You are on page 1of 6

METHODS COMMENTARY METHODS COMMENTARY

Transcription 2:3, 103-108; May/June 2011; © 2011 Landes Bioscience

Inhibiting eukaryotic transcription


Which compound to choose? How to evaluate its activity?
Olivier Bensaude
IBENS; UMR CNRS 8197; UA INSERM 1024; Paris, France

T his review first discusses ways in


which we can evaluate transcrip-
tion inhibition, describe changes in
are key issues. How can we detect if a new
compound inhibits transcription? This
review will deal first with the general
nuclear structure due to transcription questions of how to evaluate transcription
inhibition, and report on genes that are inhibition, describe changes in nuclear
paradoxically stimulated by transcrip- structure due to transcription inhibition,
tion inhibition. Next, it summarizes the and report on genes that are paradoxi-
characteristics and mechanisms of com- cally stimulated by transcription inhibi-
monly used inhibitors: α-amanitin is tion. Next, we will focus on widely used
highly selective for RNAP II and RNAP compounds (α-amanitin, actinomycin D,
III but its action is slow, actinomycin D DRB, flavopiridol) and triptolide, a new
is fast but its selectivity is poor, CDK9 compound that looks very promising.
inhibitors such as DRB and flavopiridol
are fast and reversible but many genes Evaluating Transcription I­nhibition
escape transcription inhibition. New
compounds, such as triptolide, are fast How can we determine if a compound
and selective and able to completely inhibits transcription? Quantification of
arrest transcription by triggering rapid 3
H-uridine incorporation into RNA is the
degradation of RNAP II. oldest method. 3H-uridine permeates rap-
idly into cells, is metabolized and incor-
Introduction porated into nascent RNA transcripts.
3
H-RNA accumulation results from com-
Many anti-cancer drugs inhibit transcrip- peting RNA synthesis and degradation.
tion and most transcription inhibitors Thus, short labeling time (a fraction of
have useful pharmacological properties. an hour for mammalian cells) is recom-
Many experiments require inhibition of mended to favor synthesis over degradation.
transcription. In yeast, thermosensitive However, this method does not distinguish
mutations in RNA polymerase (RNAP) between polymerases. It gives a maximal
subunits provide precious tools. A thermo- weight to RNAP I activity as rRNAs repre-
sensitive mutation has been characterized sent 60–70% of total transcript mass.
in mammalian RNAP II largest subunit, Investigating RNA levels by northern
Key words: RNA polymerase, Rpb1.1 However, transcription is main- blot, RT-Q-PCR, gene arrays on DNA
transcription, amanitin, actinomycin, tained for at least one day at non-permissive chips or massive sequencing provides infor-
DRB, flavopiridol, triptolide, CDK9, temperature, which is a major inconve- mation on specific transcripts. One may
TFIIH, Rpb1 nient. Therefore, “chemical genetics” follow the effect of drugs on short-lived
Submitted: 04/12/11 or “chemical biology” is usually pre- RNAs using RNAs with long half-lives
ferred. Among the various drugs available such as actin or rRNA as reference controls.
Accepted: 04/28/11 to inhibit transcription, how to choose However, one should keep in mind that
DOI: 10.4161/trns.2.3.16172 which one to use? Each has its advantages inhibition of transcription may enhance
*Correspondence to: Olivier Bensaude; and drawbacks (Table 1). Selectivity, effi- the stability of some mRNAs, such as those
Email: bensaude@biologie.ens.fr ciency, rapidity of action and reversibility of DNA damage-inducible (gadd) genes,

www.landesbioscience.com Transcription 103


Table 1. Overview of widely used inhibitors of transcription
Actinomycin D α-amanitin DRB Flavopiridol Triptolide
>0.01 µgml for class
-1

I genes
Concentration >2 µgml-1 100 µM >0.5 µM 1 µM
>1 µgml-1 for class II
genes
Stock solution in DMSO aqueous DMSO aqueous DMSO
Target DNA intercalation RNAPII >> RNAPIII CDK9 in P-TEFb CDK9 in P-TEFb XPB in TFIIH
RNAP II elonga- RNAP II elonga-
RNA polymerase RNA synthesis RNAP I and RNAP II
tion inhibited rRNA tion inhibited, rRNA
elongation inhibited ­inhibited ­initiation inhibited
­processing impaired ­processing impaired
Other potential target:
GC-rich DNA RNAPII & RNAPIII
Target Selectivity Other kinases inhibited Other kinases inhibited Polycystin-2 calcium
sequences only known targets
channel
Inhibition Class I >> Class II >> Class II >> Class III Class II transcription Class II transcription, Class II & Class I
Selectivity Class III transcription transcription Class I processing Class I processing ­transcription
Consequence on Proteasome-
CTD hyperphosphor- CTD serine 2 dephos- CTD serine 2 dephos-
RNA polymerase RNAPII degradation dependent RNAPII
ylation phorylation phorylation
II degradation
Reversibility Weak No Yes ? No
Rate Fast, minutes Slow, hours Fast, minutes Fast, minutes Fast, minutes

and lead to their accumulation when inhib- during global transcription inhibition. or defects in rRNA processing function as
itors are employed at moderate concentra- As a first example, transcription driven triggers of that process.20,28 An arrest in
tions.2-5 But unstable RNAs such as c-fos by the HIV-LTR is enhanced by ama- ribosome assembly releases ribosomal pro-
do not seem to be affected.6 nitin and actinomycin.11,12 The “silent” tein subunits such as RPL26. These trap
Fluorescence in situ hybridization HIV-LTR drives an efficient transcrip- the Hdm2 (human) or mdm2 (murine)
(FISH) is a direct way to observe tran- tion initiation that aborts after 60–80 E3 ubiquitin ligases. Competition with
scription of specific genes.7 Although sin- nucleotides because P-TEFb recruit- p53 binding to Hdm2/mdm2, thus pre-
gle RNA molecules might be detected and ment to the promoter is deficient and vents its degradation.29,30 Furthermore,
counted, this method is quite tricky to set cannot oppose the NELFs’ to promote the available RPL26 activates p53 mRNA
up.8 An easier, though time-consuming, a productive elongation of transcription. translation. Indeed, efficient translation of
alternative is chromatin immunopre- Amanitin and actinomycin treatments p53 mRNA relies upon binding of RPL26
cipitation (DNA ChIP) using anti-poly- enhance P-TEFb activity and release the to a cap-independent and poly(A)-inde-
merase antibodies (the Rpb3 subunit or block to elongation of transcription. This pendent interaction between its 5' and 3'
the N-terminal domain of Rpb1 are rec- effect might be consequence of a feed- UTR.31,32
ommended). The distribution of RNAP back loop regulation leading to P-TEFb
molecules on a gene determined by hyperactivation.13,14 Upon transcription Changes in Extractability
Q-PCR roughly reflects its transcription. arrest, heterogeneous nuclear ribonucleo- of Nuclear Components
However, choosing adequate controls to proteins (hnRNPs) that chaperone the
evaluate immunoprecipitation efficiencies nascent transcript are released.15 Some of Transcription inhibition is accompanied
in different samples is a major difficulty. them (hnRNP A, K, Q and R types) then by notable changes in biochemical prop-
The easiest quantitative procedure is def- trap 7SK RNA that is no more available erties of nuclear proteins such as histones
initely to monitor an inducible reporter gene to bind the HEXIM1 protein and inacti- and hnRNPs. Histone H2B ubiquitina-
such as luciferase. Tetracycline-inducible vate P-TEFb.16-18 tion and histone H1b phosphorylation
promoters are particularly convenient as A general transcription inhibition decrease in cells treated with either acti-
they respond strongly and very quickly, results in p53 accumulation, which acti- nomycin D or DRB.33,34 hnRNPs that
within a few hours, but they require the use vates transcription of p53 target genes, chaperone pre-messenger RNA are easier
of genetically engineered cell lines.9,10 such as p21CIP and Hdm2,19-21 and pro- to extract from nuclei of cells treated
motes p53 translocation into mitochon- with inhibitors of class II gene transcrip-
Transcription of a Subset dria leading to apoptosis.22 Following tion.18,35,36 In contrast, efficient extraction
of Genes is Enhanced Upon treatment with flavopiridol, DRB, amani- of the positive transcription elongation
Global Transcription Inhibition tin or actinomycin, proteins such as p53 factor (P-TEFb) subunits (CDK9 and/
accumulate because of a feedback loop or Cyclin Ts) from nuclear material is
Due to feedback loops, enhanced tran- involving enhanced synthesis23 and protein harder and requires an increase in the
scription of a small set of genes occurs stability.24-27 Inhibition of rRNA synthesis ionic strength of the extraction buffer.37

104 Transcription Volume 2 Issue 3


Changes in P-TEFb and hnRNP proper- All these changes are readily observable amanitin-resistant alleles of Rpb1 can be
ties are linked by a feedback loop involv- under the microscope and might serve as used as efficient and convenient selection
ing 7SK snRNA.13,14 reliable indicators of transcription inhibi- genes to obtain stable cell transformants.
tion. Assembly of PSF into the DNCs is They are particularly convenient to obtain
Changes in Nuclear Structure likely the most reliable indication of tran- cells expressing tagged Rpb1 subunits
scription inhibition. that replace the endogenous one targeted
Transcription inhibition results in major to degradation by α-amanitin. The low
changes in nuclear structures. The nucleo- α-Amanitin–Inhibiting RNA concentration to be used (2 μg/ml with
lus is reorganized upon transcription inhi- Polymerases II and III human cells) makes it a relatively inexpen-
bition. Low concentrations of actinomycin sive option.
D, which primarily inhibit RNAP I (i.e., α-Amanitin is a cyclic peptide isolated
rRNA transcription), result in segregation from Amanita mushrooms and respon- Triptolide–Inhibiting TFIIH
of the fibrillar center, the dense fibrillar sible for their extreme toxicity. Amanitin at Transcription Initiation
center and the granular components of binds with high specificity and high
the nucleolus.38 Blockage of rRNA tran- affinity (K i = 3–4 nM) near the catalytic Triptolide is a diterpene triepoxide
scription (by oxaliplatin, doxorubicin, active site of RNAP II.43 It traps a con- extracted from the plant Tripterygium
mitoxantrone or methotrexate) or early formation of the enzyme that prevents wilfordii, used in Chinese traditional
rRNA processing steps (by camptothecin nucleotide incorporation and translo- medicine. It has multiple interesting
or CDK9 inhibitors such as flavopiridol cation of the transcript.44,45 RNAP II is pharmacological properties including
or roscovitine) cause nucleolar disintegra- the most sensitive polymerase.46,47 RNAP anti-inflammatory, immune modula-
tion, whereas blockage of late rRNA pro- III is a hundred-fold less sensitive than tion, anti-proliferative and pro-apoptotic
cessing steps (by 5-fluorouracil, MG132 RNAP II. However, reduced expression activities (reviewed in ref. 56). Triptolide
or homoharringtonine) leave nucleoli of a number of class III genes in the pres- inhibits transcription at submicromo-
intact.39 Conditions that inhibit RNAP II ence of amanitin might be attributed to lar concentrations.57 Most of its pro-
(5 μg/ml actinomycin D, DRB or ama- their regulation by RNAP II.48,49 RNAP posed pharmacological effects relate to
nitin) result in aggregation of several pro- I is insensitive to amanitin. decreased gene expression56 and might
teins from the nucleoplasm into nucleolar α-amanitin is active on living plant, thus be attributed to inhibition of tran-
caps (for an extensive description on acti- nematode, insect and mammalian cells. scription. Triptolide binds to the XPB
nomycin effects on nucleolar caps see ref. Yeast cells are insensitive to amanitin subunit of TFIIH.58 The ATP-dependent
40). Two distinct caps are distinguished, because of deficient uptake of the drug; helicase activity of XPB is required for
the dark nucleolar caps (DNC) and the nevertheless, the S. cerevisiae enzyme is the first step in transcription to open the
light nucleolar caps (LNC). Fibrillarin highly susceptible. The organic anion- double-strand DNA and create a “tran-
might be used as a marker of LNCs. The transporting polypeptide (OATP3) has scription bubble”. Triptolide inhibits the
PTB-associated splicing factor (PSF) is been identified as the amanitin uptake ATPase activity of XPB, thus it prevents
mostly nucleoplasmic. Its assembly into transporter in human hepatic cells.50 the formation of the “transcription bub-
DNCs is spectacular and might be used Amanitin uptake is slow (several hours). ble” and hence initiation of transcription.
as a practical indicator of transcription It is unusual to observe short-term Triptolide treatment is irreversible as it
inhibition. RNAP III inhibition with effects. It must be used at much higher binds covalently to XPB and induces a
high amanitin concentrations promotes concentrations with living cells (at least fast proteasome-dependent degradation
the reorganization of a perinucleolar 2 μg/ml) than in vitro. Methyl-amanitin of RNAPII.58,59 Furthermore, class I gene
compartment (PNC) distinct from the oleate, a chemically modified derivative, transcription also relies on TFIIH and is
LNC and the PNCs.41 This structure is permeates better and can be used at lower inhibited by triptolide. This inhibitor is
enriched in polypyrimidine track binding concentrations (0.010–0.1 μg/ml with active at very low concentrations (IC50 =
protein (PTB) and several class III RNAs. human cells). 109 nM for inhibition of RNA synthe-
Following RNAP III inhibition, the PNC Amanitin is an irreversible inhibitor sis in HeLa cells). Less than an hour is
is fragmented into a dotted structure because it triggers degradation of Rpb1, required to inhibit transcription in HeLa
forming a rosette with a hollow structure. the largest RNAP II subunit.51 α-amanitin cells at 10 μM concentration (Nguyen
The splicing snRNPs usually co- promotes polyubiquitination of Rpb1 VT and Bensaude O, unpublished data).
localize with p80 coilin in coiled bodies; in a nuclear extract prepared from cells It is highly specific although it might also
but, in cells treated with inhibitors, coi- arrested in S-phase.52,53 However, the pro- bind polycystin-2, a calcium channel.60
lin clusters around the nucleolus in LNCs tease pathway has not been identified in Triptolide dissolves poorly in water, but
and snRNPs aggregate in distinct nucleo- living cells yet. promising water-soluble derivatives have
plasmic speckles.42 U1 snRNP undergoes Several mutations in Rpb1 have been developed.56 Because of its selectiv-
the most spectacular re-localization; it been isolated that confer resistance ity and its very rapid action, we expect
becomes clustered around the nucleoli in to α-amanitin.54,55 Despite mildly the use of triptolide or its water-soluble
the DNCs. affecting the enzyme performance, derivatives to be generalized.

www.landesbioscience.com Transcription 105


DRB and Flavopiridol—Kinase efficiencies of various inhibitors on a Despite the selectivity issue and limi-
Inhibitors Preventing Entry panel of kinases may be found in refer- tations due to its poor solubility, DRB
into Transcription Elongation ence 72–74. Many old studies used DRB remains a very popular inhibitor. Its major
as a casein kinase 2 inhibitor or H-8 as a advantages are common usage, rapidity of
CDK9 is required for efficient class I and PKA inhibitor, and improper conclusions action and reversibility.
class II gene expression. Transcription is a were often drawn. These compounds Flavopiridol is now becoming popu-
multistep process, RNAPs bind to DNA, efficiently target CDK9 and thus act as lar because it is the most efficient known
the dsDNA is opened to form a so-called general transcription inhibitors like DRB CDK9 inhibitor.83 Its inhibition constant
“transcription bubble” and transcription or flavopiridol. New screenings have been (K i = 3 nM) is several orders of magnitude
initiates. For most class II genes, nega- designed to provide more selective inhibi- lower than the ATP binding constant
tive elongation factors (NELFs) and Spt5, tors that might become useful pharmaco- (K m = 36 μM). The crystal structure of
also known as DRB Sensitivity Inducing logical agents.75 CDK9 complexed with flavopiridol has
Factor (DSIF), provoke RNAP II paus- One should mention the exquisitely been solved.84 It is buried into the ATP
ing shortly after initiation and prevent selective “chemical genetic” method binding site after inducing a structural
a productive elongation of transcrip- developed by Shokat and coworkers.76 change in the kinase. However, it also
tion.61 Phosphorylation of these factors by The ATP binding site of the kinase to be inhibits rather efficiently (10-fold less) the
Positive Transcription Elongation Factor investigated is enlarged by replacement of cell cycle CDK1 and CDK474 as well as
(P-TEFb) is required to overcome the a voluminous aminoacid residue selected CDK8, the mediator kinase.85 Its major
obstacle. CDK9 is the kinase subunit of from crystallographic data. The modi- advantages reside in its water solubility
P-TEFb. Hence, any CDK9 inhibitor will fied kinase becomes highly susceptible to and its efficiency at submicromolar con-
prevent productive transcription of most naphtyl-ATP, which is too large to enter centrations (IC50 = 100–300 nM) on liv-
genes. Overall, Serine 2 phosphoryla- the ATP binding site and inhibit any ing cells.
tion in the heptapeptide CTD repeats of natural kinase. This method has been suc-
RNAP II decreases readily in cells exposed cessfully used with transcription CDKs.77 Actinomycin D—DNA Intercalators
to CDK9 inhibitors.64,65 Serine 2 phos- However, it requires a gene replacement Blocking the Progression
phorylation of the CTD is required for strategy that is relatively easy to set up in of RNA Polymerases
pre-mRNA processing (splicing, termina- simple organisms such as yeast but quite
tion and polyadenylation). Thus, splic- difficult in mammals. Many DNA intercalators inhibit tran-
ing becomes impaired.66 Transcription of DRB (5,6-Dichloro-1-beta-Ribo- scription. But not all do so. For example,
short intron-less histone and <u> snRNA furanosyl Benzimidazole) has been widely Hoechst 33,342 intercalates into nuclear
encoding genes is not affected by CDK9 used as a transcription inhibitor. This DNA of living cells but does not affect
inhibition.62,63 Instead, 3' end processing compound was initially reported to inhibit its transcription significantly.6 Ethidium
of histone or U snRNA encoding genes nuclear heterogeneous RNA (hnRNA) bromide affects mitochondrial but not
is impaired.62,63 CDK9 inhibitors also synthesis.78,79 It quickly appeared to nuclear transcription.86
affect an early step in rRNA (class I gene) cause “premature” chain termination. Actinomycin D or Dactinomycin is likely
processing, thereby impairing ribosome CDK9 was identified as its major target.80 the most popular inhibitor of transcrip-
biogenesis.39 However, it also inhibits CDK7, the kinase tion. It comprises two cyclic peptides
Many compounds with potential subunit of TFIIH, with 3-fold lower effi- linked together by a phenoxazine deriva-
pharmacological applications inhibit ciency.72 The above-mentioned “DSIF” or tive. It is isolated from Streptomyces bac-
CDK9. Several CDK9 inhibitors are DRB Sensitivity Inducing Factor refers to teria. Actinomycin D is also one of the
currently under clinical trial in chemo- DRB. The crystal structure of CDK9 com- older chemotherapy drugs, commonly
therapy,67 in particular against chronic plexed with DRB has been resolved.81 The used to treat gestational trophoblas-
lymphocytic leukemia.68 Some of these chlorine atoms form halogen bonds with a tic cancer, testis cancer, Wilm’s tumor,
CDK9 inhibitors are occasionally used hinge region characteristic of CDK9 near rhabdomyosacoma and Ewing’s sarcoma.
as inhibitors of transcription (e.g., rosco- the ATP binding site. Transcription by all three eukaryotic
vitine—also known as selicilib37—the DRB has to be used at concentrations polymerases is affected. Yet, class I gene
isoquinoline sulfonamide (H-8),69 and close to its maximal solubility (100 μM). transcription is by far the most sensitive
SNS-302).70 However, DRB and flavopiri- Mother solutions in DMSO and fast (0.05 μg/ml) followed by class II gene
dol are the most popular for molecular/ homogenization in warm media are rec- transcription (0.5 μg/ml) and class III
cellular biology use (see below). ommended. RNAPII transcription arrests (around 5 μg/ml). The length of the tran-
All CDK9 inhibitors compete with within minutes following addition of DRB scription unit and its DNA sequence com-
ATP for the kinase active site. Given its to culture medium. Efficient transcription position are determinant.87 Actinomycin
conservation, the selectivity of protein resumes within minutes when the medium preferentially intercalates into GC rich
kinase inhibitors is a major issue that is replaced by fresh medium to remove sequences and stabilizes topoisomerase-I
has been largely discussed by Knight and the drug. This property has recently been DNA covalent complexes that pre-
Shokat.71 Comparison between inhibition used to measure transcription rates.82 vent RNA polymerase progression.88

106 Transcription Volume 2 Issue 3


Actinomycin D generates double-strand for critical reading of the manuscript. 18. Barrandon C, Bonnet F, Nguyen VT, Labas V,
Bensaude O. The transcription-dependent disso-
breaks in DNA and induces the synthe- The author was supported by a grant ciation of P-TEFb.HEXIM1.7SK RNA relies upon
sis of γ-histone H2AX molecules that from Ligue Nationale contre le Cancer formation of hnRNP.7SK RNA complexes. Mol Cell
Biol 2007; 27:6996-7006.
accumulate into foci.89 Transcription n°RS10/75-15.
19. Gomes NP, Bjerke G, Llorente B, Szostek SA,
is slightly recovered when actinomycin Emerson BM, Espinosa JM. Gene-specific require-
D is removed from the culture medium References ment for P-TEFb activity and RNA polymerase II
1. Sugaya K, Sasanuma S, Cook PR, Mita K. A muta- phosphorylation within the p53 transcriptional pro-
but several hours are required.90,91 gram. Genes Dev 2006; 20:601-12.
tion in the largest (catalytic) subunit of RNA poly-
Actinomycin promotes the accumulation merase II and its relation to the arrest of the cell cycle 20. Choong ML, Yang H, Lee MA, Lane DP. Specific
of CTD phosphorylated RNAPII11 likely in G(1) phase. Gene 2001; 274:77-81. activation of the p53 pathway by low dose actinomy-
2. Jackman J, Alamo IJ, Fornace AJJ. Genotoxic stress cin D: a new route to p53 based cyclotherapy. Cell
due to an enhanced P-TEFb activity.92 Cycle 2009; 8:2810-8.
confers preferential and coordinate messenger RNA
We will terminate by mentioning two stability on the five gadd genes. Cancer Res 1994; 21. Beckerman R, Prives C. Transcriptional regulation
by p53. Cold Spring Harb Perspect Biol 2011; 2:935.
compounds with interesting properties. 54:5656-62.
3. Lam LT, Pickeral OK, Peng AC, Rosenwald A, Hurt 22. Galluzzi L, Morselli E, Kepp O, Vitale I, Pinti M,
Cisplatin is a widely used anti-cancer Kroemer G. Mitochondrial liaisons of p53. Antioxid
EM, Giltnane JM, et al. Genomic-scale measurement
drug. It cross-links DNA thereby blocking of mRNA turnover and the mechanisms of action of the Redox Signal 2011; In press.
23. Mosner J, Mummenbrauer T, Bauer C, Sczakiel G,
the progression of RNA polymerase mol- anti-cancer drug flavopiridol. Genome Biol 2001; 2:41.
Grosse F, Deppert W. Negative feedback regula-
ecules.93 Yet, the cisplatin/DNA adducts, 4. Lü X, Burgan WE, Cerra MA, Chuang EY, Tsai
tion of wild-type p53 biosynthesis. EMBO J 1995;
MH, Tofilon PJ, et al. Transcriptional signature of
and hence the block, can be removed by flavopiridol-induced tumor cell death. Mol Cancer
14:4442-9.
24. An WG, Chuman Y, Fojo T, Blagosklonny MV.
the nucleotide excision repair machinery. Ther 2004; 3:861-72.
Inhibitors of transcription, proteasome inhibitors and
Trabectedin or Yondalis (ET-743) is devel- 5. Chen Q, Cao M, Xiang WL, Sun Q, Zhang J, Hou
DNA-damaging drugs differentially affect feedback
RT, et al. Study on genes with altered expression in of p53 degradation. Exp Cell Res 1998; 244:54-60.
oped as a new anti-cancer drug that has alpha-amanitin poisoned mice and evaluation on
25. Demidenko ZN, Blagosklonny MV. Flavopiridol
been isolated from the marine tunicate antagonistic effects of traditional Chinese medicines
induces p53 via initial inhibition of Mdm2 and
against toxicity of alpha-amanitin. Acta Biol Hung
Ecteinascidia turbinata. It intercalates into 2009; 60:281-91.
p21 and, independently of p53, sensitizes apoptosis-
reluctant cells to tumor necrosis factor. Cancer Res
DNA and promotes a fast proteasome- 6. White CM, Heidenreich O, Nordheim A, Beerman 2004; 64:3653-60.
dependent degradation of RNAP II.94 TA. Evaluation of the effectiveness of DNA-binding 26. Ljungman M. The transcription stress response. Cell
drugs to inhibit transcription using the c-fos serum Cycle 2007; 6:2252-7.
response element as a target. Biochemistry 2000;
Concluding Remarks 39:12262-73.
27. Radhakrishnan SK, Bhat UG, Halasi M, Gartel AL.
P-TEFb inhibitors interfere with activation of p53 by
7. Levsky JM, Singer RH. Fluorescence in situ hybrid- DNA-damaging agents. Oncogene 2008; 27:1306-9.
ization: past, present and future. J Cell Sci 2003;
To conclude, the classical inhibitors of 116:2833-8.
28. Hölzel M, Orban M, Hochstatter J, Rohrmoser M,
Harasim T, Malamoussi A, et al. Defects in 18 S or 28
transcription have advantages and draw- 8. Femino AM, Fay FS, Fogarty K, Singer RH. S rRNA processing activate the p53 pathway. J Biol
backs. Amanitin is highly selective for Visualization of single RNA transcripts in situ. Chem 2010; 285:6364-70.
Science 1998; 280:585-90. 29. Vousden KH, Prives C. Blinded by the light: the
RNAP II and RNAP III but slow, actino-
9. Gossen M, Bonin AL, Freundlieb S, Bujard H. growing complexity of p53. Cell 2009; 137:413-31.
mycin D is fast but its selectivity is poor, Inducible gene expression systems for higher eukary- 30. Zhang Y, Wang J, Yuan Y, Zhang W, Guan W, Wu Z,
CDK9 inhibitors are fast and reversible otic cells. Curr Opin Biotechnol 1994; 5:516-20. et al. Negative regulation of HDM2 to attenuate p53
10. Loew R, Heinz N, Hampf M, Bujard H, Gossen M. degradation by ribosomal protein L26. Nucleic Acids
but many genes escape transcription inhi- Res 2010; 38:6544-54.
Improved Tet-responsive promoters with minimized
bition. New inhibitors such as triptolide background expression. BMC Biotechnol 2010; 10:81. 31. Takagi M, Absalon MJ, McLure KG, Kastan MB.
are fast, selective and completely arrest 11. Cassé C, Giannoni F, Nguyen VT, Dubois MF, Regulation of p53 translation and induction after
Bensaude O. The transcriptional inhibitors, acti- DNA damage by ribosomal protein L26 and nucleo-
transcription as they trigger rapid degra- lin. Cell 2005; 123:49-63.
nomycin D and α-amanitin, activate the HIV-1
dation of RNAP II. promoter and favor phosphorylation of the RNA 32. Chen J, Kastan MB. 5'-3'-UTR interactions regulate
The physiological effects of transcrip- polymerase II C-terminal domain. J Biol Chem 1999; p53 mRNA translation and provide a target for
274:16097-106. modulating p53 induction after DNA damage. Genes
tion inhibitors have been described as the Dev 2010; 24:2146.
12. Imamichi T, Conrads TP, Zhou M, Liu X,
“transcriptional stress response”.26 They Adelsberger JW, Veenstra TD, et al. A transcription 33. Davie JR, Murphy LC. Level of ubiquitinated histone
occur after treatments such as irradia- inhibitor, actinomycin D, enhances HIV-1 replica- H2B in chromatin is coupled to ongoing transcrip-
tion through an interleukin-6-dependent pathway. tion. Biochemistry 1990; 29:4752-7.
tion that lead to DNA lesions. The stress J Acquir Defic Syndr 2005; 40:388-97. 34. Chadee DN, David Allis C, Wright JA, Davie JR.
response may have interesting pharmaco- 13. Zhou Q, Yik JH. The Yin and Yang of P-TEFb Histone H1b phosphorylation is dependent upon
logical consequences. Friend virus-trans- regulation: implications for human immunodefi- ongoing transcription and replication in normal
ciency virus gene expression and global control of cell and ras-transformed mouse fibroblasts. J Biol Chem
formed murine erythroleukemia (MEL) growth and differentiation. Microbiol Mol Biol Rev 1997; 272:8113-6.
cells differentiate when exposed to mild 2006; 70:646-59. 35. Piñol-Roma S, Dreyfuss G. Shuttling of pre-mRNA
concentrations of actinomycin D95 or 14. Barrandon C, Spiluttini B, Bensaude O. Non-coding binding proteins between nucleus and cytoplasm.
RNAs regulating the transcriptional machinery. Biol Nature 1992; 355:730-2.
DRB.96 Flavopiridol attenuates leukocyte- Cell 2008; 100:83-95. 36. Christian KJ, Lang MA, Raffalli-Mathieu F.
endothelial cell interaction.85 More gener- 15. Dreyfuss G, Kim VN, Kataoka N. Messenger-RNA- Interaction of heterogeneous nuclear ribonucleo-
ally, an arrest in transcription will lead to binding proteins and the messages they carry. Nat protein C1/C2 with a novel cis-regulatory element
Rev Mol Cell Biol 2002; 3:195-205. within p53 mRNA as a response to cytostatic drug
cell death, often through apoptosis. treatment. Mol Pharmacol 2008; 73:1558-67.
16. Hogg JR, Collins K. RNA-based affinity purification
reveals 7SK RNPs with distinct composition and 37. Biglione S, Byers SA, Price JP, Nguyen VT, Bensaude
Acknowledgments regulation. RNA 2007; 13:868-80. O, Price DH, et al. Inhibition of HIV-1 replication by
17. Van Herreweghe E, Egloff S, Goiffon I, Jady BE, P-TEFb inhibitors DRB, seliciclib and flavopiridol cor-
We are indebted to Drs. Xavier Darzacq relates with release of free P-TEFb from the large, inac-
Froment C, Monsarrat B, et al. Dynamic remodelling
Anna Smirnova and Nina Verstraete of human 7SK snRNP controls the nuclear level of tive form of the complex. Retrovirology 2007; 4:47.
active P-TEFb. EMBO J 2007; 26:3570-80.

www.landesbioscience.com Transcription 107


38. Boulon S, Westman BJ, Hutten S, Boisvert FM, 59. Vispé S, DeVries L, Créancier L, Besse J, Bréand S, 78. Sehgal PB, Derman E, Molloy GR, Tamm I, Darnell
Lamond AI. The nucleolus under stress. Mol Cell Hobson DJ, et al. Triptolide is an inhibitor of RNA JE. 5,6-Dichloro-1-ß-D-ribofuranosylbenzimidazole
2010; 40:216-27. polymerase I and II-dependent transcription lead- inhibits initiation of nuclear heterogeneous RNA
39. Burger K, Mühl B, Harasim T, Rohrmoser M, ing predominantly to downregulation of short-lived chains in HeLa cells. Science 1976; 194:431-3.
Malamoussi A, Orban M, et al. Chemotherapeutic mRNA. Mol Cancer Ther 2009; 8:2780-90. 79. Fraser NW, Sehgal PB, Darnell JE. DRB-induced
drugs inhibit ribosome biogenesis at various levels. 60. Leuenroth SJ, Okuhara D, Shotwell JD, Markowitz premature termination of late adenovirus transcrip-
J Biol Chem 2010; 285:12416-25. GS, Yu Z, Somlo S, et al. Triptolide is a tradi- tion. Nature 1978; 272:590-3.
40. Shav-Tal Y, Blechman J, Darzacq X, Montagna C, Dye tional Chinese medicine-derived inhibitor of poly- 80. Zhu Y, Pe’ery T, Peng J, Ramanathan Y, Marshall
BT, Patton JG, et al. Dynamic sorting of nuclear com- cystic kidney disease. Proc Natl Acad Sci USA 2007;
N, Marshall T, et al. Transcription elongation factor
ponents into distinct nucleolar caps during transcrip- 104:4389-94.
P-TEFb is required for HIV-1 Tat transactivation in
tional inhibition. Mol Biol Cell 2005; 16:2395-413. 61. Nechaev S, Adelman K. Pol II waiting in the start- vitro. Genes Dev 1997; 11:2622-32.
41. Pollock C, Huang S. The perinucleolar compart- ing gates: Regulating the transition from transcrip-
81. Baumli S, Endicott JA, Johnson L. Halogen bonds
ment. J Cell Biochem 2009; 107:189-93. tion initiation into productive elongation. Biochim
Biophys Acta 2011; 1809:34-45. form the basis for selective P-TEFb inhibition by
42. Carmo-Fonseca M, Pepperkok R, Carvalho MT,
DRB. Chem Biol 2010; 17:931-6.
Lamond AI. Transcription-dependent colocalization 62. Medlin J, Scurry A, Taylor A, Zhang F, Peterlin BM,
Murphy S. P-TEFb is not an essential elongation fac- 82. Singh J, Padgett RA. Rates of in situ transcription
of the U1, U2, U4/U6 and U5 snRNPs in coiled
bodies. J Cell Biol 1992; 117:1-14. tor for the intronless human U2 snRNA and histone and splicing in large human genes. Nat Struct Mol
H2b genes. EMBO J 2005; 24:4154-65. Biol 2009; 16:1128-33.
43. Bushnell DA, Cramer P, Kornberg RD. RNA poly-
merase II cocrystal at 2.8 A resolution. Proc Natl 63. Pirngruber J, Shchebet A, Schreiber L, Shema E, 83. Chao SC, Fujinaga K, Marion JE, Taube R, Sausville
Acad Sci USA 2002; 99:1218-22. Minsky N, Chapman RD, et al. CDK9 directs EA, Senderowicz AM, et al. Flavopiridol inhibits
44. Kaplan CD, Larsson KM, Kornberg RD. The RNA H2B monoubiquitination and controls replication- P-TEFb and blocks HIV-1 replication. J Biol Chem
polymerase II trigger loop functions in substrate dependent histone mRNA 3'-end processing. EMBO 2000; 275:28345-8.
selection and is directly targeted by alpha-amanitin. Rep 2009; 10:894-900. 84. Baumli S, Lolli G, Lowe ED, Troiani SLR, Bullock
Mol Cell 2008; 30:547-56. 64. Dubois MF, Bellier S, Seo SJ, Bensaude O. AN, et al. The structure of P-TEFb (CDK9/cyclin
45. Brueckner F, Cramer P. Structural basis of transcrip- Phosphorylation of the RNA polymerase II largest T1), its complex with flavopiridol and regulation by
tion inhibition by alpha-amanitin and implications subunit during heat-shock and inhibition of transcrip- phosphorylation. EMBO J 2008; 27:1907-18.
for RNA polymerase II translocation. Nat Struct Mol tion in HeLa cells. J Cell Physiol 1994; 158:417-26. 85. Schmerwitz UK, Sass G, Khandoga AG, Joore J,
Biol 2008; 15:811-8. 65. Lavoie SB, Albert AL, Handa H, Vincent M, Mayer BA, Berberich N, et al. Flavopiridol pro-
46. Kedinger C, Nuret P, Chambon P. Structural evi- Bensaude O. The peptidyl-prolyl isomerase Pin1 tects against inflammation by attenuating leuko-
dence for two alpha-amanitin sensitive RNA poly- interacts with hSpt5 phosphorylated by Cdk9. J Mol cyte-endothelial interaction via inhibition of cyclin-
merases in calf thymus. FEBS Lett 1971; 15:169-74. Biol 2001; 312:675-85. dependent kinase 9. Arterioscler Thromb Vasc Biol
47. Weinmann R, Raskas HJ, Roeder RG. Role of DNA- 66. Ip JY, Schmidt D, Pan Q, Ramani AK, Fraser AG, 2011; 31:280-8.
dependent RNA polymerases II and III in transcrip- Odom DT, et al. Global impact of RNA polymerase 86. Zylber E, Vesco C, Penman S. Selective inhibition
tion of the adenovirus genome late in productive II elongation inhibition on alternative splicing regu- of the synthesis of mitochondria-associated RNA by
infection. Proc Natl Acad Sci USA 1974; 71:3426-39. lation. Genome Res 2011; 21:390-401. ethidium bromide. J Mol Biol 1969; 44:195-204.
48. Listerman I, Bledau AS, Grishina I, Neugebauer 67. Wang LM, Ren D. Flavopiridol, the first cyclin- 87. Perry RP, Kelley DE. Inhibition of RNA synthesis by
KM. Extragenic accumulation of RNA polymerase II dependent kinase inhibitor: recent advances in com- actinomycin D: characteristc dose-response of differ-
enhances transcription by RNA polymerase III. PLoS bination chemotherapy. Mini Rev Med Chem 2010; ent RNA species. J Cell Physiol 1970; 76:127-40.
Genet 2007; 3:212. 10:1058-70. 88. Trask DK, Muller MT. Stabilization of type I topoi-
49. Raha D, Wang Z, Moqtaderi Z, Wu L, Zhong G, 68. Abou-Nassar K, Brown J. Novel agents for the treat- somerase-DNA covalent complexes by actinomycin
Gerstein M, et al. Close association of RNA poly- ment of chronic lymphocytic leukemia. Clin Adv D. Proc Natl Acad Sci USA 1988; 85:1417-21.
merase II and many transcription factors with Pol III Hematol Oncol 2010; 8:886-95.
genes. Proc Natl Acad Sci USA 2010; 107:3639-44. 89. Mischo HE, Hemmerich P, Grosse F, Zhang S.
69. Marshall NF, Peng J, Xie Z, Price DH. Control of Actinomycin D induces histone gamma-H2AX foci
50. Letschert K, Faulstich H, Keller D, Keppler D. RNA polymerase II elongation potential by a novel
Molecular characterization and inhibition of ama- and complex formation of gamma-H2AX with Ku70
carboxyl-terminal domain kinase. J Biol Chem 1996; and nuclear DNA helicase II. J Biol Chem 2005;
nitin uptake into human hepatocytes. Toxicol Sci 271:27176-83.
2006; 91:140-9. 280:9586-94.
70. Chen R, Wierda WG, Chubb S, Hawtin RE, Fox JA,
51. Nguyen VT, Giannoni F, Dubois MF, Seo SJ, 90. Schluederberg A, Hendel RC, Chavanich S.
Keating MJ, et al. Mechanism of action of SNS-032,
Vigneron M, Kédinger C, et al. In vivo degradation Actinomycin D: renewed RNA synthesis after remov-
a novel cyclin-dependent kinase inhibitor, in chronic
of RNA polymerase II largest subunit triggered by al from mammalian cells. Science 1971; 172:577-9.
lymphocytic leukemia. Blood 2009; 113:4637-45.
α-amanitin. Nucleic Acids Res 1996; 24:2924-9. 91. Hadjiolova KV, Hadjiolov AA, Bachellerie JP.
71. Knight ZA, Shokat KM. Features of Selective Kinase
52. Lee KB, Sharp PA. Transcription-dependent polyu- Actinomycin D stimulates the transcription of
Inhibitors. Chem Biol 2005; 12:621-37.
biquitination of RNA polymerase II requires lysine rRNA minigenes transfected into mouse cells. Eur J
72. Mancebo HS, Lee G, Flygare J, Tomassini J, Luu P,
63 of ubiquitin. Biochemistry 2004; 43:15223-9. Biochem 1995; 228:605-15.
Zhu Y, et al. P-TEFb kinase is required for HIV Tat
53. Jung Y, Lippard SJ. RNA polymerase II blockage by 92. Nguyen VT, Kiss T, Michels AA, Bensaude O. 7SK
transcriptional activation in vivo and in vitro. Genes
Cisplatin-damaged DNA. Stability and polyubiqui- snRNA binds to and inhibits the activity of Cdk9/
Dev 1997; 11:2633-44.
tylation of stalled polymerase. J Biol Chem 2006; cyclin T complexes. Nature 2001; 414:322-5.
281:1361-70. 73. Chao SH, Price DH. Flavopiridol inactivates P-TEFb
93. Ang WH, Myint M, Lippard SJ. Transcription
and blocks most RNA polymerase II transcription in
54. Bartolomei MS, Corden JL. Clustered alpha-amani- inhibition by platinum-DNA cross-links in live mam-
tin resistance mutations in mouse. Mol Gen Genet Vivo. J Biol Chem 2001; 276:31793-9.
malian cells. J Am Chem Soc 2010; 132:7429-35.
1995; 246:778-82. 74. Conroy A, Stockett DE, Walker D, Arkin MR,
Hoch U, Fox JA, et al. SNS-032 is a potent and 94. Aune GJ, Takagi K, Sordet O, Guirouilh-Barbat
55. Chen Y, Chafin D, Price DH, Greenleaf A. Drosophila J, Antony S, Bohr VA, et al. Von Hippel-Lindau-
RNA polymerase II mutants that affect transcription selective CDK 2, 7 and 9 inhibitor that drives target
modulation in patient samples. Cancer Chemother coupled and transcription-coupled nucleotide exci-
elongation. J Biol Chem 1996; 271:5993-9. sion repair-dependent degradation of RNA poly-
56. Liu Q. Triptolide and its expanding multiple phar- Pharmacol 2009; 64:723-32.
merase II in response to trabectedin. Clin Cancer Res
macological functions. Int Immunopharmacol 2011; 75. Wang S, Griffiths G, Midgley CA, Barnett AL,
Cooper M, Grabarek J, et al. Discovery and char- 2008; 14:6449-55.
11:377-83.
acterization of 2-anilino-4-(thiazol-5-yl)pyrimidine 95. Terada TM, Epner E, Nudel U, Salmon J, Fibach E,
57. Leuenroth SJ, Crews CM. Triptolide-induced tran-
transcriptional CDK inhibitors as anticancer agents. Rifkind RA, et al. Induction of murine erythroleu-
scriptional arrest is associated with changes in nuclear
Chem Biol 2010; 17:1111-21. kemia differentiation by actinomycin D. Proc Natl
substructure. Cancer Res 2008; 68:5257-66.
76. Bishop AC, Buzko O, Shokat KM. Magic bullets for Acad Sci USA 1978; 75:2795-9.
58. Titov DV, Gilman B, He QL, Bhat S, Low WK,
protein kinases. Trends Cell Biol 2001; 11:167-72. 96. Hensold J, Barth D, Stratton CA. RNA polymerase
Dang Y, et al. XPB, a subunit of TFIIH, is a target of
the natural product triptolide. Nat Chem Biol 2011; 77. Tietjen JR, Zhang DW, Rodríguez-Molina JB, White II inhibitor, 5,6-dichloro-1-ß-D-Ribofuranosylbenz-
7:182-8. BE, Akhtar MS, Heidemann M, et al. Chemical- imidazole (DRB) causes erythroleukemic differentia-
genomic dissection of the CTD code. Nat Struct Mol tion and transcriptional activation of erythroid genes.
Biol 2010; 17:1154-61. J Cell Physiol 1996; 168:105-13.

108 Transcription Volume 2 Issue 3

You might also like