The largest database of trusted experimental protocols

Low protein binding filter

Manufactured by Merck Group
Sourced in United States

The Low Protein Binding Filter is a laboratory filtration device designed to minimize the adsorption of proteins to the filter surface. It is intended to provide efficient separation and recovery of protein-containing samples while maintaining the integrity of the proteins. The filter is constructed using materials that exhibit low protein binding properties, enabling the preservation of protein structure and functionality during the filtration process.

Automatically generated - may contain errors

13 protocols using low protein binding filter

1

Immortalization of Primary Macrophages

Check if the same lab product or an alternative is used in the 5 most similar protocols
To immortalize macrophages, we used J2 recombinant retrovirus [110 (link)] produced from ψCREJ2 cells (a generous gift from John MacMicking, Yale University School of Medicine). The J2-expressing cells were grown to confluency in DMEM medium supplemented with 10% FBS (D10). The medium containing retroviral particles was collected and passed through 0.45 μM low protein-binding filters (Millipore). In parallel, 5 x 106 primary BMDM from AJ and C57BL/6J, obtained as described above, were thawed and grown for 2 days in DMEM medium supplemented with 10% FBS and 20% L929. After 2 days, the medium was replaced with the filtered J2-retrovirus-containing medium supplemented with 50% L929. After 24 hrs, the media was replaced with fresh D10 medium supplemented with 25% L929. Media was subsequently changed after every 24hrs with concomitant reduction in L929 until 10% L929 concentration was reached. Immortalized cells were harvested and stored in liquid nitrogen until use.
+ Open protocol
+ Expand
2

Fungal Siderophore Production under Hypoxia

Check if the same lab product or an alternative is used in the 5 most similar protocols
Extracellular siderophore production was measured by a modified form of the assay previously described (29 (link)). Briefly, fungal strains were grown in 50 ml of liquid GMM without iron under normoxic conditions at 37°C with constant shaking at 200 rpm in complete darkness; this was followed by a shift to hypoxia under the same conditions for an additional 24 h. Fungal biomass was collected via vacuum filtration and lyophilized. Culture supernatants were collected and adjusted to pH 6.5 with NaOH and/or HCl and filtered through low-protein-binding filters (0.45-µm pore size; Millipore). Five microliters of FeSO4 dissolved in 5mM HCl was added to 95 µl of culture filtrates to a final concentration of 1.5 mM iron in 96-well plates, and the plates were incubated for 30 min at room temperature. Optical density at 440 nm was measured and normalized to control wells incubated with HCl alone. Siderophore production per milligram of fungal dry weight was measured.
+ Open protocol
+ Expand
3

Lentiviral Transduction of Cathepsin D Mutants

Check if the same lab product or an alternative is used in the 5 most similar protocols
pLVX-Puro-cathDwt, LentiCRISPRv2-cathD sgRNA-GFP, or other cathD mutated constructs were transfected along with the packaging plasmid, psPax and pMD2.G envelop plasmid, into Lenti-X 293T cells at a ratio of 4:3:1. After transfection, Lenti-X 293 T cells were cultured in DMEM with high glucose (4.5 g/L) supplemented with 20% FBS for 36 h. Viral supernatants were then collected by centrifugation at 2000× g for 5 min and filtered by 0.45 μm low-protein binding filters (Merck). Harvested viral medium were added with 10 μM polybrene (Sigma-Aldrich) to improve transduction efficiency, diluted with DMEM plus 10% FBS (1:1 in volume) and added to HeLa or HEK293T cells.
After 36–48 h of lentiviral transduction, GFP-positive cells were sorted by a MoFlo Astrios EQ cell sorter (Beckman, Brea, CA) to establish stable cathD-deficient (cathD-/-) cell line. To further reintroduce cathDwt and other cathD mutants, cathD-contained lentiviral particles were added into cathD-/- HeLa cells. Transduction medium was then replaced with fresh culture medium supplemented with puromycin (Takara Bio) to a final concentration of 0.5–3 μg/mL after 48 h of lentiviral transductions to establish stable cathD-mutated cell lines.
+ Open protocol
+ Expand
4

Mycobacterial Protein Extraction Protocol

Check if the same lab product or an alternative is used in the 5 most similar protocols
Cells were harvested during the exponential phase (OD600 ~1.2 and 0.6 for M. smegmatis and M. bovis BCG, respectively) by centrifugation at 4000 g for 15 min at 4°C, washed twice with Phosphate Buffered Saline pH (7.5) (PBS). Cells were snap frozen in liquid nitrogen and stored at -80°C until needed. Frozen pellets were suspended in 800 μl lysis buffer [500 mM Tris-HCl, 0.1% (w/v) SDS, 0.15% sodium deoxycolate, 1× protease inhibitor cocktail, 1× phosphatase inhibitor cocktail (Roche, Mannheim Germany) and 50 μg/ml lysozyme (Repaske, 1956 (link))]. Cells were disrupted by sonication at maximum power for six cycles of 30 s, with 1 min cooling on ice between cycles. Cellular debris was removed by centrifugation at 4000 g for 5 min and the lysate filtered through 20 μm pore size low-protein binding filters (Merck, NJ, USA). Proteins were precipitated using the chloroform-methanol precipitation method as previously described (Wessel and Flügge, 1984 (link)). The pellet was re-suspended in urea buffer (6 M urea, 2 M thiourea and 10 mM Tris-HCl, pH 8). Protein concentration was determined using a modified Bradford assay as described by Ramagli (1999 (link)).
+ Open protocol
+ Expand
5

Disrupting HOXC13 Expression in T Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Single-guide RNA (sgRNA) targeting the predicted ORF for 5′U-HOXC13 were designed with CRISPOR tool (http://crispor.tefor.net/). sgRNA specifically binding the genomic peptide location or upstream with the highest predicted knock out (KO) efficiency were selected. Next, sgRNA were cloned into lenti-Cas9-v2 (RRID:Addgene_52961) with BsmBI.
Lentiviral supernatants were generated by co-transfecting HEK293 cells with lenti-Cas9-v2 encoding the sgRNA of interest, psPAX2 and pMD2G plasmids with PEI (Sigma) and non-supplemented DMEM media (Gibco). Media was replaced after 12 hours with Opti-MEM (Gibco) containing 2% FBS (Gibco). Supernatants were collected at 42 to 48 hours after transfection and filtered through a 0.45-μm low-protein binding filter (Millipore). Patient-derived TCL were infected with lentiviral supernatants containing polybrene (Sigma) final concentration of 8 μg/mL followed by spinoculation 900 g, 32°C, 50 minutes. Five days post-infection, T2 media (RPMI containing 10% FBS supplemented with Pen/Strep and l-Glut) was replaced with T2 media containing puromycin 1 μg/mL to select the cells efficiently transduced. Thereafter, media was replaced with fresh media containing puromycin when acidified or cells split when confluent. To evaluate the KO efficiency, puromycin-resistant cells were used as targets in coculture assays with 5′U-HOXC13 specific T cells.
+ Open protocol
+ Expand
6

Lentiviral Transduction of MCF7 Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
The overexpression (oe)-lncRNA ROR, short hairpin TIMP3 (sh-TIMP3) and sh-lncRNA ROR were synthesized by Shanghai Sangon Biological Engineering Technology & Services Co., Ltd. (Shanghai, China), which were subsequently co-transfected with three package vectors PMDLG/PRRE, vesicular stomatitis virus glycoprotein (VSVG), and respiratory syncytial virus (RSV)/respiratory entericorphan virus (REV) into HEK293T cells. After 6 h, the culture medium was replaced with fresh DMEM containing 10% FBS. The lentivirus-containing medium was harvested at both the 48 and 72 h time points post-transfection and filtered to remove cell debris using a 10 mL syringe and a 0.45 μm low protein binding filter (Millipore, Billerica, MA, USA). The virus was either immediately used for infection or stored at − 80℃ for later use. Regarding infection, the medium containing lentivirus was employed to infect the MCF7 cells in the presence of 5 μg/mL polybrene. The plates were subsequently placed in a 37℃ incubator with 5% CO2 for 20–24 h. The old medium was renewed with fresh complete medium. Finally, the infected MCF7 cells were analyzed using western blot analysis or RT-qPCR.
+ Open protocol
+ Expand
7

Lentiviral Transduction for TDO2 Modulation

Check if the same lab product or an alternative is used in the 5 most similar protocols
Lentivirus preparation and transduction were performed according to the manufacturer’s protocol. Lentivirus-based TDO2 small hairpin (sh)RNA (TRCN0000064900), control plasmid (SHC016 1EA), and lentiviral packaging mix were purchased from Sigma-Aldrich. TDO2 lentiviral vector (pLenti-GIII-CMV-Human-TDO2-GFP-2A-Puro Lentiviral Vector, LV332282) was purchased from Applied Biologic Materials (Richmond, Canada). Control, shTDO2, or TDO2 plasmids were cotransfected with the packaging plasmids into 293T cells using Lipofectamine 2000 (Invitrogen, Carlsbad, CA). Forty-eight hours after transfection, the virus-containing supernatants were harvested and centrifuged at 3000 rpm for 15 minutes and filtrated through a 0.45-μm low protein–binding filter (Millipore, Bedford, MA). Hs578T and SUM149 cells were infected with lentivirus (3 multiplicities of infection) in the presence of hexadimethrine bromide (5 μg/mL polybrene; Sigma-Aldrich) and fed with fresh complete medium the next day. Seventy-two hours after infection, the transduced cells were selected by puromycin (2 μg/ml). Efficiency of TDO knockdown or overexpression was validated by qRT-PCR and immunoblot analyses. Cells were cultured thereafter in 1.5 μg/ml puromycin and changed to puromycin-free medium 3–5 days prior to each experiment.
+ Open protocol
+ Expand
8

Metabolic Activation of Bone Marrow Macrophages

Check if the same lab product or an alternative is used in the 5 most similar protocols
Bone marrow cells were isolated from femurs of 3–6 months old male TG2 null mice and their wild type counterparts. Bone marrow macrophages (BMDMs) were differentiated in DMEM supplemented with 10% FBS (12106C), 2 mM L-glutamine (G7513), 1 mM Na-pyruvate (S8636), 50 μM 2-mercaptoethanol (M3148) and 100 U/ml penicillin/100 μg/ml streptomycin (P4333) all from Sigma-Aldrich and 10% L929 fibroblast conditioned media for 7 days. For metabolic activation, differentiated macrophages were treated with a combination of 30 mM D-glucose (G8270), 10 nM insulin (12643) and 0.4 mM sodium-palmitate (P9767) all from Sigma-Aldrich for 24 h (16). Sodium palmitate was prepared by diluting a 200 mM stock solution in 70% ethanol into 10% fatty acid-free, low-endotoxin BSA (Sigma Aldrich, A8806 adjusted to pH 7.4) to obtain a 5 mM palmitate-BSA stock solution that was filtered using a 0.22-μm low-protein binding filter (Millipore). BSA/ethanol was used in control treatments during the protocol. In some experiments during the 24 h metabolic activation BMDMs were treated with PP2 (Sigma Aldrich, 529573), a reversible ATP-competitive inhibitor of the Src family of protein tyrosine kinases, in 2 µM final concentration or 0.5 mg/ml RGD peptide (Cayman Chemical, 529573).
+ Open protocol
+ Expand
9

Generating Fluorescent Virus-Permissive Cell Lines

Check if the same lab product or an alternative is used in the 5 most similar protocols
VeroE6 GFP1-10, VeroE6-TMPRSS2 cells, VeroE6-TMPRSS2 GFP1-10 cells, and Calu-3 GFP1-10 cells were generated by retroviral transduction. To produce the retrovirus, 10 μg pQXCIH-TMPRRS2-HA or pQXCIN-GFP1-10 was co-transfected with polyethylenimine (PEI) with 6.5 μg pBS-gag-pol (Addgene #35614) and 5 μg pMD2.G (Addgene #12259) in a 10 cm dish of 70% confluent HEK-293T cells in Opti-MEM I (1×) + GlutaMAX. Retroviral particles were harvested at 72 hr post-transfection, cleared by centrifugation at 2000 × g, filtered through a 0.45 μm low protein binding filter (Millipore), and used to transduce designated cells. Polybrene (Sigma) was added at a concentration of 4 μg/mL to enhance transduction efficiency. Transduced cells were selected with hygromycin B (Invitrogen) for TMPRSS2 cells and/or geneticin (Invitrogen) for GFP1-10 cells.
+ Open protocol
+ Expand
10

Generation of KiSS1-overexpressing HepG2 Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Transfection of the KiSS1 gene into 293T cells was conducted according to the manufacturer’s manual (Origene, USA). Briefly, for transformation, 293T cells were stably packaged with lentviral vector pLenti-C-mGFP-P2A-Puro (Origene) containing KiSS1 DNA (Origene). The lentivirus-containing medium was collected from 293T packaging cell line, the medium was filtrated with 0.22 µm low protein binding filter (Millipore, Germany). At the half-confluent of HepG2 cells then, the lentivirus-containing medium transferred to HepG2 cells (50%-60% confluency) with 10 µg/ml hexadimethrine bromide (Sigma-Aldrich, USA). After infection for 18 h, the lentivirus-containing medium was replaced with fresh cell culture medium and further incubated for 48 h. Transfectants were selected with antibiotics free-medium containing 10 µg/ml puromycin (Sigma-Aldrich) for 48 h, and was repeated 3 times. In this study, the KiSS1-overexpressing HepG2 cell line gene was named HepG2-KiSS1 cells. For vector control group, mock transfection was performed without adding only KiSS1 cDNA clone.
+ 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!