The largest database of trusted experimental protocols
> Chemicals & Drugs > Amino Acid > Calreticulin, human

Calreticulin, human

Calreticulin is a multifunctional protein found in the lumen of the endoplasmic reticulum.
It plays a key role in calcium homeostasis, protein folding, and quality control.
Calreticulin is involved in a variety of cellular processes, including cell adhesion, phagocytic clearance of apoptotic cells, and immune response modulation.
Researchers can leverage PubCompare.ai's AI-driven platform to streamline Calreticulin research, effortlessly locating protocols from literature, pre-prints, and patents, while utilizing AI-powered comparisons to identify the optimal protocols and products.
This innovative solution can help accelerate Calreticulin-related discoveries and advance the field of cell biology.

Most cited protocols related to «Calreticulin, human»

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.
Publication 2008
mTagBFP2 fluorescent protein expression vectors were constructed using -C1 and -N1 (Clontech-style) cloning vectors. The mTagBFP2 cDNA was amplified with a 5′ primer encoding an AgeI site and a 3′ primer encoding either a BspEI (-C1) or NotI (-N1) site for C-terminal and N-terminal fusions (with regards to the FP), respectively. Purified and digested PCR products were ligated into similarly digested pEGFP-C1 and pEGFP-N1 cloning vector backbones. To generate targeting fusion vectors, the appropriate cloning vector and a previously assembled EGFP fusion vector were digested, either sequentially or doubly, with the appropriate enzymes and ligated together after gel purification.
Thus, to prepare mTagBFP2 C-terminal fusions (number of linker amino acids in parenthesis), the following digests were performed: human lamin B1 (10), NheI and BglII (lamin B1 cDNA source: George Patterson, NIH; NM_005573.2); 20 amino acid farnesylation signal from c-Ha-Ras (CAAX; 5), AgeI and BspEI (c-Ha-Ras cDNA source: Clontech, Mountain View, CA; NM_001130442.1); endoplasmic reticulum (5), AgeI and BspEI (calreticulin cDNA source: George Patterson, NIH; NM_004343.3); fibrillarin (7), AgeI and BspEI (fibrillarin cDNA source: Evrogen, Moscow, Russia; NM_001436.3); human light chain clathrin (15), NheI and BglII (clathrin cDNA source: George Patterson, NIH; NM_001834.2); β-actin (7), NheI and BglII (human β-actin cDNA source: Clontech, Mountain View, CA; NM_001101.3); caveolin 1 (10), NheI and BglII (human caveolin 1 cDNA source: Origene, Rockville, MD; NM_001753); vinculin (22) AgeI and EcoRI (human vinculin source: Clare Waterman, NIH; NM_003373.3); CAF1 (10), AgeI and BspEI (mouse chromatin assembly factor cDNA source: Akash Gunjan, FSU; NM_013733.3) Rab5a (7), NheI and BglII (canine Rab5a cDNA source: Vicki Allen, University of Manchester; NM_001003317.1); α-tubulin (18), NheI and BglII (human α-tubulin cDNA source: Clontech, Mountain View, CA; NM_006082); myosin IIA (18) NheI and BglII (human myosin heavy chain IIA cDNA source: DNA2.0, Menlo Park, CA; AJ312390.1); PCNA (19), AgeI and BspEI (proliferating cell nuclear antigen cDNA source: David Gilbert, FSU; NM_002592.2).
To prepare mTagBFP2 N-terminal fusions (number of linker amino acids in parenthesis), the following digests were performed: β-2 connexin-26 (7), BamHI and NotI (rat Cx26 cDNA source: Matthias Falk, Lehigh U; NM_001004099.1); TfR (20), BamHI and NotI (transferrin receptor cDNA source: George Patterson, NIH; NM_NM_003234); Golgi complex (7), BamHI and NotI (human β-galactosamide α-2,6-sialyltransferase 1cDNA source: Jennifer Lippincott-Schwartz, NIH; NM_173216.2); zyxin (6), BamHI and NotI (human zyxin cDNA source: Origene, Rockville, MD; NM_003461); vascular epithelial cadherin (10), AgeI and NotI (human VE cadherin cDNA source: Origene, Rockville, MD; NM_001795.3); mitochondria (7), BamHI and NotI (human mitochondrial targeting sequence, cytochrome c oxidase cDNA source: Clontech, Mountain View, CA; NM_004074.2); centromere protein B (22), BamHI and NotI (human CENPB cDNA source: Alexey Khodjakov, Wadsworth Center, Albany, NY; NM_001810.5); α-actinin (19), BamHI and EcoRI (human α-actinin cDNA source: Tom Keller, Florida State University, Tallahassee; NM_001130005.1); c-src sarcoma (7), BamHI and EcoRI (chicken c-src cDNA source: Marilyn Resh, Sloan-Kettering, New York; XM_001232484.1); Lifeact (7), BamHI and NotI (Lifeact cDNA source: IDT, Coralville, IA); vimentin (7), BamHI and NotI (human vimentin cDNA source: Robert Goldman, Northwestern University; NM_003380.3).
All DNA for transfection was prepared using the Plasmid Maxi kit (QIAGEN, Valencia, CA). To ensure proper localization, mTagBFP2 fusion proteins were characterized by transfection in HeLa cells (CCL2 line; ATCC, Manassas, VA) using Effectene (QIAGEN) and 1 µg vector. Transfected cells were grown on coverslips in DMEM/F12, fixed after 48 hours, and mounted with Gelvatol. Epifluorescence images (Figure 4) were taken with a Nikon 80i microscope using widefield illumination and an Omega QMax Blue filter set to confirm proper localization.
Full text: Click here
Publication 2011

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2017
Base Sequence Calmodulin-Dependent Protein Kinase II calreticulin, human Cloning Vectors Deoxyribonuclease EcoRI Mice, Laboratory Mutagenesis, Site-Directed PHluorin Plasmids Proteins Retention (Psychology) RNA Interference Short Hairpin RNA Signal Peptides STIM1 protein, human Synapsins Synaptophysin Transfection VAMP regimen
Rabbit polyclonal antibody against a synthetic peptide corresponding to residues 324–340 of human STING was obtained from EvoQuest Custom Antibody Services (Invitrogen, CA). Other antibodies were obtained from following sources: SEC61β (Upstate, NY), β-Actin, HA, FLAG (Sigma, MO), COX IV, Calreticulin, IRF3 and RIG-I (Abcam, MA).
Publication 2008
Actins Antibodies Calreticulin DDX58 protein, human Homo sapiens Immunoglobulins IRF3 protein, human Peptides Rabbits
The amino-acid sequence of super-folder mCherry (sfCherry) was obtained from the published literature18 (link). sfCherry was split between 208D and 209Y at the middle of the loop between β-strands 10 and 11 (Fig. 1a). For the nucleotide sequence of sfCherry1-10 and sfCherry11, see Supplementary Tables 1 and 2.
The DNAs of GFP11 (ref. 5 (link)) and sfCherry11 were directly synthesized. The DNAs of H2B, zyxin, clathrin light chain, β-actin were subcloned from the corresponding sfGFP or mEmerald fusion plasmids (cDNA source: the Michael Davidson Fluorescent Protein Collection at the UCSF Nikon Imaging Center). We performed the following restriction enzyme digestion (amino-acid linker length shown in parentheses for each): histone H2B (10 a.a.): sfGFP sequence between AgeI and BglII (sfGFP-H2B C-10); zyxin (6 a.a.): sfGFP sequence between BamHI and NotI (sfGFP-zyxin-6); clathrin light chain (15 a.a.): mEmerald sequence between NheI and BglII (mEmerald-clathrin-15); β-actin (18 a.a.): mEmerald sequence between AgeI and BglII (mEmerald-actin-C-18). PCR-amplified GFP11 as well as sfCherry11 fragments were then inserted into the digested vectors using In-Fusion assembly (Life Technologies).
For the cloning of GFP11-tagged Drosophila calreticulin, GFP11 was inserted at the nineteenth a.a. position where the signal peptide ends. GFP11::calreticulin was inserted into Drosophila expression pACUH vectors (source: Yuh-Nung Jan) at the EcoRI/XbaI sites. For the cloning of GFP11-tagged human β2AR, the signalling peptide (5HT3R: MRLCIPQVLLALFLSMLTGPGEGS), β2AR and GFP11 were synthesized and cloned into pcDNA3.1 vectors at the BamHI and XhoI sites.
For the expression of GFP1-10, GFP1-10 was synthesized and cloned into pACUH vectors, pcDNA3.1 vectors as well as modified lentiviral pHR-SFFV vector32 (link). For GFP1-10 sequence information, see Supplementary Table 1. For the expression of sfCherry1-10, sfCherry1-10 was synthesized and cloned into pcDNA3.1 vectors.
To generate an ER-localized GFP1-10 (GFP1-10ER), a signal peptide (5′- ATGATGTGGTG CAAAACAGTGATAGTGTTGCTGGCGACAGTCGGCTTTATTAGTGCC -3′) and an ER retention sequence (5′- AGCGAACACGACGAATTG -3′) were fused to the N-terminal and the C-terminal of GFP1-10, respectively. The SP::GFP1-10::SEHDEL fragment was cloned into pACUH vectors. For mammalian expression, we cloned SP::GFP1-10 into pcDNA3.1 vectors.
For CFP and YFP imaging, we introduced point mutations in GFP1-10. It has been previously reported that the introduced point mutations Y66W and T203Y in GFP alter the GFP spectral properties to CFP and YFP, respectively. We generated the corresponding mutations in GFP1-10 with the Q5 site-Directed Mutagenesis Kit (NEB). The primers were designed using the NEB online primer design software (NEBaseChanger). The used primers were as follows: YFP_forward (5′- CTACCTCTCAtatCAAACAGTCCTGAGCAAAGATC -3′), YFP_reverse (5′- TGATTATCAGGAAGAAGTACC -3′), CFP_forward (5′- ACGCTTACGTggGGAGTTCAGTGC -3′), and CFP_reverse (5′- TGTTACGAGAGTCGGCCA -3′). We fused these colour variants to the N terminus of Drosophila β-actin and cloned them into pACUH vectors.
To prepare GFP11 repeat arrays constructs, we first synthesized GFP11x1, x3 or x7 with long (15 a.a.) or short (5 a.a.) linkers (see the sequence information in Supplementary Table 2). Then, these GFP11 fragments were fused to β-tubulin, mouse IFT20 and dCas9. Specifically, for the cloning of GFP11x1,x3 or x7::mCherry::β-tubulin, Drosophila β-tubulin (cDNA source: R.Vale) was cloned into pACUH vectors. We then inserted mCherry-fused GFP11x1,x3 or x7 at the EcoRI site. For the cloning of GFP11x1 or x7::Ift20, we first fused Ift20 with GFP11x1 or x7, and then we inserted it into the EcoRI/NotI sites of pEGFP-N1 vectors. For the cloning dCas9:: GFP11x7, we modified our SunTag vector (pHRdSV40-NLS-dCas9-24xGCN4_v4-NLS-P2A-BFP-dWPRE (addgene #60910)) (ref. 2 (link)). 24xGCN4 was cut out by BamHI and NotI, and GFP11x7 was inserted into these sites.
For the cloning of sfCherry::β-actin, we synthesized full-length sfCherry and cloned it into the vector containing β-actin fragment (see above) at the BglII/NheI site. To construct sfCherry11x4::β-actin, we also synthesized sfCherry11x4, and replaced the sfCherry fragment to the tandem one at the BglII/NheI site. For the sequence information of sfCherry11x4, see Supplementary Table 2.
For our CRISPRa assay in Fig. 5, we fused an NLS sequence to the C-terminal of GFP1-10::VP64 and cloned into modified pHR vectors. For the GFP1-10::VP64::NLS sequence information, see Supplementary Table 3.
Full text: Click here
Publication 2016

Most recents protocols related to «Calreticulin, human»

In our experiments, phosphate buffer solution (PBS) was purchased from Hangzhou Haotian Biotechnology Co., Ltd., and RA patient serum was provided by Shaoxing Second Hospital. The human calreticulin (CRT) enzyme-linked immunosorbent assay kit was purchased from Hangzhou Sodium Magnesium Technology Co., Ltd.
Publication 2024
Tetrabutylammonium
tetrafluoroborate (TBAPF4), sodium perchlorate/lithium
perchlorate (NaClO4/LiClO4), and tetrabutylammonium
hexafluorophosphate (TBAPF6) were from Sigma-Aldrich. Anti-resistin
antibody (0.75 mg/mL), resistin from mouse (25 ug), neuron-specific
enolase human (NSE, 10 μg), vascular endothelial growth factor
(VEGF), calreticulin human (CALR, 10 μg), p53 protein human
(50 μg), GM2-activator protein (GM2A, 3.70 mg/mL), human serum,
and bovine serum albumin (BSA) were from Sigma-Aldrich. Elabscience
Biotechnology Inc. (USA) provided the ELISA Resistin kit.
Full text: Click here
Publication 2024
Human (MKN7, MKN45, and KATOIII) and mouse (T3-2D) GC cells were infected with OBP-702 (0, 10, or 50 MOI) for 24 h (n = 5), and levels of extracellular ATP in supernatants were measured using an ENLITEN ATP assay (Promega, Madison, WI, USA), according to the protocols from the manufacturer. HMGB-1 and calreticulin assays were performed as follows. Cells were incubated with Fc block, followed by CD16/32 antibodies (Thermo Fisher Scientific, Waltham, MA, USA). Cells were stained with LIVE/DEAD Fixable Aqua Dead Cell Stain Kit (Thermo Fisher Scientific) to detect live cells. Flow Cytometry was performed on a FACSArray (BD Biosciences, San Jose, CA, USA), and analyzed by the FlowJo program (BD Biosciences, Franklin Lakes, NJ, USA). The antibodies used for flow cytometry were mouse anti-HMGB-1 Ab (MA5-16263, Invitrogen, Waltham, MA, USA) and mouse anti-Calreticulin Ab (ab22683, Abcam plc).
Full text: Click here
Publication 2024
Chemicals were purchased from Sigma‐Aldrich or ThermoFisher. Recombinant human proinsulin (expressed in E. coli) was purchased from Creative Biomart (Catalog # INS‐315H). Antibodies in this study include rabbit anti‐Myc (Immunology Consultants, RRID:AB_2921297); guinea pig anti‐insulin (Covance, RRID:AB_10013624); rabbit anti‐cyclophilin B (ThermoFisher, RRID:AB_2169138); rabbit polyclonal anti‐calreticulin (Abcam RRID:AB_303402); rabbit mAb anti‐Hsp90 (Cell Signaling, RRID:AB_2233307); rabbit anti‐GM130 (Abcam, RRID:AB_880266); mouse mAb anti‐human proinsulin B‐C junction sequence PKTRREAEDLQVGQ (Abmart, RRID:AB_2921300); mouse mAb anti‐rat proinsulin (CCI‐17, Novus Biologicals RRID:AB_1107982); mouse anti‐rat C‐peptide CII‐29 (Advanced Immunochemical Inc., Catalog ID: 1‐CP‐r); mouse anti rat C‐Peptide I 6H1 (Biorad Catalog # MCA2857) and as a back‐up, a custom‐made rabbit polyclonal antibody against mouse C‐peptide‐2 sequence PQVAQLELGGGPGAGDLQT (Biogot, RRID:AB_2921302).
Publication 2024
For making transgenic flies, DNA for Rpr, Rpr-MTS, Rpr-Golgi, or Rpr-ER was synthesized by GenScript and cloned into the pUASTattB vector. For Rpr-MTS, the last 24 amino acids of the HID protein were added to the C-terminus of Rpr as previously described (Sandu et al, 2010 (link)). For Rpr-ER, a 17–amino acid calreticulin signal peptide was added to the N-terminus of Rpr along with a KDEL sequence at the C-terminal end. For Rpr-Golgi, the N-terminal 81 amino acids of human beta-1,4-galactosyltransferase (GalT) signal peptide were added to the N-terminus of Rpr. Transgenic flies were generated by inserting the plasmid into the attP2 on the third chromosome by BestGene.
Full text: Click here
Publication 2024

Top products related to «Calreticulin, human»

Sourced in United States, Germany, United Kingdom, China, Canada, Japan, Italy, France, Belgium, Singapore, Uruguay, Switzerland, Spain, Australia, Poland, India, Austria, Denmark, Netherlands, Jersey, Finland, Sweden
The FACSCalibur is a flow cytometry system designed for multi-parameter analysis of cells and other particles. It features a blue (488 nm) and a red (635 nm) laser for excitation of fluorescent dyes. The instrument is capable of detecting forward scatter, side scatter, and up to four fluorescent parameters simultaneously.
CRT mAb is a monoclonal antibody that recognizes the calreticulin (CRT) protein. Calreticulin is a chaperone protein involved in the regulation of calcium homeostasis and the folding of newly synthesized glycoproteins in the endoplasmic reticulum.
The HSP70 mAb is a monoclonal antibody that specifically recognizes the Heat Shock Protein 70 (HSP70). HSP70 is a highly conserved molecular chaperone that plays a crucial role in protein folding, trafficking, and the cellular stress response. The HSP70 mAb can be used as a tool for the detection and study of HSP70 in various experimental and research applications.
The HSP90 mAb is a monoclonal antibody specifically targeting the Heat Shock Protein 90 (HSP90). HSP90 is a molecular chaperone that plays a critical role in the folding, stability, and function of various client proteins. The HSP90 mAb can be used in various applications, such as Western blotting, immunoprecipitation, and immunohistochemistry, to detect and study the expression and localization of HSP90 in biological samples.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in United States, United Kingdom, Germany, Canada, Japan, France, Switzerland, Panama, Spain, China, Italy
Vectashield is a non-hardening, aqueous-based mounting medium designed for use with fluorescent-labeled specimens. It is formulated to retard photobleaching of fluorescent dyes and provides excellent preservation of fluorescent signals.
Sourced in United States, Germany, United Kingdom
Anti-calreticulin is a laboratory reagent used to detect the presence and distribution of calreticulin protein in biological samples. Calreticulin is a calcium-binding protein involved in various cellular processes. This antibody can be used in techniques such as Western blotting, immunohistochemistry, and immunofluorescence to analyze calreticulin expression and localization.
Sourced in United States
Sec31A is a laboratory equipment product designed for specialized applications. It is a multi-purpose device capable of performing various analytical and processing tasks. The core function of Sec31A is to provide reliable and precise results for researchers and scientists working in diverse fields. Further details about its intended use or specific capabilities are not available at this time.
Sourced in United States
TANGO1 is a laboratory equipment product from Merck Group. It is designed for the detection and analysis of specific biomolecules. The core function of TANGO1 is to provide accurate and reliable results for researchers and scientists working in the fields of biochemistry and molecular biology.
Sourced in United States
The TGN46 is a lab equipment product offered by Bio-Rad. It is a device used for the detection and analysis of specific proteins in biological samples. The core function of the TGN46 is to perform Western blot analysis, a widely used technique in molecular biology and biochemistry.

More about "Calreticulin, human"