The largest database of trusted experimental protocols

Latest AI and User-Annotated Protocols for Enhanced Reproducibility

As authors may omit details in methods from publication, our AI will look for missing critical information across the 5 most similar protocols.

Our protocols are annotated on demand by our users. Below you will find the list of the last 30 protocols annotated by our AI tool

Last 30 annotated protocols

Phenol-chloroform extraction method was used to obtain total RNA. 10 ml of cells were quickly chilled in an ice water bath and harvested by centrifugation at 3000 g for 5 min. Cell pellets were re-suspended in 500 µl of 0.3 M sodium acetate and 10 mM EDTA pH 4.5. Re-suspended cells were mixed with 500 µl of acetate-saturated phenol-chloroform pH 4.5 and 500 µl of acid-washed glass beads (G1277, Sigma). The mixture was shaken in a vortexer for 3 min and then clarified by centrifugation at 21,000 g for 10 min. The samples were maintained at 4°C through this step. The aqueous layer was extracted twice with acetate-saturated phenol-chloroform pH 4.5 and once with chloroform. Total RNA was precipitated with an equal volume of isopropanol, washed with 70% ethanol, and finally re-suspended in 200 µl of RNase-free 10 mM Tris pH 7.0. 200 ng of the total RNA was treated with DNase I (M0303S, NEB) to remove residual DNA contamination (manufacturer’s instructions were followed). The DNA-free RNA was column-purified (Quick-RNA Miniprep, Zymo Research, Irvine, CA).
Publication 2017
Acids Bath Cells Centrifugation Chloroform Deoxyribonuclease I DNA Contamination Edetic Acid Ethanol Ice Isopropyl Alcohol Pellets, Drug Phenol phenyl acetate ribonuclease PH Sodium Acetate Tromethamine
Wild type (C57BL/10) male and female mice 8–10 weeks-of-age were anesthetized with isoflurane, and hair on the distal portion of both lower legs was removed with Nair Lotion (Church and Dwight Co., Ewing, NJ). The leg was rinsed with sterile water and dried. The mice were injected (Becton Dickinson, Franklin Lakes, NJ, 3/10 cc U-100 Insulin syringe, 30G × 3/8″needle) intramuscularly into the middle portion of the mouse’s left tibialis anterior (TA) muscle with 50 μL of sterile 1.2% barium chloride (Sigma-Aldrich, St. Louis, MO, B0750) diluted in sterile water as previously described (Hauck et al., 2019 (link)). To serve as a control, the right TA muscle was injected with 50 μL of sterile saline. Animals were put into a warm chamber for recovery, prior to being transferred back into regular housing cages with food at the cage bed. Their health was monitored each day post injury. No animal in this study met early removal criteria. Mice were euthanized, and TA muscles were harvested 4 days post-injury. All procedures were approved by The Ohio State University’s Institutional Animal Care and Use Committee.
Publication 2024
Frequency of T cell subsets, macrophage subsets, and myeloid-derived suppressor cells (MDSC) in the peripheral blood was analyzed via flow cytometry. PBMCs procured from pre-therapy and cycle 4 day 1 blood draws were stained with anti-CD3-V450, anti-CD8-PE, anti-CD4-FITC, anti-Foxp3-PE, anti-CD56-APC, anti-CD16-APC Cy7, anti-NKG2C, anti-CD33-APC, anti-HLA-DR-PECy7, anti-CD11b-PE, anti-CD14-V450, anti-CD15-FITC, anti-CD80-FITC, anti-CD1630-PE, anti-CD206-APC and/or anti-CD69-PE-Texas Red (Beckman Coulter, Brea, CA). Immune cell subsets were defined as follows: CD4 T cells CD3+/CD4+, CD8 T cells CD3+/CD8+, Treg CD3+/CD4+/Foxp3+, M1 macrophages CD14+/CD80+/CD163/CD206+, M2 macrophages CD14+/CD80/CD163+/CD206+, NK cells CD3/CD56+/CD16+, granulocytic MDSC CD33+/HLA-DR/CD11b+/CD15+, monocytic MDSC CD33+/HLA-DR/CD11b+/CD14+. Data was acquired using a LSRII flow cytometer (BD Biosciences, San Jose, CA).
Publication 2019
Blood Cells CD4 Positive T Lymphocytes CD8-Positive T-Lymphocytes CD163 protein, human Cells Flow Cytometry Fluorescein-5-isothiocyanate galiximab Granulocyte HLA-DR Antigens ITGAM protein, human Macrophage Monocytes Muromonab-CD3 Myeloid-Derived Suppressor Cells Natural Killer Cells Phlebotomy T-Lymphocyte Subsets Therapeutics
The liposome formulation combined chloroform dissolved samples of cholesterol, DOPE, DSPC, DSPE-(PEO)4-cRGDfK, DSPE-mPEG2000, and DiO (6:6:6:1:1:0.5 molar ratio). The chloroform was evaporated under argon gas and the dried lipid film was hydrated with a solution containing 10 mg of human serum albumin in 1 ml phosphate buffer pH 7.4 for a minimum of 30 minutes. The solution was vortexed for 1 minute to remove any adhering lipid film and sonicated in a bath sonicator (ULTRAsonik 28X) for 1 minute at room temperature to produce multilamellar vesicles. These vesicles were then sonicated with a Ti-probe (Branson 450 sonifier) for 2 minutes to produce small unilamellar nanoliposomes filled with the cross-linked human serum albumin gel as indicated by the formation of a translucent solution. To reduce the size of the filled nanoliposomes, stepwise extrusion was carried out with the final step being extrusion through a polycarbonate filter with 200-nm pore size (Whatman). The nanoliposomes are then purified by size exclusion chromatography on sepharose CL-4B columns to remove free albumin. The final concentration, size distribution, and zeta potentials were the same as the empty nanoliposomes described in section 2.3.
Publication 2015
1,2-distearoylphosphatidylethanolamine Albumins Argon Bath Buffers Chloroform Cholesterol Lipids Liposomes Molar Molecular Sieve Chromatography Phosphates polycarbonate Sepharose CL 4B Serum Albumin, Human
Liposomes were prepared by thin-film hydration (TFH) method, followed by extrusion and by microfluidic technique with Nanoassemblr apparatus. They were formulated with 1,2 distearoyl-sn-glycero-3-phosphocholine (DSPC) and cholesterol (Chol) with a molar ratio of DSPC: Chol 50:50, since preliminary experiments showed this was the more stable composition (see Supplementary data Table S4) by both preparation methods tested. The liposome compositions showing the best properties in terms of stability and particle size were selected for further encapsulation of either bovine serum albumin (BSA) or myoglobin (Myo). These proteins were used as models of protein drugs, such as growth factors (i.e., hepatocyte growth factor), with different molecular weights.
Liposome preparation by TFH was carried out as follows. The stock solutions of DPSC (10 mg/mL) and cholesterol (10 mg/mL) were prepared by solubilizing the lipids with a mixture of chloroform:methanol 9:1 volume ratio. Different volumes were taken from each stock solution to achieve the exact lipid DSPC:Chol 50:50 molar ratio and placed inside the round-bottom flask. The organic solvent was evaporated under vacuum using Heidolph® Hei Vap Rotavapor (Heidolph, Germany) to obtain a homogeneous dry lipid film around the bottom flask wall. Then, the dry lipid film was rehydrated with phosphate-buffered saline (PBS) solution (pH 7.4) at 45 ± 3 °C to obtain placebo liposomes. Temperature set up (45 ± 3 °C) was chosen because it is close to DSPC (55 °C) and cholesterol (37 °C) main transition temperatures (Tm), making lipid chains more mobile and flexible. A round-bottom flask filled with PBS solution (1.3 mL) was vortexed (Advanced Vortex Mixer Zx3 Velp scientifica®, Italy) for 3 min at 24,000 rpm to suspend the lipid film and, subsequently, it was sonicated (SONICA® ultrasonic cleaner Soltec®, Milan, Italy) for 3 min at 36–37 kHz to reduce the liposome size. Subsequently, the round-bottom flask with liposomes suspension was left 30 min in a warm water bath at 45 ± 3 °C to improve hydration and promote liposome formation. Finally, liposome suspension was manually extruded under mild heating (35 ± 3 °C) to produce a homogenous population of unilamellar liposomes through three polyvinylidene difluoride (PVDF) filter membranes of 0.45 µm, 0.22 µm and 0.1 µm. A total of 10 extrusion cycles were performed for each filter. Each filter was connected to two 5 mL (Luer-lock) syringes. The same procedure described above was applied to obtain myoglobin- and BSA-loaded liposomes, but, during rehydration phase, Myo solution in PBS buffer (0.15 mg/mL) or BSA solution in PBS buffer (10 mg/mL) were added. Figure 8a schematizes the thin-film hydration methods followed for the preparation methods of BSA- and Myo-loaded liposomes.
NanoAssemblrTM platform provided by Precision NanoSystems Inc. (Vancouver, Canada), characterized by a staggered herringbone micromixer (SHM), was employed for the liposomes’ preparation by microfluidic technique [29 (link)]. Micromixer cartridge dimensions are 6.6 × 5.5 × 0.8 cm (w × d × h); it is made of polypropylene, viton and cyclic olefin copolymer. The cartridge’s mixing channel is 200 × 79 μm (w × h) and the herringbone structure is 31 μm high and 50 μm thick. There is an angle of 45 between the ridges and the long axis of the channel. The microfluidic device consists of a Y-junction, known as a staggered herringbone, followed by a staggered mixing region. The staggered herringbone structures induce rapid mixing by chaotic advection [30 (link)]. Channel 1 was loaded with PBS (pH 7.4) or BSA solution in PBS (10 mg/mL) or Myo solution in PBS buffer (0.15 mg/mL), while DSPC:Chol 50:50 ethanol solution was loaded in channel 2 (see Figure 8b). Both inlet streams are controlled by syringe pumps connected to a computer that controls the process [31 (link)]. Stock solutions of DSPC (4.3 mg/mL) and cholesterol (2 mg/mL) were prepared by solubilizing lipids in ethanol (3 mL). Liposome preparation was carried out using a total flow rate (TFR) of 8 mL/min and a flow rate ratio (FRR) of aqueous phase: lipids phase 3:1 for 1 mL total volume. The preparation process was performed at room temperature (25 ± 3 °C). Preliminary experiments moving TFR from 8 mL/min to 12 mL/min and FRR between 5:1 and 3:1 were carried out to set up the process parameters (see Supplementary data Table S4).
In order to increase BSA EE%, BSA loading into liposomes prepared by microfluidic technique was also investigated, modifying the formulation and process parameters as follows:

Liposomes’ lipid molar ratio was changed from 50:50 to DSPC:Chol 70:30;

Total flow rate (TFR) was increased from 8 mL/min to 12 mL/min;

Different amount of trehalose (10% w/w, 20% w/w and 40% w/w) were added to BSA aqueous solution to increase ionic strength.

BSA was solubilized in purified water (pH 7.4 adjusted by NaOH 0.1 M) to prevent its dimerization and maintain Mw 66 kDa.

The batches produced are reported in Table 4.
Publication 2022
1,2-distearoyllecithin Bath Bos taurus Chloroform Cholesterol Cycloalkenes Dimerization Epistropheus Ethanol Growth Factor Hepatocyte Growth Factor Homozygote Lipid A Lipids Liposomes Methanol Microchip Analytical Devices Molar Myoglobin Pharmaceutical Preparations Phosphates Placebos Polypropylenes polyvinylidene fluoride Proteins Rehydration Saline Solution Serum Albumin Serum Albumin, Bovine Solvents Syringes Tissue, Membrane Trehalose Ultrasonics Unilamellar Liposomes Vacuum
Liposomes were produced by the thin-film hydration technique, followed by subsequent agitation and freeze-thaw cycles18 (link),36 (link). A lipid mix containing Ph-DPPC, DOPC, DSPG, and cholesterol in a 3:3:2:3 molar ratio was dissolved in a solution of 90% chloroform and 10% methanol. This mixture underwent evaporation at low pressure, after which the lipid content was reconstituted in tert-butanol and subjected to freeze-drying. The resulting lipid cake was then rehydrated with different payloads, including Sulforhodamine B (SRho), tetrodotoxin (TTX), polyethylene glycol (PEG), and albumin–fluorescein isothiocyanate conjugate (FITC-Ab) in PBS buffer (pH=7.4) or bupivacaine hydrochloride (Bup), or doxorubicin hydrochloride (Dox) in saline. After 10 cycles of freezing and thawing, the liposomal solution was subjected to a 48-h dialysis against either PBS or water. The molecular mass cut-off of the dialysis devices was 1000 kDa (Spectra/Por™ Float-A-Lyzer™ G2 Dialysis Devices). The dialysis fluid was routinely replaced with fresh PBS or saline approximately every 12 h. To determine the drug and dye content across different formulations, liposomes were disrupted using octyl-β-D-glucopyranoside (100 mM, twice the volume of liposomal solution) and analyzed.
Publication 2023
1,2-oleoylphosphatidylcholine Albumins Buffers Bupivacaine Hydrochloride Chloroform Cholesterol Dialysis Dialysis Solutions distearoyl phosphatidylglycerol Fluorescein Fluorescein-5-isothiocyanate Hydrochloride, Doxorubicin isothiocyanate Lipids Liposomes lissamine rhodamine B Medical Devices Methanol Molar Pharmaceutical Preparations Polyethylene Glycols Pressure Saline Solution tert-Butyl Alcohol Tetrodotoxin
Heat shock competent cells (50 µL) were removed from the freezer and placed on ice for 20 min (4 tubes, 1
for each ligation product and 1 pYT354 plasmid control). Two µL of ligation products (and 1 µL of 10 ng/µL pYT354 plasmid) were added to each tube and placed on ice for an additional 20 min. Cells were then heat shocked for 45 s at 42 o C. Heat shocked cells were placed on ice for 2 min. One mL of super optimal broth (SOC) medium was added to each tube and the tubes were incubated at 37 o C, 200 rpm for 1 h. Cells were pelleted by centrifugation at 4000 g for 2 min at 4 o C. The supernatant was removed (950 µL) and the remaining culture was plated on LB agar containing ampicillin (100 µg/mL). Plates were incubated over night at 37 o C. Successful transformants were inoculated into LB containing ampicillin (100 µg/mL) and incubated overnight. Freezer stocks were made the following morning for triparental conjugation.
Publication 2022
The Caco-2 cell line was purchased from the Institute of Biochemistry and Cell Biology (SIBS, CAS, China). Caco-2 cell culture was performed using the method reported by Fernández et al. [32 (link)], with modifications. Briefly, Caco-2 cells were inoculated into Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum and 1% (v/v) antibiotics (100 U/mL penicillin sodium, 1.0 μg/mL streptomycin) and incubated at 37°C (5% CO2). The culture medium was replaced the next day. After 15–18 days, the Caco-2 cells were transferred into six-well plates and grown to confluence. A monolayer of Caco-2 cells (5 × 105 CFU/mL/cm2) was used for adhesion assays after washing twice with PBS (pH 7.2).
The wild-type and mutant strains of KLDS1.0391 were routinely grown overnight in MRS broth. Then, the cells were collected (8000 ×g, 4°C, 10 min), washed twice with PBS (pH 7.2), and resuspended in DMEM to the final concentration of 108 CFU/mL. The suspensions (5 mL) were added to the abovementioned wells containing Caco-2 cells and incubated for 2 h (37°C, 5% CO2). At the end of the incubation period, the Caco-2 cell cultures were washed twice with prewarmed PBS (37°C, pH 7.2) to remove the nonadherent cells and then treated with Triton X100 (1%, 10 min) to release the adhered bacterial cells. The adhesion ratio of bacterial cells was calculated by comparing the viable count on MRS agar plates before and after adhesion.
Publication 2018
Agar Antibiotics Bacteria Bacterial Adhesion Biological Assay Caco-2 Cells Cell Lines Fetal Bovine Serum Penicillins Sodium Strains Streptomycin Triton X-100
The Caco-2 cell line, known as ATCC HTB-37 (Rio de Janeiro Cell Bank, Rio de Janeiro, Brazil), was routinely cultured (29-31 days) in Dulbecco's modified Eagle's minimum medium (DMEM) (Sigma-Aldrich), supplemented with 20 % (v/v) heat-inactivated fetal bovine serum (Cultilab, Campinas, Brazil), a mixture of penicillin (100 IU/ml) and streptomycin (100 μg/ml) solution (Sigma-Aldrich), and 1 % (v/v) non-essential amino acid solution (Sigma-Aldrich) at 37 °C in an atmosphere of 5 % CO 2 and 95 % air. The adhesion assay was performed as described by Argyri et al. (2013) , with modifications. Caco-2 cells were seeded at a concentration of 10 5 cells per well into 24-well tissue culture plates (NEST) and incubated at 37 °C in an atmosphere of 5 % CO 2 and 95 % air (Thermo Fisher Scientific, Waltham, MA, USA) until a confluent monolayer was formed (15-17 days). One day before the performance of the adhesion assays, the medium was replaced, but without antibiotics. Before adhesion, the monolayer was washed once with phosphate-buffered saline (PBS, pH 7.4) to remove all traces of the medium. The L. mesenteroides subsp. mesenteroides SJRP55 was grown overnight, until it reached the stationary phase in MRS at 30 °C, and it was then washed twice with sterile PBS. Subsequently, approximately 10 8 colony forming units (CFU)/ml was transferred to post-confluent monolayers of Caco-2 cells in the 24-well tissue culture plates and incubated at 37 °C in 5 % CO 2 95 % air atmosphere for 2 h. Cells were then washed at least three times with PBS, in order to remove both the non-adherent bacteria and the cells with adherent bacteria from each well with the addition of 1 ml of Triton X-100 (0.5 %, v/v) (Sigma-Aldrich). The suspension (1 ml) from each well was then transferred to a tube containing 9 ml of sterile saline, serially diluted, and plated on MRS agar in duplicate, in order to determine adhesion ability. Adherence (expressed as a percentage) was calculated using the ratio of the number of bacterial cells that remained attached to the total number of bacterial cells added initially to each well. The experiment was performed in triplicate on two separate occasions.
Publication 2014
Caco-2 cells were cultured in DMEM supplemented with 10% heat-inactivated fetal bovine serum, 1% nonessential amino acid and 1% penicillin-streptomycin solution at 37°C in 5% CO 2 incubator, as previously described (Li et al., 2014) (link). The culture medium was changed every second day and regularly checked to ensure normal morphology of cells. For the adhesion assay, Caco-2 cells were seeded at 1.0 × 10 6 cells/well in 6-well culture plates (Corning Inc., Corning, NY), then 1 mL of the L. gasseri suspension (1.0 × 10 8 cfu/mL) was added to each well, which was incubated at 37°C in a 5% CO 2 atmosphere. After 2 h of incubation, the monolayers were washed thrice with 1 mL of PBS (pH 7.4) and nonadherent cells were removed. Subsequently, the Caco-2 cells adhered by L. gasseri were digested with trypsin-EDTA and serially diluted. The number of viable cell-adhered L. gasseri was assessed by plating on MRS agar.
Publication 2020
Caco-2 cell was obtained from Shanghai Zhongqiaoxinzhou Biotechnology Co.,Ltd.(Shanghai, China). Cells were cultured in DMEM (Basalmedia, Shanghai, China) supplemented with 20% (v/v) fetal bovine serum (FBS, NEWZERUM, China) and 5% antibiotics (penicillin and streptomycin, Basalmedia, Shanghai, China). Caco-2 cells were seeded at a concentration of 5 × 104 cells per well in 24-well or 96-well cell culture plates to obtain a monolayer of differentiated cells.
The adhesion and invasion experiments refer to the previous studies with minor modifications (Schierack et al., 2011 (link)). After incubating A.hydrophila with EGCG for 24 hours, the mixture was centrifuged and resuspended in DMEM to adjust the bacterial concentration to 1×108 CFU/mL. The bacterial suspension was then added to a monolayer of differentiated Caco-2 cells and incubated for 1 hour. In the adhesion experiments, the culture medium was discarded, and the cells were washed three times with PBS (pH 7.4) before adding 0.25% trypsin for digestion. After gradient dilution, bacterial counting was performed. In the invasion experiments, the culture medium was discarded, and 10% antibiotics (penicillin and streptomycin) were added for 1 hour before bacterial counting was performed. LDH release was tested according to the manufacturer’s instructions (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China).
Publication 2023
Antibiotics Bacteria Caco-2 Cells Cell Culture Techniques Cells Culture Media Digestion epigallocatechin gallate Penicillins Streptomycin Technique, Dilution Trypsin
Human colorectal cancer cell lines (HCT 116 and RKO) were acquired from the Cell Bank of the Shanghai Institute of Cell Biology. The HCT 116 cells were cultured in McCoy’s 5A medium (Gibco, Carlsbad, CA, USA) with 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin mixture added to the above medium. The RKO cells were maintained in MEM with NEAA (Meilun Biotechnology, Dalian, China) supplemented with 10% FBS and 1% penicillin–streptomycin mixture. The cells were cultured in a 37 °C incubator (Thermo Fisher, Waltham, MA, USA) with 5% CO2. The synthesis of small interfering RNA (siRNA) was entrusted to GenePharma (Shanghai, China). The cells were placed into 6-well plates (1.5 × 105 cells/well), and the siNC group was transfected with the scrambled siRNA (negative control), and the siTMED1 group was transfected with the siRNA specific for TMED1 (Table S1). All siRNA transfections were performed with GP-transfect-Mate at a 75 nM final concentration following the manufacturer’s protocol. The transfected cells were collected for the following assays 48 h after transfection. Gene knockdown efficiency was determined by real-time quantitative PCR.
Publication 2024
Not available on PMC !
LOX activity was assayed by monitoring the increase in the absorbance at 234 nm by the change of the cis,cis-1,4 pentadiene of linoleic acid into the conjugated hydroperoxyldiaene derivative cis-trans- (2) . The unit of the enzyme defines as the amount of enzyme that converts one µmol of the substrate (linoleic) to the product in a min (35) .
Publication 2022
Total genomic DNAs of each population were extracted with a modified cetyltrimethylammonium bromide (CTAB) protocol (Doyle and Doyle., 1987 ). To confirm the species, rbcL genes were determined before genome sequencing. The rbcL marker, which encodes the ribulose-1,5-bisphosphate carboxylase large subunit, is the most widely used in species identification in the red algae (Saunders and Moore, 2013 (link)). The rbcL primers were F57 and rbcLrevNew described in Freshwater and Rueness (1994) (link) and Saunders and Moore (2013) (link). Amplified DNAs were purified using the LaboPass™ PCR Purification Kit (COSMO Genetech, Seoul, Republic of Korea). PCR products were sent for sequencing to Macrogen (Seoul, Republic of Korea). After a quality check, the forward and reverse sequences were merged using Geneious prime 2020.0.4 (https://www.geneious.com). For phylogenetic analysis, published rbcL sequences of Rhodymeniophycidae and Ahnfeltia were obtained from NCBI (see accession number of each taxon in Supplementary Table S1), and alignments for all sequences were conducted with MAFFT v7.310 (Katoh and Standley, 2013 (link)). A maximum likelihood (ML) phylogenetic tree was reconstructed by IQ-TREE v.1.6.8 with 1,000 ultrafast bootstrap replications (-bb 1000) and model test option (-m TEST) (Nguyen et al., 2015 (link)).
Publication 2021
Cetrimonium Bromide DNA DNA Replication Genes Genome Oligonucleotide Primers Protein Subunits Rhodophyta ribulose Sequence Alignment Trees
Neural differentiation medium: DMEM/F12, 2% serum replacement, 1% N2 supplement, 1× Glutamax, 50 unit/ml penicillin-streptomycin, 80 ng/mL Noggin, 10 μ M SB431542, 2 μ M dorsomorphin (instead combination of 10 μ M SB431542, 2 μ M dorsomorphin and 0.5 μ M LDN-193189 can be used).
Neuronal progenitor medium: DMEM/F12, 2% B27 supplement, 1× Glutamax, 50 unit/mL penicillin-streptomycin, 100 ng/mL Shh, 100 ng/mL FGF8 and 2 μ M purmorphamine.
NB maturation medium: Neurobasal, 2% B27 supplement, 1× Glutamax, 50 unit/mL penicillin-streptomycin, 20 ng/mL BDNF, 20 ng/mL GDNF, 200 μ M ascorbic acid and 4 μ M forskolin.
NBA maturation medium: Neurobasal-A, 2% B27 supplement, 1× Glutamax, 50 unit/mL penicillin-streptomycin, 20 ng/mL BDNF, 20 ng/mL GDNF, 200 μ M ascorbic acid and 10 μ M forskolin.
Publication 2023
4-(5-benzo(1,3)dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)benzamide 4-methoxy-N-butylamphetamine Ascorbic Acid Colforsin Dietary Supplements dorsomorphin FGF8 protein, human GDNF protein, human LDN 193189 Nervousness Neurons noggin protein Penicillins purmorphamine Serum Streptomycin
ReN cell VM human neural progenitor cells (NPCs) were purchased from EMD Millipore (Billerica, MA, USA). The cells were plated onto BD Matrigel® (BD Biosciences, San Jose, CA, USA) coated T25 cell culture flasks (BD Biosciences, San Jose, CA, USA) and maintained in DMEM/F12 (Life Technologies, Grand Island, NY, USA) media supplemented with 2 mg heparin (StemCell Technologies, Vancouver, Canada), 2% (v/v) B27 neural supplement (Life Technologies, Grand Island, NY, USA), 20 mg EGF (Sigma-Aldrich, St Louis, MO, USA), 20 mg bFGF (Stemgent, Cambridge, MA, USA) and 1% (v/v) penicillin/streptomycin/amphotericin-B solution (Lonza, Hopkinton, MA, USA) in a CO2 cell culture incubator. Cell culture media were changed every 3 days until cells were confluent. For 2D neuron/astrocyte differentiation, the cells were plated onto Matrigel®-coated microfluidic devices with DMEM/F12 differentiation media supplemented with 2 mg heparin, 2% (v/v) B27 neural supplement, and 1% (v/v) penicillin/streptomycin/amphotericin-B solution without growth factors. One half volume of the differentiation media was changed every 3–4 days for 3–9 weeks. LPS-RS was purchased from InvivoGen (San Diego, CA). LPS-RS was resuspended in dimethylsulphoxide (DMSO; 0.1 mg in 10 ml) followed by 1:10 dilution in differentiation culture medium. This stock solution of 22.2 mM was then diluted for cell treatment experiments.
Publication 2018
Amphotericin Amphotericin B Astrocytes Cell Culture Techniques Cells Culture Media Dietary Supplements Fifth Cranial Nerves Growth Factor Heparin Homo sapiens matrigel Microchip Analytical Devices Neural Stem Cells Neurons Penicillins Penicillin V phenethicillin Stem Cells Streptomycin Sulfoxide, Dimethyl Technique, Dilution Therapies, Investigational
For immunophenotyping using flow cytometry, cells were harvested, centrifuged for 10 min at 400× g, washed once with DPBS/EDTA containing 0.05% bovine serum albumin (BSA, Sigma-Aldrich), and stained with different panels of monoclonal antibodies. In this study, CD4+ T cells were stained with mouse anti-chicken CD4-phycoerythrin (PE) or CD4-SpectralRed (SPRD) (CT-4, SouthernBiotech, Birmingham, Alabama, USA). CD8+ T cells were labeled with CD8-allophycocyanin (APC) (CT-8, ThermoFisher Scientific). In addition, human anti-chicken CD25-FITC (AbD13504, Bio-Rad Laboratories, Hercules, California, USA) and CD28-PE (AV7, SouthernBiotech) were used to investigate the activation of CD4+ T-helper cells (CD4+CD25+, CD4+CD28+) and CD8+ cytotoxic T cells (CD8+CD25+, CD8+CD28+). All CD3+ T cells were labeled with the pan-T-cell marker CD3-APC (CT-3, ThermoFisher Scientific). Bu-1+ B cells were stained with Bu-1-FITC (AV20, ThermoFisher Scientific). As a viability marker, 1.5 µL of 1 mg/mL 4′, 6-diamino-2-phenylindole (DAPI, ThermoFisher Scientific) was used. 20,000 vital PBMCs were recorded on a BD FACSCanto™ II (Becton, Dickinson and Company, Franklin Lakes, NJ, USA) flow cytometer.
Publication 2023
allophycocyanin B-Lymphocytes CD3 Antigens CD4 Positive T Lymphocytes CD8-Positive T-Lymphocytes Cells Chickens Cytotoxic T-Lymphocytes DAPI Edetic Acid Flow Cytometry Fluorescein-5-isothiocyanate Helper-Inducer T-Lymphocyte Homo sapiens IL2RA protein, human Monoclonal Antibodies Mus Phycoerythrin Serum Albumin, Bovine T-Lymphocyte
ReNcell VM human neural precursor (ReN) cells were purchased from EMD Millipore (Billerica, MA, USA). The cells were plated onto BD Matrigel (BD Biosciences, San Jose, CA, USA)-coated T25 cell culture flask (BD Biosciences, San Jose, CA, USA) and maintained in DMEM/F12 (Life Technologies, Grand Island, NY, USA) media supplemented with 2 μg/ml Heparin (Stemcell Technologies, Vancouver, Canada), 2% (v/v) B27 neural supplement (Life Technologies, Grand Island, NY, USA), 20 μg/ml EGF (Sigma-Aldrich, St. Louis, MO, USA), 20 μg/ml bFGF (Stemgent, Cambridge, MA, USA), and 1% (v/v) Penicillin/Streptomycin/Amphotericin b solution (Lonza, Hopkinton, MA, USA) in CO2 cell culture incubator. The cell culture media were changed every 3 days until the cells were confluent. For 2D neuronal/glial differentiation, the cells were plated onto either Matrigel-coated 24-well or 6-well plates with DMEM/F12 differentiation media supplemented with 2 μg/ml Heparin, 2% (v/v) B27 neural supplement, and 1% (v/v) Penicillin/Streptomycin/Amphotericin b solution without growth factors. A half volume of the differentiation media was changed every 3 days for 3–7 weeks. DAPT, Compound E and BACE inhibitor IV were purchased from EMD Millipore, N-Lauroylsarcosine (Sarkosyl) from Sigma-Aldrich. Hematoxylin QS from Vector Laboratories (Burlingame, CA, USA), and Amylo-Glo from Biosensis (Thebarton, Australia). SGSM41 is an aminothiazole-bridged heterocycle-containing soluble γ-secretase modulator (SGSM) similar in structure to those published recently19 (link). SGSM 41 has the typical characteristics of this series of SGSM molecules that potently inhibit the production of Aβ42 and to a lesser degree Aβ40 while concomitantly increasing the generation of shorter Aβ peptide species such as Aβ38 and Aβ37. The structures and the detailed properties are included in Extended Data Table 1.
Publication 2014
1,2-dilinolenoyl-3-(4-aminobutyryl)propane-1,2,3-triol Amphotericin Amphotericin B Cardiac Arrest Cell Culture Techniques Cells Cloning Vectors Cortisone Culture Media Dietary Supplements Fifth Cranial Nerves Growth Factor Hematoxylin Heparin Homo sapiens matrigel N-lauroylsarcosine Nervousness Neuroglia Neurons Penicillins Penicillin V Peptides phenethicillin Secretase sodium lauroyl sarcosinate Stem Cells Streptomycin
Methanol (>99.9% pure) and dipotassium hydrogen phosphate (anhydrous) were purchased from Fisher Chemical (Fair Lawn, NJ, USA). Sodium 2,2-dimethyl- 2-silapentane-5-sulfonate (DSS-d6, 98%) and deuterium oxide (D2O, 98%) were purchased from Cambridge Isotope Laboratories, Inc. (Tewksbury, MA, USA).
Publication 2020
Alkanesulfonates Deuterium Oxide Isotopes Methanol potassium phosphate, dibasic Sodium
Use of 1H NMR spectra alone cannot directly determine the differences in metabolites between the two groups. Therefore, we performed principal component analysis (PCA) analysis to identify differences and similarities in serum metabolism. Data were analyzed using SIMCA software (version 14.1; MKS Data Analytics Solutions, Umea, Sweden) for multivariate statistical analysis. Metabolite signals in the 1H NMR serum profiles were first examined by unsupervised PCA, which reduced the dimensionality of the data and summarized the similarities and differences between the two groups using score plots. The interpretability and predictability of the model (R2X; presents the interpretation rate of X variable and predictive Q2) were calculated and evaluated.
Subsequently, to explore the specific discriminant information between the two groups, orthogonal partial least squares discriminant analysis (OPLS‐DA) was performed to filter out orthogonal variables in metabolites that were not associated with categorical variables and to analyze nonorthogonal and orthogonal variables separately.19, 20 To evaluate the reliability of the OPLS‐DA model, we performed 200 random permutations. The statistically significant metabolites were then analyzed and summarized by calculating the corresponding correlation coefficients, as previously described.21 Briefly, we multiplied the loading value with the square root of its SD, which was compared with the Corresponding Correlation Coefficient Critical Value Table. The metabolites that had significantly different levels between the groups were identified.
Publication 2020
1H NMR Metabolism Plant Roots Serum
LC‒MS/MS analysis was supported by Jingjie PTM BioLabs (Hangzhou, China). In brief, the tryptic peptides were dissolved in solvent A (0.1% formic acid, 2% acetonitrile/in water) and separated by a homemade reversed-phase analytical column (25 cm length, 100 μm i.d.) on a nanoElute UHPLC system (Bruker Daltonics). After being subjected to a capillary source, the peptides were subjected to timsTOF Pro (Bruker Daltonics) mass spectrometry for further analysis in parallel accumulation serial fragmentation (PASEF) mode. The MS/MS data were processed by the MaxQuant search engine (v.1.6.6.0). Tandem mass spectra were searched using the human SwissProt database (20,366 entries) and reverse decoy database. FDR < 1%. The relative quantitative values of modified peptides in different samples were obtained by centralizing the signal intensity values in different samples. After filtering lysine lactylation sites (localization probability > 0.75), the relative quantitative values of each sample were obtained by two experiments. All the ratios of quantified lysine lactylation peptides were normalized according to their corresponding protein expression levels. The protein pathways were annotated using the KEGG database. The STRING database was used to identify protein–protein interactions of the DELPs.
Publication 2023
acetonitrile Capillaries formic acid Homo sapiens Lysine Mass Spectrometry Peptides Proteins Solvents Tandem Mass Spectrometry Trypsin
Not available on PMC !
A live cell, high-throughput, microplate-based ROS assay that utilized the cell permeable substrate, 2',7'-dichlorofluorescin diacetate, (DCF-DA) a reliable fluorogenic marker for ROS detection was used as previously described [10] . Upon ROS generation, the highly fluorescent dye, 2',7'-dichlorofluorescein is produced, with EX (excitation): 495 nm and EM (emission): 530 nm. In brief, DLD-1.ApoL6 cells were plated in 96-well tissue culture plates at a density of 100,000 cells/well and then loaded with DCF-DA at 20 μM final concentration for 30 min before treating with the indicated compounds at 2, 10 and 50 μM in D.0 for 8 hrs. Both end point and area-scan readings were taken using the Synergy H4 Plate Reader at 485 nm with emission at 528 nm using a 20 nm bandwidth, according to the manufacturer's instruction.
Publication 2020
Cells were plated at a density of 1×104 cells in 6 replicates in 96-well dish. Twelve hours later, the cells were incubated with a cocktail of 5 μM concentration of CellROX Deep Red Reagent (C10422, Invitrogen) and 2 μg/ml Hoechst 33342(Invitrogen) for 45 minutes at 37°C. Images were captured using a Zeiss fluorescent microscope and Axiovision Image 4.5 software. Quantification of the percent ROS was obtained using a high-throughput immunofluorescence plate reader (Celigo)40 (link).
Publication 2014
Cells Fluorescent Antibody Technique HOE 33342 Hyperostosis, Diffuse Idiopathic Skeletal Microscopy
TEM was performed as described previously with some modifications (Yuan et al., 2013 (link)). Briefly, small pieces of WT and miR-dKO testes were fixed in 0.1 M cacodylate buffer (pH 7.4) containing 3% paraformaldehyde and 3% glutaraldehyde plus 0.2% picric acid for 2 h in 4°C, then for 1 h at RT. Following washes with 0.1 M cacodylate buffer, the samples were post-fixed with 1% OsO4 for 1 h at RT. Dehydration was performed using 30%, 50%, 70%, 90% and 100% ethanol solutions sequentially, followed by infiltration of propylene oxide and Eponate with BDMA overnight at RT. After infiltration, samples were embedded in Eponate mixture (Electron Microscopy Sciences, Hatfield, PA, USA) and polymerized at 60°C for 24 h. Ultrathin sections (60–70 nm in thickness) were cut with a diamond knife using an ultra-microtome (Leica). The sections were collected on collodion covered electron microscope nickel grids and stained with uranyl acetate and lead citrate. The ultrastructure of the samples was observed and photographed using a transmission electron microscope (Phillips CM10) at 80 kV.
Publication 2015
1-naphthol-8-amino-3,6-disulfonic acid Buffers Cacodylate Citrate Collodion Dehydration Diamond Electron Microscopy Ethanol Glutaral Microtomy N-benzyl-N,N-dimethylamine Nickel paraform propylene oxide Testis Transmission Electron Microscopy uranyl acetate
JTP-74057, a highly specific and potent MEK1/2 inhibitor [35 (link)], and GDC-0994, a highly selective ERK1/2 inhibitor [36 (link)], were used to prevent the activation of the MEK/ERK pathway. Cells were treated with 1 nmol/L JTP-74057 or GDC-0994 in a humidified incubator for 24 h at 37 °C with 5% CO2.
Doxorubicin is a chemotherapy medication used to treat cancer. Cells were treated with 5 μmol/L doxorubicin for 24 h, followed by the cell counting kit-8 (CCK-8) assay and western blotting.
Publication 2021
Biological Assay Cells Doxorubicin GDC-0994 JTP 74057 Malignant Neoplasms MAP2K1 protein, human MEK-ERK Pathway Mitogen-Activated Protein Kinase 3 Pharmaceutical Preparations Pharmacotherapy
The following antibodies were used for immunofluorescence (IF) and immunoblotting (IB): mouse antibodies against ACTB/β-ACTIN (Sigma-Aldrich, A5441; IB, 1:10,000), DDIT3 (Cell Signaling Technology, 2895; IB, 1:1,000), NFKBIA (Cell Signaling Technology, 4814; IB, 1:1,000), VPS37A (Santa Cruz Biotechnology, sc-376978; IB, 1:100); rabbit antibodies against ATF3 (Cell Signaling Technology, 18665; IB, 1:1,000), ATF4 (Cell Signaling Technology, 11815; IB, 1:1,000), ATG5 (Cell Signaling Technology, 12994; IB, 1:1,000), ATG7 (Cell Signaling Technology, 8558; IB, 1:1,000), BCL2L1 (Cell Signaling Technology, 2762; IB, 1:1,000), HSPA5/BiP (Cell Signaling Technology, 3177; IB, 1:1,000), CHMP2A (Proteintech, 10477-1-AP; IB, 1:1,000), cleaved CASP3 (Cell Signaling Technology, 9661; IB, 1:1,000), cleaved CASP8 (Cell Signaling Technology, 9496; IB, 1:1,000), CASP9 (Cell Signaling Technology, 9502; IB, 1:1,000), TNFRSF10B/DR5 (Cell Signaling Technology, 8074; IB, 1:1,000), GAPDH (Proteintech, 60004-1-lg; IB, 1:50,000), HRK (Novus, NBP1-76414; IB, 1:400), LMNB1 (Proteintech, 12987-1-AP; IB, 1:5,000), MAP1LC3B (Novus, NB100-2220; IB, 1:5,000) (Cell Signaling Technology, 3868; IF, 1:200), PARP (Cell Signaling Technology, 9542; IB, 1:1,000), PMAIP1 (Cell Signaling Technology, 14766; IB, 1:1,000), CASP8 (Abcam, ab108333; IB, 1:1,000), RELA (Cell Signaling Technology, 8242; IB, 1:1,000), TRIB3 (Proteintech, 13300-1-AP; IB, 1:1,000); guinea pig antibody against SQSTM1/p62 (American Research Products, 03-GP62-C; IB, 1:4,000; IF, 1:400). SMARTvector Inducible Lentiviral shRNAs and ON-TARGETplus SMART Pool siRNAs listed in Table S7 were purchased from Horizon Discovery. sgCASP8-Cas9-2A-GFP plasmid was purchased from Santa Cruz Biotechnology (sc-400147). epiCRISPR (135960) [52 (link)], LentiCRISPR v2 (52961) [53 (link)], LRG (Lenti_sgRNA_EFS_GFP, 65656) [54 (link)] and pSI-Check2-hRluc-NFkB-firefly (106979) [55 (link)] were obtained from Yongming Wang, Feng Zhang, Christopher Vakoc and Qing Deng, respectively, through Addgene. For CRISPR-Cas9-mediated gene silencing, each gRNA listed in Table S5 was subcloned into the BsmB1 site of lentiCRISPRv2 and the Sap1 site of epiCRISPR. LRG plasmids encoding three distinct VPS37A gRNAs (LRG-gVPS37As) were generated as previously described [20 (link)] and used to generate ATG7 VPS37A and CASP8 VPS37A DKO cells. pCDH1-myc-CASP8(C360A)-KN151-SV40-hygro and pCDH1-HA-CASP8(C360A)-LC151-SV40-hygro were previously described [19 (link)]. All other reagents were obtained from the following sources: 5Z-7-oxozeaenol (MedChemExpress, HY-12686); 7-amino-actinomycin D (7-AAD; BioLegend, 420404); APC Annexin-V (BioLegend, 640941); BMS-345541 (Sigma-Aldrich, B9935); bovine serum albumin (BSA; EMD Millipore, 126575); Caspase-3/7 Apoptosis Assay Reagent (Essen Biosciences, 4704); Caspase-Glo 3 Assay kit (Promega, G8090); Caspase-Glo 8 Assay kit (Promega, G8200); deoxycholate (Sigma-Aldrich, D6750); digitonin (Sigma-Aldrich, 11024-24-1); dimethyl sulfoxide (DMSO; Sigma-Aldrich, D2438); iTaq Universal SYBR Green Supermix (BIO-RAD, 1725121); Dual-Glo Luciferase Assay System (Promega, E2920); Dulbecco’s Modification of Eagle’s Medium (DMEM; CORNING, 10–013-CV); GSK2656157 (MedChem Express, HY-13820); Incucyte Caspase-3/7 Red Dye (Essen Biosciences, 4704); jetPRIME (Polyplus, 101000015); Lipofectamine RNAiMAX (Thermo Fisher Scientific, 13778075); NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Fisher Scientific, 78833); normal goat serum (Sigma-Aldrich, G9023); paraformaldehyde (Electron Microscopy Sciences, 15710); Phosphatase Inhibitor Cocktail 2/3 (Sigma-Aldrich, P5726/P0044); Protease Inhibitor Cocktail (Sigma-Aldrich, P8340); QuantSeq 3’ mRNA-Seq Library Prep Kit (Lexogen, 015); QuantiTect Reverse Transcription Kit (Qiagen, 205311); RNeasy Plus Mini Kit (Qiagen, 74134); Thapsigargin (Sigma-Aldrich, T9033); Triton X-100 (Sigma-Aldrich, T8532); tunicamycin (Sigma-Aldrich, T7765); YOYO-3 iodide (Thermo Fisher Scientific, Y3606).
Publication 2023
Cells were plated in a 24 well plate (Corning, Corning, NY) and were allowed to adhere over night, at 37°C and 5% CO2. The following day, apoptosis was induced by several methods: Staurosporine- cells were incubated with 0.5 µM Staurosporine (Sigma-Aldrich) for 1.5 h and then washed with fresh media. γ-Irradiation- cells were irradiated (10Gy in GammaCell 220 Excel, MDS Nordion). Peroxide treatment- medium was aspirated and cells were washed with PBS with Ca2+ and Mg2+ (Biological Industries). Cells were incubated with H2O2 (30%, Sigma, Steinheim, Germany) diluted in PBS to a final concentration of 200 µM, at 37°C. After 1 h, cells were washed and fresh medium was added. Anti-FAS mAb- cells were treated with 1 µg/ml of anti-human FAS mAb (MBL, Naka-ku Nagoya, Japan). The number of apoptotic cells in the cultures was determined at different time points following apoptosis induction using Annexin/PI kit (MBL) according to manufacturer instructions.
Publication 2013
Annexins Apoptosis Biopharmaceuticals Cells Homo sapiens Peroxide, Hydrogen Peroxides Staurosporine
Liver specimens from mice that were injected i.v. with the green lipophilic dye PKH67 (Sigma, St Louis, MO, USA)-labelled RL-male-1 cells (5 × 10 6 ) and either LAD-1 (200 µg) or control rat IgG (200 µg) were fixed in periodate-lysine-paraformaldehyde fixative, frozen, and sections cut (8 µm) with a cryostat as described previously [17] . The sections were blocked with phosphate buffered saline (PBS) containing 5% bovine serum albumin, and incubated for 6 h at 4°C with biotinylated anti-CD80 mAb and for 30 min at room temperature with Texas red-avidin (BD Pharmingen). A biotinylated hamster anti-KLH mAb (BD Pharmingen) was used as a negative control. The sections were examined by fluorescence microscopy (Olympus, Tokyo, Japan). Peritoneal exudate cells (PEC), obtained by rinsing the peritoneal cavity with PBS, were cultured for 3 h with PKH67labelled RL-male-1 cells at a ratio of 1 : 1 in the presence of either LAD-1 or control M17/4 or 30-H12 IgG (10 µg each). The cells were then harvested and labelled with anti-CD80 mAb, as described above. In some experiments, cells were labelled with PE-anti-CD80 mAb and analysed with a confocal microscope as described previously [18] as well as by flow cytometry.
Publication 2005
Chromogenic immunohistochemistry was performed mainly using the automated Ventana Discovery ULTRA (Roche). The slides were steamed for 32 min in CC1 buffer (950-124, Roche) and incubated in the corresponding primary antibody at room temperature for 36 min. Anti-MTCO1 antibody was used at 1:8000 dilution (ab14705, abcam, Cambridge, MA) and Anti-COX IV antibody was used at 1:8000 dilution (3E11, 4850, Cell Signaling, Danvers, MA) as primary antibodies. The slides were developed using Discovery HQ HRP hapten-linked multimer detection kit (Roche).
Page 10
Publication 2019
IHC was performed on 4 µm formalin-fixed, paraffin-embedded tissue sections using BRG1 (SMARCA4), AR, FOXA1, ERG and C-Myc. The IHC process was performed on the Ventana ULTRA automated slide staining system using the Omnimap and Ultraview Universal DAB detection kit. The antibody details are provided in SI Appendix, Table S1. The following commercial kits from Roche-Ventana Medical System were used: Discovery CC1 (Cat No. 950-500), Discovery CC2 (Cat No. 950-123), OptiView Universal DAB Detection Kit (Cat No. 760-700), and OmniMap Universal DAB Detection Kit (Cat No. 760-149).
Publication 2024

Frequently Asked Questions (FAQ)

About annotations

We welcome your feedback to improve our services. Use the voting system to indicate the usefulness of an annotation. For additional feedback or to report issues, please contact us through our support page or email us at contact@pubcompare.ai.

Discover the latest AI and user-annotated protocols below, contribute your own annotations, and help enhance the reproducibility of research worldwide!