ArticlePDF Available

In Situ Detection of Mature Micrornas by Labeled Extension on Ultramer Templates

Authors:

Abstract and Figures

We describe a new method for the in situ detection of a mature microRNA (miRNA) in formalin-fixed, paraffin-embedded tissues. The method involves the labeled extension of miRNA hybridized to an approximately 100-nucleotide-long ultramer template containing the complementary sequence of the miRNA at its 3' terminus. Pretreatment of the tissue involves incubation with protease to expose the genomic DNA to DNase digestion, thereby eliminating the ultramer-independent DNA synthesis process inherent in paraffin-embedded tissue. By direct comparison with real-time reverse transcriptase (RT)-PCR, RT in situ PCR, and standard in situ hybridization using a locked nucleic acid (LNA) probe, it was evident that the ultramer extension method detects only the mature miRNA, is easier to optimize, results generally in a stronger signal, and is much less expensive than the LNA probe method currently used.
Content may be subject to copyright.
In situ detection of mature microRNAs by labeled extension on
ultramer templates
Gerard J. Nuovo1, Eun Joo Lee2, Sean Lawler3, Jakub Godlewski3, and Thomas D.
Schmittgen2
1Department of Pathology, Ohio State University Medical Center, Columbus, OH, USA
2College of Pharmacy, Ohio State University Medical Center, Columbus, OH, USA
3Dardinger Laboratory for Neuro-oncology and Neurosciences, Ohio State University Medical Center,
Columbus, OH, USA
Abstract
We describe a new method for the in situ detection of a mature microRNA (miRNA) in formalin-
fixed, paraffin-embedded tissues. The method involves the labeled extension of miRNA hybridized
to an approximately 100-nucleotide–long ultramer template containing the complementary sequence
of the miRNA at its 3 terminus. Pretreatment of the tissue involves incubation with protease to expose
the genomic DNA to DNase digestion, thereby eliminating the ultramer-independent DNA synthesis
process inherent in paraffin-embedded tissue. By direct comparison with real-time reverse
transcriptase (RT)–PCR, RT in situ PCR, and standard in situ hybridization using a locked nucleic
acid (LNA) probe, it was evident that the ultramer extension method detects only the mature miRNA,
is easier to optimize, results generally in a stronger signal, and is much less expensive than the LNA
probe method currently used.
Keywords
microRNA; in situ hybridization; in situ PCR; LNA probe
Introduction
MicroRNAs (miRNAs) are small, noncoding sequences of 20–23 nucleotides that regulate
cell processes by binding to the 3 untranslated region (UTR) of mRNAs resulting in its
translation repression; it is predicted that they regulate thousands of human genes (1–3).
miRNAs are transcribed as long primary precursor molecules (pri-miRNA) that are
subsequently processed by the nuclear enzyme Drosha to the precursor miRNA (pre-miRNA).
The pre-miRNA, in turn, is processed by Dicer to generate the mature miRNA (4). miRNA
expression is critical in oncogenesis (2,5–7). Increased miRNA expression in cancers is
associated with the down-regulation of tumor suppressors while miRNAs that are reduced in
cancer may normally suppress oncogenes (2,5–7).
miRNA expression analyses have used real-time RT-PCR (8–10), Northern blot analysis (1,
4), or microarrays (2) that can detect either the precursor (8–10) or, more commonly, the mature
Address correspondence to Gerard J. Nuovo, Department of Pathology, Ohio State University, Medical Center, 81 HLRI, 473 W. 12th
Avenue, Columbus, OH 43210. email: E-mail: gnuovomd@pol.net.
The authors declare no competing interests.
NIH Public Access
Author Manuscript
Biotechniques. Author manuscript; available in PMC 2009 June 9.
Published in final edited form as:
Biotechniques. 2009 February ; 46(2): 115–126. doi:10.2144/000113068.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
form of the molecule (10). Relatively few studies on miRNA expression have used in situ–
based techniques (5,11–14). Thus, there is evidence lacking to show that, in cancers, only the
malignant cells—and not the adjacent normal tissue—are expressing the miRNA of interest.
RT in situ PCR can be used to detect the miRNA precursors: since it is very sensitive, a negative
result would effectively rule out the production of miRNA precursors (5,14,15). The in situ
detection of miRNAs has been assisted by the use of locked nucleic acid (LNA)–modified
nucleotides (13,16). The LNA nucleotides are much more rigid in three-dimensional space,
which results in a substantial increase in the Tm of the small LNA-modified oligonucleotide
probe hybridized to its target miRNA (17). It has been demonstrated by some that the LNA-
modified probe can detect either the miRNA precursors or mature miRNA depending on which
specific sequence in the pre-miRNA is targeted (18,19).
We have developed a novel method for the in situ detection of the mature miRNA based on
the extension of the molecule after its hybridization to an ultramer template. This method, like
RNA-primed, array-based, Klenow enzyme (RAKE) analysis, is based on the RNA molecule
acting as a primer to initiate the reaction (12). However, unlike RAKE, which is only effective
in solution phase reactions, our system allows for the in situ analysis of the mature miRNA.
This novel method—along with the two established in situ methods of RT in situ PCR and in
situ hybridization with LNA probes—were used to study the spatial patterns of different
microRNAs in a variety of malignant cell lines, as well as in normal and cancerous tissues.
Materials and methods
Solution phase extension of miR-376a
Mature miR-376a oligoribonucleotide was synthesized by IDT (Coralville, IA, USA) and gel-
purified. miR-376a oligomer (1 µg) was end-labeled with γ-P32 ATP and T4 kinase. The labeled
miR-376a oligomer was used to prime a miR-376a ultramer, and the oligos were extended
using a variety of enzymes including rTth reverse transcriptase (Applied Biosystems, Foster
City, CA, USA), Taq DNA polymerase (Promega, Madison, WI, USA), MMLV reverse
transcriptase (Invitrogen, Carlsbad, CA, USA), MultiScribe reverse transcriptase (Applied
Biosystems), and T7 RNA polymerase (Epicentre, Madison, WI, USA), according to each
company’s protocol. The extension products were verified on a 12% polyacrylamide, 6 M urea
denaturing gel.
Cell lines and tissue samples
Tissue samples (from the files of G.J.N.) and cell preparations (HL-60 and Jurkat cell lines,
provided by colleagues at Ohio State University Medical Center)were immediately fixed in
10% buffered formalin for 4–12 h at room temperature; buffered formalin is the optimal fixative
for RNA in situ hybridization (15). All tissues were obtained via an approved protocol from
the Ohio State University Medical Center Internal Review Board. We focused on cardiac and
central nervous system (CNS) tissues, as these have been well associated with particular
miRNAs [miR-1 (20), and miR-128a (21) (S. Lawler, unpublished data)], respectively.
Furthermore, since mature miR-128a is decreased in the malignant counterpart (glioblastoma
multiforme) (S. Lawler, unpublished data), this allowed us to study the in situ correlates of
mature miRNA down-regulation in these cancerous tissues.
Ultramer extension method
General principle—The ultramer is at least 100 nucleotides in size. At its 5 end, it consists
of a series of four 20-nucleotide repeats (GACCCCTTAATGCGTCTAAA), ending at the 3
end with the complementary sequence of the miRNA of interest. A DNA or RNA polymerase
will extend the miRNA using the ultramer as the template (see Figure 1, panel A with miR-1
as the example). As the miRNA is being extended, the newly synthesized DNA incorporates
Nuovo et al. Page 2
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the reporter nucleotide digoxigenin (Figure 1A). The repetitive sequence allows the use of a
complementary oligonucleotide (TTTAGACGCATTAAGGGGTC) that can be added at the
onset of the reaction that will inhibit the extension of the miRNA. This serves as an additional
negative control to document the specificity of the reaction.
Both the RT in situ PCR and ultramer extension methods are optimized for formalin-fixed,
paraffin-embedded tissue. Heating the paraffin to 65°C during the embedding process induces
nicks and short gaps in the genomic DNA, whereas formalin fixation causes DNA- and RNA-
protein crosslinks (Figure 1, B1) (15). Any DNA polymerase will utilize these nicks and short
gaps to synthesize DNA that will incorporate the reporter nucleotide, either through gap repair
or—if the polymerase has 5–3 exonuclease activity—via nick translation (Figure 1, B2). This
will result in a nonspecific nuclear signal as previously shown with in situ PCR in paraffin
tissue sections (5,14–16). In order to prevent this, it is necessary to degrade the genomic DNA
by DNase pre-treatment to the point where it will no longer support this nonspecific DNA
synthesis pathway (Figure 1, B3). Efficient DNase pre-treatment requires the removal by
protease digestion of the formalin-induced DNA-protein adducts in the genomic DNA, which
may sterically interfere with DNase activity. Optimal protease digestion is defined by the
generation of an intense nuclear signal in the extension/labeling reaction of a test tissue section
or cell preparation, reflecting loss of the steric interference of polymerase activity by the DNA-
protein adducts (5,14–16). If the protease digestion is not sufficient, residual DNA-protein
crosslinks will allow the persistence of sufficiently intact DNA not degraded by DNase,
resulting in a nonspecific nuclear signal (Figure 1, B4). If protease digestion is too strong, then
the morphology of the tissue or cell preparation is lost and thus the results are not interpretable
(15). The key components of the ultramer extension method are as follows:
Protease digestion—Cells or deparaffinized tissue sections are digested in protease (2 mg/
mL pepsin in RNase-free water) at room temperature. The optimal protease time is defined as
the number of minutes of digestion that will elicit an intense nuclear-based signal in all cell
types, in a test tissue section or cell preparation with direct incorporation of the reporter
nucleotide in an extension/labeling reaction. This signal is completely eliminated by overnight
digestion with RNase-free DNase (5,15).
DNase digestion—After optimal pro tease digestion, approximately 200 U of RNase-free
DNase I (Roche Diagnostics, Indianapolis, IN, USA) is applied to each tissue section or cell
preparation and incubated for 15 h at 37°C (15,16). The DNase is removed by successive
washes in RNase-free water and 100% ethanol.
miRNA extension solution—The ultramer extension solution consisted of the following:
10 µl rTth buffer (Applied Biosystems), 1.6 µl of each of the four dNTPS (10 mM), 1.6 µl of
2% BSA, 12.4 µl of 10 mM mangenese acetate, 2 µl of the ultramer (500 pmol), 0.6 µl of 1
mM digoxigenin dUTP, 2 µl of rTth DNA polymerase, and 15.0 µl of RNase free water.
In situ microRNA extension—After DNase digestion, the tissue section or cell preparation
is covered with the miRNA extension solution and then with a polypropylene coverslip and
mineral oil to prevent evaporation. The extension reaction proceeds for 30–60 min at 60°C.
The incorporated digoxigenin is detected with an alkaline phosphatase conjugated to an
antidigoxigenin antibody (75 U/100 µl; Roche Diagnostics) diluted 1:150 with Tris buffer (pH
7.0). As with the RT in situ PCR method, the chromogen is nitroblue tetrazolium (NBT; 300
µg/mL) and bromochloroindolyl phosphate (BCIP; 200 µg/mL) and the counterstain is nuclear
fast red (Enzo Life Sciences, Farmingdale, NY).
An alternative method can be used to confirm the specificity of the ultramer-extended miRNA.
The ultramer extension can proceed as described above, but minus the reporter nucleotide. The
Nuovo et al. Page 3
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
ultramer can be end-tailed with biotin (17) and used as a probe to detect the unlabeled extended
miRNA using the method described below, under “In situ hybridization with LNA-DNA
probes.”
Interpretation—A successful reaction with direct incorporation of the reporter nucleotide is
defined as a strong nuclear signal in the no-DNase control, and its complete elimination in the
DNase control in which either no ultramer or (for miR-128a) a scrambled ultramer was used.
The latter negative control section should be on the same slide as the test section in order to
assure uniformity of variables such as protease digestion. As with RT in situ PCR, these two
results indicate optimal protease digestion and successful degradation of the genomic DNA to
the point that it can no longer support detectable DNA synthesis (Figure 1B, 2 and 3). If a
signal is seen in the DNase negative control (no ultramer), which is nuclear-based, the reaction
must be redone with increased protease digestion time. If the tissue morphology has been
destroyed and, hence, there is no signal, the reaction must be redone with decreased protease
digestion time (17).
In situ hybridization with LNA-DNA probes
Our protocol for detection of miRNAs by in situ hybridization has been previously published
(13,16). In brief, tissue sections or cells were digested in pepsin (2 mg/mL) for 30 min at room
temperature. The sequences of the different miRNAs studied by in situ hybridization with the
LNA modified probes are provided in Table 1. The probes (250 pmol) were made by Exiqon
(Woburn, MA, USA), and include multiple modified LNA nucleotides dispersed throughout
the sequence. The probes were labeled with the 3 oligonucleotide tailing kit (Enzo Diagnostics,
Farmingdale, NY) using biotin as the reporter nucleotide. The labeled probe was diluted in the
in situ hybridization buffer (Enzo Life Sciences), added to the slide and denatured at 60°C for
5 min, and then hybridized at 37°C for 15 h. After a 10-min wash in 0.2× salt sodium citrate
(SSC)/2% bovine serum albumin (BSA) at 5°C, the probe-target complex is visualized by the
action of alkaline phosphatase (as part of the streptavidin complex; Enzo Life Sciences) on the
chromogen NBT/BCIP. Nuclear fast red served as the counterstain. The negative controls
included omission of the probe and the use of a scrambled LNA probe (for miR-128a; see Table
1). Furthermore, miR-96 was chosen as an additional negative control for each analysis as this
miRNA is not expressed in most normal tissues, including brain, heart, and placenta (22). As
with the ultramer extension method and RT in situ PCR, the negative control with the LNA in
situ method was done on the same slide as the test section, since at least 2 sections (4 microns
thick) were placed on each slide.
RT in situ PCR
The protocol we used has been previously described (5,15). The miRNA precursors tested were
miR-1, and -128a using the same primers employed for solution phase RT-PCR analysis, as
recently described (14). The controls for the RNA-based signal included omission of the
primers, as lack of signal demonstrated loss of the nonspecific DNA synthesis pathway in the
intact cells (5,15; see Figure 1B).
Nuclear and cytoplasmic fractionization and real-time RT-PCR
HL60 and Jurkat cells were collected with a packed cell volume of 20 µl and nuclear, and
cytoplasmic fractions were isolated using NE-PER Nuclear and Cytoplasmic extraction
reagents (Pierce, Rockford, IL, USA) according to the manufacturer’s protocol. Total RNA
was isolated from both fractions using Trizol LS reagent (Invitrogen, Carlsbad, CA), and RNA
pellets were dissolved in an equal volume of water. One microliter of RNA from the nuclear
or cytoplasmic fraction was reverse transcribed using the protocol described in the TaqMan
assay (Applied Biosystems), and mature and miR-150 precursor expression was measured by
Nuovo et al. Page 4
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
real-time RT PCR as previously described (14). The mean of triplicate cDNAs was determined
and the data are presented as 2Ct, as described (20).
Results
Solution phase extension of mature miRNAs
Before testing the ultramer extension method in situ, we used an ultramer to miR-376 [chosen
because of prior work with this miRNA in pancreatic cancer (5)] to demonstrate that the mature
form of this miRNA could be extended in solution by a variety of polymerases. As evident
from Figure 2, rTth RT/DNA polymerase, Taq DNA polymerase, MMLV RT, Multiscribe RT,
and T7 RNA polymerase were each able to extend the mature miR-376. The increased size of
the Taq DNA polymerase product was likely due to the template-independent terminal
transferase activity of the polymerase that adds a single deoxyadenosine to the 3 ends of PCR
products.
Cell lines: correlation of real-time RT-PCR with in situ–based data in cell lines
It should be noted that the primers designed to anneal to the miRNA hairpin for real-time RT-
PCR and RT in situ PCR will amplify both the pri-and pre-microRNA since the hairpin is
contained within each (14). For this reason, we shall consider the amplification of miRNA
precursors to represent both the pri- and pre-miRNA. We analyzed the cancer cell lines HL60
and Jurkat for miR-150 expression (precursor and mature forms) by real-time RT-PCR (Figure
3) based on prior work with this miRNA that showed it was variably expressed in different
malignant cell lines (14). Note that both cell lines had high levels of the miR-150 precursors
(Figure 3A). However, the mature miR-150 was barely detectable in the HL60 cell line whereas
it was present at a high level in the Jurkat cell line (Figure 3B). This data allowed us to test
whether the ultramer extension system would detect only the mature miR-150 in these two cell
lines. No signal was seen using the ultramer extension method for miR-150 in the HL60 cells
(Figure 4B) whereas a strong, primarily cytoplasmic signal was seen in the Jurkat cells (Figure
4A). In situ hybridization with an LNA probe for miR-150 yielded an intense, primarily
cytoplasmic signal for the Jurkat cell line (Figure 4C). Using the same hybridization conditions,
no signal was seen in the HL60 cells (Figure 4D). Using RT in situ PCR for miR-150 precursors,
the Jurkat cells showed a cytoplasmic signal (Figure 4F) whereas the HL60 cells showed a
signal present in the entire cell (Figure 4E). The in situ data was then compared with cell
fractionation data using real-time RT-PCR to compare the miRNA copy numbers in the nucleus
versus the cytoplasm. As seen in Figure 3C, mature miR-150 had increased expression in the
cytoplasm of the Jurkat cells compared with the nuclear fraction and was undetectable in either
compartment in the HL60 cells; this is consistent with the LNA and ultramer extension data.
The miR-150 precursors were present in both the nucleus and cytoplasm of the Jurkat cells
(Figure 3C), predominating in the latter, which is consistent with the RT in situ PCR data.
However, the miR-150 precursors were mostly cytoplasmic in the HL60 cells (Figure 3C)
whereas the RT in situ PCR demonstrated a nuclear- and cytoplasmic-based signal (Figure 4E).
miR-128a and miR-1
miR-128a is highly expressed in normal brain (21,22). As demonstrated through real-time RT-
PCR, normal brain tissue expressed a high copy number of mature and miR-128a precursors
(Figure 5B). In comparison, whereas glioblastoma multiforme had equivalent amounts of
miR-128a precursors, this malignant tumor did not express detectable mature miR-128a
(Figure 5, A and B). Since the mature sequence of miR-128a is located at the 3 end of the
precursor, the premiR-128a could theoretically prime the ultramer and be extended. Thus,
normal versus malignant brain tissue allowed us to examine the specificity of the in situ
ultramer extension method for the mature miRNA in this type of situation. We examined five
normal brain tissues and three glioblastoma multiformes for mature and precursor miR-128a.
Nuovo et al. Page 5
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Using an LNA probe for mirR-128a on normal brain tissue, a strong cytoplasmic signal was
seen in neurons, but not other cell types such as endothelium and microglial cells (Figure 5C).
The negative controls (scrambled probes or miR-96 probe) gave no signal (data not shown).
Also, no signal was seen in the majority of cells in the glioblastoma samples with the LNA
probe (Figure 5D). Rare glioblastoma cells showed a nuclear signal with the LNA probe that
was slightly above background; the significance of this signal is unclear (data not shown). The
ultramer extension method for miR-128a demonstrated a predominantly cytoplasmic signal in
neurons in each of the five normal brain tissues (Figure 5E), but not in the three cases of
glioblastoma multiforme (Figure 5F). RT in situ PCR for miR-128a precursors in normal brain
tissue showed a predominantly cytoplasmic signal in cells with the cytologic features of
neurons (Figure 5G), while in glioblastoma cells it showed a mostly nuclear signal (Figure
5H). Thus, the normal brain/glioblastoma data provided further evidence that the ultramer
extension method is capable of only detecting the mature form of the miRNA and that extension
of the pre-miRNA on the ultramer template is not occurring.
Finally, we examined normal cardiac tissue for miR-1, as the mature form of this miR is
abundantly expressed in this tissue (22,23). The miR-1 ultramer-based signal was intense in
the heart tissue where it was present in over 95% of the myocytes and not evident in the
surrounding cells (Figure 6C). The signal was evident in a banding-type pattern that
corresponded to the A and H bands and the M line of the sarcomere (Figure 6C), providing
internal control evidence on the extension’s specificity. The ultramer-based signal was
eliminated if the inhibitor was added at the start of the reaction (Figure 6D, inset) or if the
ultramer was omitted (Figure 6D). A similar cytoplasmic localization for the miR-1 precursors
using RT in situ PCR was noted in cardiac tissue (Figure 6A), which was lost if the primers
were omitted (Figure 6B). The ultramer extension method was done on five heart tissues in
which the reporter nucleotide digoxigenin dUTP was omitted during the miR extension
process. After the ultramer extension process occurred, in situ hybridization was done using
the 101-nucleotide ultramer labeled with biotin. After in situ hybridization, a strong signal was
evident in the myofibrils of cardiac tissue and not the surrounding cell types (Figure 6E). This
signal was lost if the ultramer extension was omitted (Figure 6F).
Direct comparison of the ultramer extension and LNA in situ methods for miRNA detection
To compare the relative signal intensity of the LNA versus the ultramer extension method,
serial sections from normal placenta blocks were chosen and analyzed at the same time for
miRNAs 128a, 29b, and 16. As detailed by Liang et al. (22), miR-128a is not expressed in
placenta, miR-29b is expressed in low copy number, and the copy number of miR-16 is sixty
times higher than miR-29b. The relative intensity of the signals was then scored for six different
400× fields per slide, blinded to the miRNA being tested using Photoshop CS3 (Adobe
Systems, Inc., San Jose, CA, USA) and the eyedrop RGB function on the entire field. The mean
scores (± standard error) were as follows: miR-128a (LNA: 10.0 ± 2.8; ultramer: 8.1 ± 2.4),
miR-29b (LNA: 63.0 ± 6.6, ultramer: 64.3 ± 7.6), miR-16 (LNA: 115.1 ± 6.9, ultramer: 122.3
± 5.9). There was a significant difference between the signal intensities of miR-128a compared
with miR-29b (P = 0.002) and miR-16 (P = 0.0001) for both the LNA probes and ultramer
extension methods. There was also a significant difference between the intensities for low copy
(miR-29b) and high copy (miR-16) microRNA for each method (P = 0.002). However,
although the ultramer extension method gave less background and a stronger signal in each
class, the differences were not statistically significant. Finally, the signal intensities for the
LNA method were highly dependent on stringency conditions, whereas this was not the case
with the ultramer extension method. Specifically, increasing the post-hybridization wash to
50°C, which is typical with LNA probes and miRNA detection (11,18) eliminated the LNA-
based signal for miR-29b (11.0 ± 2.2) but had no effect on the signal for miR-29b as detected
by the ultramer extension method (ultramer 71.2 ± 5.6).
Nuovo et al. Page 6
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Discussion
We have described a new method for the in situ detection of mature microRNAs. The major
advantages of the ultramer extension method over the LNA probe method include that it is
specific for the mature, active form of the miRNA, it is easier to optimize with a broader
window between signal and background, is less sensitive to changes in stringency, and it is
much less expensive. For example, changes in stringency of around 50°C with the LNA probe
method could result in the eradication of the signal, as shown in this study and by Obernosterer
et al. (18), or the emergence of background (16). Under the same changes in stringency, the
ultramer extension method signal is not affected. Our cost for the LNA-labeled probe (250
pmol) was $40 per test. In comparison, the cost for the ultramer (6000 pmol) was $4 per test,
although there would be the additional charge for the reporter nucleotide and polymerase. The
potential for nonspecific DNA synthesis underlines the importance of performing the negative
control after DNase digestion for RT in situ PCR (no PCR primers) or the ultramer extension
reaction (no ultramer) on the same slide as the test section, and assuring that it shows no nuclear
signal. Similarly, we routinely perform the negative control for the LNA in situ hybridization
on a section on the same slide as the test, which underscores the importance of placing multiple
sections per slide. Another important internal control for the interpretation of the specificity
of in situ–based miRNA analysis is the well-documented cell and tissue specificity associated
with many miRNAs (22).
The ultramer extension method yielded a signal in those cells and tissues for which real-time
RT-PCR or published Northern blot analysis (22) supported the fact that the cells contained
the mature form of the given miRNA. A reason that we chose to study cardiac tissue and normal
brain tissues was because the abundant production of mature miRNAs that occur in these tissues
(miR-1 and -128a, respectively) is well-established (21,22). Conversely, the ultramer extension
method did not show any signal in HL60 cells for miR-150 or glioblastoma multiforme tissues
for miR-128a, where real-time RT-PCR showed that these samples did contain the miRNA
precursors, but not the mature form of the specific miR. This provides further evidence that
this method is specific for mature miRNA.
Direct comparisons of in situ hybridization with an LNA-modified probe, RT in situ PCR, and
the ultramer extension method provide insights into miRNA modulation. Specifically, the
observation that miR-150 and miR-128a precursors tend to be nuclear when there is no
detectable mature form (in HL 60 and malignant brain cells, respectively), but cytoplasmic in
the corresponding cell types when the mature form is evident, suggests that precursor
sequestration in the nucleus and its transport into the cytoplasm may play a role in the
modulation of miRNA expression. We reported a similar phenomenon in various cancer cell
lines (14). The data with miR-1, where the precursors are localized to the perimeter of
myofibrils and the mature form was seen in the A/H bands of the myofibrils, provides further
evidence that precursor transport to a specific compartment of the cell may be an important
factor in the processing of the precursor by Dicer into the mature, active form.
Acknowledgments
This work was supported by a grant from the Lewis Foundation (to G.J.N) and the National Institutes of Health (NIH;
grant no. R21 CA114304, to T.D.S). We thank Dr. Terry Elton for helpful discussions regarding miR-1 and miR-155.
We also greatly appreciate the support of Ventana Medical Systems, which provided some of the reagents for the
microRNA studies. We thank Dr. Margaret Nuovo for excellent and patient photographic work. This paper is subject
to the NIH Public Access Policy.
Nuovo et al. Page 7
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
References
1. Lewis BP, Burge CB, Bartel DP. Conserved seed paring, often flanked by adenosines, indicates that
thousands of human genes are microRNA targets. Cell 2005;120:15–20. [PubMed: 15652477]
2. Garzon R, Fabbri M, Cimmino A, Calin GA, Croce CM. MicroRNA expression and function in cancer.
Trends Mol. Med 2006;12:580–587. [PubMed: 17071139]
3. Wienholds E, Kloosterman WP, Miska E, Alvarez-Saavedra E, Berezikov E, de Bruijn E, Horvitz HR,
Kauppinen S, Plasterk RH. MicroRNA expression in zebrafish embryonic development. Science
2005;309:310–311. [PubMed: 15919954]
4. Ruby JG, Jan CH, Bartel DP. Intronic microRNA precursors that bypass Drosha processing. Nature
2007;448:83–86. [PubMed: 17589500]
5. Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, Morgan DL, Postier RG, et al.
Expression profiling identifies distinct microRNA signature in pancreatic cancer. Int. J. Cancer
2007;120:1046–1054. [PubMed: 17149698]
6. Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, et al.
Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell
2006;9:189–198. [PubMed: 16530703]
7. Jiang J, Lee EJ, Schmittgen TD. Increased expression of microRNA-155 in Epstein-Barr virus
transformed lymphoblastoid cell lines. Genes Chromosomes Cancer 2006;45:103–106. [PubMed:
16175574]
8. Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expression profiling of microRNA precursors in
human cancer cell lines. Nucleic Acids Res 2005;33:5394–5403. [PubMed: 16192569]
9. Schmittgen TD, Jiang J, Liu Q, Yang L. A high-throughput method to monitor the expression of
microRNA precursors. Nucleic Acids Res 2004;32:e43. [PubMed: 14985473]
10. Chen C, Ridzon DA, Zhou Z, Lee DH, Nguyen JT, Barbisin M, Xu NL, Mahuvakkar VR, et al. Real
time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res 2005;33:e179.
[PubMed: 16314309]
11. Politz JC, Zhang F, Pederson T. MicroRNA-206 colocalizes with ribosome-rich regions in both the
nucleolus and cytoplasm of rat myogenic cells. Proc. Natl. Acad. Sci. USA 2006;103:18957–18962.
[PubMed: 17135348]
12. Nelson PT, Baldwin DA, Kloosterman WP, Kauppinen S, Plasterk RH, Mourelatos Z. RAKE and
LNA-ISH reveal microRNA expression and localization in archival human brain. RNA 2006;12:187–
191. [PubMed: 16373485]
13. Martin MM, Buckenberger JA, Jiang J, Malana GE, Nuovo GJ, Chotani M, Feldman DS, Schmittgen
TD, Elton TS. The human angiotensin II type 1 receptor +1166 A/C polymorphism attenuates
miR-155 binding. J. Biol. Chem 2007;282:24262–24269. [PubMed: 17588946]
14. Lee EJ, Back M, Gusev Y, Brackett DJ, Nuovo GJ, Schmittgen TD. Systematic evaluation of
microRNA processing patterns in tissues, cell lines, and tumors. RNA 2008;14:35–42. [PubMed:
18025253]
15. Nuovo, GJ. RT in situ PCR. In: Nuovo, GJ., editor. PCR in situ hybridization: Protocols and
Applications. Vol. 3rd edition. Baltimore, MD: Lippincott Williams and Wilkins Press; 1997. p.
271-333.
16. Nuovo GJ. In situ detection of precursor and mature microRNAs in paraffin embedded, formalin
fixed tissues and cell preparations. Methods 2008;44:39–46. [PubMed: 18158131]
17. Christensen U, Jacobsen N, Rajwanshi VK, Wengel J, Koch T. Stopped-flow kinetics of locked
nucleic acid (LNA)-oligonucleotide duplex formation: studies of LNA-DNA and DNA-DNA
interactions. Biochem. J 2001;354:481–484. [PubMed: 11237851]
18. Obernosterer G, Martinez J, Alenius M. Locked nucleic acid-based in situ detection of microRNAs
in mouse tissue sections. Nat. Protocols 2007;2:1508–1514.
19. Obernosterer G, Leuschner PJ, Alenius M, Martinez J. Post-transcriptional regulation of microRNA
expression. RNA 2006;12:1161–1167. [PubMed: 16738409]
20. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat.
Protocols 2008;3:1101–1108.
Nuovo et al. Page 8
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
21. Wulczyn FG, Smirnova L, Rybak A, Brandt C, Kwidzinski E, Ninnemann O, Strehle M, Seiler A, et
al. Post-transcriptional regulation of the let-7 microRNA during neural cell specification. FASEB J
2007;21:415–426. [PubMed: 17167072]
22. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, et al.
MicroRNA-133 controls cardiac hypertrophy. Nat. Med 2007;13:613–618. [PubMed: 17468766]
23. Liang Y, Ridzon D, Wong L, Chen C. Characterization of microRNA expression profiles in normal
human tissues. BMC Genomics 2007;8:166. [PubMed: 17565689]
Nuovo et al. Page 9
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Schematic representation of (A) the ultramer extension method and (B) the non-specific
in situ DNA synthesis process in paraffin-embedded, formalin-fixed tissues
(A) An ultramer that has the cDNA of the microRNA (upper panel; mature miR-1 is used in
this example) at its 3 end preceded by a series of 20 nucleotide repeats (5-
GACCCCTTAATGCGTCTAAA-3) serves as the template for miR extension. As the miR is
being extended, a reporter nucleotide such as digoxigenin-dUTP is incorporated into the
synthesized DNA (lower panel) which can then be detected using an anti-digoxigenin antibody.
(B) Fixation in buffered formalin creates extensive crosslinking between proteins and nucleic
acids in the cell. The high temperature (60–70°C) used in paraffin embedding the formalin-
fixed tissues for tissue sections induces small nicks and short gaps in the DNA (part 1), which
Nuovo et al. Page 10
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
are the initiation points for non-specific DNA synthesis by a DNA polymerase. This DNA
repair–like process is facilitated by protease digestion, which removes the proteins crosslinked
to the DNA (part 2). Optimal protease digestion also allows the genomic DNA to be completely
digested by DNase (part 3) such that it can no longer support this nonspecific DNA synthesis
pathway. If too many protein-DNA crosslinks persist due to inadequate protease digestion,
then the DNase cannot eliminate this DNA repair–like pathway (part 4).
Nuovo et al. Page 11
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. Solution phase extension of miR-376a ultramer
A miR-376a ultramer was primed by a 32P-labeled miR-376a oligoribonucleotide. The oligos
were extended using a variety of polymerases and the products were verified by gel
electrophoresis. Each column represents either a positive or negative enzymatic reaction: 1,
rTth polymerase; 2, no rTth polymerase control; 3, no Taq DNA polymerase control; 4, Taq
DNA polymerase; 5, MMLV RT; 6, no MMLV RT control; 7, Multiscribe RT; 8, no
Multiscribe RT control; 9, T7 RNA polymerase; 10, no T7 RNA polymerase control. The
molecular size was estimated by comparing the migration to that of a 10-nucleotide (nt) RNA
ladder.
Nuovo et al. Page 12
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. Real-time RT-PCR for miR-150 precursors and mature molecules in two cancer cell lines
Real-time RT-PCR shows that miR-150 precursors were each present in the cell lines HL60
and Jurkat (A), while only the mature form of miR-150 was detectable in the Jurkat cell line
(B). The cell fractionation experiments showed that the mature miR-150 was present in much
higher amounts in the cytoplasm (black bar) than the nucleus (gray bar) in the Jurkat cells (C)
and, thus, was consistent with the ultramer extension data (see Figure 4).
Nuovo et al. Page 13
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4. In situ detection of miR-150 precursors and mature molecules in two cancer cell lines
The ultramer extension method for mature miR-150 yielded a strong cytoplasmic signal in the
Jurkat cells (A, arrow) and no signal with the HL60 cells (B) under identical conditions. Using
in situ hybridization with an LNA probe, a strong cytoplasmic signal was seen in the Jurkat
cells (C, arrow) and no signal was seen in the majority of the HL 60 cells (D). The inset in D
shows the relative sizes of the nuclear and cytoplasmic compartment in these cells. E and F
show the results for RT in situ PCR using the primers for miR-150 precursors; note that the
Jurkat signal (F) is primarily cytoplasmic. The signal for the miR-150 precursors in the HL60
cells (E) shows an intense signal that involves the entire cell. In such cases, it is not possible
Nuovo et al. Page 14
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
to determine if the signal is cytoplasmic- and nuclear-based, or primarily nuclear-based (D,
inset).
Nuovo et al. Page 15
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 5. The in situ localization of miR-128a in normal versus malignant brain tissues
(A) Equivalent copy numbers by qRTPCR are shown for the miR-128a precursors in normal
(premiR128N) versus malignant brain tissue (premiR128T), while the mature form of
miR-128a was produced in normal brain tissue (miR128N) but not in the brain cancer cells
(miR128T) (B). The LNA probe for miR-128a produced a strong cytoplasmic signal in neurons
in normal brain (C) and no signal in the majority of glioblastoma multiforme cells (D). A
cytoplasmic signal is seen after ultramer extension in normal brain, and it is localized to the
same compartment of neurons as the LNA probe (E; the lower inset is the negative control of
adding the inhibitor to the ultramer reaction and the upper insert is the control of no DNase).
Malignant brain tissue showed the absence of any signal with the ultramer extension method
Nuovo et al. Page 16
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
(F). G and H show the RT in situ PCR results for the miR-128a precursors in normal and
malignant brain tissue, respectively; note the predominantly cytoplasmic signal seen in neurons
(G) and the nuclear signal present in most malignant brain cells (H).
Nuovo et al. Page 17
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 6. The in situ localization of miR-1
(A) Depiction of the miR-1 precursors in myocytes as detected by RT in situ PCR; note that
the signal localizes to the periphery of each myofibril. The signal was lost if the primers were
omitted (B); the insert shows the control of no DNase digestion where the signal is nuclear-
based. With the ultramer extension method for the mature miR-1, the signal was seen in the
myofibril, but now in the A and H bands (C); the arrow depicts a negative fibroblast which
serves as an internal control. The ultramer extension signal was lost if the ultramer was omitted
(D) or if the inhibitor DNA sequence was added at the onset of the extension (D, inset). E
shows the results with the ultramer extension where the reporter nucleotide was omitted, and
Nuovo et al. Page 18
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the miR-1 extended product detected by in situ hybridization with a biotin labeled probe; the
signal was lost if the ultramer was omitted during extension (F)
Nuovo et al. Page 19
Biotechniques. Author manuscript; available in PMC 2009 June 9.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Nuovo et al. Page 20
Table 1
Sequence of the LNA-Modified Probes and Ultramers Used in This Study
microRNA Probe sequence
miRNA-1 TACATACTTCTTTACATTCCA
miR-16 TAGCAGCACGTAAATATTGGCG
miR-29b GAACACCAGGAGAAATCGGTCA
miR-96 AGCAAAAATGTGCTAGTGCCAAA
miRNA-128a AAAGAGACCGGTTCACTGTGA
miRNA-150 CACTGGTACAAGGGTTGGGAGA
Scrambled probe (miR-128a) CGTATAGGCCCAAGAATTAGG
Ultramer miR-1 GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
TACATACTTCTTTACATTCCA
Ultramer miR-16 GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA TAGCAGCACGTAAATATTGGCG
Ultramer miR-29b GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GAACACCAGGAGAAATCGGTCA
Ultramer miR-128a GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
AAAGAGACCGGTTCACTGTGA
Ultramer miR-150 GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
CACTGGTACAAGGGTTGGGAGA
Scrambled ultramer miR-128a- GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
GACCCCTTAATGCGTCTAAA GACCCCTTAATGCGTCTAAA
CGTATAGGCCCAAGAATTAGG
Inhibitor TTTAGACGCATTAAGGGGTC
The underlined sequence is complementary to the mature miRNA.
Biotechniques. Author manuscript; available in PMC 2009 June 9.
... Endothelial cells (ECs) also up-regulate miR-155 under inflammatory conditions, and miR-155 has been implicated in BBB dysfunction caused by neuro-inflammation [15]. The in situ detection of miR-155 (and other microRNAs) is well established and can be performed by any diagnostic pathology laboratory [16]. ...
... ISH for miR-155 was performed by Phylogeny Inc. (Powell, OH), as previously described [16]. Sections were evaluated blindly by the pathologist. ...
... Co-expression analyses were done using the Nuance system (CRI) as previously published [16]. In brief, a given tissue was tested for two different targets using fast red, NBT/BCIP or DAB as the chromogens. ...
Article
Spinal cord paralysis is relatively common after surgical repair of thoraco-abdominal aortic aneurysm (TAAA) and its etiology is unknown. The present study was designed to examine the histopathology of the disease and investigate whether miR-155 ablation would reduce spinal cord ischemic damage and delayed hindlimb paralysis induced by aortic cross-clamping (ACC) in our mouse model. The loss of locomotor function in ACC-paralyzed mice correlated with the presence of extensive gray matter damage and central cord edema, with minimal white matter histopathology. qRTPCR and Western blotting showed that the spinal cords of wild-type ACC mice that escaped paralysis showed lower miR-155 expression and higher levels of transcripts encoding Mfsd2a, which is implicated in the maintenance of blood-brain barrier integrity. In situ based testing demonstrated that increased miR-155 detection in neurons was highly correlated with the gray matter damage and the loss of one of its targets, Mfsd2a, could serve as a good biomarker of the endothelial cell damage. In vitro, we demonstrated that miR-155 targeted Mfsd2a in endothelial cells and motoneurons and increased endothelial cell permeability. Finally, miR-155 ablation slowed the progression of central cord edema, and reduced the incidence of paralysis by 40%. In sum, the surgical pathology findings clearly indicated that the epicenter of the ischemic-induced paralysis was the gray matter and that endothelial cell damage correlated to Mfsd2a loss is a good biomarker of the disease. MiR-155 targeting therefore offers new therapeutic opportunity for edema caused by traumatic spinal cord injury and diagnostic pathologists, by using immunohistochemistry, can clarify if this mechanism also is important in other ischemic diseases of the CNS, including stroke.
... Different subtypes of a particular cancer can be discriminated by gene expression profiling based upon microarray platforms [164]. Numerous methods detecting miRNAs have been developed, including quantitative real-time polymerase chain reaction (RT-qPCR), in situ hybridization [165,188], and high throughput sequencing [189]. As technology, creativity, and pathophysiology comprehension advances, so will our capability to diagnosis CRC earlier and more accurately [3]. ...
Article
Full-text available
Colorectal cancer (CRC) is the third most lethal malignancy, with pathogenesis intricately dependent upon microRNAs (miRNAs). miRNAs are short, non-protein coding RNAs, targeting the 3′-untranslated regions (3′-UTR) of certain mRNAs. They usually serve as tumor suppressors or oncogenes, and participate in tumor phenotype maintenance. Therefore, miRNAs consequently regulate CRC carcinogenesis and other biological functions, including apoptosis, development, angiogenesis, migration, and proliferation. Due to its differential expression and distinct stability, miRNAs are regarded as molecular biomarkers (for diagnosis/prognosis) and therapeutic targets for CRC. Recently, a remarkable number of miRNAs have been discovered with implications via incompletely understood mechanisms in CRC. As further study of relevant miRNAs continues, it is hopeful that novel miRNA-based therapeutic strategies may be available for CRC patients in the future.
... Over the past few years, an increasing number of methods had emerged to detect short oligonucleotide sequences based on quantitative real-time polymerase chain reaction [15][16][17], in situ hybridization [18][19][20] and high throughput sequencing [21,22]. These methods are however expensive and in situ hybridization is technically challenging and semi-quantitative. ...
Article
We report on the preparation of a sensing platform for the fast multiplex detection of short oligonucleotide sequences. Monodispersed micro-sized polystyrene beads (average diameter of ca. 4.72 μm) were prepared and subsequently coated with nanosized gold nanoparticles. The micro-sized beads are large enough to scatter light and allow the detection of specific analytes using flow cytometry. Fluorescently labeled DNA hairpins are assembled onto the gold nanoparticles. In the absence of the target sequence, the metal particles quench the probe signal. Upon hybridization, the fluorescent signal of the hairpin is turned on and is readily detected by the flow cytometer with a detection limit of 5 nM. The sensing platform was able to sense and discriminate in parallel two specific genetic markers of Vaccinia virus and Hepatitis B Virus. Viral sequences spiked in an artificial serum were readily detected with comparable sensitivity to the buffer solution. The assay time was optimized from the assembly to testing to take less than 2 hours including a hybridization of 30 mins.
... High copy numbers levels have been observed for miR-31 in normal astrocytes (340 copies/pg small RNA) (Wu et al., 2009) and miR-128 in normal brain tissue (Nuovo et al., 2009) (Most of our experiments, cell culture and mouse studies, were conducted by transducing lenti-vectors expressing miRNAs at MOI of 1e2 (1e2 copies per cell)). In respect to glioblastoma (U87), consistent with other research (Skalsky and Cullen, 2011;Ciafre et al., 2005) we demonstrate very low miR-128 expression levels in comparison with breast cancer cells (MCF7) ( Figure S11B). ...
Article
Oncolytic measles virus strains are currently being evaluated in several clinical trials, as a promising novel oncolytic platform. Poliovirus receptor-related 4 (PVRL4) was recently identified as a potent measles virus (MV) receptor; however, its regulation is not yet understood. Increased levels of PVRL4 protein were observed in cell membrane, cytoplasm and nuclei of glioblastoma, breast and ovarian tumor clinical samples with no significant change in PVRL4 mRNA levels in glioblastoma and breast cancer compared with their corresponding control samples, suggesting that PVRL4 is likely post-transcriptionally regulated. Therefore, we sought to investigate the potential role of miRNAs in PVRL4 regulation and thus MV infectivity. We demonstrated that miR-31 and miR-128 can bind to the 3’UTR of PVRL4 and decrease PVRL4 levels while anti-miR-31/128 increase PVRL4 levels suggesting that PVRL4 is miRNA targeted. Furthermore, miR-31/128 expression levels were down-regulated in glioblastoma and breast tumor samples and showed significant negative correlations with PVRL4 levels. Infection with an MV strain that exclusively utilizes PVRL4 as its receptor showed that over-expression of miR-31/128 decreases MV infectivity while inhibition of the respective miRNAs via anti-miRs increase MV infectivity and reduces tumor size in mouse xenograft models of glioblastoma, breast and ovarian cancer. Additionally, miR-128 levels showed significant correlations with MV infection and in vivo anti-tumor effect, while MV infection increased miR-31 expression and thereby contributed to the observed decrease in PVRL4 levels. This study suggests that PVRL4 is post-transcriptionally regulated by miR-128 and miR-31 and harbors possible miRNA targets that could modulate MV infectivity and in turn enhance MV based oncolytic therapeutic strategies.
... Sections (5 mm thick) of carotid arteries and aortic roots were treated with DNase (Roche) for 15-17 h at 37°C. One-step reverse transcriptase in situ PCR was performed using gene-specific Taq in situ primers (Sigma-Aldrich; Supplementary Table 3), SuperScript One-Step RT-PCR with PlatinumTaq (Life Technologies), and digoxigenin-11-dUTPs (Roche) 68 . After two washes with saline-sodium citrate buffer buffer and blocking of biotin/ avidin-binding sites (Blocking Kit; Vector Laboratories), the sections were incubated with horseradish peroxidase-conjugated anti-digoxigenin sheep F'ab fragments (Roche) for 1 h at 37°C. ...
Article
Full-text available
MicroRNAs regulate the maladaptation of endothelial cells (ECs) to naturally occurring disturbed blood flow at arterial bifurcations resulting in arterial inflammation and atherosclerosis in response to hyperlipidemic stress. Here, we show that reduced endothelial expression of the RNAse Dicer, which generates almost all mature miRNAs, decreases monocyte adhesion, endothelial C-X-C motif chemokine 1 (CXCL1) expression, atherosclerosis and the lesional macrophage content in apolipoprotein E knockout mice (Apoe-/-) after exposure to a high-fat diet. Endothelial Dicer deficiency reduces the expression of unstable miRNAs, such as miR-103, and promotes Krüppel-like factor 4 (KLF4)-dependent gene expression in murine atherosclerotic arteries. MiR-103 mediated suppression of KLF4 increases monocyte adhesion to ECs by enhancing nuclear factor-κB-dependent CXCL1 expression. Inhibiting the interaction between miR-103 and KLF4 reduces atherosclerosis, lesional macrophage accumulation and endothelial CXCL1 expression. Overall, our study suggests that Dicer promotes endothelial maladaptation and atherosclerosis in part by miR-103-mediated suppression of KLF4.
Chapter
Organophosphorus Chemistry provides a comprehensive and critical review of the recent literature. Coverage includes phosphines and their chalcogenides, phosphonium salts, low coordination number phosphorus compounds, penta- and hexa- coordinated compounds, quiquevalent phosphorus acids, nucleotides and nucleic aicds, ylides and related compounds, phosphazenes and the application of physical methods in the study of organophosphorus compounds. This is the 40th in a series of volumes which first appeared in 1970 under the editorship of Stuart Trippett and which covered the literature of organophosphorus chemistry published in the period from January 1968 to June 1969, citing some 1370 publications. The present volume covers the literature from January 2009 to January 2010, citing more than 2200 publications, continuing our efforts to provide an up to date survey of progress in an area of chemistry that has expanded significantly over the past 40 years.
Chapter
Surface-enhanced Raman scattering (SERS) in combination with advanced multivariate methods for pattern recognition has been used for rapid, sensitive, and accurate identification of miRNAs. The strength of the SERS-based sensor is its sensitivity to detect extremely low levels of analyte and specificity to provide the molecular fingerprint of the analyte. The SERS spectra of related and unrelated miRNAs can be detected in near-real time; that detection is sequence dependent, and that SERS spectra can be used to classify miRNA patterns with high accuracy. This technique requires oblique-angle deposition (OAD) fabrication of nanostructured SERS substrate, spotting of RNA or miRNA onto the SERS substrate, and measurement of SERS spectra, followed by classification of the SERS spectra using partial least squares discriminate analysis (PLS–DA). The first application of this technique to miRNA profiling was made recently by Driskell et al. from the Department of Infectious Diseases, Center for Disease Intervention, University of Georgia (AHRC, USA) (Biosens Bioelectron 24:923–928, 2008). This technology is well suited for routine miRNA expression profiling in clinic laboratory. However, the SERS technique requires sophisticated read-out system, complicated analysis skill, and convoluted data interpretation and verification, which requires highly trained professionals to handle.
Chapter
The in situ detection of microRNAs (miRNAs) has been greatly facilitated by several recent technologic advances. These include the locked nucleic acid (LNA)-modified nucleotide, in situ extension of the mature miRNA, and RT in situ PCR amplification of the pre- and pri-microRNA. Furthermore, marked increases in the sensitivity of automated immunohistochemistry allow for co-localization of a given miRNA and its possible target. Key variables for successful LNA in situ detection of miRNAs include probe concentration, miRNA copy number, and stringency of the hybridization and wash. Success with RT in situ PCR and ultramer extension is highly dependent on adequate protease digestion. The key variables with immunohistochemistry include antibody concentration and pretreatment conditions. With co-labeling, miRNA detection is done first; the antigen can be co-localized if the immunohistochemistry reaction requires pretreatment in either the protease or antigen retrieval.
Chapter
In situ hybridization is an important tool for analyzing gene expression and developing hypotheses about gene functions. Normal hybridization requires the isolation of DNA or RNA, separating it on a gel, blotting it onto nitrocellulose, and probing it with a complementary sequence. For hybridization histochemistry, sample cells and tissues are usually treated to fix the target transcripts in place and increase access of the probe. As noted above, the probe is either a labeled complementary DNA or, now most commonly, a complementary RNA. The probe hybridizes to the target sequence at elevated temperature, and then the excess probe is washed away. Solution parameters such as temperature, salt, and/or detergent concentration can be manipulated to remove any non-identical interactions. The probe that was labeled with either radio-, fluorescent- or antigen-labeled bases is then localized and quantitated in the tissue using autoradiography, fluorescence microscopy, or immunohistochemistry, respectively. ISH can also use two or more probes, labeled with radioactivity or the other non-radioactive labels, to simultaneously detect two or more transcripts. Alenius and colleagues have applied LNA-modified miRNA capture oligo probe to ISH to enhance the detection sensitivity and specificity, compared to normal DNA probes (RNA 12:1161–1167, 2006; Nature Protocols 2:1508–1514, 2007). LNA-modified oligonucleotide probes display markedly increased hybridization affinities toward complementary RNAs compared to traditional RNA or DNA based probes (Biochemistry 43:13233–13241, 2004). This property allows for very stringent hybridization conditions, increasing the specificity and sensitivity of miRNA detection (Nucleic Acids Res 32:e175, 2004; Biochemistry 43:13233–13241, 2004). Most recently, Schmittgen and colleagues from Ohio State University Medical Center (Columbus, OH, USA) (Methods 46:115–126, 2009) have developed a novel method for in situ detection of the mature miRNA, based on the extension of the molecule after its hybridization, to an ultramer template in formalin-fixed, paraffin-embedded tissues. The major advantages of the ultramer extension method over the LNA probe method is that it is specific to the mature, active form of the miRNA, is easier to optimize with a broader window between signal and background, is less sensitive to changes in stringency, and is much less expensive.
Article
Full-text available
The locked nucleic acid (LNA) monomer is a conformationally restricted nucleotide analogue with an extra 2'-O,4'-C-methylene bridge added to the ribose ring. Oligonucleotides that contain LNA monomers have shown greatly enhanced thermal stability when hybridized to complementary DNA and RNA and are considered most promising candidates for efficient recognition of a given mixed sequence in a nucleic acid duplex and as an antisense molecule. Here the kinetics and thermodynamics of a series of oligonucleotide duplex formations of DNA-DNA and DNA-LNA octamers were studied using stopped-flow absorption measurements at 25 degrees C and melting curves. The reactions of the DNA octamer 5'-CAGGAGCA-3' with its complementary DNA octamer 5'-TGCTCCTG-3', and with the LNA octamers 5'-T(L)GCTCCTG-3' (LNA-1), 5'-T(L)GCT(L)CCTG-3' (LNA-2) and 5'-T(L)GCT(L)CCT(L)G-3'(LNA-3), containing respectively one, two or three thymidine 2'-O,4'-C-methylene-(D-ribofuranosyl) nucleotide monomers, designated T(L), were studied. In all cases were seen fast second-order association reactions with k(obs)=2x10(7) M(-1)s(-1). At 25 degrees C the dissociation constants of the duplexes obtained from melting curves were: DNA-DNA, 10 nM; DNA-LNA-1, 20 nM; DNA-LNA-2, 2 nM; and DNA-LNA-3, 0.3 nM; thus the greatly enhanced duplex stability induced by LNA is confirmed. Since the association rates were all equal this increase in stability is due to slower rates of dissociation of the complexes.
Article
Full-text available
Hundreds of small RNAs of approximately 22 nucleotides, collectively named microRNAs (miRNAs), have been discovered recently in animals and plants. Although their functions are being unravelled, their mechanism of biogenesis remains poorly understood. miRNAs are transcribed as long primary transcripts (pri-miRNAs) whose maturation occurs through sequential processing events: the nuclear processing of the pri-miRNAs into stem-loop precursors of approximately 70 nucleotides (pre-miRNAs), and the cytoplasmic processing of pre-miRNAs into mature miRNAs. Dicer, a member of the RNase III superfamily of bidentate nucleases, mediates the latter step, whereas the processing enzyme for the former step is unknown. Here we identify another RNase III, human Drosha, as the core nuclease that executes the initiation step of miRNA processing in the nucleus. Immunopurified Drosha cleaved pri-miRNA to release pre-miRNA in vitro. Furthermore, RNA interference of Drosha resulted in the strong accumulation of pri-miRNA and the reduction of pre-miRNA and mature miRNA in vivo. Thus, the two RNase III proteins, Drosha and Dicer, may collaborate in the stepwise processing of miRNAs, and have key roles in miRNA-mediated gene regulation in processes such as development and differentiation.
Article
Full-text available
microRNAs (miRNAs) are small, functional, non‐coding RNAs. miRNAs are transcribed as long primary transcripts (primary precursors) that are processed to the ∼75 nt precursors (pre‐miRNAs) by the nuclear enzyme Drosha. The ∼22 nt mature miRNA is processed from the pre‐miRNA by the RNase III Dicer. The vast majority of published studies to date have used northern blotting to detect the expression of miRNAs. We describe here a sensitive, high throughput, real‐time PCR assay to monitor the expression of miRNA precursors. Gene‐specific primers and reverse transcriptase were used to convert the primary precursors and pre‐miRNAs to cDNA. The expression of 23 miRNA precursors in six human cancer cell lines was assayed using the PCR assay. The miRNA precursors accumulated to different levels when compared with each other or when a single precursor is compared in the various cell lines. The precursor expression profile of three miRNAs determined by the PCR assay was identical to the mature miRNA expression profile determined by northern blotting. We propose that the PCR assay may be scaled up to include all of the 150+ known human miRNA genes and can easily be adaptable to other organisms such as plants, Caenorhabditis elegans and Drosophila.
Article
Full-text available
MicroRNAs (miRNAs) are small noncoding RNAs, about 21 nucleotides in length, that can regulate gene expression by base-pairing to partially complementary mRNAs. Regulation by miRNAs can play essential roles in embryonic development. We determined the temporal and spatial expression patterns of 115 conserved vertebrate miRNAs in zebrafish embryos by microarrays and by in situ hybridizations, using locked-nucleic acid–modified oligonucleotide probes. Most miRNAs were expressed in a highly tissue-specific manner during segmentation and later stages, but not early in development, which suggests that their role is not in tissue fate establishment but in differentiation or maintenance of tissue identity.
Article
Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative CT method also referred to as the 2−ΔΔCT method. This protocol provides an overview of the comparative CT method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.
Article
The locked nucleic acid (LNA) monomer is a conformationally restricted nucleotide analogue with an extra 2′-O,4′-C-methylene bridge added to the ribose ring. Oligonucleotides that contain LNA monomers have shown greatly enhanced thermal stability when hybridized to complementary DNA and RNA and are considered most promising candidates for efficient recognition of a given mixed sequence in a nucleic acid duplex and as an antisense molecule. Here the kinetics and thermodynamics of a series of oligonucleotide duplex formations of DNA-DNA and DNA-LNA octamers were studied using stopped-flow absorption measurements at 25 °C and melting curves. The reactions of the DNA octamer 5′-CAGGAGCA-3′ with its complementary DNA octamer 5′-TGCTCCTG-3′, and with the LNA octamers 5′-TLGCTCCTG-3′ (LNA-1), 5′-TLGCTLCCTG-3′ (LNA-2) and 5′-TLGCTLCCTLG-3′(LNA-3), containing respectively one, two or three thymidine 2′-O,4′-C-methylene-(D-ribofuranosyl) nucleotide monomers, designated TL, were studied. In all cases were seen fast second-order association reactions with kobs = 2 × 107 M-1·s-1. At 25 °C the dissociation constants of the duplexes obtained from melting curves were: DNA-DNA, 10 nM; DNA - LNA-1, 20 nM; DNA - LNA-2, 2 nM; and DNA - LNA-3, 0.3 nM; thus the greatly enhanced duplex stability induced by LNA is confirmed. Since the association rates were all equal this increase in stability is due to slower rates of dissociation of the complexes.
Article
Smith [Smith, M., 1996. Shareholder activism by institutional investors: evidence from CALPERS. Journal of Finance 51, 227-252] and Wahal [Wahal, S., 1996. Public pension fund activism and firm performance. Journal of Financial and Quantitative Analysis 31, 1-23] identify significant positive abnormal returns surrounding the announcement of performance targetings by the California Public Employees Retirement System (CalPERS), dubbed the “CalPERS effect.” More recent studies suggest that this “CalPERS effect” continues in later samples. While I confirm the early period results, I find the results reported in studies examining later periods are driven by the inclusion of early 1992–1993 targetings and from a significant bias in the market model parameters caused by estimation during periods of known under-performance. Additionally, these results are partially driven by the failure to control for contaminating events and the use unnecessarily long event windows. Contrary to previous studies, after addressing these methodological concerns, I find no evidence to support the continued existence of a “CalPERS effect”.
Article
We predict regulatory targets of vertebrate microRNAs (miRNAs) by identifying mRNAs with conserved complementarity to the seed (nucleotides 2-7) of the miRNA. An overrepresentation of conserved adenosines flanking the seed complementary sites in mRNAs indicates that primary sequence determinants can supplement base pairing to specify miRNA target recognition. In a four-genome analysis of 3' UTRs, approximately 13,000 regulatory relationships were detected above the estimate of false-positive predictions, thereby implicating as miRNA targets more than 5300 human genes, which represented 30% of our gene set. Targeting was also detected in open reading frames. In sum, well over one third of human genes appear to be conserved miRNA targets.