Real-time quantification of microRNAs by stem–loop RT–PCR (original) (raw)

Journal Article

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

,

Applied Biosystems 850 Lincoln Centre Drive, Foster City, CA 94404, USA

Search for other works by this author on:

... Show more

Published:

01 January 2005

Revision received:

08 July 2005

Accepted:

25 October 2005

Cite

Caifu Chen, Dana A. Ridzon, Adam J. Broomer, Zhaohui Zhou, Danny H. Lee, Julie T. Nguyen, Maura Barbisin, Nan Lan Xu, Vikram R. Mahuvakar, Mark R. Andersen, Kai Qin Lao, Kenneth J. Livak, Karl J. Guegler, Real-time quantification of microRNAs by stem–loop RT–PCR, Nucleic Acids Research, Volume 33, Issue 20, 1 November 2005, Page e179, https://doi.org/10.1093/nar/gni178
Close

Navbar Search Filter Mobile Enter search term Search

Abstract

A novel microRNA (miRNA) quantification method has been developed using stem–loop RT followed by TaqMan PCR analysis. Stem–loop RT primers are better than conventional ones in terms of RT efficiency and specificity. TaqMan miRNA assays are specific for mature miRNAs and discriminate among related miRNAs that differ by as little as one nucleotide. Furthermore, they are not affected by genomic DNA contamination. Precise quantification is achieved routinely with as little as 25 pg of total RNA for most miRNAs. In fact, the high sensitivity, specificity and precision of this method allows for direct analysis of a single cell without nucleic acid purification. Like standard TaqMan gene expression assays, TaqMan miRNA assays exhibit a dynamic range of seven orders of magnitude. Quantification of five miRNAs in seven mouse tissues showed variation from less than 10 to more than 30 000 copies per cell. This method enables fast, accurate and sensitive miRNA expression profiling and can identify and monitor potential biomarkers specific to tissues or diseases. Stem–loop RT–PCR can be used for the quantification of other small RNA molecules such as short interfering RNAs (siRNAs). Furthermore, the concept of stem–loop RT primer design could be applied in small RNA cloning and multiplex assays for better specificity and efficiency.

INTRODUCTION

MicroRNAs (miRNAs) are naturally occurring, highly conserved families of transcripts (18–25 nt in length) that are processed from larger hairpin precursors (1,2). miRNAs are found in the genomes of animals (39) and plants (1012). To date, there are ∼1000 unique transcripts, including 326 human miRNAs in the Sanger Center miRNA registry (13).

miRNAs regulate gene expression by catalyzing the cleavage of messenger RNA (mRNA) (1419) or repressing mRNA translation (1921). They are believed to be critical in cell development, differentiation and communication (2). Specific roles include the regulation of cell proliferation and metabolism (22), developmental timing (23,24), cell death (25), haematopoiesis (26), neuron development (27), human tumorigenesis (28) and DNA methylation and chromatin modification (29).

Although miRNAs represent a relatively abundant class of transcripts, their expression levels vary greatly among species and tissues (30). Less abundant miRNAs routinely escape detection with technologies such as cloning, northern hybridization (31) and microarray analysis (32,33). Here, we present a novel real-time quantification method for accurate and sensitive detection of miRNAs and other small RNAs. This method expands the real-time PCR technology for detecting gene expression changes from macromolecules (e.g. mRNAs) to micro molecules (e.g. miRNAs).

MATERIALS AND METHODS

Targets, primers and probes (Supplementary Data)

Seventeen miRNA genes were selected from the Sanger Center miRNA Registry at http://www.sanger.ac.uk/Software/Rfam/mirna/index.shtml. All TaqMan miRNA assays are available through Applied Biosystems (P/N: 4365409). Standard TaqMan® assays for pri-miRNA precursors, pri-let-7a-3 and pri-miR-26b and pre-miRNA precursor pre-miR-30a were designed using PrimerExpress® software (Applied Biosystems, Foster City, CA). All sequences are available in the section of the Supplementary Data. Synthetic miRNA oligonucleotides were purchased from Integrated DNA Technologies (IDT, Coralville, IA).

Tissue RNA samples, cells, cell lysates and total RNA preparation

Mouse total RNA samples from brain, heart, liver, lung, thymus, ovary and embryo at day 10–12 were purchased from Ambion (P/N: 7810, 7812, 7814, 7816, 7818, 7824, 7826 and 7968). Ambion's mouse total RNAs are derived from Swiss Webster mice. All RNA samples were normalized based on the TaqMan® Gene Expression Assays for human or mouse glyceraldehyde-3-phosphate dehydrogenase (GAPDH) endogenous controls (P/N: 4310884E and 4352339E, Applied Biosystems).

Two cell lines, HepG2 and OP9, were cultured using Gibco MEM (P/N: 12492–021, Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum (FBS) (P/N: SH30070.01, HyClone, Logan, UT). Trypsinized cells were counted with a hemocytometer. Approximately 2.8 × 106 suspended cells were pelleted by centrifugation (Allegra 6, Beckman Coulter, Fullerton, CA) at 1500 r.p.m. for 5 min, washed with 1 ml Dulbecco's phosphate-buffered saline (PBS) without MgCl2 and CaCl2 (P/N: 14190078, Invitrogen, Carlsbad, CA). The cell pellets were re-suspended in 140 µl PBS and processed with three different sample preparation methods. With the first method, a 50 µl sample (106 cells) was mixed with an equal amount of Nucleic Acid Purification Lysis Solution (P/N: 4305895; Applied Biosystems) by pipetting up and down 10 times, and then spun briefly. The lysate was diluted 1/10 with 1 U/µl RNase inhibitor solution (P/N: N8080119; Applied Biosystems) before adding the solution to an RT reaction. In the second method, a 50 µl sample (106 cells) was used to purify total RNA using the mirVana™ miRNA Isolation Kit (P/N: 1560, Ambion, Austin, TX) according to the manufacturer's protocol. Purified total RNA was eluted in 100 µl of elution buffer. The third method involved diluting cells 1/2 with 1× PBS, heating at 95°C for 5 min, and immediately chilling on ice before aliquotting directly into RT reactions.

miRNA detection using mirVana™ miRNA detection kit

Solution hybridization-based miRNA analysis was carried out using the mirVana™ miRNA Detection Kit (Cat. #: 1552, Ambion) according to the manufacturer's protocol. RNA probes were synthesized by IDT. The radioisotope labeled RNA fragments were detected and quantitated with a Cyclone Storage Phosphor System (PerkinElmer, Boston, MA).

Reverse transcriptase reactions

Reverse transcriptase reactions contained RNA samples including purified total RNA, cell lysate, or heat-treated cells, 50 nM stem–loop RT primer (P/N: 4365386 and 4365387, Applied Biosystems), 1× RT buffer (P/N: 4319981, Applied Biosystems), 0.25 mM each of dNTPs, 3.33 U/µl MultiScribe reverse transcriptase (P/N: 4319983, Applied Biosystems) and 0.25 U/µl RNase inhibitor (P/N: N8080119; Applied Biosystems). The 7.5 µl reactions were incubated in an Applied Biosystems 9700 Thermocycler in a 96- or 384-well plate for 30 min at 16°C, 30 min at 42°C, 5 min at 85°C and then held at 4°C. All Reverse transcriptase reactions, including no-template controls and RT minus controls, were run in duplicate.

PCR

Real-time PCR was performed using a standard TaqMan® PCR kit protocol on an Applied Biosystems 7900HT Sequence Detection System (P/N: 4329002, Applied Biosystems). The 10 µl PCR included 0.67 µl RT product, 1× TaqMan® Universal PCR Master Mix (P/N: 4324018, Applied Biosystems), 0.2 µM TaqMan® probe, 1.5 µM forward primer and 0.7 µM reverse primer. The reactions were incubated in a 384-well plate at 95°C for 10 min, followed by 40 cycles of 95°C for 15 s and 60°C for 1 min. All reactions were run in triplicate. The threshold cycle (_C_T) is defined as the fractional cycle number at which the fluorescence passes the fixed threshold. TaqMan®_C_T values were converted into absolute copy numbers using a standard curve from synthetic lin-4 miRNA.

The method for real-time quantification of pri-miRNA precursors, let-7a-3 and miR-26b, and pre-miRNA precursor miR-30a was described elsewhere (34).

RESULTS

We proposed a new real-time RT–PCR scheme for miRNA quantification (Figure 1). It included two steps: RT and real-time PCR. First, the stem–loop RT primer is hybridized to a miRNA molecule and then reverse transcribed with a MultiScribe reverse transcriptase. Next, the RT products are quantified using conventional TaqMan PCR.

The dynamic range and sensitivity of the miRNA quantification scheme were first evaluated using a synthetic cel-lin-4 target. Synthetic RNA was quantified based on the A260 value and diluted over seven orders of magnitude. The cel-lin-4 TaqMan miRNA assay showed excellent linearity between the log of target input and _C_T value, demonstrating that the assay has a dynamic range of at least 7 logs and is capable of detecting as few as seven copies in the PCR reaction (Figure 2).

Eight additional miRNA assays were also validated using mouse lung total RNA. The RNA input ranged from 0.025 to 250 ng (Figure 3). The _C_T values correlated to the RNA input (_R_2 > 0.994) over four orders of magnitude. A negative control assay, cel-miR-2, did not give a detectable signal, even in reactions with 250 ng mouse total RNA.

The expression profile of five miRNAs was determined in seven different mouse tissues to create a miRNA expression map. The copy number per cell was calculated based on the input total RNA (assuming 15 pg/cell) and the standard curve of synthetic lin-4 target. Several interesting observations were made from this expression map. First, miRNAs are very abundant, averaging 2390 copies per cell in these tissues. The level of expression ranged from less than 10 to 32 090 copies per cell. Of the 12 miRNAs, miR-16 and miR-323 were the most and least abundant miRNAs, respectively, across all tissues. In addition, each tissue had a distinctive signature of miRNA expression. The overall level of miRNA expression was highest in mouse lung and lowest in embryos. Finally, the dynamic range of miRNA expression varied greatly from less than 5-fold (let-7a) to more than 2000-fold (miR-323) among these seven tissues (Table 1).

To assess the need for RNA isolation, we added cell lysates directly to miRNA assays. The equivalent of 2.5–2500 cells were added directly to 7.5 µl RT reactions. When detected, the _C_T values correlated (_R_2 > 0.998) to the number of cells in the RT reactions over at least three orders of magnitude (Figure 4).

The effect of non-specific genomic DNA on TaqMan miRNA assays was also tested for 12 assays. Results showed no difference in _C_T values in the presence or absence of 5 ng of human genomic DNA added to the RT reactions, suggesting that the assays are highly specific for RNA targets (data not shown). Based on this observation, we added heat-treated cells directly to miRNA quantification assays. Figure 5 illustrates the comparison of miRNA quantification using purified total RNA, cell lysates and heat-treated cells derived from an equal number of HepG2 cells. Adding heat-treated cells directly to the miRNA assays produced the lowest _C_T values, and good concordance was observed among all three different sample preparation methods.

The reproducibility of TaqMan miRNA assays was examined by performing12 miRNA assays with 16 replicates performed by two independent operators (data not shown). The standard deviation of the _C_Ts averaged 0.1, demonstrating the high precision of the assays.

Solution hybridization-based miRNA northern analysis was used as an independent technology to compare with TaqMan miRNA assays (Figure 6). We observed that hybridization-based miRNA analyses were less reproducible and that concordance with TaqMan assays varied from target to target. There was a general concordance between the two methods (_R_2 = 0.916) for miR-16 across five mouse tissue samples. However, correlations were relatively low for less abundant miRNAs, such as miR-30 (_R_2 = 0.751).

Hybridization methods can lack specificity for the mature miRNAs. We investigated the ability of the TaqMan miRNA assays to differentiate between the mature miRNAs and their longer precursors, using synthetic targets for pri-miRNA precursors, pri-miR-26b and pri-let-7a and pre-miRNA precursor pre-miR-30a (Table 2). TaqMan assays designed to detect either precursors or mature miRNAs were tested with synthetic targets averaging 1.5 × 108 copies per RT reaction (1.3 × 107 copies per PCR). TaqMan miRNA analyses with only pri-miRNA precursor molecules produced _C_T values at least 11 cycles higher than analyses with mature miRNA ones. This result implies that if mature miRNA and precursor were at an equal concentration, the latter would contribute <0.05% background signal to the assay of mature target. For pre-miR-30a where the mature miRNA miR-30a-3p is located at the 3′ end of the pre-miR-30a sequence, a difference of 8.4 _C_T was observed. The results showed that TaqMan miRNA assays are specific to mature miRNAs. However, the assay specificity is better if the miRNA is located at the 5′ strand of the pre-miRNA precursor. Experiments analyzing total RNA instead of synthetic targets indicated that the precursors are at least two orders of magnitude less abundant than mature miRNAs, based on _C_T differences of 7 or more for miR-26b-1 and let-7a-2 precursors. Considered together, these results suggest that the TaqMan miRNA assays are highly specific for the mature miRNAs.

The ability of the TaqMan miRNA assays to discriminate miRNAs that differ by as little as a single nucleotide was tested with the five synthetic miRNAs of let-7a, let-7b, let-7c, let-7d and let-7e (Figure 7). Each miRNA assay was examined against each miRNA. Relative detection efficiency was calculated from _C_T differences between perfectly matched and mismatched targets, assuming 100% efficiency for the perfect match. Very low levels of non-specific signal were observed, ranging from zero to 0.3% for miRNAs with 2–3 mismatched bases and only 0.1–3.7% for the miRNAs that differed by a single nucleotide. Most cross-reactions resulted from G–T mismatches during the RT reaction (let-7a assay versus let-7c target etc.). Only the targeted miRNA was detected if more than three mismatched bases between any two miRNAs were present.

We compared the discrimination ability of the TaqMan miRNA assays to that of solution-based hybridization analysis (Figure 8). In our hands, the hybridization method discriminated well between let-7a and let-7b. However, poor or no discrimination was observed among let-7a, let-7c and let-7d, which differ by 1–3 nt.

We speculated that stem–loop primers might provide better RT efficiency and specificity than linear ones. Base stacking of the stem might enhance the thermal stability of the RNA–DNA heteroduplex. Furthermore, spatial constraint of the stem–loop would likely improve the assay specificity in comparison to conventional linear RT primers. We compared the sensitivity and specificity of the stem–loop and linear RT primers using synthetic miRNAs for let-7a (Figure 9). We observed several advantages for the stem–loop RT. First, in the presence of the synthetic let-7a target, the _C_T values between linear and stem–loop RT methods differed by 7, indicating that the efficiency of stem–loop RT was at least 100 times higher. Secondly, stem–loop RT discriminated better between miRNAs that differ by two bases based on Δ_C_T values. Finally, the stem–loop RT was at least 100 times better able to discriminate between the mature miRNA and its precursor, based on the Δ_C_T (precursor versus mature) of 7.

DISCUSSION

Since the discovery of miRNAs, remarkable advances in the characterization of these gene families have delineated the mechanism for their functions in gene regulation (35). As a result, extensive surveys have begun to identify miRNA biomarkers specific for tissue types or disease status. These studies will benefit from methods that allow for both accurate identification and quantification of miRNAs.

Current methods for detection and quantification of miRNAs are largely based on cloning, northern blotting (5), or primer extension (36). Although microarrays could improve the throughput of miRNA profiling, the method is relatively limited in terms of sensitivity and specificity (32,33). Low sensitivity becomes a problem for miRNA quantification because it is difficult to amplify these short RNA targets. Furthermore, low specificity may lead to false positive signal from closely related miRNAs, precursors and genomic sequences. More recently, a modified Invader assay has been reported for the quantification of several miRNAs (37). However, Invader assays have limited specificity and sensitivity, requiring at least 50 ng total RNA, or 1000 lysed cells, per assay.

Real-time PCR is the gold standard for gene expression quantification (38,39). It has been a long challenge for scientists to design a conventional PCR assay from miRNAs averaging ∼22 nt in length. We developed a novel scheme to design TaqMan PCR assays that specifically quantify miRNA expression levels with superior performance over existing conventional detection methods. We have designed and validated assays for 222 human miRNAs (Chen et al., unpublished data). These assays combine the power of PCR for exquisite sensitivity, real-time monitoring for a large dynamic range and TaqMan assay reporters to increase the specificity. In our hands, miRNA precursors were at least 2000 times less effective targets than mature miRNAs (Table 2). Because these assays are insensitive to precursors or genomic DNA, we were able to add heat-treated cells directly to the assays, eliminating the need for sample preparation. For applications where both mature miRNAs and their precursors need to be assayed, conventional TaqMan assays can be used in parallel to specifically detected precursors.

We observed the better specificity and sensitivity of stem–loop RT primers than conventional linear ones likely due to the base stacking and spatial constraint of the stem–loop structure (Figure 9). The base stacking could improve the thermal stability and extend the effective footprint of RT primer/RNA duplex that may be required for effective RT from relatively shorter RT primers. The spatial constraint of the stem–loop structure may prevent it from binding double-strand genomic DNA molecules and, therefore, eliminate the need of TaqMan miRNA assays for RNA sample preparation. Stem–loop RT primers can potentially be used for multiplex RT reactions and small RNA cloning for possibly better efficiency and specificity.

There is an increasing need for sensitive and specific whole miRNA profiling. The ability to effectively profile miRNAs could lead to the discoveries of disease- or tissue-specific miRNA biomarkers, as well as contribute to the understanding of how miRNAs regulate stem cell differentiation. Our stem–loop RT–PCR method should provide a practical solution for these studies. We are currently developing multiplex approaches that should further increase the utility of this method.

SUPPLEMENTARY DATA

Supplementary Data are available at NAR Online.

Schematic description of TaqMan miRNA assays, TaqMan-based real-time quantification of miRNAs includes two steps, stem–loop RT and real-time PCR. Stem–loop RT primers bind to at the 3′ portion of miRNA molecules and are reverse transcribed with reverse transcriptase. Then, the RT product is quantified using conventional TaqMan PCR that includes miRNA-specific forward primer, reverse primer and a dye-labeled TaqMan probes. The purpose of tailed forward primer at 5′ is to increase its melting temperature (Tm) depending on the sequence composition of miRNA molecules.

Figure 1

Schematic description of TaqMan miRNA assays, TaqMan-based real-time quantification of miRNAs includes two steps, stem–loop RT and real-time PCR. Stem–loop RT primers bind to at the 3′ portion of miRNA molecules and are reverse transcribed with reverse transcriptase. Then, the RT product is quantified using conventional TaqMan PCR that includes miRNA-specific forward primer, reverse primer and a dye-labeled TaqMan probes. The purpose of tailed forward primer at 5′ is to increase its melting temperature (Tm) depending on the sequence composition of miRNA molecules.

Dynamic range and sensitivity of the TaqMan lin-4 miRNA assay. (A) Amplification plot of synthetic lin-4 miRNA over seven orders of magnitude. Synthetic RNA input ranged from 1.3 × 10−3 fM (equivalent to 7 copies per reaction) to 1.3 × 104 fM (7 × 107 copies per reaction) in PCR; (B) Standard curve of the lin-4 miRNA.

Figure 2

Dynamic range and sensitivity of the TaqMan lin-4 miRNA assay. (A) Amplification plot of synthetic lin-4 miRNA over seven orders of magnitude. Synthetic RNA input ranged from 1.3 × 10−3 fM (equivalent to 7 copies per reaction) to 1.3 × 104 fM (7 × 107 copies per reaction) in PCR; (B) Standard curve of the lin-4 miRNA.

Correlation of total RNA input to the threshold of cycle (CT) values for eight miRNA assays. Mouse lung total RNA input ranged from 0.025 to 250 ng per RT reaction. A Caenorhabditis elegans miRNA (miR-2) was included as a negative control assay.

Figure 3

Correlation of total RNA input to the threshold of cycle (_C_T) values for eight miRNA assays. Mouse lung total RNA input ranged from 0.025 to 250 ng per RT reaction. A Caenorhabditis elegans miRNA (miR-2) was included as a negative control assay.

Table 1

Expression profiles of five miRNAs across seven mouse tissues

miRNA ID Copy number per cell cFold-change
Brain Heart Liver Lung Thymus Ovary Embryo Average
let-7a 2010 1420 700 2390 1420 3120 1050 1730 5
miR-16 10 240 13 520 3890 22 080 32 090 11 100 5210 14 020 6
miR-20 70 300 130 580 1990 420 620 590 28
miR-21 670 2540 4450 7970 3550 5310 390 3550 20
miR-22 290 1020 310 590 130 560 40 420 26
Average 2240 2040 1140 4430 3540 2600 750 2390 17
miRNA ID Copy number per cell cFold-change
Brain Heart Liver Lung Thymus Ovary Embryo Average
let-7a 2010 1420 700 2390 1420 3120 1050 1730 5
miR-16 10 240 13 520 3890 22 080 32 090 11 100 5210 14 020 6
miR-20 70 300 130 580 1990 420 620 590 28
miR-21 670 2540 4450 7970 3550 5310 390 3550 20
miR-22 290 1020 310 590 130 560 40 420 26
Average 2240 2040 1140 4430 3540 2600 750 2390 17

Mouse or human total RNA samples from brain, heart, liver, lung, thymus, ovary and embryo (at day 10–12) were purchased from Ambion. Copy number per cell is estimated based on standard curve of lin-4 synthetic miRNA. A total of 150 ng RNA (or equivalent to approximately 10 000 cells assuming 15 pg of total RNA per cell) was added to each RT reaction. RNA input was normalized based on TaqMan GAPDH endogenous control (P/N: 4352339E).

Table 1

Expression profiles of five miRNAs across seven mouse tissues

miRNA ID Copy number per cell cFold-change
Brain Heart Liver Lung Thymus Ovary Embryo Average
let-7a 2010 1420 700 2390 1420 3120 1050 1730 5
miR-16 10 240 13 520 3890 22 080 32 090 11 100 5210 14 020 6
miR-20 70 300 130 580 1990 420 620 590 28
miR-21 670 2540 4450 7970 3550 5310 390 3550 20
miR-22 290 1020 310 590 130 560 40 420 26
Average 2240 2040 1140 4430 3540 2600 750 2390 17
miRNA ID Copy number per cell cFold-change
Brain Heart Liver Lung Thymus Ovary Embryo Average
let-7a 2010 1420 700 2390 1420 3120 1050 1730 5
miR-16 10 240 13 520 3890 22 080 32 090 11 100 5210 14 020 6
miR-20 70 300 130 580 1990 420 620 590 28
miR-21 670 2540 4450 7970 3550 5310 390 3550 20
miR-22 290 1020 310 590 130 560 40 420 26
Average 2240 2040 1140 4430 3540 2600 750 2390 17

Mouse or human total RNA samples from brain, heart, liver, lung, thymus, ovary and embryo (at day 10–12) were purchased from Ambion. Copy number per cell is estimated based on standard curve of lin-4 synthetic miRNA. A total of 150 ng RNA (or equivalent to approximately 10 000 cells assuming 15 pg of total RNA per cell) was added to each RT reaction. RNA input was normalized based on TaqMan GAPDH endogenous control (P/N: 4352339E).

Dynamic range of eight TaqMan miRNA assays using OP9 cell lysates. The number of cell input ranged from 3 to 2500 cells per RT. A Caenorhabditis elegans miRNA (miR-2) was used as a negative control.

Figure 4

Dynamic range of eight TaqMan miRNA assays using OP9 cell lysates. The number of cell input ranged from 3 to 2500 cells per RT. A Caenorhabditis elegans miRNA (miR-2) was used as a negative control.

Comparison of heat-treated cells, cell lysate and total RNA for real-time quantitation of 10 miRNAs. The level of miRNA expression is measured in the threshold cycles (CT). Approximately 400 HepG2 cells were analyzed per PCR.

Figure 5

Comparison of heat-treated cells, cell lysate and total RNA for real-time quantitation of 10 miRNAs. The level of miRNA expression is measured in the threshold cycles (_C_T). Approximately 400 HepG2 cells were analyzed per PCR.

Comparison of TaqMan miRNA miR-16 assay to solution-based northern hybridization analysis. Total RNAs from mouse kidney, liver, lung, spleen and testicle tissues were used.

Figure 6

Comparison of TaqMan miRNA miR-16 assay to solution-based northern hybridization analysis. Total RNAs from mouse kidney, liver, lung, spleen and testicle tissues were used.

Table 2

Discrimination between mature miRNAs and their pri- or pre-miRNA precursors

ID Synthetic miRNA (No. of copies) Synthetic precursor (No. of copies) Total RNA (ng) C T miRNA Precursor
miR-26b 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 27.4 18.7
0 0 7.5 21.9 28.7
0 0 0 ND ND
let-7a 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 29.5 19.4
0 0 7.5 19.9 34.7
0 0 0 ND ND
miR-30a-3p 1.5 × 108 0 0 15.8 ND
0 1.5 × 108 0 24.2 17.2
0 0 7.5 25.4 30.3
0 0 0 ND ND
miR-30a-5p 1.5 × 108 0 0 16.3 ND
0 1.5 × 108 0 29.1 18.9
0 0 7.5 22.1 30.3
0 0 0 39.6 ND
ID Synthetic miRNA (No. of copies) Synthetic precursor (No. of copies) Total RNA (ng) C T miRNA Precursor
miR-26b 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 27.4 18.7
0 0 7.5 21.9 28.7
0 0 0 ND ND
let-7a 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 29.5 19.4
0 0 7.5 19.9 34.7
0 0 0 ND ND
miR-30a-3p 1.5 × 108 0 0 15.8 ND
0 1.5 × 108 0 24.2 17.2
0 0 7.5 25.4 30.3
0 0 0 ND ND
miR-30a-5p 1.5 × 108 0 0 16.3 ND
0 1.5 × 108 0 29.1 18.9
0 0 7.5 22.1 30.3
0 0 0 39.6 ND

Note: ND represents no detectable PCR products after 40 cycles. The copy number of synthetic miRNAs in RT was estimated based on the _A_260 values. Only 9% of RT product was added to PCR. Total RNA from human lung was used. Pre-miRNA precursors, pri-let-7a-3 and pri-miR-26b, and pre-miRNA precursor pre-miR-30a were examined.

Table 2

Discrimination between mature miRNAs and their pri- or pre-miRNA precursors

ID Synthetic miRNA (No. of copies) Synthetic precursor (No. of copies) Total RNA (ng) C T miRNA Precursor
miR-26b 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 27.4 18.7
0 0 7.5 21.9 28.7
0 0 0 ND ND
let-7a 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 29.5 19.4
0 0 7.5 19.9 34.7
0 0 0 ND ND
miR-30a-3p 1.5 × 108 0 0 15.8 ND
0 1.5 × 108 0 24.2 17.2
0 0 7.5 25.4 30.3
0 0 0 ND ND
miR-30a-5p 1.5 × 108 0 0 16.3 ND
0 1.5 × 108 0 29.1 18.9
0 0 7.5 22.1 30.3
0 0 0 39.6 ND
ID Synthetic miRNA (No. of copies) Synthetic precursor (No. of copies) Total RNA (ng) C T miRNA Precursor
miR-26b 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 27.4 18.7
0 0 7.5 21.9 28.7
0 0 0 ND ND
let-7a 1.5 × 108 0 0 16.5 ND
0 1.5 × 108 0 29.5 19.4
0 0 7.5 19.9 34.7
0 0 0 ND ND
miR-30a-3p 1.5 × 108 0 0 15.8 ND
0 1.5 × 108 0 24.2 17.2
0 0 7.5 25.4 30.3
0 0 0 ND ND
miR-30a-5p 1.5 × 108 0 0 16.3 ND
0 1.5 × 108 0 29.1 18.9
0 0 7.5 22.1 30.3
0 0 0 39.6 ND

Note: ND represents no detectable PCR products after 40 cycles. The copy number of synthetic miRNAs in RT was estimated based on the _A_260 values. Only 9% of RT product was added to PCR. Total RNA from human lung was used. Pre-miRNA precursors, pri-let-7a-3 and pri-miR-26b, and pre-miRNA precursor pre-miR-30a were examined.

Discrimination power of let-7 miRNA assays. Relative detection (%) calculated based on CT difference between perfectly matched and mismatched targets. A total of 1.5 × 108 copies of synthetic RNA was added to RT reaction. The concentration was estimated based on the A260 values.

Figure 7

Discrimination power of let-7 miRNA assays. Relative detection (%) calculated based on _C_T difference between perfectly matched and mismatched targets. A total of 1.5 × 108 copies of synthetic RNA was added to RT reaction. The concentration was estimated based on the _A_260 values.

Poor discrimination of miRNAs with solution hybridization-based (northern) analysis.

Figure 8

Poor discrimination of miRNAs with solution hybridization-based (northern) analysis.

Specificity of TaqMan miRNA assays between stem–loop and linear RT primers. Mature let-7a-specific assay was tested against let-7a, let-7e and pri-miR precursor let-7a-3. ΔCT represents the CT difference between two targets or methods. A total of 1.5 × 108 copies of synthetic targets were added to each RT reaction.

Figure 9

Specificity of TaqMan miRNA assays between stem–loop and linear RT primers. Mature let-7a-specific assay was tested against let-7a, let-7e and pri-miR precursor let-7a-3. Δ_C_T represents the _C_T difference between two targets or methods. A total of 1.5 × 108 copies of synthetic targets were added to each RT reaction.

We greatly thank Kelly McDonald, Fenton Williams, Will Bloch, Neil Straus, and Victor Ambros for critical reading of the manuscript. Funding to pay the Open Access publication charges for this article was provided by Applied Biosystems.

Conflict of interest statement. None declared.

REFERENCES

1

Ambros, V.

2004

The functions of animal microRNAs

Nature

431

350

–355

2

Bartel, D.P.

2004

MicroRNAs: genomics, biogenesis, mechanism, and function

Cell

116

281

–297

3

Mourelatos, Z., Dostie, J., Paushkin, S., Sharma, A., Charroux, B., Abel, L., Rappsilber, J., Mann, M., Dreyfuss, G.

2002

miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs

Genes Dev.

16

720

–728

4

Lau, N.C., Lim, L.P., Weinstein, E.G., Bartel, D.P.

2001

An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans

Science

294

858

–862

5

Lagos-Quintana, M., Rauhut, R., Lendeckel, W., Tuschl, T.

2001

Identification of novel genes coding for small expressed RNAs

Science

294

853

–858

6

Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., Tuschl, T.

2002

Identification of tissue-specific microRNAs from mouse

Curr. Biol.

12

735

–739

7

Lagos-Quintana, M., Rauhut, R., Meyer, J., Borkhardt, A., Tuschl, T.

2003

New microRNAs from mouse and human

RNA

9

175

–179

8

Lai, E.C.

2002

Micro RNAs are complementary to 3′-UTR sequence motifs that mediate negative post-transcriptional regulation

Nature Genet.

30

363

–364

9

Lee, R.C. and Ambros, V.

2001

An extensive class of small RNAs in Caenorhabditis elegans

Science

294

862

–864

10

Llave, C., Xie, Z., Kasschau, K.D., Carrington, J.C.

2002

Cleavage of Scarecrow-like mRNA targets directed by a class of Arabidopsis miRNA

Science

297

2053

–2056

11

Park, W., Li, J., Song, R., Messing, J., Chen, X.

2002

CARPEL FACTORY, a Dicer homolog, and HEN1, a novel protein, act in microRNA metabolism in Arabidopsis thaliana

Curr. Biol.

12

1484

–1495

12

Reinhart, B.J., Weinstein, E.G., Rhoades, M.W., Bartel, B., Bartel, D.P.

2002

MicroRNAs in plants [Erratum (2002) Genes Dev., 16, 2313.]

Genes Dev.

16

1616

–1626

13

Griffiths-Jones, S.

2004

The microRNA Registry

Nucleic Acids Res.

32

D109

–D111

14

Hutvagner, G. and Zamore, P.D.

2002

A microRNA in a multiple-turnover RNAi enzyme complex

Science

297

2056

–2060

15

Zhang, H., Kolb, F.A., Brondani, V., Billy, E., Filipowicz, W.

2002

Human Dicer preferentially cleaves dsRNAs at their termini without a requirement for ATP

EMBO J.

21

5875

–5885

16

Zhang, H., Kolb, F.A., Jaskiewicz, L., Westhof, E., Filipowicz, W.

2004

Single processing center models for human Dicer and bacterial RNase III

Cell

118

57

–68

17

Zhang, S. and Semino, C.E.

2003

Design peptide scaffolds for regenerative medicine

Adv. Exp. Med. Biol.

534

147

–163

18

Doench, J.G., Petersen, C.P., Sharp, P.A.

2003

siRNAs can function as miRNAs

Genes Dev.

17

438

–442

19

Doench, J.G. and Sharp, P.A.

2004

Specificity of microRNA target selection in translational repression

Genes Dev.

18

504

–511

20

Wightman, B., Ha, I., Ruvkun, G.

1993

Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C.elegans

Cell

75

855

–862

21

Olsen, P.H. and Ambros, V.

1999

The lin-4 regulatory RNA controls developmental timing in Caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation

Dev. Biol.

216

671

–680

22

Brennecke, J., Hipfner, D.R., Stark, A., Russell, R.B., Cohen, S.M.

2003

Bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila

Cell

113

25

–36

23

Reinhart, B.J., Slack, F.J., Basson, M., Pasqulnelli, A.E., Bettinger, J.C., Rougvie, A.E., Horvitz, H.R., Ruvkun, G.

2000

The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans

Nature

403

901

–906

24

Ambros, V.

2003

MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing [Erratum (2003) Cell, 114, 269.]

Cell

113

673

–676

25

Baehrecke, E.H.

2003

miRNAs: micro managers of programmed cell death

Curr. Biol.

13

R473

–R475

26

Chen, C.-Z., Li, L., Lodish, H.F., Bartel, D.P.

2004

MicroRNAs modulate hematopoietic lineage differentiation

Science

303

83

–86

27

Johnston, R.J. and Hobert, O.

2003

A microRNA controlling left/right neuronal asymmetry in Caenorhabditis elegans

Nature

426

845

–849

28

Michael, M.Z., O'Connor, S.M., van Holst Pellekaan, N.G., Young, G.P., James, R.J.

2003

Reduced accumulation of specific microRNAs in colorectal neoplasia

Mol. Cancer Res.

1

882

–891

29

Bao, N., Lye, K.-W., Barton, M.K.

2004

MicroRNA binding sites in Arabidopsis class III HD-ZIP mRNAs are required for methylation of the template chromosome

Dev. Cell

7

653

–662

30

Kim, J., Krichevsky, A., Grad, Y., Hayes, G.D., Kosik, K.S., Church, G.M., Ruvkun, G.

2004

Identification of many microRNAs that copurify with polyribosomes in mammalian neurons

Proc. Natl Acad. Sci. USA

101

360

–365

31

Lim, L.P., Glasner, M.E., Yekta, S., Burge, C.B., Bartel, D.P.

2003

Vertebrate microRNA genes

Science

299

1540

32

Krichevsky, A.M., King, K.S., Donahue, C.P., Khrapko, K., Kosik, K.S.

2003

A microRNA array reveals extensive regulation of microRNAs during brain development [Erratum (2004) RNA, 10, 551.]

RNA

9

1274

–1281

33

Liu, C.-G., Calin, G.A., Meloon, B., Gamliel, N., Sevignani, C., Ferracin, M., Dumitru, C.D., Shimizu, M., Zupo, S., Dono, M., et al.

2004

An oligonucleotide microchip for genome-wide microRNA profiling in human and mouse tissues

Proc. Natl Acad. Sci. USA

101

9740

–9744

34

Schmittgen, T.D., Jiang, J., Liu, Q., Yang, L.

2004

A high-throughput method to monitor the expression of microRNA precursors

Nucleic Acids Res.

32

e43

35

He, L. and Hannon, G.J.

2004

MicroRNAs: small RNAs with a big role in gene regulation

Nature Rev. Genet.

5

522

–531

36

Zeng, Y. and Cullen, B.R.

2003

Sequence requirements for micro RNA processing and function in human cells

RNA

9

112

–123

37

Allawi, H.T., Dahlberg, J.E., Olson, S., Lund, E., Olson, M., Ma, W.-P., Takova, T., Neri, B.P., Lyamichev, V.I.

2004

Quantitation of microRNAs using a modified Invader assay

RNA

10

1153

–1161

38

Livak, K.J. and Schmittgen, T.D.

2001

Analysis of relative gene expression data using real-time quantitative PCR and the 2[-Delta Delta C(T)] Method

Methods

25

402

–408

39

Heid, C.A., Stevens, J., Livak, K.J., Williams, P.M.

1996

Real time quantitative PCR

Genome Res.

6

986

–994

© The Author 2005. Published by Oxford University Press. All rights reserved The online version of this article has been published under an open access model. Users are entitled to use, reproduce, disseminate, or display the open access version of this article for non-commercial purposes provided that: the original authorship is properly and fully attributed; the Journal and Oxford University Press are attributed as the original place of publication with the correct citation details given; if an article is subsequently reproduced or disseminated not in its entirety but only in part or as a derivative work this must be clearly indicated. For commercial re-use, please contact journals.permissions@oxfordjournals.org

Supplementary data

I agree to the terms and conditions. You must accept the terms and conditions.

Submit a comment

Name

Affiliations

Comment title

Comment

You have entered an invalid code

Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.

Citations

Views

Altmetric

Metrics

Total Views 62,970

46,670 Pageviews

16,300 PDF Downloads

Since 11/1/2016

Month: Total Views:
November 2016 13
December 2016 29
January 2017 219
February 2017 492
March 2017 448
April 2017 275
May 2017 350
June 2017 329
July 2017 341
August 2017 294
September 2017 300
October 2017 311
November 2017 400
December 2017 840
January 2018 1,082
February 2018 1,172
March 2018 1,098
April 2018 1,066
May 2018 1,020
June 2018 944
July 2018 1,107
August 2018 833
September 2018 630
October 2018 720
November 2018 931
December 2018 682
January 2019 645
February 2019 565
March 2019 701
April 2019 630
May 2019 792
June 2019 621
July 2019 692
August 2019 624
September 2019 557
October 2019 638
November 2019 593
December 2019 575
January 2020 547
February 2020 451
March 2020 462
April 2020 306
May 2020 355
June 2020 566
July 2020 533
August 2020 703
September 2020 739
October 2020 890
November 2020 868
December 2020 651
January 2021 784
February 2021 745
March 2021 799
April 2021 799
May 2021 900
June 2021 804
July 2021 742
August 2021 751
September 2021 758
October 2021 786
November 2021 733
December 2021 753
January 2022 788
February 2022 762
March 2022 820
April 2022 757
May 2022 776
June 2022 711
July 2022 703
August 2022 786
September 2022 673
October 2022 647
November 2022 700
December 2022 556
January 2023 588
February 2023 735
March 2023 876
April 2023 858
May 2023 728
June 2023 642
July 2023 649
August 2023 713
September 2023 719
October 2023 835
November 2023 680
December 2023 565
January 2024 739
February 2024 688
March 2024 767
April 2024 674
May 2024 722
June 2024 583
July 2024 596
August 2024 541
September 2024 409

×

Email alerts

Citing articles via

More from Oxford Academic