Nonidet is a nonionic detergent commonly used in biochemical applications, such as cell lysis, protein extraction, and enzyme assays.
It is a polyoxyethylene alkyl phenol that effectively solubilizes membrane proteins and disrupts protein-protein interactions while maintaining protein structure and function.
PubCompare.ai's AI-powered platform helps researchers effortlessly identify the best Nonidet protocols and products from literature, preprints, and patents, leveraging comparison tools to enhance reproducibility and accuracy.
Take the guesswork out of your Nonidet-based research with PubCompare.ai's powerful suite of tools.
This method modifies a previously published protocol based on cultured cells [1] (link), expanding it to allow for the collection of subcellular fractions from fresh tissue. In addition, the buffer compositions (Table 1) were optimized to minimize nuclear protein loss via the addition of 1 M hexylene glycol, which helps to further stabilize the membranes, especially that of the nucleus, and has been previously shown to yield highly enriched nuclear fractions [2] (link). HEPES is an organic buffer that stabilizes the pH of the solution while NaCl maintains the ionic strength [2] (link). The basis of this method (Fig. 1) is the sequential lysis of cell membranes by increasing the detergent strength of lysis buffers to obtain proteins from each fraction. Lysis buffer A is meant to release cytosolic proteins and its main component is digitonin. Digitonin is a steroidal saponin that permeabilizes the plasma membrane by binding with cholesterol and other β-hydroxysterols, thereby leading to the formation of pores in the membrane and its subsequent disruption. The advantage of lysing cells with digitonin is that it is unable to disrupt the membranes of cellular organelles, as the cholesterol composition of these membranes is lower [3] (link). Lysis buffer B releases the proteins from all membrane bound organelles except the nucleus. The main component of this buffer is igepal, which is a non-ionic, non-denaturing detergent chemically equivalent to Nonidet P-40 [4] (link). It is used at a low concentration to allow permeabilization of the endoplasmic reticulum, Golgi and mitochondria membranes, while keeping the nuclear membrane intact. Lysis buffer C is meant to permeabilize the nuclear membrane and release the nuclear proteins. Among its components, lysis buffer C contains sodium deoxycholate, a mild, non-ionic and non-denaturing biological detergent (a constituent of bile). Sodium dodecyl sulphate is an anionic detergent that is extremely effective in membrane solubilization [5] . The combination of sodium deoxycholate and sodium dodecyl sulphate thus creates an effective nuclear lysis buffer. Benzonase is added prior to the isolation of nuclear fractions and digests DNA and RNA, facilitating the complete release of all nuclear proteins [1] (link). Equipment required for this method:
End-over-end tube rotator
Handheld homogenizer (rotor/stator type)
Qiagen QIAshredder columns (Qiagen, 79656)
Microcentrifuge
Fractionation protocol for isolated tissue (Fig. 1):
Mince fresh, unfrozen tissue into 2–4 mm pieces, wash with 1 mL of ice cold phosphate buffered saline solution (10 mM Na2HPO4, 2 mM KH2PO4, 137 mM NaCl, 2.7 mM KCl, pH 7.4).
Add 40–60 mg of tissue into a 1.5 mL microtube.
Add 500 μL of ice cold lysis buffer A supplemented with 5 μL protease inhibitor cocktail.
Disrupt the tissue for 5 s using a hand held tissue homogenizer (rotor/stator type).
Transfer the tissue suspension to QIAshredder homogenizer (Qiagen, 79656) and centrifuge at 500 × g for 10 min at 4 °C to filter the homogenate.
Discard the top filter containing trapped tissue particles, and resuspend the pellet in the filtrate by gently pipetting up and down. Transfer into a new tube.
Add 500 μL of ice cold lysis buffer A (1.5 mL if working with brain tissue), and supplement with 5 μL of protease inhibitor cocktail.
Incubate the homogenate on an end-over-end rotator for 10 min at 4 °C
Centrifuge at 4000 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the cytosolic proteins.
Using a 1 mL pipette and tip, resuspend the pellet by gently pipetting up and down in 1 mL of ice cold lysis buffer B supplemented with 10 μL of protease inhibitor cocktail. Incubate for 30 min on an end-over-end rotator at 4 °C.
Centrifuge at 6000 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the proteins from membrane-bound organelles (mitochondria, endoplasmic reticulum, Golgi, etc.) except those from the nucleus.
Add 500 units of benzonase (Sigma, E1014) to 20 μL of water and combine it with the pellet from step 12.
Resuspend the pellet by gently flicking the bottom of the tube and incubate at room temperature for 15 min.
Add 500 μL of ice cold lysis buffer C with 5 μL of protease inhibitor cocktail to the benzonase-digested pellet and incubate on an end-over-end rotator for 10 min at 4 °C.
Pellet the insoluble material by centrifuging at 6800 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the nuclear proteins.
Pellet contains nuclear proteins and protein complexes that resist extraction and typically include active RNA polymerases and regulatory proteins. These can be solubilized with lysis buffer C supplemented with 8 M urea for analysis, or discarded.
Fractionation protocol for cultured cells (Fig. 1):The following protocol is optimized for cultured cells grown on a 100 mm diameter dish (55 cm2 surface area).
Remove culture medium and wash the cells with room temperature phosphate buffered saline solution.
Trypsinize the cells by adding 800 μL of 0.25% Trypsin: 0.9 mM EDTA: phenol red solution (Gibco Life Technologies, 25200) and incubating the cells at 37 °C for 2 min or until cells are detached.
Add 5 mL of culture medium containing 10% fetal bovine serum to inhibit trypsin activity, and collect the cells.
Centrifuge at 500 × g for 10 min at 4 °C to pellet the cells.
Using a 1 mL pipette and tip, discard the supernatant and resuspend the pellet by pipetting up and down in 500 μL of ice cold PBS.
Centrifuge for at 500 × g for 10 min at 4 °C to pellet the cells.
Discard the supernatant and add 400 μL of ice cold lysis buffer A supplemented with 4 μL of protease inhibitor cocktail.
Incubate on end-over-end rotator for 10 min at 4 °C.
Centrifuge at 2000 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the cytosolic proteins.
Add 400 μL of ice cold lysis buffer B supplemented with 4 μL of protease inhibitor cocktail and resuspend the pellet by vortexing.
Incubate on ice (or at 4 °C) for 30 min.
Centrifuge at 7000 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the proteins from membrane-bound organelles (mitochondria, endoplasmic reticulum, Golgi, etc.) except those from the nucleus.
Add 400 μL of ice cold lysis buffer C containing 7 μL of Benzonase and 4 μL of protease inhibitor cocktail.
Incubate on an end-over-end rotator for 30 min at 4 °C to allow complete solubilization of nuclei and digestion of genomic DNA.
Centrifuge at 7800 × g for 10 min at 4 °C.
Collect the supernatant. This fraction contains the nuclear proteins.
Pellet contains nuclear proteins and protein complexes that resist extraction and typically include active RNA polymerases and regulatory proteins. These can be solubilized with lysis buffer C supplemented with 8 M urea for analysis, or discarded.
Using the technique developed for fractionation of isolated tissue, freshly isolated rat hearts perfused free of blood with Krebs-Henseleit solution at 37 °C using the Langendorff technique [6] (link), were fractionated into cytosolic (C), membrane bound organelle (M) and nuclear (N) fractions. The purity of the fractions was assessed by western blotting against specific markers (Fig. 2). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH, antibody 14C10, Cell Signaling Technology, Beverly, MA, USA) was used as the major cytosolic marker [7] (link). The sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2, antibody ab137020, Abcam, Cambridge, UK) and voltage-dependent anion channel (VDAC, antibody ab15895, Abcam) were used as membrane markers since they are associated with the sarco/endoplasmic reticulum and mitochondria, respectively [8] (link). Lamin A/C (antibody 2032S, Cell Signaling Technology, Beverly, MA, USA) a major structural protein of the nuclear membrane, was used as a nuclear marker [1] (link). GAPDH can be found in the mitochondria and small vesicular structures of the cell when exposed to stressors which cause a dynamic subcellular redistribution of GAPDH [7] (link). Consistent with this, while the bulk of GAPDH is found in the cytosolic fraction, a much smaller amount is present in the membrane fraction. GAPDH is not seen in the nuclear fraction of hearts (Fig. 2A) or human fibrosarcoma HT1080 cells (Fig. 2B), confirming nuclear fraction purity. Both SERCA2 and VDAC were present in the membrane fraction from hearts, but absent from the cytosolic and nuclear fractions (Fig. 2A). Lamin A/C was found exclusively in the nuclear fractions isolated from hearts (Fig. 2A) and primarily in the nuclear fraction isolated from HT1080 cells, with substantially less appearing in the membrane fraction (Fig. 2B). The presence of the nuclear membrane-associated protein lamin A/C mostly in the nuclear fraction indicates that the nuclear membrane is contained in this fraction. These results confirm the purity of the fractions collected from tissue and cells with the protocol presented above.
Baghirova S., Hughes B.G., Hendzel M.J, & Schulz R. (2015). Sequential fractionation and isolation of subcellular proteins from tissue or cultured cells. MethodsX, 2, 440-445.
Yeast Strains and Constructs—The following yeast strains were used: BY4741 (MATa his3Δ1 leu2Δ met15Δ ura3Δ) and NDY257 (BY4741 rtn1::kanMX4 rtn2::kanMX4 yop1::kanMX) (6 (link)). Strains expressing GFP fusions to the chromosomal alleles of YOP1 and RTN1 were obtained from Invitrogen. The plasmid encoding Sec63-GFP (pJK59) has been previously described (12 (link)). To make the plasmid encoding Rtn1-GFP (pCV19), the SEC63 portion of pJK59 was removed by digestion with XbaI and XhoI. The RTN1 gene, including 400 bp upstream of the start site, was PCR-amplified from yeast chromosomal DNA and inserted into the same sites. Mammalian Plasmid Constructs—HA-DP1 was described previously (6 (link)). HA-Rtn3c was cloned by PCR amplifying Rtn3c (NCBI accession number: BC036717) from mouse cDNA with primers containing an N-terminal HA tag and inserted into pcDNA3.1D (Invitrogen). For Rtn4a-GFP, human Rtn4a was PCR-amplified from Rtn4a-Myc (described in a previous study (6 (link))) and ligated into the pAcGFP-N1 backbone (Clontech) using the XhoI and KpnI restriction sites at the 5′ and 3′ ends, respectively. For GFP-Rtn3c, Rtn3c was PCR-amplified from HA-Rtn3c and ligated into the pAcGFP-C1 backbone (Clontech) using the XhoI and EcoRI restriction sites. To clone GFP-Rtn4HD, the region encoding amino acids 961–1192 was PCR-amplified from human Rtn4a-Myc and inserted into pAcGFP-C1 using the XhoI/EcoRI restriction sites. GFP-DP1 was subcloned by PCR-amplifying mouse DP1 from HA-DP1 (described in a previous study (6 (link))) and inserting into pAc-GFP C1 using SacI/BamHI restriction sites. For GFP-Climp63, Climp63 was PCR-amplified from mouse cDNA and cloned into pAcGFP-C1 using the XhoI/EcoRI sites. Climp63Δlum-GFP was cloned by PCR amplifying the region encoding amino acids 1–115 (as described in (13 (link))) from GFP-Climp63 and inserted into pAcGFP-N1 using XhoI/EcoRI restriction sites. LBR-GFP was PCR-amplified from plasmid containing human LBR (14 (link)) and cloned into pAcGFP-N1 using the XhoI/BamHI restriction sites. For GFP-Sec61β, human Sec61β was PCR-amplified from the pcDNA3.1/GFP-Sec61β construct described previously (6 (link)), and inserted into pAcGFP-C1 using the BglII/EcoRI restriction sites. RFP-Sec61β was subcloned from GFP-Sec61β using the same restriction sites as above and inserted into an mRFP1 vector (pEGFP-C1 vector backbone where pEGFP has been replaced with mRFP1). Microscopy of Yeast—Yeast strains were grown in synthetic complete medium (0.67% yeast nitrogen base and 2% glucose) and imaged live at room temperature using an Olympus BX61 microscope, UPlanApo 100×/1.35 lens, QImaging Retiga EX camera, and IPlabs version 3.6.4 software. Screen for Mutations in Yeast RTN1 That Affect Localization—Error-prone PCR on RTN1 was performed using the GeneMorphII Random Mutagenesis Kit (Stratagene). The product of this reaction and pJK59 cut with XbaI and XhoI were used to transform wild-type yeast. Transformants were visually screened for those that showed perinuclear GFP localization. Tissue Culture, Indirect Immunofluorescence, and Confocal Microscopy of COS-7 Cells—Cells were grown at 37 °C with 5% CO2 in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum and subcultured every 2–3 days. Transfection of DNA into cells was performed using Lipofectamine 2000 (Invitrogen). After 5 h of transfection, cells were split onto acid-washed No. 1 coverslips and allowed to spread for an additional 24–36 h before being processed for indirect immunofluorescence. For immunofluorescence, transfected cells were fixed in PBS containing 4% paraformaldehyde (Electron Microscopy Sciences) for 15 min, washed twice, and permeabilized in 0.1% Triton X-100 (Pierce) in PBS for 5–15 min. Cells were washed twice again and then probed with primary antibodies for 45 min in PBS containing 1% calf serum, at the following concentrations: rat anti-HA antibody (Roche Applied Science) at 1:200 dilution; mouse anti-αtubulin (Sigma) at 1:500 dilution; and rabbit anti-calreticulin antibody (Abcam) at 1:500 dilution. Cells were washed three times in PBS, and then incubated with various fluorophore-conjugated secondary antibodies for an additional 45 min (Alexafluor 488 or 555 anti-mouse at 1:250 dilution, Alexafluor 647 anti-rabbit 1:500 dilution, and Alexafluor 488 anti-rat 1:200 dilution (all from Invitrogen)). Cells were then washed and mounted onto slides using Fluoromount-G mounting medium (Southern Biotech). All imaging for indirect immunofluorescence was captured using a Yokogawa spinning disk confocal on a Nikon TE2000U inverted microscope with a 100× Plan Apo numerical aperture 1.4 objective lens, and acquired with a Hamamatsu ORCA ER cooled charge-coupled device camera using MetaMorph 7.0 software. For image presentation, brightness and contrast were adjusted across the entire image using Adobe Photoshop 7.0, and images were converted from 12 to 8 bits. Transmission Electron Microscopy—COS-7 cells expressing GFP-Rtn4HD were sorted in a MoFlo cell sorter (Cytomation). The resulting cell pellet was fixed for 1 h in a mixture of 2.5% glutaraldehyde and 2% paraformaldehyde in 0.1 m sodium cacodylate buffer (pH 7.4), washed in 0.1 m cacodylate buffer, and postfixed with a mixture of 1% OsO4 and 1.5% KFeCN6 for 30 min. The pellet was then washed in water and stained in 1% aqueous uranyl acetate for 30 min followed by dehydration in grades of alcohol (50%, 70%, and 95%, 2 × 100%). Next, the pellet was infiltrated in a 1:1 mixture of propylene oxide and TAAB Peon (Maria Canada Inc.) for 2 h, placed in pure TAAB Epon in a silicon-embedding mold, and polymerized at 65 °C for 48 h. Ultrathin sections (∼60–80 nm) were cut on a Reichert Ultracut-S microtome, placed onto copper grids, and stained with 0.2% lead citrate. Specimens were examined on a Tecnai G Spirit BioTWIN transmission electron microscope, and images were acquired with a 2k AMT charge-coupled device camera. Fluorescence Recovery after Photobleaching—Transfected COS-7 cells were imaged in phenol red-free HyQ DME (HyClone) supplemented with 25 mm Hepes, pH 7.4, and 1% fetal bovine serum. FRAP experiments were conducted on a Zeiss LSM 510 NLO laser scanning inverted microscope using a Plan-Neofluor 100×/1.3 oil objective with argon laser line 488 nm (optical slices <1.2 mm for COS-7 and 4.2 μm for yeast). Mammalian cell experiments were done at 37 °C using an objective heater (Bioptechs) and an enclosed stage incubator (Zeiss). LSM 510 software version 3.2 was used for image acquisition and analysis. Magnification, laser power, and detector gains were identical across samples. For all mammalian experiments, COS-7 cells were treated with 0.5 μm nocodazole, and all data were collected during the first 5–30 min of nocodazole addition. For photobleaching all constructs, except for LBR-GFP, the tubular ER was magnified using the 3× zoom function so that individual tubules could be seen clearly. For LBR-GFP, the microscope was focused onto the bottom of the nuclear envelope. Images taken for 5-s prebleaching, whereupon a region of interest of 65 × 65 pixels was photobleached at 100% laser power. After the photobleaching, images were taken at 1-s intervals for 75–300 s. Yeast cells were treated similarly except that the region of interest was 17 × 17 pixels, and images were taken every 2–4 s at room temperature. Raw data were quantitated using Zeiss LSM510Meta software. For analysis, the fluorescence intensity of three regions of interest was measured: the photobleached region (PR), a region outside of the cell to check for overall background fluorescence (BR), and a region within the cell that was not photobleached to check for overall photobleaching and fluorescence variation (CR), for the entire course of the experiment. Microsoft Excel was used to normalize the relative fluorescence intensity, I, for each individual FRAP experiment using Equation 1. For data presentation, the mean averages of the normalized data for each set of FRAP experiments were plotted using GraphPad Prism 5.0, and fluorescence recovery curves were shown for the first 80–140 s of each experiment. Estimated half-times of recovery and mobile fraction values were calculated using the standard Michaelis-Menten equation. Sucrose Gradient Centrifugation—For yeast sucrose gradient analysis, crude membranes were isolated from yeast strains expressing GFP-fused proteins at endogenous levels as follows: 200 ml of culture were grown to OD ∼1, pelleted and then resuspended in TKMG lysis buffer (50 mm Tris, pH 7.0, 150 mm KCl, 2 mm MgCl2, 10% glycerol, 1 mm EDTA, 1 mm PMSF, 1 mm 4-(2-aminoethyl)benzenesulfonylfluoride hydrochloride), flash frozen in liquid nitrogen, and ground using a mortar and pestle. Cell debris was separated from the lysate by low speed centrifugation for 5 min at ∼2,000 × g. Membranes were then pelleted by ultracentrifugation for 15 min at 100,000 × g and solubilized in 200 μl of TKMG buffer containing 1% digitonin. Solubilized lysate was centrifuged for 10 min at 12,000 × g to separate out any remaining cell debris. 100-μl of lysate were run on 5–30% w/v sucrose gradients for 4 h at 166,000 × g at 25 °C on a Beckman TLS55 rotor. Twenty gradient fractions were collected from top to bottom and analyzed by SDS-PAGE and immunoblotting with anti-GFP antibody (Roche Applied Science). 50 mg of apoferritin, catalase, and aldolase was used as molecular weight standards. Xenopus washed membrane fractions were prepared in MWB (50 mm Hepes, pH 7.5, 2.5 mm MgCl2, 250 mm sucrose, and 150 mm potassium acetate) as previously described (6 (link)), incubated for 60 min at 25 °C in MWB containing 200 mm KCl and 0.5 mm GTP, and then solubilized for 30 min at 25 °C with either 2% Nonidet P-40 or 1.25% digitonin. Samples were pelleted for 15 min at 12,000 rpm, and the soluble fraction was loaded onto a 10–30% w/v sucrose gradient made with MWB containing 200 mm KCl, 0.1 mm GTP, and either 0.1% Nonidet P-40 or 0.1% digitonin, respectively. The sucrose gradient was centrifuged for 3 h, 45 min at 55,000 rpm. Sixteen gradient fractions were collected and analyzed by SDS-PAGE and immunoblotted with antibody against Xenopus Rtn4 (described in a previous study (6 (link))). For mammalian sucrose gradient analysis, COS-7 cells transiently transfected with HA-DP1 or GFP-Sec61β were harvested by scraping and then lysed and solubilized in HKME buffer (25 mm Hepes, pH 7.8, 150 mm potassium acetate, 2.5 mm magnesium acetate, 1 mm EDTA, and 2 mm PMSF) containing 1% digitonin for 1 h. The lysate was clarified by centrifugation at 10,000 × g for 10 min, and 100 μl of clarified lysate was sedimented on 5–30% w/v sucrose gradients under the same conditions as yeast. Fractions were analyzed by SDS-PAGE and immunoblotting with anti-HA antibody or anti-Sec61β antibody (described in a previous study (15 (link))). Chemical Cross-linking Experiments—Yeast crude membrane fractions were resuspended in buffer containing 50 mm Hepes, pH 7.0, 150 mm KCl, and 1 mm PMSF. Ethylene glycobis(succinimidylsuccinate) (EGS, Pierce), was dissolved in anhydrous DMSO and diluted to the desired concentration. 1 μl of EGS was added into every 20 μl of protein-containing sample for 30 min at room temperature. The reactions were quenched for 15 min with 2 μl of 1 m Tris, pH 7.5. Samples were analyzed on a 4–20% SDS-PAGE and immunoblotted using standard procedures with mouse anti-His or rat anti-HA antibody conjugated to peroxidase (Sigma). For mammalian cross-linking experiments, transfected COS-7 cells were grown in a 10-cm plate to ∼80% confluency and then lysed using a standard hypotonic lysis protocol. Briefly, cells were harvested in PBS, washed, incubated in hypotonic buffer (10 mm Hepes, pH 7.8, 10 mm potassium acetate, 1.5 mm magnesium acetate, 2 mm PMSF) for 10 min, and then passed through a 25-gauge syringe ten times. Nuclei and any remaining intact cells were separated from the lysate by centrifugation for 5 min at 3,000 × g, and the supernatant was then centrifuged for 10 min at 100,000 × g to pellet the membrane fraction. The membrane pellet was washed in HKM buffer (25 mm Hepes pH 7.8, 150 mm potassium acetate, 2.5 mm magnesium acetate, and 2 mm PMSF), repelleted at 100,000 × g, and resuspended to a final volume of 60 μl in HKM buffer. 10-μl membrane aliquots were used for each cross-linking reaction using the same conditions as above. Samples were analyzed on a 4–20% SDS-PAGE and immunoblotted using standard procedures with anti-HA antibody.
Rtn1p and Yop1p have slow diffusional mobility in the ER of yeast cells.A, typical FRAP of Sec63-GFP or Rtn1-GFP in S. cerevisiae cells expressed at endogenous levels. Images were taken before and then after the photobleach for the times indicated. The boxed region shows the area that was photobleached. B, fluorescence intensities normalized to prebleach values of FRAP analyses on yeast Sec63-GFP, Rtn1-GFP, and Yop1-GFP were plotted over time. Error bars indicate ± S.E.; n = 4 cells. C, fluorescence intensities normalized to prebleach values plotted over time of FRAP analyses on yeast Rtn1p in ATP-depleted (green) or non-depleted (orange) cells, compared with that of Sec63p-GFP (ATP depleted in blue; non-depleted in red). Error bars indicate ± S.E., n = 4 cells.
ATP Depletion Experiments—For yeast experiments, ATP was depleted by the addition of 10 mm 2-deoxy-d-glucose and 10 mm sodium azide (both from Sigma) for 2–5 min, and FRAP experiments were performed using the same parameters as described above. Similarly, for mammalian cell experiments, COS-7 cells were depleted of ATP as follows: transfected cells were washed twice in Opti-Mem serum-free media (Invitrogen) and then incubated with 50 mm 2-deoxy-d-glucose and 0.02% sodium azide in glucose-free imaging buffer (50 mm Hepes, pH 7.4, 150 mm potassium acetate, 2.5 mm magnesium acetate, and 1% fetal bovine serum). FRAP experiments were conducted in the same medium and completed within 5–30 min of treatment using the same parameters as above.
Shibata Y., Voss C., Rist J.M., Hu J., Rapoport T.A., Prinz W.A, & Voeltz G.K. (2008). The Reticulon and Dp1/Yop1p Proteins Form Immobile Oligomers in the Tubular Endoplasmic Reticulum. The Journal of Biological Chemistry, 283(27), 18892-18904.
Equal amounts of recombinant prion proteins (100ng for Figure 5A and 10ng for Figure 5C) were mixed with loading dye and loaded on a 12% NuPAGE gel (Invitrogen, running with MES buffer). Mouse brain homogenates were prepared in PBS, 0.05% sodium deoxycholate, and 0.05% Nonidet P-40. For PK digestion 25 µg/ml of enzyme was used to digest PrPC. Samples corresponding to 40 µg of total proteins (with or without prior PK digestion) were mixed with loading dye and loaded on a 12% NuPAGE gel. Proteins were then transferred onto a nitrocellulose membrane incubated with hybridoma cell supernatant or purified monoclonal anti-PrP antibodies and horseradish peroxidase (HRP)-conjugated rabbit anti-mouse IgG (gamma) antibody (Zymed). For analysis of samples acquired from immunoprecipitation, blots were incubated with a biotinylated anti-PrP (POM1) antibody and HRP-labeled avidin (Pharmingen). Blots were finally incubated with HRP substrate (ECL, Pierce) and exposed on photosensitive film (Kodak) or with Versadoc 3000 imaging system (Bio-Rad).
Polymenidou M., Moos R., Scott M., Sigurdson C., Shi Y.Z., Yajima B., Hafner-Bratkovič I., Jerala R., Hornemann S., Wuthrich K., Bellon A., Vey M., Garen G., James M.N., Kav N, & Aguzzi A. (2008). The POM Monoclonals: A Comprehensive Set of Antibodies to Non-Overlapping Prion Protein Epitopes. PLoS ONE, 3(12), e3872.
Reagents—The αIIbβ3 antagonist lotrafiban was supplied by GlaxoSmithKline (King of Prussia, PA). The anti-Rac (23A8) monoclonal antibody was purchased from Upstate Biotechnology (TCS Biologicals, Bucks, UK). Anti-Rac2 polyclonal antibody and anti-Rac3 polyclonal antibody were generously provided from Gary Bokoch (Scripps Institute, La Jolla, CA) and Ivan de Curtis (San Rafaele Scientific Institute, Milan, Italy), respectively. The cDNA for the GST-CRIB domain of PAK1 prepared as described previously (21 (link)) and the active form of Rac (L61Rac) were the kind gifts from Dr. Doreen Cantrell (Imperial Cancer Research Fund, London, UK). D-Phenyl-alanyl-1-prolyl-1 arginine chloromethyl ketone was purchased from Calbiochem. Fibrinogen depleted of plasminogen, VWF, and fibronectin was from Kordia Laboratory Supplies, Leiden, Netherlands. VWF was a generous gift from Michael C. Berndt (Monash University, Clayton, Australia). All other reagents were from Sigma or previously named sources (22 (link), 23 (link)). Preparation of Human Washed Platelets—Human venous blood was drawn by venipuncture from healthy volunteers into sodium citrate and acid/citrate/dextrose as described previously (23 (link)). Platelet-rich plasma (PRP) was prepared by centrifugation of whole blood at 200 × g for 20 min. The platelets were then isolated from PRP by centrifugation at 1000 × g for 10 min in the presence of prostacyclin (0.1 μg/ml). The pellet was resuspended in modified HEPES/Tyrodes buffer (in mM: 129 NaCl, 0.34 Na2HPO4, 2.9 KCl, 12 NaHCO3, 20 HEPES, 5 glucose, 1 MgCl2; pH 7.3) containing 0.1 μg/ml prostacyclin. The platelets were washed once via centrifugation (1000 × g for 10 min) and resuspended at the desired concentration with HEPES/Tyrode buffer. Preparation of Murine Washed Platelets—The generation of mice bearing a conditional loxP-flanked allele of Rac1, Rac1flox, has been described previously (6 (link)). To induce expression of the Mx1-Cre trans-gene, the mice were given a 150-μl intraperitoneal injection of synthetic double-stranded RNA polyinosinic-polycytidylic acid (2 mg/ml) every other day for a total of three injections, and blood was taken at least 14 days after the last injection to ensure a complete turnover of platelets. The protein expression of Rac1 and Rac2 was verified for both control and Rac-deficient mice for each experiment (data not shown). The number of platelets in whole blood from Rac1-/-, Rac2-/-, or Rac1-/-Rac2-/- mice was no different from wild type (data not shown). Bleeding problems, such as the intraperitoneal hemorrhage seen in Syk- and SLP-76-deficient mice, were not observed for these mice. Murine blood was drawn from CO2terminally anesthetized mice by cardiac puncture and taken into 100 μl of acid/citrate/dextrose. PRP was obtained by centrifugation at 200 χ g for 6 min. Washed platelets were prepared via centrifugation of PRP at 1000 × g in the presence of prostacyclin (0.1 μg/ml) for 6 min. The pellet was resuspended in modified HEPES/Tyrode buffer to the desired platelet level. All animals were maintained using housing and husbandry in accordance with local and national legal regulations. In separate experiments, human or murine platelet suspensions were treated with 10 μM cytochalasin D, 0.1-10 μg/ml CRP, 1-10 μM ADP, 0.04-1 units/ml thrombin, 10 μM lotrafiban or 1 mM adenosine 3′,5′-diphosphate (A3P5P), and 1 μM AR-C67085 for 10-30 min before use in the assays. Unless otherwise stated, all experiments were performed in the presence of 2 units/ml apyrase and 10 μM indomethacin and in the absence of exogenously added Ca2+. Measurement of Rac Activity—Rac activity was measured essentially as described in Pearce et al. (18 (link)) using the CRIB domain of PAK1 (amino acids 67-150), which binds the GTP-bound form of Rac. Following stimulation of platelet suspensions (3 × 108/ml), reactions were stopped with an equal volume of 2× lysis buffer (2% (v/v) Nonidet P-40, 2% (w/v) N-octyl glucoside, 300 mM NaCl, 20 mM Tris/HCl, 2 mM EGTA, 20 mM MgCl2, 1 mM phenylmethylsulfonyl fluoride, 10 μg/ml leupeptin, 10 μg/ml aprotinin, 1 μg/ml pepstatin A, pH 7.4, and 2 mM orthovana-date). Alternatively, platelets (3 × 108/ml) were incubated for 10-45 min in dishes coated with fibrinogen, collagen, laminin, or BSA2in the absence or presence of thrombin (1 units/ml) and apyrase (2 units/ml). Unbound platelets were removed by two washes with phosphate-buffered saline followed by aspiration, and adherent cells were solubilized with 1× lysis buffer. A sample of the suspension over BSA was taken and used as a control. Insoluble material was then removed by centrifugation (5 min, 13,000 rpm), and GST-PAK1, previously incubated with glutathione agarose beads, stored in glycerol at -80 °C, and washed with 1× lysis buffer, was added to the lysates and incubated for 1 h at 4 °C. Beads were then washed with 1× lysis buffer, and the bound protein was taken up into Laemmli buffer. The resulting samples were separated by 12% SDS-PAGE, transferred to polyvinylidene difluoride membranes, and immunoblotted with a Rac-specific antibody and horseradish peroxidase-conjugated secondary antibodies (Amersham Biosciences). Protein was detected using ECL (Amersham Biosciences). Measurement of Filamentous Actin Content—Filamentous actin content of washed platelets was measured using a modification of the method of Machesky and Hall (24 (link)). Basal or activated platelets (2 × 108/ml) were fixed with an equal volume of 3.7% formaldehyde containing a saturating amount of FITC-phalloidin (20 mM KH2PO4, 10 mM Pipes, 5 mM EGTA, 2 mM MgCl2, 0.1% Triton X-100, 3.7% formalde-hyde, 2 μM FITC-phalloidin) and incubated for1hat room temperature on a nutator. The platelets were then pelleted for 2 min in a microcentrifuge, and pellets were washed in 0.1% saponin, 20 mM KH2PO4, 10 mM Pipes, 5 mM EGTA, 2 mM MgCl2. Pellets were then resuspended in methanol to extract the FITC-phalloidin and incubated for 1 h on a nutator at room temperature. FITC-phalloidin binding was measured for each sample with the fluorescence emission at 520 nm and excitation at 488 nm. Alternatively, F-actin levels were assessed via flow cytometry after fixation, permeabilization, and staining of platelets with FITCphalloidin as described previously (18 (link)). Filamentous actin content was expressed as a comparison with values obtained for untreated cells processed in parallel on the same day. Platelet Aggregation and Shape Change—A quantity of 300 μl of PRP or washed platelets (2 × 108/ml) was used for aggregation. Stimulation of platelets was performed in a PAP-4 aggregometer (Bio/Data Corp., Horsham, PA) with continuous stirring at 1200 rpm at 37 °C for the times shown. Aggregation of platelets was monitored by measuring changes in light transmission. Static Adhesion Assays—Coverslips were incubated with a suspension of fibrinogen (100 μg/ml), collagen (100 μg/ml), or laminin (50 μg/ml) overnight at 4 °C. Surfaces were then blocked with denatured BSA (5 mg/ml) for 1 h at room temperature followed by subsequent washing with phosphate-buffered saline before use in spreading assays. Quiescent platelets failed to bind to surfaces coated with denatured BSA (data not shown). Platelet spreading (2 × 107/ml) was imaged in real time using Köhler illuminated Nomarski differential interference contrast optics with a Zeiss 63× oil immersion 1.40 NA plan-apochromat lens on a Zeiss Axiovert 200M microscope. Time-lapse events were captured by a Hamamatsu Orca 285 cooled digital camera (Cairn Research, Kent, UK) using Slidebook 4.0 (Intelligent Imaging Innovations, Inc., Denver, CO). To compute the length and thickness of filopodia and surface area of spreading platelets, images were manually outlined and quantitated by determining the number of pixels within each outline using a Java plugin for the Image J software package as described previously (23 (link)). Imaging a graticule under the same conditions allowed the conversion of pixels size to microns. Flow Adhesion Studies—For flow adhesion studies using collagen, mouse blood was drawn into sodium heparin (10 IU/ml) and D-phenylalanyl-1-prolyl-1 arginine chloromethyl ketone (40 μM). Alternatively, mouse blood was drawn into sodium citrate (0.38% w/v) for immobilized VWF adhesion studies. Glass capillary tubes (Camlab, Cambridge, UK) were coated with 100 μg/ml type I collagen from equine tendon (Horm, Nycomed, Munich, Germany) or 100 μg/ml VWF, 100 units/ml thrombin for1hat room temperature. The capillaries were washed and blocked with phosphate-buffered saline containing 5 mg/ml BSA for 1 h at room temperature before being mounted on the stage of an inverted microscope (DM IRB; Leica, Milton Keynes, UK). Anticoagulated whole blood was perfused through the chamber for 4 min at a wall shear rate of 1000 s-1, followed by washing for 3 min at the same shear rate with modified Tyrodes buffer before being fixed with 3.7% paraformaldehyde for 30 min and imaged using DIC microscopy. In selected experiments, fixed samples were incubated overnight with the fluorescent dye DiOC6(2 μM; Molecular Probes Inc., Eugene, Oregon) before being imaged using confocal microscopy (DM IRE2; Leica, Milton Keynes, UK). In separate experiments, whole blood was fluorescently labeled with DiOC6(2 μM, 10 min at 37 °C), and the accumulation of DiOC6-labeled platelets was monitored in real time using fluorescence microscopy (CoolSnap ES, Photometrics, Huntington Beach, CA). Image analysis was performed off-line using ImageJ. Platelet adhesion results are expressed as the percentage of surface area covered by platelets. Laser-induced Vessel Wall Injury—All procedures were undertaken with approval from the United Kingdom Home Office in accordance with the Animals (Scientific Procedures) Act of 1986. Male mice were anesthetized with ketamine (100 mg/kg Vetalar; Amersham Biosciences and Upjohn Ltd., UK) and 2% xylazine (20 mg/kg; Millpledge Pharmaceuticals, UK). The cremaster, a transparent muscle surrounding the testicle, was exteriorized and continuously superfused with a bicarbonate-buffered saline (36 °C) aerated with 5% CO2, 95% N2. High speed intravital microscopy experiments were performed as described previously by Falati et al. (25 (link)). Arterioles with a diameter of 25-35 μm were selected for study, and endothelial injury was induced using a pulsed nitrogen dye laser (coumarin 440 nm) focused on the luminal surface. To label platelets fluorescently, 20 μl of Alexa Fluor 488 conjugated to goat anti-rat antibody (Molecular Probes, Eugene, OR) and 5 μl of rat anti-mouse CD41 antibody (Pharmingen) were added to 70 μl of saline and infused via the carotid cannula. Multiple thrombi in each cremaster preparation were generated upstream to previous injuries in the same or similar sized arterioles. The background fluorescence intensity, predominantly because of freely circulating platelets, was determined and subtracted from the fluorescence intensity of the developing thrombus. The resulting value was multiplied by the sum of all pixels above background to give a value for integrated intensity at each time point. This integrated intensity value was directly proportional to the size of the developing thrombus and when plotted against time provided a graph that illustrated the dynamic kinetics of platelet accumulation. Analysis of Data—Experiments were carried out on at least three occasions, and images shown are representative data from one experiment. Where applicable, results are shown as mean ± S.E. Statistical significance of differences between the means was determined by analysis of variance. If the means were shown to be significantly different, multiple comparisons were performed by the Tukey test. Probability values of p < 0.01 were selected to be statistically significant.
McCarty O.J., Larson M.K., Auger J.M., Kalia N., Atkinson B.T., Pearce A.C., Ruf S., Henderson R.B., Tybulewicz V.L., Machesky L.M, & Watson S.P. (2005). Rac1 Is Essential for Platelet Lamellipodia Formation and Aggregate Stability under Flow. The Journal of biological chemistry, 280(47), 39474-39484.
HeLa cells were co-transfected with pcDNA3.1-MS2 vector or pcDNA3.1-U3-MS2, and pMS2-GFP using Lipofectamine 2000. Twenty-four hours after transfection, cells were synchronized by 50 ng/mL nocodazole for 12 h. Cells were harvested and lysed in Buffer A (25 mM Tris-Cl pH 7.5, 150 mM KCl, 1 mM DTT, 2 mM EDTA, 0.5 mM PMSF, 0.2% Nonidet P-40 and fresh protease inhibitor cocktail). Anti-GFP antibody was coupled with a 50% suspension of protein A-Sepharose beads (Cytiva) in IPP500 (500 mM NaCl, 10 mM Tris-Cl pH8.0, 0.2% Nonidet P-40) for 2 h at 4 °C. After washing, the coupled beads were incubated with 5 mg cell lysates for 4 h at 4 °C. After washing with Buffer B (10 mM Tris-Cl pH 8.0, 150 mM KCl, 5 mM MgCl2, 0.1% Nonidet P-40) five times, the precipitants were subjected to SDS-PAGE and visualized by silver staining. The bands were cut from SDS-PAGE gel, fully trypsinized, and analyzed by Q-Extractive liquid chromatography-tandem mass spectrometry (LC-MS/MS).
Jiang Y., Sun S., Liu X., Su K., Zhang C., Zhang P., Zhao Z., Su Y., Wang C, & Du X. (2024). U3 snoRNA inter-regulates with DDX21 in the perichromosomal region to control mitosis. Cell Death & Disease, 15(5), 342.
Total proteins were extracted from one gram of co-inoculated leaves of N. benthamiana by using 2 mL of ice-cold immunoprecipitation buffer (10% [v/v] glycerol, 25 mM Tris-HCI, pH 7.5, 150 mM NaCl, 10 mM DTT, 1 mM EDTA, 1 × Protease Inhibitor Cocktail, For Plant Cell (Sangon Biotech), and 0.15% [v/v] Nonidet P-40). Protein extracts were incubated with GFP-Trap beads (ChromoTek) for 1h at 4°C. The beads were collected and washed with the buffer (10 mM Tris-HCl pH 7.5, 150 mM NaCl, 0.05% Nonidet P40, 0.5 mM EDTA). Total protein extracts prior to (Input) and after immunoprecipitation (IP) were analyzed by immunoblotting using anti-GFP and anti-Myc polyclonal antibodies (Abcam), essentially as previously described [63 (link)].
Qin L., Liu H., Liu P., Jiang L., Cheng X., Li F., Shen W., Qiu W., Dai Z, & Cui H. (2024). Rubisco small subunit (RbCS) is co-opted by potyvirids as the scaffold protein in assembling a complex for viral intercellular movement. PLOS Pathogens, 20(3), e1012064.
Cells were harvested by trypsinization and washed once in ice-cold phosphate-buffered saline (pH 7.2). The suspension was centrifuged at 400× g for 7 min at 25 °C. The pellet was resuspended in buffer A (20 mM HEPES, pH 7.9, 25 mM KCl, 10% glycerol, 0.1% Nonidet P-40), supplemented with a commercially available protease inhibitor cocktail (ThermoFisher), and incubated on ice for 7 min. Nuclei were isolated by centrifugation at 645× g for 10 min at 4 °C. Pelleted nuclei were resuspended in lysis buffer (50 mM HEPES, pH 7.5, 150 mM NaCl, 0.5% Nonidet P-40, supplemented with protease inhibitor), rotated at 4 °C for 30 min, then the extracts were cleared by centrifugation at 21,000× g at 4 °C for 30 min. Lysate concentration was assessed using bicinchoninic acid assay (BCA).
Innis S., Alpsoy A., Crodian J., Tseng Y.C., Dykhuizen E., Cabot B, & Cabot R. (2024). Identification of SWI/SNF Subcomplex GBAF Presence, Intra-Complex Interactions, and Transcriptional Dynamics during Early Porcine Development. Animals : an Open Access Journal from MDPI, 14(5), 773.
293FT cells transfected with each of the plasmid using FuGENE6 (Roche Applied Science, Upper Bavaria, Germany) were washed with ice-cold PBS and lysed with lysis buffer (50 mM Tris-HCl, pH 8.0, 50 mM NaCl, 1% Nonidet P-40) containing protease inhibitor cocktail (Pierce), 1 mM Na3VO4, 1 mM NaF, 1 mM phenylmethylsulfonyl fluoride, and 60 mM octyl glucoside. Immunoprecipitation was carried out by incubating the same amount of cell lysates with magnetic beads (Magnosphere MS300/Carboxyl, COSMO BIO, Tokyo, Japan) coated with each antibody at 4 °C overnight. Beads were washed with wash buffer (50 mM Tris, pH 8.0, 50 mM NaCl, 1.0% Nonidet P-40, 1 mM NaF) five times and the precipitated proteins were separated by SDS–PAGE, transferred to a polyvinylidene difluoride membrane, and probed with each antibody.
Tomita S., Nakanishi N., Ogata T., Higuchi Y., Sakamoto A., Tsuji Y., Suga T, & Matoba S. (2024). The Cavin-1/Caveolin-1 interaction attenuates BMP/Smad signaling in pulmonary hypertension by interfering with BMPR2/Caveolin-1 binding. Communications Biology, 7, 40.
For in vitro ubiquitination assay, cells carrying plasmid ptifHis-ubi (expressing His6-tagged ubiquitin under control of the tif51 promoter) were grown for 12 h and then treated with BZ for 12 h. Cells were then harvested and resuspended in lysing buffer (50 mM Tris–HCl, pH 7.5, 150 mM NaCl, 5 mM MgCl2, 1% Nonidet P-40, 10 mM N-ethylmaleimide, 1 mM PMSF, and a complete protease inhibitor mixture) and lysed using a FastPrep-24 bead beater (MP Biomedical). The cell lysates were incubated with Ni-NTA agarose beads overnight at 4 °C. Beads were washed with washing buffer (50 mM Tris–HCl, pH 7.5, 150 mM NaCl, 5 mM MgCl2, 0.1% Nonidet P-40, 10 mM N-ethylmaleimide, 1 mM PMSF, and a complete protease inhibitor mixture), and bound proteins were eluted by boiling in SDS-PAGE sample buffer and were analyzed by Western blotting.
Yao R., Li R., Wu X., Jin T., Luo Y., Li R, & Huang Y. (2024). E3 ubiquitin ligase Hul6 modulates iron-dependent metabolism by regulating Php4 stability. The Journal of Biological Chemistry, 300(3), 105670.
Sourced in United States, Germany, China, United Kingdom, Italy, Japan, Sao Tome and Principe, France, Canada, Macao, Switzerland, Spain, Australia, Israel, Hungary, Ireland, Denmark, Brazil, Poland, India, Mexico, Senegal, Netherlands, Singapore
The Protease Inhibitor Cocktail is a laboratory product designed to inhibit the activity of proteases, which are enzymes that can degrade proteins. It is a combination of various chemical compounds that work to prevent the breakdown of proteins in biological samples, allowing for more accurate analysis and preservation of protein integrity.
Sourced in United States, Switzerland, Germany, China, United Kingdom, France, Canada, Japan, Italy, Australia, Austria, Sweden, Spain, Cameroon, India, Macao, Belgium, Israel
Protease inhibitor cocktail is a laboratory reagent used to inhibit the activity of proteases, which are enzymes that break down proteins. It is commonly used in protein extraction and purification procedures to prevent protein degradation.
Sourced in United States, Germany, China, United Kingdom, Morocco, Ireland, France, Italy, Japan, Canada, Spain, Switzerland, New Zealand, India, Hong Kong, Sao Tome and Principe, Sweden, Netherlands, Australia, Belgium, Austria
PVDF membranes are a type of laboratory equipment used for a variety of applications. They are made from polyvinylidene fluoride (PVDF), a durable and chemically resistant material. PVDF membranes are known for their high mechanical strength, thermal stability, and resistance to a wide range of chemicals. They are commonly used in various filtration, separation, and analysis processes in scientific and research settings.
Sourced in United States, Germany, Italy, Switzerland, China, Canada, Japan, France, Ireland
Nonidet P-40 is a non-ionic detergent used in various laboratory applications. It is a polyoxyethylene derivative that can be used for cell lysis, protein extraction, and other biochemical procedures. The product is designed to maintain the native structure and function of biomolecules during processing.
Sourced in United States, Germany, Switzerland, United Kingdom, China, France, Japan, Canada, Spain, Belgium, Australia, Sweden, Italy, Ireland, Macao
The Complete Protease Inhibitor Cocktail is a laboratory product designed to inhibit a broad spectrum of proteases. It is a concentrated solution containing a mixture of protease inhibitors effective against a variety of protease classes. This product is intended to be used in research applications to preserve the integrity of target proteins by preventing their degradation by proteolytic enzymes.
Sourced in United States, China, Germany, United Kingdom, Japan, Belgium, France, Switzerland, Italy, Canada, Australia, Sweden, Spain, Israel, Lithuania, Netherlands, Denmark, Finland, India, Singapore
The BCA Protein Assay Kit is a colorimetric detection and quantification method for total protein concentration. It utilizes bicinchoninic acid (BCA) for the colorimetric detection and quantification of total protein. The assay is based on the reduction of Cu2+ to Cu1+ by protein in an alkaline medium, with the chelation of BCA with the Cu1+ ion resulting in a purple-colored reaction product that exhibits a strong absorbance at 562 nm, which is proportional to the amount of protein present in the sample.
Sourced in United States, Switzerland, Germany, China, United Kingdom, Canada, France, Japan, Italy, Australia, Spain, India, Macao, Netherlands, Denmark, Belgium, Cameroon
Protease inhibitors are a class of laboratory equipment used in the field of biochemistry and molecular biology. These inhibitors are designed to specifically target and inactivate proteases, which are enzymes that break down proteins. Protease inhibitors play a crucial role in various experimental and analytical procedures, such as protein extraction, purification, and stabilization.
Sourced in United States, United Kingdom, Germany, China, Canada, Japan, Macao, Italy, Sao Tome and Principe, Israel, Spain, Denmark, France, Finland, Australia, Morocco, Ireland, Czechia, Sweden, Uruguay, Switzerland, Netherlands, Senegal
β-actin is a protein that is found in all eukaryotic cells and is involved in the structure and function of the cytoskeleton. It is a key component of the actin filaments that make up the cytoskeleton and plays a critical role in cell motility, cell division, and other cellular processes.
Sourced in United States, Germany, Italy, United Kingdom, Canada, France, China, Switzerland, Japan, Spain, Australia, Sweden, Portugal, Israel, Netherlands, Belgium
Nitrocellulose membranes are a type of laboratory equipment designed for use in protein detection and analysis techniques. These membranes serve as a support matrix for the immobilization of proteins, enabling various downstream applications such as Western blotting, dot blotting, and immunodetection.
Sourced in United States, Germany, United Kingdom, Italy, Canada, China, France, Switzerland, Austria, Spain, Japan, Australia, Brazil, Ireland, Sweden
The Bradford assay is a colorimetric protein assay used to measure the concentration of protein in a solution. It is based on the color change of the Coomassie Brilliant Blue G-250 dye in response to various concentrations of protein.
Nonidet is a nonionic detergent that is widely used in biochemical applications, such as cell lysis, protein extraction, and enzyme assays. It effectively solubilizes membrane proteins and disrupts protein-protein interactions while maintaining protein structure and function. Nonidet is particularly useful for studies involving membrane proteins and other hydrophobic biomolecules.
One of the key challenges with Nonidet is identifying the optimal concentration and protocol for a specific application. The detergent's effectiveness can vary depending on the target proteins, experimental conditions, and other factors. Researchers may also face difficulties in ensuring consistent and reproducible results when using Nonidet-based protocols.
PubCompare.ai's AI-powered platform can greatly assist researchers in optimizing their use of Nonidet. The platform allows you to efficiently screen protocol literature, leveraging AI to pinpoint critical insights. PubCompare.ai can help you identify the most effective Nonidet protocols for your specific research goals, highlighting key differences in protocol effectiveness and enabling you to choose the best option for reproducibility and accuracy. This helps take the guesswork out of your Nonidet-based research.
Yes, there are several different Nonidet variants, each with slightly different properties and applications. For example, Nonidet P-40 (NP-40) is a commonly used Nonidet variation that is particularly effective at solubilizing membrane proteins, while Nonidet P-10 (NP-10) is more gentle and better suited for preserving protein-protein interactions. Researchers should carefully consider the specific properties of each Nonidet variant to select the most appropriate one for their experiments.
More about "Nonidet"
Nonidet, a versatile nonionic detergent, plays a crucial role in various biochemical applications such as cell lysis, protein extraction, and enzyme assays.
Derived from polyoxyethylene alkyl phenol, Nonidet effectively solubilizes membrane proteins and disrupts protein-protein interactions, all while maintaining the structure and function of the proteins being studied.
Researchers can leverage PubCompare.ai's AI-powered platform to effortlessly identify the best Nonidet protocols and products from literature, preprints, and patents.
The platform's comparison tools enhance reproducibility and accuracy, taking the guesswork out of Nonidet-based research.
Complementary techniques like Protease inhibitor cocktails, PVDF membranes, Nonidet P-40, Complete protease inhibitor cocktail, BCA protein assay kits, Protease inhibitors, β-actin, Nitrocellulose membranes, and Bradford assays can be seamlessly integrated into Nonidet-based workflows to ensure optimal protein extraction, purification, and analysis.
PubCompare.ai's powerful suite of tools empowers researchers to navigate the complexities of Nonidet-related protocols and products, ultimately enhancing the reproducibility and accuracy of their findings.
With the AI-driven platform, researchers can confidently optimize their Nonidet-based research and unlock new insights with ease.