RNA Technologies

Ribonucleic acids (RNAs) are essential for transfer of genetic information and cell regulation. About 1-2% of the transcribed genome (transcriptome) consists of protein-coding messenger RNAs (mRNAs). The vast majority however, are non-coding RNAs (ncRNAs) of various length and function1-5protein synthesis-related housekeeping RNAs (transfer RNA (tRNA), ribosomal RNA (rRNA)) and regulatory RNAs (> 200 nt: long non-coding RNA (lncRNA), < 200 nt: e.g. micro RNA (miRNA), small interfering RNA (siRNA) or PIWI-interacting RNA (piRNA)). Circular RNAs (circRNAs) have initially been categorized as non-coding RNAs4 but contain protein-coding functions5 as well.

RNA Synthesis

In vitro synthesis of RNA (> 20 nt up to several thousand nt) is catalyzed by bacteriophage RNA polymerases using linear DNA as a template (in vitro transcription). T7 RNA polymerase is the most efficient and widely used RNA polymerase. A modified version (T7 P&L RNA polymerase) with proline 266 replaced by leucine (P266L) has been associated with decreased abortive transcription[1], increased 5′ homogeneity of transcripts synthesized from A-initiating phi2.5 promoter[2], increased 5′ incorporation efficiency of GTP analogs[3].

140 – 160 µg RNA are synthesized after 30 min incubation with our HighYield formulation (1 μg T7 control template, 1.4 kb RNA transcript).

Products & Ordering
HighYield T7 RNA Synthesis Kit RNT-101 RNA synthesis via in vitro transcription with T7 RNA Polymerase HighYield T7 P&L RNA Synthesis Kit RNT-201 RNA synthesis via in vitro transcription with a modified T7 RNA Polymerase (P266L)

Selected References

[1] Guillerez et al. (2005) A mutation in T7 RNA polymerase that facilitates promoter clearance. Natl. Acad. Sci. U.S.A102:5958.
[2] Salvail-Lacoste et al. (2018) Affinity purification of T7 RNA transcripts with homogeneous ends using ARiBo and CRISPR tags. RNA19:1003.
[3] Lyon et al. (2018) A T7 RNA Polymerase Mutant Enhances the Yield of 5′-Thienoguanosine-Initiated RNAs. ChemBioChem19:142.

Synthetic mRNAs are widely used as alternative molecules to plasmid DNA for modulating protein levels in a therapeutic and research context. Their biological functionality depends on both a 5’ Cap structure and nucleotide modifications that ensure efficient translation and reduced immunogenicity [1]-[16]. The classic combination of modifications consists of ARCA (Cap 0), 5-methylcytidine and pseudouridine which are conveniently introduced by correspondingly modified nucleotides via in vitro transcription.

Beyond this classic combination, other mRNA modifications (e.g. N1-Methylpseudouridine [10], N4-Acetyl-Cytidine [5,6]) or the introduction of a Cap 1 moiety [15,16]) have been demonstrated to increase translation efficiency and/or to reduce immunogenicity as well. The optimal combination of Cap moiety & nucleotide modification however, needs to be individually determined for each mRNA target. Investigations can conveniently be performed with HighYield T7 mRNA Testkits.

Table 1: Overview on available (m)RNA synthesis kit configurations

Modified nucleotide w/o cap analog ARCA ( m27,3‘-OGP3G) Cap 0, G-initiating Cap 1 AG (3‘-OMe) (m27,3‘-OGP3(2‘-OMe)ApG) Cap 1, A-initiating
none HighYield T7 RNA Synthesis Kit HighYield T7 ARCA mRNA Synthesis Kit HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit
HighYield T7 Cap Analog Testkit
Pseudo-UTP HighYield T7 mRNA Synthesis Kit (Ψ-UTP) HighYield T7 ARCA mRNA Synthesis Kit (Ψ-UTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (Ψ-UTP)
N1-Methylpseudo-UTP HighYield T7 mRNA Synthesis Kit (me1Ψ-UTP) HighYield T7 ARCA mRNA Synthesis Kit (me1Ψ-UTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (me1Ψ-UTP
5-Methoxy-UTP HighYield T7 mRNA Synthesis Kit (5moUTP) HighYield T7 ARCA mRNA Synthesis Kit (5moUTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (5moUTP)
2-Thio-UTP HighYield T7 mRNA Synthesis Kit (s2UTP) HighYield T7 ARCA mRNA Synthesis Kit (s2UTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (s2UTP)
5-Methyl-CTP HighYield T7 mRNA Synthesis Kit (m5CTP) HighYield T7 ARCA mRNA Synthesis Kit (m5CTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (m5CTP)
N4-Acetyl-CTP HighYield T7 mRNA Synthesis Kit (ac4CTP) HighYield T7 ARCA mRNA Synthesis Kit (ac4CTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (ac4CTP)
N6-Methyl-ATP HighYield T7 mRNA Synthesis Kit (m6ATP) HighYield T7 ARCA mRNA Synthesis Kit (m6ATP) n/a
N1-Methyl-ATP HighYield T7 mRNA Synthesis Kit (m1ATP) HighYield T7 ARCA mRNA Synthesis Kit (m1ATP) n/a
5-Methyl-CTP & Pseudo-UTP HighYield T7 mRNA Synthesis Kit (m5CTP/Ψ-UTP) HighYield T7 ARCA mRNA Synthesis Kit (m5CTP/Ψ-UTP) HighYield T7 Cap 1 AG (3‘-OMe) mRNA Synthesis Kit (m5CTP/Ψ-UTP)
Pseudo-UTP N1-Methylpseudo-UTP 5-Methoxy-UTP 2-Thio-UTP HighYield T7 Uridine Modification Testkit
5-Methyl-CTP N4-Acetyl-CTP HighYield T7 Cytidine Modification Testkit
Pseudo-UTP N1-Methylpseudo-UTP 5-Methoxy-UTP 2-Thio-UTP 5-Methyl-CTP N4-Acetyl-CTP N6-Methyl-ATP N1-Methyl-ATP HighYield T7 mRNA Modification Testkit
Products & Ordering
HighYield T7 RNA Synthesis Kit RNT-101 RNA synthesis via in vitro transcription with T7 RNA Polymerase HighYield T7 ARCA mRNA Synthesis Kit RNT-102 Synthesis of ARCA-capped (m)RNA
HighYield T7 ARCA mRNA Synthesis Kit (m5CTP/Ψ-UTP) RNT-103 Synthesis of ARCA-capped, 5-methylcytidine & pseudouridine-modified (m)RNA HighYield T7 mRNA Synthesis Kit (m5CTP/Ψ-UTP) RNT-104 Synthesis of 5-methylcytidine and pseudouridine-modified (m)RNA
Poly(A) Tailing Enzyme Testkit RNT-004 in vitro Polyadenylation of (m)RNA with E. coli and Yeast Poly(A) Polymerase HighYield T7 ARCA mRNA Synthesis Kit (Ψ-UTP) RNT-114 Synthesis of ARCA-capped & pseudouridine-modified (m)RNA
HighYield T7 ARCA mRNA Synthesis Kit (me1Ψ-UTP) RNT-115 Synthesis of ARCA-capped & N1-Methylpseudouridine-modified (m)RNA HighYield T7 ARCA mRNA Synthesis Kit (5moUTP) RNT-116 Synthesis of ARCA-capped & 5-methoxyuridine-modified (m)RNA
HighYield T7 ARCA mRNA Synthesis Kit (s2UTP) RNT-117 Synthesis of ARCA-capped & 2-thiouridine-modified (m)RNA HighYield T7 ARCA mRNA Synthesis Kit (m5CTP) RNT-118 Synthesis of ARCA-capped & 5-methylcytidine-modified (m)RNA
HighYield T7 ARCA mRNA Synthesis Kit (ac4CTP) RNT-119 Synthesis of ARCA-capped & N4-acetylcytosine-modified (m)RNA HighYield T7 ARCA mRNA Synthesis Kit (m6ATP) RNT-120 Synthesis of ARCA-capped & N6-methyladenosine-modified (m)RNA
HighYield T7 ARCA mRNA Synthesis Kit (m1ATP) RNT-121 Synthesis of ARCA-capped & N1-methyladenosine-modified (m)RNA HighYield T7 mRNA Synthesis Kit (Ψ-UTP) RNT-106 Synthesis of pseudouridine-modified (m)RNA
HighYield T7 mRNA Synthesis Kit (me1Ψ-UTP) RNT-107 Synthesis of N1-Methylpseudouridine-modified (m)RNA HighYield T7 mRNA Synthesis Kit (5moUTP) RNT-108 Synthesis of 5-methoxyuridine-modified (m)RNA
HighYield T7 mRNA Synthesis Kit (s2UTP) RNT-109 Synthesis of 2-thiouridine-modified (m)RNA HighYield T7 mRNA Synthesis Kit (m5CTP) RNT-110 Synthesis of 5-methylcytidine-modified (m)RNA
HighYield T7 mRNA Synthesis Kit (ac4CTP) RNT-111 Synthesis of N4-acetylcytosine-modified (m)RNA HighYield T7 mRNA Synthesis Kit (m6ATP) RNT-112 Synthesis of N6-methyladenosine-modified (m)RNA
HighYield T7 mRNA Synthesis Kit (m1ATP) RNT-113 Synthesis of N1-methyladenosine-modified (m)RNA

Selected References

[1] Karikó et al.(2005) Suppression of RNA Recognition by Toll-like Receptors: The Impact of Nucleoside Modification and the Evolutionary Origin of RNA. Immunity23:165.
[2] Karikó et al.(2008) Incorporation of Pseudouridine into mRNA Yields Superior Nonimmunogenic Vector With Increased Translational Capacity and Biological Stability. Mol. Ther.16(11):1833.
[3] Kormann et al.(2011) Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nature Biotechnology29(2):154.
[4] Warren et al.(2011) Highly Efficient Reprogramming to Pluripotency and Directed Differentiation of Human Cells with Synthetic Modified mRNA. Cell Stem Cell7:618.
[5] Svitkin et al.(2017) N1-methyl-pseudouridine in mRNA enhances translation through eIF2alpha-dependent and independent mechanisms by increasing ribosome density. Nucleic Acid Res45(10):6023.
[6] Andies et al.(2015) N1-methylpseudouridine-incorporated mRNA outperforms pseudouridine-incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice. J. Control. Release217:337.
[7] Li et al.(2016) Effects of Chemically Modified Messenger RNA on Protein Expression. Bioconjugate Chem.27:849.
[8] Arango et al.(2018) Acetylation of Cytidine in mRNA Promotes Translation Efficiency. Cell175(7):1872.
[9] Sinclair et al.(2017) Profiling Cytidine Acetylation with Specific Affinity and Reactivity. ACS Chem. Neurosci.12(12):2922.
[10] Dominissini et al.(2016) The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature530:441.
[11] Wienert et al. (2018) In vitro transcribed guide RNAs trigger an innate immune response via RIG-I pathway. PLoS Biol. 16 (7) :e2005840.
[12] Kim et al. (2018) CRISPR RNAs trigger innate immune responses in human cells. Genome Res. 28 (3):367.
[13] Badieyan et al. (2019) Concise Review: Application of Chemically Modified mRNA in Cell Fate Conversion and Tissue Engineering. Stem Cells Translational Medicine8:833.
[14] Hadas et al. (2019) Optimizing Modified mRNA In Vitro Synthesis Protocol for Heart Gene Therapy. Molecular Therapy: Methods & Clinical Development 14:300.
[15] Shatkinet al. (1976) Capping of eukaryotic mRNAs. Cell 9(4):645.
[16] Gallowayet al.(2019) mRNA cap regulation in mammalian cell function and fate. Biochimica et Biophysica Acta 1862(3):270.

In vitro synthesis of RNA is catalyzed by bacteriophage (SP6, T7 or T3) RNA polymerases using linear DNA as a template. Ribonucleoside triphosphates (NTPs) are the building blocks of these in vitro transcription reactions.

Some naturally occuring RNAs such as messenger RNAs (mRNAs) or long non-coding RNAs (lncRNAs) contain a variety of modifications e.g. 5‘-end guanosinemethylation (5‘-Cap), 3‘-end adenosine (poly(A)) tail or several internal base modifications which markedly influence their biological function (e.g. translation efficiency, stability or immunogenicity).

These structural features can be introduced during in vitro transcription via correspondingly modified NTPs and cap analoga to obtain biologically functional synthetic RNAs.

Kits for enzymatic RNA Synthesis and poly(A) tailing are available as well.

Table 1: Raw materials for RNA Synthesis

Modified NTPsUnmodified NTPsCap AnalogaProteins
5-Methyl-CTP (100 mM)NTP bundle (4x 100 mM ATP, CTP, GTP, UTP)m7GP3G (100 mM)T7 RNA Polymerase (HC)
N4-Acetyl-CTP (100 mM)m7,3‘-OGP3G (ARCA) (100 mM)T7 P&L RNA Polymerase (HC)
Pseudo-UTP (100 mM)ATP (100 mM)m7,3‘-OGP3(2’OMe)ApG (100 mM)T7 3M RNA Polymerase (HC)
N1-Methylpseudo-UTP (100 mM)CTP (100 mM)RNAse Inhibitor - recombinant
5-Methoxy-UTP (100 mM)GTP (100 mM)
2-Thio-UTP (100 mM)UTP (100 mM)
N6-Methyl-ATP (100 mM)
Products & Ordering
m7GP3G (Monomethylated Cap Analog) – Solution NU-852 5-Methyl-CTP NU-1138 m5CTP
m27,3′-OGP3G (ARCA Cap Analog) – Solution NU-855 N4-Acetyl-CTP NU-988 ac4CTP
Pseudo-UTP NU-1139 Ψ-UTP N1-Methylpseudo-UTP NU-890 me1Ψ-UTP
5-Methoxy-UTP NU-972 5moUTP 2-Thio-UTP NU-1151 s2UTP
N6-Methyl-ATP NU-1101 m6ATP NTP Bundle NU-1014 4 x 100 mM (ATP, CTP, GTP, UTP)
ATP – Solution NU-1010-SOL 100 mM Sodium salt solution CTP – Solution NU-1011-SOL 100 mM Sodium salt solution
GTP – Solution NU-1012-SOL 100 mM Sodium salt solution UTP – Solution NU-1013-SOL 100 mM Sodium salt solution
T7 RNA Polymerase HC RNT-008 Highly concentrated T7 RNA Polymerase for set-up of high yield in vitro transcription reactions T7 P&L RNA Polymerase HC RNT-018 Highly concentrated and modified T7 RNA Polymerase for set-up of high yield in vitro transcription reactions
T7 3M RNA Polymerase HC RNT-019 Highly concentrated triple mutant T7 RNA Polymerase for set-up of high yield in vitro transcription reactions

HighYield T7 RNAi Kit is designed to produce large amounts of double-stranded RNA (dsRNA) > 200 bp via T7 RNA Polymerase-mediated in vitro transcription. The resulting dsRNA can subsequently be used for RNA interference (RNAi) applications.

Products & Ordering
HighYield T7 RNAi Kit RNT-134 dsRNA synthesis for RNAi applications via T7 RNA Polymerase-mediated in vitro transcription

RNA Labeling & Modification

Random labeled single-stranded RNA probes can be synthesized by in vitro transcription. The reaction is catalyzed by bacteriophage T7 RNA polymerase that incorporates labeled NTPs (mostly UTP) as substitute for their natural counterpart using linear, RNA probe-encoding DNA as template.

The optimal ratio of labeled NTP/NTP in terms of product yield and labeling efficiency depends on both the type of labeled NTP and final application. Individual optimization of labeled NTP/NTP ratio can easily be achieved with the single nucleotide format of our HighYield T7 RNA Labeling Kits. They offer maximum optimization flexibility in contrast to fixed NTP labeling mixes combined with high product yields. The combination of (poly)-HRP-conjugated Biotin/Digoxigenin detection reagents with AF 488 (Alexa Fluor® 488)-, AF546 (Alexa Fluor® 546)- or AF594 (Alexa Fluor® 594)-labeled tyramide further increases detection sensitivity of up to 100-fold.

HaptenFluorophoreCopper-free CLICK
HighYield T7 Digoxigenin RNA Labeling KitHighYield T7 Fluorescein RNA Labeling KitHighYield T7 Azide RNA Labeling Kit
HighYield T7 Biotin16 RNA Labeling KitHighYield T7 Cy3 RNA Labeling Kit
HighYield T7 Biotin11 RNA Labeling KitHighYield T7 Cy5 RNA Labeling Kit
HighYield T7 AF405 RNA Labeling Kit
HighYield T7 Atto 488 RNA Labeling Kit
HighYield T7 AF488 RNA Labeling Kit
HighYield T7 AF555 RNA Labeling Kit
HighYield T7 AF594 RNA Labeling Kit
HighYield T7 AF647 RNA Labeling Kit
Products & Ordering
HighYield T7 Digoxigenin RNA Labeling Kit RNT-101-DIGX Preparation of randomly Digoxigenin-modified RNA probes by in vitro transcription with Dioxigenin-11-UTP HighYield T7 Biotin16 RNA Labeling Kit RNT-101-BIO16 Preparation of randomly Biotin-modified RNA probes by in vitro transcription with Biotin-16-UTP
HighYield T7 Biotin11 RNA Labeling Kit RNT-101-BIOX Preparation of randomly Biotin-modified RNA probes by in vitro transcription with Biotin-11-UTP HighYield T7 Biotin11 RNA Labeling Kit RNT-101-BIOX Preparation of randomly Biotin-modified RNA probes by in vitro transcription with Biotin-11-UTP
HighYield T7 Cy3 RNA Labeling Kit RNT-101-CY3 Preparation of randomly Cy3-modified RNA probes by in vitro transcription with UTP-X-CY3 HighYield T7 AF647 RNA Labeling Kit RNT-101-AF647 Preparation of randomly AF647-modified RNA probes by in vitro transcription with UTP-PEG5-AF647
HighYield T7 Cy5 RNA Labeling Kit RNT-101-CY5 Preparation of randomly Cy5-modified RNA probes by in vitro transcription with UTP-X-CY5 HighYield T7 Azide RNA Labeling Kit RNT-101-AZ Preparation of randomly Azide-modified RNA probes by in vitro transcription with 5-Azido-C3-UTP
HighYield T7 AF405 RNA Labeling Kit RNT-101-AF405 Preparation of randomly AF488-modified RNA probes by in vitro transcription with UTP-PEG5-AF405 AF488 tyramide reagent RNT-012 also known as Alexa Fluor® 488 tyramide
HighYield T7 Atto488 RNA Labeling Kit RNT-101-488 Preparation of randomly Atto488-modified RNA probes by in vitro transcription with UTP-ATTO-488 AF546 tyramide reagent RNT-013 also known as Alexa Fluor® 546 tyramide
HighYield T7 AF488 RNA Labeling Kit RNT-101-AF488 Preparation of randomly AF488-modified RNA probes by in vitro transcription with UTP-PEG5-AF488 AF594 tyramide reagent RNT-014 also known as Alexa Fluor® 594 tyramide
HighYield T7 AF555 RNA Labeling Kit RNT-101-AF555 Preparation of randomly AF555-modified RNA probes by in vitro transcription with UTP-PEG5-AF555 AF488 Streptavidin Conjugate RNT-015 Streptavidin, Alexa Fluor® 488 Conjugate
HighYield T7 AF594 RNA Labeling Kit RNT-101-AF594 Preparation of randomly AF594-modified RNA probes by in vitro transcription with UTP-PEG5-AF594 AF594 Streptavidin Conjugate RNT-016 Streptavidin, Alexa Fluor® 594 Conjugate

One-step labeling of short RNA is achieved via T4 RNA Ligase 1-mediated incorporation of labeled pCps.

AF (Alexa Fluor®)- and ATTO® dyes are hydrophilic fluorescent dyes with remarkable photostability and thus ideally suited for fluorescence imaging applications. Biotin- and Desthiobiotin labeling allows efficient RNA pull-down.

Products & Ordering
pCp-ATTO-488 NU-1706-488 pCp-Cy5 NU-1706-CY5
pCp-AF488 NU-1706-AF488 pCp-Biotin NU-1706-BIO
pCp-Cy3 NU-1706-CY3 pCp-Desthiobiotin NU-1706-Desthiobio
pCp-AF555 NU-1706-AF555 pCp-Amine NU-1706
pCp-AF594 NU-1706-AF594 pCp-Azide NU-1708
pCp-ATTO-643 NU-1706-643 T4 RNA Ligase 1 RNT-007 recombinant, E. coli
pCp-AF647 NU-1706-AF647 RNase Inhibitor – recombinant PCR-392 Mus musculus, recombinant
Products & Ordering
E. coli Poly(A) Polymerase RNT-005 recombinant, E. coli overexpressing E. coli Poly(A) Polymerase Yeast Poly(A) Polymerase RNT-006 recombinant, E. coli overexpressing Saccharomyces cerevisiae Poly(A) Polymerase
Poly(A) Tailing Enzyme Testkit RNT-004 in vitro Polyadenylation of (m)RNA with E. coli and Yeast Poly(A) Polymerase T4 RNA Ligase 1 RNT-007 recombinant, E. coli

RNA Analysis & Detection

RNA aptamers are specific RNA sequences that can be detected by cell-permeable small-molecule dyes. Dye fluorescence is significantly enhanced upon complex formation thus aptamers are versatile tags for imaging and functional analysis of coding and non-coding RNA both in vitro and in live cells [1-4]. Attachement of a specific aptamer sequence to a RNA of interest can be performed e.g. by in vitro transcription.

The most commonly used aptamer/dye systems are based on dye derivatives of the GFP chromophore 4-Hydroxybenzlidene (HBI). These 3,5-difluor-HBI dyes (DFHBI, DFHBI-1T and DFHO) show negligible fluorescence in aqueous solutions but light-up upon aptamer complex formation (Tab. 1).

Table 1: Spectroscopic properties of aptamer/dye complexes

Dye Light-up aptamer Exc.max/Em.max [nm] of aptamer/dye complex ɛmax [M-1cm-1] of aptamer/dye complex Reference
DFHBI Spinach 469/501 24271 [5]
Spinach 2 447/501 22000 [6]
DFHBI-1T Spinach 2 482/505 31000 [6]
Broccoli 472/507 29600 [7]
DFHO Corn 505/545 29000 [8]
Products & Ordering
DFHBI RNT-001 Mimic of green fluorescent protein (GFP) fluorophore DFHBI-1T RNT-002 Mimic of green fluorescent protein (GFP) fluorophore DFHO RNT-003 Mimic of red fluorescent protein (RFP) fluorophore

Selected References

[1] Neubacher et al. (2019) RNA Structure and Cellular Applications of Fluorescent Light-Up Aptamers. Angew. Chem. Int. Ed. 58:1266.
[2] Ouellet (2016) RNA Fluorescence with Ligth-Up Aptamers. Front. Chem 4:29.
[3] George et al. (2018) Intracelluar RNA-tracking methods. Open Biol 8:180104.
[4] Dologosheina et al. (2016) Fluorophore Binding RNA aptamers and their application. Wires RNA 7(6):843.
[5] Paige et al. (2011) RNA mimics of green fluorescent protein. Science 333(6042):642.
[6] Song et al. (2014) Plug-and-Play fluorophores extend the Spectral Properties of Spinach. J. Am. Chem. Soc. 136:1198.
[7] Filonov et al. (2014) Broccoli: Rapid Selection of an RNA Mimic of Green Fluorescent Protein by Fluorescence-Based Selection and Directed Evolution. J. Am. Chem. Soc. 136:16299.
[8] Song et al. (2017) Imaging RNA polymerase III transcription using a photostable RNA-fluorophore complex. Nat. Chem. Biol. 13(11):1187.

Northern Blot analysis is a reliable hybridization technique frequently used for the detection of a specific RNA transcript (e.g. mRNA) within a complex mixture. Historically, mainly radioactively labeled hybridization probes have been used however, non-radioactive labels such as Digoxigenin, Biotin or near-infrared fluorescent dyes are attractive alternatives in terms of probe stability, handling convenience and improved safety profiles.[1-3]

Using RNA probes (“riboprobes”) generated by in vitro transcription instead of double-stranded DNA probes results in significantly higher assay sensitivity due to increased riboprobe affinity to RNA target and a higher thermodynamic stability of RNA:RNA complex.[4-6] Riboprobes thus allow a reduced exposure time and more stringent washing conditions resulting in a lower background signal. The combination of (poly)-HRP-conjugated detection reagents with AF 488 (Alexa Fluor® 488)-, AF546 (Alexa Fluor® 546)- or AF594 (Alexa Fluor® 594)-labeled tyramide further increases detection sensitivity of up to 100-fold.

Products & Ordering
HighYield T7 Digoxigenin RNA Labeling Kit RNT-101-DIGX Preparation of randomly Digoxigenin-modified RNA probes by in vitro transcription with Dioxigenin-11-UTP HighYield T7 Biotin16 RNA Labeling Kit RNT-101-BIO16 Preparation of randomly Biotin-modified RNA probes by in vitro transcription with Biotin-16-UTP
HighYield T7 Azide RNA Labeling Kit RNT-101-AZ Preparation of randomly Azide-modified RNA probes by in vitro transcription with 5-Azido-C3-UTP AF488 tyramide reagent RNT-012 also known as Alexa Fluor® 488 tyramide
AF546 tyramide reagent RNT-013 also known as Alexa Fluor® 546 tyramide AF488 Streptavidin Conjugate RNT-015 Streptavidin, Alexa Fluor® 488 Conjugate
AF594 tyramide reagent RNT-014 also known as Alexa Fluor® 594 tyramide AF594 Streptavidin Conjugate RNT-016 Streptavidin, Alexa Fluor® 594 Conjugate

Selected References

[1] Miller et al. (2018) Near-infrared fluorescent northern blot. RNA 24(12):1871.
[2] O’Neill JW et al. (1994) Double-label in situ hybridization using biotin and digoxigenin-tagged RNA probes. Biotechniques 17(5):870.
[3] Brauburger et al. (2017) Nonradioactive Northern Blot Analysis to Detect Ebola Virus Minigenomic mRNA. Methods in Molecular Biology 1628: DOI 10.1007/978-1-4939-7116-9_11.
[4] Melton et al. (1984) Efficient in vitro synthesis of biologicaly active RNA and RNA hybridization probes from plasmids containing a bacteriophage SP6 promotor. Nucleic Acids Research 12(18):7035.
[5] Srivastava et al. (2000) Analysis of RNA by Northern Blotting Using Riboprobes. The Nucleic Acid Protocols Handbook 38:249.
[6] Srivastava et al. (1991) Use of riboprobes for Northern blotting analysis. Biotechniques 11:584.

RNA structure determination can be performed by liquid state nuclear magnetic resonance (NMR) spectroscopy that requires millimolar amounts of isotopically labeled RNA to obtain well-resolved signals.

A successful approach to generate sufficient isotopically labeled RNA amounts is the in vitro transcription-mediated synthesis of 19F-labeled RNA using fluorinated NTPs[1]. 2F-ATP or 5F-UTP are incorporated instead of their natural counterparts in a non-perturbing way (intact base-pairing properties) and simultaneously function as a sensitive NMR reporter group (larger chemical shift dispersion than 1H)[1].

Selected References

[1] Graber et al. (2008) 19F NMR Spectroscopy for the Analysis of RNA Secondary Structure Populations. J. Am. Chem. Soc. 130 (51):17230.
[2] Sochor et al. (2016) S(19)F-labeling of the adenine H2-site to study large RNAs by NMR spectroscopy. J. Biomol. NMR 64 (1):63.
[3] Scott et al. (2004) Enzymatic synthesis and 19F NMR studies of 2-fluoroadenine-substituted RNA. J. Am. Chem. Soc. 126 (38):11776.

Products & Ordering
HighYield T7 P&L RNA NMR Kit (5F-UTP) RNT-202 Synthesis of 5-Fluoro-modified RNA HighYield T7 P&L RNA NMR Kit (2F-ATP) RNT-203 Synthesis of 2-Fluoro-modified RNA
Products & Ordering
HighYield T7 RNA Crosslinking Kit (4-Thio-UTP) RNT-135 Synthesis of 4-Thio-modified RNA

Double-stranded (ds)RNA formation is a hallmark of viral infections that is essential for the induction of innate immunity. dsRNA is also involved in gene silencing and produced as side product during in vitro transription-based RNA synthesis. Anti-dsRNA monoclonal antibodies are efficient tools for the detection of dsRNA in cell culture & tissues such as FFPE samples as well as in vitro transcribed (m)RNA preparations [1-7] (Tab. 1) e.g.

  • for characterization & detection of viruses with dsRNA genomes or intermediates (including SARS, Hepatitis C, Dengue or West Nile Virus).
  • as diagnostic tool for determination whether an unknown pathogen is of viral or bacterial origin.
  • for quality control of in vitro transcribed (m)RNA preparations.

 

Table 1: Overview on available dsRNA detection products

Product Description Application Selection guide
Anti-dsRNA monoclonal antibody J2 Mouse, IgG2a, kappa light chain ELISA, IF, FACS, IHC, IP, Dot Blot, ChIP, affinity purification, immunoelectron microscopy Gold standard for dsRNA detection Recommended for quality control of in vitro transcribed (m)RNA
Anti-dsRNA monoclonal antibody K1 Mouse, IgG2a, kappa light chain Recommended for Poly I:C detection J2 alternative in case of cross reactions
Anti-dsRNA monoclonal antibody K2 Mouse, IgM, kappa light chain ELISA, IHC, Dot Blot Isotype alternative to J2 & K1 Recommended for (Sandwich-) ELISA
dsRNA 142 bp synthetic dsRNA Positive control for Anti-dsRNA monoclonal antibodies J2, K1 and K2 n/a

Selected References

[1] Schönborn et al. (1991) Monoclonal antibodies to double-stranded RNA as probes of RNA structure in crude nucleic acid extracts. Nucleic Acids Res.19: 2993.
[2] Lukacs (1994) Detection of virus infection in plants and differentiation between coexisting viruses by monoclonal antibodies to double-stranded RNA. J. Virol. Methods47: 255.
[3] Lukacs (1997) Detection of sense:antisense duplexes by structure-specific anti-RNA antibodies. In: Antisense Technology. A Practical Approach, C. Lichtenstein and W. Nellen (eds), pp. 281-295. IRL Press, Oxford
[4] Weber et al. (2006) Double-Stranded RNA is produced by positive-strand RNA viruses and DNA viruses but not in detectable amounts by negative-strand RNA viruses. Journal of Virology 80(10): 5059.
[5] Knoops et al. (2008) SARS-Coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum. PLOS Biology 6(9): e226.
[6] Richardson et al. (2010) Use of antisera directed against dsRNA to detect viral infections in formalin-fixed paraffin-embedded tissue. Journal of Clinical Virology 49: 180.
[7] Karikó et al. (2011) Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA. Nucleic Acids Research 39(21): e142.

Products & Ordering
Anti-dsRNA monoclonal antibody J2 RNT-SCI-10010 mouse, IgG2a, kappa chain Anti-dsRNA monoclonal antibody K1 RNT-SCI-10020 mouse, IgG2a, kappa chain
Anti-dsRNA monoclonal antibody K2 RNT-SCI-10030 mouse, IgM, kappa chain dsRNA 142 bp RNT-SCI-10080100 Positive control for dsRNA detection by Anti-dsRNA monoclonal J2, K1 und K2 antibodies

Poly (A) carrier RNA improves recovery of low RNA and DNA amounts from biological samples presumably by enhanced aggregation. Poly (A) carrier RNA-purified RNA or DNA is compatible with subsequent downstream analyses such as (RT-)qPCR or digital droplet PCR (ddPCR).

Products & Ordering
Poly (A) carrier RNA RNT-017 Polyadenylic acid, potassium salt
Products & Ordering
HighYield T73M Aptamer Synthesis Kit (2’F-dUTP) RNT-304 Synthesis of 2′-Fluoro-modified RNA HighYield T73M Aptamer Synthesis Kit (2’F-dGTP) RNT-303 Synthesis of 2′-Fluoro-modified RNA
HighYield T73M Aptamer Synthesis Kit (2’F-dCTP) RNT-302 Synthesis of 2′-Fluoro-modified RNA HighYield T73M Aptamer Synthesis Kit (2’F-dATP) RNT-301 Synthesis of 2′-Fluoro-modified RNA

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) are the hallmark of a bacterial defense system that forms the basis for CRISPR/Cas sequence-specific gene targeting technology [1-5].

CRISPR/Cas systems typically consists of two components:

1.

     RNA-guided CRISPR-associated (Cas) endonuclease(e.g. class II endonuclease Cas9)

2.

Sequence-specific guide RNA

     (e.g. sgRNA)

There are several ways to introduce Cas endonucleases and sequence-specific guide RNA into cells however, delivery as ribonucleoprotein (RNP) complex or RNA molecules shows the highest efficiency in most cases.[6-7]

Component 1Component 2
Cas endonucleases(s)gRNA Synthesis

CRISPR/Cas principle:
Complexation of guide RNA and Cas endonuclease is required to exert their function: The guide RNA directs the binding of this ribonucleoprotein (RNP) complex to the complementary gene region of interest that is site-specifically cleaved prior to gene editing by a Cas endonuclease upstream of a Cas endonuclease-specific recognition sequence[1].
Cas endonucleases can conveniently be programmed to target different gene sites by altering the guide RNA sequence only. Due to this simplicity, CRISPR/Cas systems are the most flexible and efficient site-directed gene targeting tools currently available [2,3].

Selected References

[1] Jinek et al. (2012) A programmable dual-RNA-guided, DNA endonuclease in adaptive bacterial immunity. Science 337(6096):816.
[2] Wang et al. (2016) CRISPR/Cas9 in Genome Editing and Beyond. Annu. Rev. Biochem. 85:227.
[3] Gaj et al. (2013) ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 31(7):397.
[4] Aldi et al. (2018) The CRISPR tool kit for genome editing and beyond. Nature Communications 9:1911.
[5] Pickar-Oliver et al. (2018) The next generation of CRISPR–Cas technologies and applications. Nature Reviews Molecular Cell Biology 20:507.
[6] DeWitt et al. (2017) Genome editing via delivery of Cas9 ribonucleoprotein. Methods 121:9.
[7] Kim et al. (2014) Highly efficient RNA-guided genome editing in humancells via delivery of purified Cas9 ribonucleoproteins. Genome Research 24:1012.

T7 RNA Polymerase-mediated in vitro transcription is an easy, fast and currently the most cost-efficient way of guide RNAs synthesis.

HighYield T7 sgRNA Synthesis Kit (SpCas9) allows the cloning-free preparation of SpCas9-specific single-guide RNA (sgRNA).[1,2] Other (s)gRNA-encoding T7 DNA templates (e.g with a different SpCas9 scaffold or for different Cas endonucleases) can efficiently be in vitro transcribed with the HighYield T7 RNA Synthesis Kit.

Selected References

[1] Jinek et al. (2012) A programmable dual-RNA guided DNA Endonuclease in adaptive bacterial immunity. Science 337:816.
[2] Modzelewski et al. (2018) Efficient mouse genome engineering by CRISPR-EZ technology. Nature Protocols 13 (6) :1253.
[3] Hendel et al. (2015) Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nature Biotechnology 33 (9):985.

Products & Ordering
HighYield T7 sgRNA Synthesis Kit (SpCas9) RNT-105 Cloning-free preparation of SpCas9-specific sgRNA by in vitro transcription HighYield T7 RNA Synthesis Kit RNT-101 RNA synthesis via in vitro transcription with T7 RNA Polymerase

Selected References

[1] Cech et al. (2014) The noncoding RNA revolution – trashing old rules to forge new ones. Cell 157:17.
[2] Morris et al. (2014) The rise of regulatory RNA. Nature Reviews Genetics 15:423.
[3] Uszczynska-Ratajczak et al. (2018) Towards a complete map of the human long non-coding RNA transcriptome. Nature Reviews Genetics 19:423.
[4] Wiluz et al. (2013) A circuitous route to noncoding RNA. Science 340 (6131):440.
[5] Pamudurti et al. (2017) Translation of circRNAs. Molecular Cell 66:9.

SUPPORT

outstanding technical support

PRODUCT

we offer a full product guarantee

DELIVERY

we offer free delivery to UK universities and non profit organisations