The largest database of trusted experimental protocols

Transfection

Transfection is the process of introducing nucleic acids, such as DNA or RNA, into cells using various methods.
This technique is widely used in biological research to study gene expression, protein function, and cellular pathways.
Transfection can be achieved through physical, chemical, or biological means, including lipid-based reagents, electroporation, and viral vectors.
The efficiency and toxicity of transfection methods can vary, and optimal protocols must be carefully selected to ensure successful gene delivery and minimized cellular stress.
PubCompare.ai revolutionizes transfection optimzation by leveraging AI-driven comparisons of research protocols, helping researchers identify the best methods from literature, pre-prints, and patents, and improve the reproducibility of their experiments.
By harnessing the power of artificial intellignce, PubCompare.ai enables seamless protocol discovery and optimization, empowering researchers to find the optimal transfection methods and products for their specific needs.

Most cited protocols related to «Transfection»

Targeting constructs were generated using a combined gene synthesis (GenScript Corp.) and molecular cloning approach. Briefly, to target the Rosa26 locus, a cassette containing the following components was constructed: FRT – LoxP – Stop codons – 3x SV40 polyA – LoxP – EYFP – WPRE – bGH polyA – AttB – PGK promoter – FRT – Neo – PGK polyA – AttP. For most targeting vectors, this cassette was cloned into a Rosa-CAG targeting vector3 (link), downstream of the CAG promoter and upstream of the 3′ arm, to generate the final EYFP targeting vector. Unique restriction sites flanking the EYFP gene were used to replace EYFP with alternative reporter genes. For the Ai2 vector, which lacks the WPRE, the CAG promoter was inserted between the first FRT and LoxP sites, and the cassette was cloned immediately downstream of the 5′ homology arm. The final targeting vectors contained 5′ and 3′ homology arms of 1.1 kb and 4.3 kb, as well as a PGK-DTA cassette for negative selection. Targeting constructs for knock-in Cre lines inserted into other gene loci were constructed in similar ways.
The targeting vectors were linearized and transfected into the 129/B6 F1 hybrid ES cell line G442 (link) using an Amaxa electroporator. G418-resistant ES clones were screened by Southern blot analysis of HindIII digested DNA, which was probed with a 1.1 kb genomic fragment from immediately upstream of the 5′ arm. We observed a recombination rate of about 25% for the four constructs. Positive ES clones were injected into C57BL/6J blastocysts to obtain chimeric mice following standard procedures. Both ES cell transfections and blastocyst injections were performed by the University of Washington Transgenic Resources Program. Due to the robustness of the G4 cells, high-percentage chimeras and high rates of germline transmission were routinely obtained. Chimeric mice were bred with either C57BL/6J mice to obtain germline transmission or various Cre-driver lines for direct characterization.
An Ai9 ES cell clone with strong germline transmission potency was used in subsequent transfections for the Flp-mediated exchange strategy outlined in Supplementary Figure 4 online. Ai9 ES cells were co-transfected using a Bio-Rad electroporator with 100 μg of pCAGGS-FLPe (Open Biosystems) and 40 μg of an incoming replacement vector. After 8 to 10 days of Hygromycin B selection, surviving colonies that also appeared green by fluorescence microscopy were picked and screened by PCR using primer sets designed to confirm a correct insertion of the incoming vector at the 5′ and 3′ FRT recombinase sites.
Publication 2009
Anabolism Animals, Transgenic antibiotic G 418 Blastocyst Cells Chimera Clone Cells Cloning Vectors Codon, Terminator Embryonic Stem Cells Genes Genes, Reporter Genetic Loci Genome Germ Cells Germ Line Hybrid Cells Hygromycin B Mice, Inbred C57BL Microscopy, Fluorescence Mus N-fluoresceinylphosphatidylethanolamine Oligonucleotide Primers Poly A Recombinase Recombination, Genetic Rosa Simian virus 40 Southern Blotting Transfection Transmission, Communicable Disease
Targeting constructs were generated using a combined gene synthesis (GenScript Corp.) and molecular cloning approach. Briefly, to target the Rosa26 locus, a cassette containing the following components was constructed: FRT – LoxP – Stop codons – 3x SV40 polyA – LoxP – EYFP – WPRE – bGH polyA – AttB – PGK promoter – FRT – Neo – PGK polyA – AttP. For most targeting vectors, this cassette was cloned into a Rosa-CAG targeting vector3 (link), downstream of the CAG promoter and upstream of the 3′ arm, to generate the final EYFP targeting vector. Unique restriction sites flanking the EYFP gene were used to replace EYFP with alternative reporter genes. For the Ai2 vector, which lacks the WPRE, the CAG promoter was inserted between the first FRT and LoxP sites, and the cassette was cloned immediately downstream of the 5′ homology arm. The final targeting vectors contained 5′ and 3′ homology arms of 1.1 kb and 4.3 kb, as well as a PGK-DTA cassette for negative selection. Targeting constructs for knock-in Cre lines inserted into other gene loci were constructed in similar ways.
The targeting vectors were linearized and transfected into the 129/B6 F1 hybrid ES cell line G442 (link) using an Amaxa electroporator. G418-resistant ES clones were screened by Southern blot analysis of HindIII digested DNA, which was probed with a 1.1 kb genomic fragment from immediately upstream of the 5′ arm. We observed a recombination rate of about 25% for the four constructs. Positive ES clones were injected into C57BL/6J blastocysts to obtain chimeric mice following standard procedures. Both ES cell transfections and blastocyst injections were performed by the University of Washington Transgenic Resources Program. Due to the robustness of the G4 cells, high-percentage chimeras and high rates of germline transmission were routinely obtained. Chimeric mice were bred with either C57BL/6J mice to obtain germline transmission or various Cre-driver lines for direct characterization.
An Ai9 ES cell clone with strong germline transmission potency was used in subsequent transfections for the Flp-mediated exchange strategy outlined in Supplementary Figure 4 online. Ai9 ES cells were co-transfected using a Bio-Rad electroporator with 100 μg of pCAGGS-FLPe (Open Biosystems) and 40 μg of an incoming replacement vector. After 8 to 10 days of Hygromycin B selection, surviving colonies that also appeared green by fluorescence microscopy were picked and screened by PCR using primer sets designed to confirm a correct insertion of the incoming vector at the 5′ and 3′ FRT recombinase sites.
Publication 2009
Anabolism Animals, Transgenic antibiotic G 418 Blastocyst Cells Chimera Clone Cells Cloning Vectors Codon, Terminator Embryonic Stem Cells Genes Genes, Reporter Genetic Loci Genome Germ Cells Germ Line Hybrid Cells Hygromycin B Mice, Inbred C57BL Microscopy, Fluorescence Mus N-fluoresceinylphosphatidylethanolamine Oligonucleotide Primers Poly A Recombinase Recombination, Genetic Rosa Simian virus 40 Southern Blotting Transfection Transmission, Communicable Disease
DNA extracted from cancer specimens and normal tissue was labeled and hybridized to the Affymetrix 250K Sty I array to obtain signal intensities and genotype calls. Signal intensities were normalized against data from 1480 normal samples. Copy-number profiles were inferred using GLAD48 (link) and changes of > 0.1 copies in either direction were called SCNAs. The significance of focal SCNAs (covering < 0.5 chromosome arms) was determined using GISTIC18 (link), with modifications to score SCNAs directly proportional to amplitude and to allow summation of non-overlapping deletions affecting the same gene. Peak region boundaries were determined so that the change in the GISTIC score from peak to boundary had < 5% likelihood of occurring by random fluctuation. P-values for Figures 2b and 4 were determined by comparing the gene densities of SCNAs and fraction overlap of peak regions respectively to the same quantities calculated from random permutations of the locations of these SCNAs and peak regions. RNAi was performed by inducible and stable expression of shRNA lentiviral vectors and by siRNA transfection. Proliferation in inducible shRNA experiments was measured in triplicate every half-hour on 96-well plates by a real time electric sensing system (ACEA Bioscience) and in stable shRNA expression and siRNA transfection experiments by CellTiterGlo (Promega). Apoptosis was measured by immunoblot against cleaved PARP and FACS analysis of cells stained with antibody to annexin V and propidium iodide. Tumor growth in nude mice was measured by caliper twice weekly. Expression of MYC, MCL1, and BCL2L1 was performed with retroviral vectors in lung epithelial cells immortalized by introduction of SV40 and hTERT49 (link).
Full methods are described in Supplementary Methods.
Publication 2010
Annexin A5 Apoptosis Arm, Upper bcl-X Protein Cells Chromosomes Cloning Vectors Electricity Epithelial Cells Gene Deletion Genes Genotype Immunoblotting Immunoglobulins Lung Malignant Neoplasms MCL1 protein, human Mice, Nude Neoplasms Promega Propidium Iodide Retroviridae RNA, Small Interfering RNA Interference Short Hairpin RNA Simian virus 40 Tissues Transfection
DNA extracted from cancer specimens and normal tissue was labeled and hybridized to the Affymetrix 250K Sty I array to obtain signal intensities and genotype calls. Signal intensities were normalized against data from 1480 normal samples. Copy-number profiles were inferred using GLAD48 (link) and changes of > 0.1 copies in either direction were called SCNAs. The significance of focal SCNAs (covering < 0.5 chromosome arms) was determined using GISTIC18 (link), with modifications to score SCNAs directly proportional to amplitude and to allow summation of non-overlapping deletions affecting the same gene. Peak region boundaries were determined so that the change in the GISTIC score from peak to boundary had < 5% likelihood of occurring by random fluctuation. P-values for Figures 2b and 4 were determined by comparing the gene densities of SCNAs and fraction overlap of peak regions respectively to the same quantities calculated from random permutations of the locations of these SCNAs and peak regions. RNAi was performed by inducible and stable expression of shRNA lentiviral vectors and by siRNA transfection. Proliferation in inducible shRNA experiments was measured in triplicate every half-hour on 96-well plates by a real time electric sensing system (ACEA Bioscience) and in stable shRNA expression and siRNA transfection experiments by CellTiterGlo (Promega). Apoptosis was measured by immunoblot against cleaved PARP and FACS analysis of cells stained with antibody to annexin V and propidium iodide. Tumor growth in nude mice was measured by caliper twice weekly. Expression of MYC, MCL1, and BCL2L1 was performed with retroviral vectors in lung epithelial cells immortalized by introduction of SV40 and hTERT49 (link).
Full methods are described in Supplementary Methods.
Publication 2010
Annexin A5 Apoptosis Arm, Upper bcl-X Protein Cells Chromosomes Cloning Vectors Electricity Epithelial Cells Gene Deletion Genes Genotype Immunoblotting Immunoglobulins Lung Malignant Neoplasms MCL1 protein, human Mice, Nude Neoplasms Promega Propidium Iodide Retroviridae RNA, Small Interfering RNA Interference Short Hairpin RNA Simian virus 40 Tissues Transfection
CN34 tumour cells were isolated from the pleural effusion of a breast cancer patient treated at our institution, after written consent in accordance with Institutional Review Board (IRB) regulations. Brain metastatic populations from these cells and MDA-MB-231cells were obtained by consecutive rounds of in vivo selection in 6–7-week-old beige nude and athymic mice, respectively. All animal work was done in accordance with the MSKCC Institutional Animal Care and Use Committee. Methods for RNA extraction, labelling and hybridization for DNA microarray analysis have been described previously17 (link). Bioinformatics analyses with detailed descriptions can be found in the Methods. Knockdown and overexpression of candidate genes, and cetuximab inhibitor studies were performed as previously described6 (link). The in vitro BBB model was set up as previously described25 (link), and modified to enable tumour cell counting. Sambucus nigra lectin staining was performed using standard histochemical techniques, and quantified using Metamorph software analysis. The Methods section provides further information, including malignant cell isolation from pleural fluids, tumour cell extraction and cell culture protocols, animal inoculation and bioluminescence imaging, generation of retroviral gene knockdown and overexpression vectors, transfections and infections, RNA and protein expression, in vitro BBB transmigration assay, endothelial cell adhesion assay, and metastatic tissue staining and quantification.
Publication 2009
Animals Biological Assay Brain Breast Carcinoma Cell Adhesion Cell Culture Techniques Cells Cell Separation Cetuximab Cloning Vectors Crossbreeding DNA Chips Endothelial Cells Endothelium Ethics Committees, Research Gene Knockdown Techniques Genes Infection Institutional Animal Care and Use Committees Lectin Mice, Nude Microarray Analysis Neoplasms Patients Pleura Pleural Effusion Population Group Proteins Retroviridae Sambucus nigra Tissues Transfection Vaccination

Most recents protocols related to «Transfection»

Example 17

To further validate the activity of the DMPK siRNAs, many of the sequences that showed the best activity in the initial screen were selected for a follow-up evaluation in dose response format. Once again, two human cell lines were used to assess the in vitro activity of the DMPK siRNAs: first, SJCRH30 human rhabdomyosarcoma cell line; and second, Myotonic Dystrophy Type 1 (DM1) patient-derived immortalized human skeletal myoblasts. The selected siRNAs were transfected in a 10-fold dose response at 100, 10, 1, 0.1, 0.01, 0,001, and 0.0001 nM final concentrations or in a 9-fold dose response at 50, 5.55556, 0.617284, 0.068587, 0.007621, 0.000847, and 0.000094 nM final concentrations. The siRNAs were formulated with transfection reagent Lipofectamine RNAiMAX (Life Technologies) according to the manufacturer's “forward transfection” instructions. Cells were plated 24 h prior to transfection in triplicate on 96-well tissue culture plates, with 8500 cells per well for SJCRH30 and 4000 cells per well for DM1 myoblasts. At 48 h (SJCRH30) or 72 h (DM1 myoblasts) post-transfection cells were washed with PBS and harvested with TRIzol® reagent (Life Technologies). RNA was isolated using the Direct-zol-96 RNA Kit (Zymo Research) according to the manufacturer's instructions. 10 μl of RNA was reverse transcribed to cDNA using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) according to the manufacturer's instructions. cDNA samples were evaluated by qPCR with DMPK-specific and PPIB-specific TaqMan human gene expression probes (Thermo Fisher) using TaqMan® Fast Advanced Master Mix (Applied Biosystems). DMPK values were normalized within each sample to PPIB gene expression. The quantification of DMPK downregulation was performed using the standard 2−ΔΔCt a method. All experiments were performed in triplicate, with Tables 16A-B, 17A-B, and 18A-B presenting the mean values of the triplicates as well as the calculated IC50 values determined from fitting curves to the dose-response data by non-linear regression.

TABLE 16A
sense strandSEQantisense strandSEQ
sequence (5′-3′)IDsequence (5′-3′)ID
ID #1Passenger Strand (PS)NO:Guide Strand (GS)NO:
535GGGCGAGGUGUCGUGCUUA9349UAAGCACGACACCUCGCCC12053
584GACCGGCGGUGGAUCACGA9398UCGUGAUCCACCGCCGGUC12102
716AUGGCGCGCUUCUACCUGA9530UCAGGUAGAAGCGCGCCAU12234
1028CAGACGCCCUUCUACGCGA9842UCGCGUAGAAGGGCGUCUG12546
1276UUUCGAAGGUGCCACCGAA10090UUCGGUGGCACCUUCGAAA12794
1825UGCUCCUGUUCGCCGUUGA10639UCAACGGCGAACAGGAGCA13343
1945CCCUAGAACUGUCUUCGAA10759UUCGAAGACAGUUCUAGGG13463
2529CUUCGGCGGUUUGGAUAUA11343UAUAUCCAAACCGCCGAAG14047
2558GUCCUCCGACUCGCUGACA11372UGUCAGCGAGUCGGAGGAC14076
2628CCGACAUUCCUCGGUAUUA11442UAAUACCGAGGAAUGUCGG14146
2636CCUCGGUAUUUAUUGUCUA11450UAGACAAUAAAUACCGAGG14154
119mer position in NM_001288766.1

TABLE 16B
IC50
ID #1qPCR2qPCR3qPCR4qPCR5qPCR6qPCR7qPCR8(nM)
535111.9105.4106.382.436.729.535.70.165
58490.590.284.767.838.025.828.30.190
71688.985.281.962.032.619.320.30.181
102888.581.883.061.332.727.331.50.127
127687.085.084.066.140.534.036.40.150
182585.185.983.769.136.225.225.00.259
194585.081.774.444.922.917.717.20.070
252983.381.875.350.624.617.517.70.103
255884.381.174.345.423.413.311.80.088
262885.384.079.559.830.323.525.10.140
263686.386.974.344.019.812.413.00.070
2SJCRH30; 0.0001 nM; % DMPK mRNA
3SJCRH30; 0.001 nM; % DMPK mRNA
4SJCRH30; 0.01 nM; % DMPK mRNA
5SJCRH30; 0.1 nM; % DMPK mRNA
6SJCRH30; 1 nM; % DMPK mRNA
7SJCRH30; 10 nM; % DMPK mRNA
8SJCRH30; 100 nM; % DMPK mRNA

TABLE 17A
sense strandSEQantisense strandSEQ
sequence (5′-3′)IDsequence (5′-3′)ID
ID #1Passenger Strand (PS)NO:Guide Strand (GS)NO:
2600CAAUCCACGUUUUGGAUGA11414UCAUCCAAAACGUGGAUUG14118
2636CCUCGGUAUUUAUUGUCUA11450UAGACAAUAAAUACCGAGG14154
2675CCCCGACCCUCGCGAAUAA11489UUAUUCGCGAGGGUCGGGG14193
2676CCCGACCCUCGCGAAUAAA11490UUUAUUCGCGAGGGUCGGG14194
2679GACCCUCGCGAAUAAAAGA11493UCUUUUAUUCGCGAGGGUC14197
2680ACCCUCGCGAAUAAAAGGA11494UCCUUUUAUUCGCGAGGGU14198
2681CCCUCGCGAAUAAAAGGCA11495UGCCUUUUAUUCGCGAGGG14199
2682CCUCGCGAAUAAAAGGCCA11496UGGCCUUUUAUUCGCGAGG14200
119mer position in NM_001288766.1

TABLE 17B
IC50
ID #1qPCR2qPCR3qPCR4qPCR5qPCR6qPCR7(nM)
2600107.5107.6108.1106.3103.172.731.31
263681.181.174.047.225.711.50.073
267588.188.384.364.638.120.70.151
267688.978.984.472.744.935.60.204
267984.087.382.753.331.413.50.091
268087.485.385.168.544.539.60.110
268187.085.477.649.626.516.00.061
268282.483.977.150.827.331.10.047
2SJCRH30; 0.000094 nM; % DMPK mRNA
3SJCRH30; 0.000847 nM; % DMPK mRNA
4SJCRH30; 0.007621 nM; % DMPK mRNA
5SJCRH30; 0.068587 nM; % DMPK mRNA
6SJCRH30; 0.617284 nM; % DMPK mRNA
7SJCRH30; 5.55556 nM; % DMPK mRNA

TABLE 18A
sense strandSEQantisense strandSEQ
sequence (5′-3′)IDsequence (5′-3′)ID
ID #1Passenger Strand (PS)NO:Guide Strand (GS)NO:
584GACCGGCGGUGGAUCACGA9398UCGUGAUCCACCGCCGGUC12102
716AUGGCGCGCUUCUACCUGA9530UCAGGUAGAAGCGCGCCAU12234
1265UUUACACCGGAUUUCGAAA10079UUUCGAAAUCCGGUGUAAA12783
1297AUGCAACUUCGACUUGGUA10111UACCAAGUCGAAGUUGCAU12815
1945CCCUAGAACUGUCUUCGAA10759UUCGAAGACAGUUCUAGGG13463
1960CGACUCCGGGGCCCCGUUA10774UAACGGGGCCCCGGAGUCG13478
2529CUUCGGCGGUUUGGAUAUA11343UAUAUCCAAACCGCCGAAG14047
2530UUCGGCGGUUUGGAUAUUA11344UAAUAUCCAAACCGCCGAA14048
2531UCGGCGGUUUGGAUAUUUA11345UAAAUAUCCAAACCGCCGA14049
2554CCUCGUCCUCCGACUCGCA11368UGCGAGUCGGAGGACGAGG14072
2628CCGACAUUCCUCGGUAUUA11442UAAUACCGAGGAAUGUCGG14146
2629CGACAUUCCUCGGUAUUUA11443UAAAUACCGAGGAAUGUCG14147
2681CCCUCGCGAAUAAAAGGCA11495UGCCUUUUAUUCGCGAGGG14199
119mer position in NM_001288766.1

TABLE 18B
IC50
ID #1qPCR2qPCR3qPCR4qPCR5qPCR6qPCR7(nM)
58490.877.097.771.945.029.70.228
71696.582.577.064.643.333.90.080
126568.580.968.057.137.525.70.146
129771.467.269.453.540.525.40.171
194571.862.341.729.822.415.30.006
196063.065.462.145.831.128.30.068
252963.558.749.231.122.921.90.017
253069.366.753.143.238.824.50.016
253169.972.457.340.235.425.60.018
255468.270.151.243.032.117.30.043
262869.767.962.538.431.617.10.042
262972.165.669.042.134.413.70.078
268182.491.587.655.529.319.60.084
2DM1 myoblasts; 0.000094 nM; % DMPK mRNA
3DM1 myoblasts; 0.000847 nM; % DMPK mRNA
4DM1 myoblasts; 0.007621 nM; % DMPK mRNA
5DM1 myoblasts; 0.068587 nM; % DMPK mRNA
6DM1 myoblasts; 0.617284 nM; % DMPK mRNA
7DM1 myoblasts; 5.55556 nM; % DMPK mRNA

Patent 2024
Cell Lines Cells DNA, Complementary Down-Regulation Gene Expression Homo sapiens Lipofectamine Myoblasts Myoblasts, Skeletal Myotonic Dystrophy NM-107 Patients PPIB protein, human Reverse Transcription Rhabdomyosarcoma RNA, Messenger RNA, Small Interfering Tissues Transfection trizol

Example 1

The sequence coding for the light chain variable region of the antibody was inserted into vector pFUSE2ss-CLIg-hK (Invivogen, Catalog Number: pfuse2ss-hclk) using EcoRI and BsiWI restriction sites to construct a light chain expression vector. The sequence coding for the heavy chain variable region of the antibody was inserted into vector pFUSEss-CHIg-hG2 (Invivogen, Catalog Number: pfusess-hchg2) or vector pFUSEss-CHIg-hG4 (Invivogen, Catalog Number: pfusess-hchg4) using EcoRI and NheI restriction sites to construct a heavy chain expression vector.

The culture and transfection of Expi293 cells were performed in accordance with the handbook of Expi293™ Expression System Kit from Invitrogen (Catalog Number: A14635). The density of the cells was adjusted to 2×106 cells/ml for transfection, and 0.6 μg of the light chain expression vector as described above and 0.4 μg of the heavy chain expression vector as described above were added to each ml of cell culture, and the supernatant of the culture was collected four days later.

The culture supernatant was subjected to non-reduced SDS-PAGE gel electrophoresis in accordance with the protocol described in Appendix 8, the Third edition of the “Molecular Cloning: A Laboratory Manual”.

Pictures were taken with a gel scanning imaging system from BEIJING JUNYI Electrophoresis Co., LTD and in-gel quantification was performed using Gel-PRO ANALYZER software to determine the expression levels of the antibodies after transient transfection. Results were expressed relative to the expression level of control antibody 1 (control antibody 1 was constructed according to U.S. Pat. No. 7,186,809, which comprises a light chain variable region as set forth in SEQ ID NO: 10 of U.S. Pat. No. 7,186,809 and a heavy chain variable region as set forth in SEQ ID NO: 12 of U.S. Pat. No. 7,186,809, the same below) (control antibody 2 was constructed according to U.S. Pat. No. 7,638,606, which comprises a light chain variable region as set forth in SEQ ID NO: 6 of U.S. Pat. No. 7,638,606 and a variable region as set forth in SEQ ID NO: 42 of U.S. Pat. No. 7,638,606, the same below). See Tables 2a-2c below for the results.

TABLE 2a
Expression levels of the antibodies of the present
invention after transient transfection (antibodies whose
expression levels are significantly higher than that of control antibody 1):
Number ofExpression level vsNumber of Expression level vs
the antibodycontrol antibody 1the antibodycontrol antibody 1
L1021H10002.08L1000H10281.27
L1020H10001.58L1000H10151.19
L1000H10271.56L1000H10321.18
L1000H10241.51L1000H10261.15
L1000H10251.48L1021H10291.12
L1001H10001.48L1000H10301.1
L1021H10161.43L1024H10311.08
L1000H10141.35L1000H10161.05

TABLE 2b
Expression levels of the antibodies of the present
invention after transient transfection (antibodies whose
expression levels are slightly lower than that of control antibody 1):
Number of Expression level vsNumber of Expression level vs
the antibodycontrol antibody 1the antibodycontrol antibody 1
L1000H10310.99L1017H10000.85
L1021H10310.99L1020H10160.84
L1020H10290.96L1000H10090.81
control anti-0.93L1000H10070.8
body 2
L1012H10000.89L1000H10230.8
L1019H10000.87L1020H10270.78
L1020H10310.87L1024H10070.77
L1021H10200.87L1000H10130.75
L1000H10290.86L1020H10070.74
L1008H10000.86L1021H10070.74
L1000H10010.85L1000H10210.71

TABLE 2c
Expression levels of the antibodies of the present
invention after transient transfection (antibodies whose
expression levels are significantly lower than that of control antibody 1):
Number ofExpression level vsNumber of Expression level vs
the antibodycontrol antibody 1the antibodycontrol antibody 1
L1000H10200.69L1024H10000.52
L1010H10000.69L1000H10080.51
L1000H10220.67L1000H10370.5
L1000H10120.64L1007H10000.49
L1022H10000.64L1016H10000.49
L1011H10000.63L1000H10170.47
L1000H10110.62L1000H10350.46
L1000H10330.62L1012H10270.46
L1020H10200.61L1018H10000.44
L1000H10360.6L1023H10000.43
L1021H10270.6L1012H10160.42
L1012H10070.59L1013H10000.41
L1009H10000.57L1000H10340.4
L1012H10200.57L1000H10180.35
L1012H10310.56L1000H10190.34
L1000H10380.54L1015H10000.27
L1012H10290.54L1014H10000.17
L1000H10100.53

Example 4

6-8 week-old SPF Balb/c mice were selected and injected subcutaneously with antibodies (the antibodies of the present invention or control antibody 2) in a dose of 5 mg/kg (weight of the mouse). Blood samples were collected at the time points before administration (0 h) and at 2, 8, 24, 48, 72, 120, 168, 216, 264, 336 h after administration. For blood sampling, the animals were anesthetized by inhaling isoflurane, blood samples were taken from the orbital venous plexus, and the sampling volume for each animal was about 0.1 ml; 336 h after administration, the animals were anesthetized by inhaling isoflurane and then euthanized after taking blood in the inferior vena cava.

No anticoagulant was added to the blood samples, and serum was isolated from each sample by centrifugation at 1500 g for 10 min at room temperature within 2 h after blood sampling. The collected supernatants were immediately transferred to new labeled centrifuge tubes and then stored at −70° C. for temporary storage. The concentrations of the antibodies in the mice were determined by ELISA:

1. Preparation of Reagents

sIL-4Rα (PEPRO TECH, Catalog Number: 200-04R) solution: sIL-4Rα was taken and 1 ml ddH2O was added therein, mixed up and down, and then a solution of 100 μg/ml was obtained. The solution was stored in a refrigerator at −20° C. after being subpacked.

Sample to be tested: 1 μl of serum collected at different time points was added to 999 μl of PBS containing 1% BSA to prepare a serum sample to be tested of 1:1000 dilution.

Standard sample: The antibody to be tested was diluted to 0.1 μg/ml with PBS containing 1% BSA and 0.1% normal animal serum (Beyotime, Catalog Number: ST023). Afterwards, 200, 400, 600, 800, 900, 950, 990 and 1000 μl of PBS containing 1% BSA and 0.1% normal animal serum were respectively added to 800, 600, 400, 200, 100, 50, 10 and 0 μl of 0.1 μg/ml antibodies to be tested, and thus standard samples of the antibodies of the present invention were prepared with a final concentration of 80, 60, 40, 20, 10, 5, 1, or 0 ng/ml respectively.

2. Detection by ELISA

250 μl of 100 μg/ml sIL-4Rα solution was added to 9.75 ml of PBS, mixed up and down, and then an antigen coating buffer of 2.5 μg/ml was obtained. The prepared antigen coating buffer was added to a 96-well ELISA plate (Corning) with a volume of 100 μl per well. The 96-well ELISA plate was incubated overnight in a refrigerator at 4° C. after being wrapped with preservative film (or covered). On the next day, the 96-well ELISA plate was taken out and the solution therein was discarded, and PBS containing 2% BSA was added thereto with a volume of 300 μl per well. The 96-well ELISA plate was incubated for 2 hours in a refrigerator at 4° C. after being wrapped with preservative film (or covered). Then the 96-well ELISA plate was taken out and the solution therein was discarded, and the plate was washed 3 times with PBST. The diluted standard antibodies and the sera to be detected were sequentially added to the corresponding wells, and three duplicate wells were made for each sample with a volume of 100 μl per well. The ELISA plate was wrapped with preservative film (or covered) and incubated for 1 h at room temperature. Subsequently, the solution in the 96-well ELISA plate was discarded and then the plate was washed with PBST for 3 times. Later, TMB solution (Solarbio, Catalog Number: PR1200) was added to the 96-well ELISA plate row by row with a volume of 100 μl per well. The 96-well ELISA plate was placed at room temperature for 5 minutes, and 2 M H2SO4 solution was added in immediately to terminate the reaction. The 96-well ELISA plate was then placed in flexstation 3 (Molecular Devices), the values of OD450 were read, the data were collected and the results were calculated with Winnonlin software. The pharmacokinetic results were shown in FIG. 1 and Table 6 below.

TABLE 6
Pharmacokinetic results of the antibodies of the present invention in mouse
Area
TimeUnder the
HalftoPeakdrug-timeVolume ofClearance
lifepeakconcentrationCurvedistributionrate
Numberhhμg/mlh*μg/mlml/kgml/h/kg
L1020H1031Mean269.347233.797679.28138.920.38
value
Standard105.730.000.42163.9122.480.09
deviation
L1012H1031Mean167.274845.59852.391.30.38
value
Standard8.520.001.86448.345.580.00
deviation
ControlMean56.67367.881132.68288.923.79
antibody 2value
Standard25.8416.970.2594.4249.451.12
deviation

Example 5

A series of pharmacokinetic experiments were carried out in Macaca fascicularises to further screen antibodies.

3-5 year-old Macaca fascicularises each weighting 2-5 Kg were selected and injected subcutaneously with antibodies (the antibodies of the present invention or control antibody 2) in a dose of 5 mg/kg (weight of the Macaca fascicularis). The antibody or control antibody 2 to be administered was accurately extracted with a disposable aseptic injector, and multi-point injections were made subcutaneously on the inner side of the thigh of the animal, and the injection volume per point was not more than 2 ml. Whole blood samples were collected from the subcutaneous vein of the hind limb of the animal at the time points before administration (0 h) and at 0.5, 2, 4, 8, 24, 48, 72, 120, 168, 240, 336 h, 432 h, 504 h, 600 h, 672 h after administration. The blood volume collected from each animal was about 0.1 ml each time.

No anticoagulant was added to the blood samples, and serum was isolated from each sample by centrifugation at 1500 g for 10 min at room temperature within 2 h after blood sampling. The collected supernatants were immediately transferred to new labeled centrifuge tubes and then stored at −70° C. for temporary storage. The concentrations of the antibodies in the Macaca fascicularises were determined according the method as described in Example 4. The pharmacokinetic results are shown in FIG. 2 and Table 7 below.

TABLE 7
Pharmacokinetic results of the antibodies of the present invention in macaca fascicularis
Area
TimeUnder the
HalftoPeakdrug-timeVolume ofClearance
lifepeakconcentrationCurvedistributionrate
Numberhhμg/mlh*μg/mlml/kgml/h/kg
L1020H1031Mean254.9548.0089.6522189.9175.940.22
value
Standard44.5733.9444.298557.1522.950.10
deviation
L1012H1031Mean185.75486516185.7373.410.28
value
Standard42.5433.944.52506.980.810.06
deviation
ControlMean37.031637.822773.2193.971.78
antibody 2value
Standard18.0311.316.75155.8442.470.07
deviation

Example 10

In vivo pharmacokinetics of the antibodies of the invention are further detected and compared in this Example, in order to investigate the possible effects of specific amino acids at specific positions on the pharmacokinetics of the antibodies in animals. The specific experimental method was the same as that described in Example 4, and the results are shown in Table 9 below.

TABLE 9
Detection results of in vivo pharmacokinetics of the antibodies of the present invention
Area
TimeUnder the
HalftoPeakdrug-timeVolume ofClearance
lifepeakconcentrationCurvedistributionrate
hhug/mlh*ug/mlml/kgml/h/kg
L1020H1031Mean185.494038.948188.8114.280.43
value
Standard18.5213.862.33510.476.50.05
deviation
L1012H1001Mean161.2648.0012.362491.19332.791.47
value
Standard54.300.002.26165.1676.910.20
deviation
L1001H1031Mean171.4156.0042.749273.7399.170.40
value
Standard6.1213.867.381868.6618.690.07
deviation
L1020H1001Mean89.0064.0020.113481.40164.141.30
value
Standard16.7013.862.14268.3922.860.20
deviation

From the specific sequence, the amino acid at position 103 in the sequence of the heavy chain H1031 (SEQ ID NO. 91) of the antibody (in CDR3) is Asp (103Asp), and the amino acid at position 104 is Tyr (104Tyr). Compared with antibodies that have no 103Asp and 104Tyr in heavy chain, the present antibodies which have 103Asp and 104Tyr have a 2- to 4-fold higher area under the drug-time curve and an about 70% reduced clearance rate.

The expression levels of the antibodies of the present invention are also detected and compared, in order to investigate the possible effects of specific amino acids at specific positions on the expression of the antibodies. Culture and transfection of Expi293 cells were conducted according to Example 1, and the collected culture supernatant was then passed through a 0.22 μm filter and then purified by GE MabSelect Sure (Catalog Number: 11003494) Protein A affinity chromatography column in the purification system GE AKTA purifier 10. The purified antibody was collected and concentrated using Amicon ultrafiltration concentrating tube (Catalog Number: UFC903096) and then quantified. The quantitative results are shown in Table 10 below.

TABLE 10
Detection results of the expression
levels of the antibodies of the present invention
Expression level
Antibody(×10−2 mg/ml culture medium)
L1020H10318.39
L1001H10311.79
L1020H10014.04
L1012H10015.00
L1023H10014.63
L1001H10011.75

From the specific sequence, the amino acid at position 31 in the sequence of the light chain L1012 (SEQ ID NO. 44), L1020 (SEQ ID NO. 55) or L1023 (SEQ ID NO. 51) of the antibody (in CDR1) is Ser (31Ser). Compared with antibodies that have no 31Ser in light chain, the present antibodies which have 31Ser have a 2- to 5-fold higher expression level.

The above description for the embodiments of the present invention is not intended to limit the present invention, and those skilled in the art can make various changes and variations according to the present invention, which are within the protection scope of the claims of the present invention without departing from the spirit of the same.

Patent 2024
Amino Acids Animals Antibodies Anticoagulants Antigens Asepsis BLOOD Blood Volume Buffers Cell Culture Techniques Cells Centrifugation Chromatography Chromatography, Affinity Cloning Vectors Culture Media Deoxyribonuclease EcoRI Drug Kinetics Electrophoresis Enzyme-Linked Immunosorbent Assay Hindlimb Human Body Immunoglobulin Heavy Chains Immunoglobulin Light Chains Immunoglobulins Interleukin-1 Isoflurane Light Macaca Macaca fascicularis Medical Devices Metabolic Clearance Rate Mice, Inbred BALB C Mus Open Reading Frames Pharmaceutical Preparations Pharmaceutical Preservatives SDS-PAGE Serum Staphylococcal Protein A Technique, Dilution Thigh Transfection Transients Ultrafiltration Veins Vena Cavas, Inferior
Not available on PMC !

Example 2

iPS cells were prepared according to protocols known in the art and seeded in a Geltrex®-Matrix coated 12-well culture dish. Transfection was performed in iPSCs with 3 ul of Lipofectamine® 2000 or 3 ul of Lipofectamine® 3000 as indicated and according to manufacturer's instructions, to deliver a GeneArt® CRISPR Nuclease vector targeting the HPRT locus. Transfection was also performed with GeneArt® CRISPR Nuclease RNA editing system targeting the HPRT locus and 3 ul of Formulation 21 lipid aggregate complex. RNA editing system utilizes a Cas9 mRNA, which was prepared via in vitro transcription with the Ambion® mMESSAGE mMACHINE® Kit, and a gRNA which was transcribed using the Ambion® MEGAshortscript™ Kit. Cells were harvested 72-hours post-transfection and cleavage efficiency was determined using the GeneArt® Genomic Cleavage Detection Kit.

Results are shown in FIG. 2A and FIG. 2B, which clearly demonstrate that using an mRNA based form of Cas9 with a guide-RNA for gene editing with the lipid aggregates described herein for transfection results in at least 4-fold more targeted cleavage of the host cell genome when compared to standard DNA based editing approaches.

Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

Patent 2024
Cells Cloning Vectors Clustered Regularly Interspaced Short Palindromic Repeats Cytokinesis Genes Genome Hyperostosis, Diffuse Idiopathic Skeletal Induced Pluripotent Stem Cells Lipids Lipofectamine lipofectamine 2000 RNA, Messenger Transcription, Genetic Transfection
Not available on PMC !

Example 5

Further to what has been shown in Example 4, it was then tested whether a psEON, as outlined in detail herein, could also yield pseudouridylation using the substrate GL-IDUA swap plasmids after transfection in cells. For this, HEK293T cells were transfected at 90-100% confluency, using PEI in a 6-well dish, with 500 ng GL-IDUA swap substrate plasmid and 2.5 μg the pugIntron-IDUA guide RNA expressing plasmid or transfected with 100 pmol Cy3-IDUA-A psEON oligonucleotide. Four days after transfection cells were washed and incubated at for 24 h. Total RNA was isolated as described and RT-PCR was performed as outlined above, except that 21 cycles were performed for all samples. RT-PCR products were separated by gel electrophoresis. Results are shown in FIG. 18. These indicate that when no DNA was transfected (meaning no plasmid or psEON, on top of the transfected substrate plasmid) that no GL39 RT-PCR product was detectable, although the 5S control was abundant. However, after co-transfection of the pugIntron-IDUA guide RNA-expressing plasmid and also after co-transfection with the Cy3-iDUA-A psEON, the product was detectable, indicating that read-through of the mRNA occurred, and that NMD was inhibited. This shows that the inventors of the present invention were able to obtain pseudouridylation not only by using intronically-embedded guide RNAs, but also with the short psEONs of the present invention.

Patent 2024
Cells Electrophoresis Hyperostosis, Diffuse Idiopathic Skeletal IDUA protein, human Introns Inventors Oligonucleotides Plasmids Reverse Transcriptase Polymerase Chain Reaction RNA RNA, Messenger Transfection

Example 8

To evaluate which lipid composition within the dendrimer nanoparticles lead to improved siRNA delivery, the identity and concentration of different phospholipids and PEG-lipids were varied. Three different cell lines (HeLa-Luc, A549-Luc, and MDA-MB231-Luc) were used. The cells were present at 10K cells per well and a 24 hour incubation. The readout was determined 24 hours post transfection. In the nanoparticles, DSPC and DOPE were used as phospholipids and PEG-DSPE, PEG-DMG, and PEG-DHD were used as PEG-lipids. The compositions contain a lipid or dendrimer:cholesterol:phospholipid:PEG-lipid mole ratio of 50:38:10:2. The mole ratio of lipid/dendrimer to siRNA was 100:1 with 100 ng dose being used. The RiboGreen, Cell-titer Fluor, and OneGlo assays were used to determine the effectiveness of these compositions. Results show the relative luciferase activity in HeLa-Luc cells (FIG. 17A), A549-Luc (FIG. 17B), and MDA-MB231-Luc (FIG. 17C). The six formulations used in the studies include: dendrimer (lipid)+cholesterol+DSPC+PEG-DSPE (formulation 1), dendrimer (lipid)+cholesterol+DOPE+PEG-DSPE (formulation 2), dendrimer (lipid)+cholesterol+DSPC+PEG-DMG (formulation 3), dendrimer (lipid)+cholesterol+DOPE+PEG-DMG (formulation 4), dendrimer (lipid)+cholesterol+DSPC+PEG-DSPE (formulation 5), and dendrimer (lipid)+cholesterol+DOPE+PEG-DHD (formulation 6).

Further experiments were run to determine which phospholipids showed the increased delivery of siRNA molecules. A HeLa-Luc cell line was used with 10K cells per well, 24 hour incubation, and readout 24 hours post transfections. The compositions contained either DOPE or DOPC as the phospholipid with PEG-DHD as the PEG-lipid. The ratio of lipid (or dendrimer):cholesterol:phospholipid:PEG-lipid was 50:38:10:2 in a mole ratio with the mole ratio of dendrimer (or lipid) to siRNA of 200:1. These compositions was tested at a 50 ng dose using the Cell-titer Fluor and OneGlo assays. These results are shown in FIGS. 18A & 18B.

Patent 2024
1,2-oleoylphosphatidylcholine Biological Assay Cell Lines Cells Cholesterol Dendrimers Figs HeLa Cells Lipid Nanoparticles Lipids Luciferases Nevus Obstetric Delivery Phospholipids polyethylene glycol-distearoylphosphatidylethanolamine RNA, Small Interfering Transfection

Top products related to «Transfection»

Sourced in United States, China, Germany, United Kingdom, Canada, Japan, France, Italy, Switzerland, Australia, Spain, Belgium, Denmark, Singapore, India, Netherlands, Sweden, New Zealand, Portugal, Poland, Israel, Lithuania, Hong Kong, Argentina, Ireland, Austria, Czechia, Cameroon, Taiwan, Province of China, Morocco
Lipofectamine 2000 is a cationic lipid-based transfection reagent designed for efficient and reliable delivery of nucleic acids, such as plasmid DNA and small interfering RNA (siRNA), into a wide range of eukaryotic cell types. It facilitates the formation of complexes between the nucleic acid and the lipid components, which can then be introduced into cells to enable gene expression or gene silencing studies.
Sourced in United States, China, Germany, Japan, United Kingdom, France, Canada, Italy, Australia, Switzerland, Denmark, Spain, Singapore, Belgium, Lithuania, Israel, Sweden, Austria, Moldova, Republic of, Greece, Azerbaijan, Finland
Lipofectamine 3000 is a transfection reagent used for the efficient delivery of nucleic acids, such as plasmid DNA, siRNA, and mRNA, into a variety of mammalian cell types. It facilitates the entry of these molecules into the cells, enabling their expression or silencing.
Sourced in United States, China, Germany, United Kingdom, Switzerland, Japan, France, Italy, Spain, Austria, Australia, Hong Kong, Finland
The Dual-Luciferase Reporter Assay System is a laboratory tool designed to measure and compare the activity of two different luciferase reporter genes simultaneously. The system provides a quantitative method for analyzing gene expression and regulation in transfected or transduced cells.
Sourced in United States, China, United Kingdom, Germany, Japan, France, Canada, Switzerland, Denmark, Belgium, Italy, Australia, Singapore, Spain, Colombia, Sweden, Netherlands, New Zealand, Poland, Pakistan, Lithuania
Lipofectamine RNAiMAX is a transfection reagent designed for efficient delivery of small interfering RNA (siRNA) and short hairpin RNA (shRNA) into a wide range of cell types. It is a cationic lipid-based formulation that facilitates the uptake of these nucleic acids into the target cells.
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, China, Japan, Canada, France, Switzerland, Italy, Australia, Belgium, Spain, Denmark, Ireland, Netherlands, Holy See (Vatican City State), Israel
Opti-MEM is a cell culture medium designed to support the growth and maintenance of a variety of cell lines. It is a serum-reduced formulation that helps to reduce the amount of serum required for cell culture, while still providing the necessary nutrients and growth factors for cell proliferation.
Sourced in United States, China, Germany, Canada, United Kingdom, Japan, France, Italy, Australia, Switzerland, Spain, Netherlands, Singapore, Cameroon, Colombia, Denmark, Lithuania
Lipofectamine 2000 reagent is a cationic lipid-based transfection reagent used for the delivery of nucleic acids, such as DNA and RNA, into eukaryotic cells. It facilitates the uptake of these molecules by the cells, enabling efficient gene expression or gene silencing studies.
Sourced in United States, China, United Kingdom, Germany, France, Australia, Canada, Japan, Italy, Switzerland, Belgium, Austria, Spain, Israel, New Zealand, Ireland, Denmark, India, Poland, Sweden, Argentina, Netherlands, Brazil, Macao, Singapore, Sao Tome and Principe, Cameroon, Hong Kong, Portugal, Morocco, Hungary, Finland, Puerto Rico, Holy See (Vatican City State), Gabon, Bulgaria, Norway, Jamaica
DMEM (Dulbecco's Modified Eagle's Medium) is a cell culture medium formulated to support the growth and maintenance of a variety of cell types, including mammalian cells. It provides essential nutrients, amino acids, vitamins, and other components necessary for cell proliferation and survival in an in vitro environment.
Sourced in United States, China, United Kingdom, Germany, Canada, Austria, Japan, Netherlands, Sweden, Spain, France, Switzerland, Italy, Lithuania
Lipofectamine 2000 is a cationic lipid-based transfection reagent used for the introduction of nucleic acids, such as DNA and RNA, into a variety of eukaryotic cell types. It facilitates the uptake and delivery of these molecules into the cells.
Sourced in United States, China, Germany, United Kingdom, Japan, Italy, France, Denmark, Belgium, Canada, Australia, Moldova, Republic of, Dominican Republic, Sweden, Singapore
Lipofectamine RNAiMAX is a transfection reagent designed for efficient delivery of small interfering RNA (siRNA) and other nucleic acids into mammalian cells. It is optimized to facilitate cellular uptake and intracellular trafficking of siRNA, leading to effective gene silencing.

More about "Transfection"

Transfection is the process of introducing genetic material, such as DNA or RNA, into cells using various techniques.
This powerful method is widely employed in biological research to investigate gene expression, protein function, and cellular pathways.
Transfection can be achieved through physical, chemical, or biological approaches, including the use of lipid-based reagents like Lipofectamine 2000 and Lipofectamine 3000, electroporation, and viral vectors.
The efficiency and toxicity of transfection methods can vary, and it's crucial to carefully select the optimal protocols to ensure successful gene delivery and minimize cellular stress.
Researchers often utilize tools like the Dual-Luciferase Reporter Assay System to assess transfection efficiency and optimize their experiments.
Lipofectamine RNAiMAX, a transfection reagent, is commonly used for the delivery of small interfering RNA (siRNA) and microRNA (miRNA) into cells, enabling the study of gene silencing and knockdown.
Supplemental media components, such as FBS and Opti-MEM, can also play a role in enhancing transfection outcomes.
PubCompare.ai revolutionizes the process of transfection optimization by leveraging AI-driven comparisons of research protocols.
This innovative platform helps researchers identify the best methods from literature, preprints, and patents, improving the reproducibility of their experiments.
By harnessing the power of artificial intelligence, PubCompare.ai enables seamless protocol discovery and optimization, empowering researchers to find the optimal transfection methods and products for their specific needs.
Whether working with Lipofectamine 2000 reagent, DMEM, or other transfection-related tools and techniques, PubCompare.ai's AI-powered insights can streamline the experimental process and enhance the overall quality of transfection-based research.
Experience the power of seamless protocol discovery and optimization with PubCompare.ai.