frost et al., 2016 (link)) and used at indicated concentrations and times. The following chemicals were added to cell media at indicated concentrations and times: MLN4924 (Active Biochem), MG132 (Abcam), Bafilomycin-A1 (Enzo Life Sciences), MRT68921 (MRC PPU Reagents and Services), SBI-0206965 (Sigma-Aldrich). Cells were synchronised in mitosis using the Eg5 inhibitor S-trityl-L-cysteine (STLC, Sigma-Aldrich, 5 μM, 16 hr) (Fulcher et al., 2019 (link)). Following incubation, mitotic cells were isolated through shake-off. For amino acid starvation, cells were washed twice in Earle’s balanced salt solution (EBSS, Gibco) and incubated in EBSS for 2 hr.
+ Open protocol
+ Expand
9

Cell Cycle Synchronization Techniques

Check if the same lab product or an alternative is used in the 5 most similar protocols
To synchronise in G1, cells were placed in serum-free media for 24 hours. To arrest cells in S-phase, cells were incubated for 16 hours with 2 mM thymidine, released by washing three times with PBS and placed into fresh medium for 6 hours, followed by a second incubation with 2 mM thymidine for 16 hours. Cells in the G2 phase of the cell cycle were obtained 4 hours after release from double thymidine block. Where nocodazole was used to arrest cells in mitosis, treatment was with 50 μg/ml nocodazole (Sigma) for 16 hours. Where Eg5 inhibitor was used to arrest cells in mitosis, cells were incubated with S-trityl-L-cysteine (STLC, Sigma, 5 μM) or monastrol (Tocris, 100 μM) for 16 hours unless otherwise stated. Release from monastrol was achieved by washing cells 3 times in PBS and replacing media. Cdk1 inhibitor (RO-3306, Alexis) was used at 10 μM. Treatment with the Pak inhibitor FRAX597 (a kind gift from Jonathan Chernoff, Fox Chase Cancer Center, Philadelphia, U.S.) was at 2 μM for 6 hours, and with NSC 23766 for 24 hours at 100 μM. For dox-inducible expression of shRNA or Pak2-CFP or Tiam1-HA constructs, cells were treated with doxycyline (1 μg/μl) for 3-4 days (shRNA), or 24 hours (Pak2-CFP, Tiam1-HA constructs).
+ Open protocol
+ Expand
10

Cell Culture and Drug Treatment Protocol

Check if the same lab product or an alternative is used in the 5 most similar protocols
HeLa, hTERT-RPE1, U2OS, A549, DLD-1, and NIH/3T3 cell lines were cultured in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and streptomycin, and 2 mM L-glutamine at 37°C with 5% CO2. The HEC-6 cell line was cultured in DMEM supplemented with 15% FBS and 2 mM L-glutamine. Doxycycline-inducible cell lines were cultured in medium containing FBS certified tetracycline-free. spCas9 expression in inducible CRISPR/Cas9 cell lines was induced with 1 μg/ml doxycycline hyclate (Sigma) at 24 hr intervals for 2 days. All other doxycycline-inducible constructs were induced with 10 ng/ml doxycycline hyclate, unless indicated in figure legend. Other drugs used on human cells were Nocodazole (Sigma, 330 nM), S-trityl-L-cysteine (STLC; Sigma, 10 μM unless otherwise indicated), Paclitaxel (Taxol; Invitrogen, 1 μM), GSK923295 (CENP-E inhibitor; Selleck Chemicals, 100 nM), proTAME (APC/Ci; R&D Systems, 12 μM), AZ-3146 (Mps1i; Selleck Chemicals, 4 μM), cycloheximide (CHX; Sigma, 50 μg/ml), MG-132 (MG132; Enzo Life Sciences, 10 μM) unless concentration indicated otherwise in figure legend. Cells were enriched in mitosis with treatment with 330 nM Nocodazole for 16-17 hrs or if indicated, 10 μM STLC for 18 hrs. The antibodies used in this study are described in the relevant methods and in Supplementary Table 1.
+ 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!