1. Trang chủ
  2. » Luận Văn - Báo Cáo

Báo cáo khoa học: Investigating RNA polymerase II carboxyl-terminal domain (CTD) phosphorylation ˆ Benoıt Palancade and Olivier Bensaude pptx

12 250 0

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

THÔNG TIN TÀI LIỆU

Nội dung

Eur J Biochem 270, 3859–3870 (2003) Ó FEBS 2003 doi:10.1046/j.1432-1033.2003.03794.x REVIEW ARTICLE Investigating RNA polymerase II carboxyl-terminal domain (CTD) phosphorylation ˆ Benoıt Palancade and Olivier Bensaude Ge´ne´tique Mole´culaire, UMR 8541 CNRS, Ecole Normale Supe´rieure, Paris, France Phosphorylation of RNA polymerase II’s largest subunit C-terminal domain (CTD) is a key event during mRNA metabolism Numerous enzymes, including cell cycledependent kinases and TFIIF-dependent phosphatases target the CTD However, the repetitive nature of the CTD prevents determination of phosphorylated sites by conventional biochemistry methods Fortunately, a panel of monoclonal antibodies is available that distinguishes between phosphorylated isoforms of RNA polymerase II’s (RNAP II) largest subunit Here, we review how successful these tools have been in monitoring RNAP II phosphorylation changes in vivo by immunofluorescence, chromatin immunoprecipitation and immunoblotting experiments The CTD phosphorylation pattern is precisely modified as RNAP II progresses along the genes and is involved in sequential recruitment of RNA processing factors One of the most popular anti-phosphoCTD Igs, H5, has been proposed in several studies as a landmark of RNAP II molecules engaged in transcription Finally, we discuss how global RNAP II phosphorylation changes are affected by the physiological context such as cell stress and embryonic development The C-terminal domain (CTD) of RNA polymerase II’s (RNAP II) largest subunit is essential for transcription [1–3], for its function as enhancer [4,5], for organization of transcription foci within the nucleus [6] and for pre-mRNA processing [7,8] The CTD consists of multiple repeats of a seven amino acid motif [9,10] (Fig 1) This motif has been conserved during evolution in eukaryotes, but the number of repeats varies depending on the species: 26 in yeast, 45 in flies and 52 in mammals Five out of seven amino acids in the consensus motif are phosphate acceptors and indeed phosphorylation is a major post-translational modification of the CTD in vivo [11] Serine O-glycosylation has also been reported as a minor modification with unknown functional significance [12] In contrast, CTD phosphorylation plays a major role in the transcriptional process [13–16] A large variety of kinases have been reported to phosphorylate the CTD of RNAP II in vitro [17,18] Of particular significance, CDK7, CDK8 and CDK9 are subunits of the TFIIH general transcription factor, of the mediator complex and of the positive transcription elongation factor (P-TEFb), respectively Interactions with the unphosphorylated CTD are involved in assembly of RNAP II with the mediator complex to form a holoenzyme or with general transcription factors to form a preinititiation complex of transcription Phosphorylation of the CTD is required to disrupt these interactions at elongation of transcription and to assist the recruitment of pre-mRNA modification enzymes [16,19] Initial studies generally considered CTD phosphorylation as an all or nothing process Here, we review recent evidence that multiple forms of CTD-phosphorylated RNAP II are present in cells These forms have been characterized using monoclonal antibodies that distinguish between phosphorylated amino acid residues within the CTD The phosphorylation site specificity of kinases and phosphatases that target the CTD is discussed next Variations in RNAP II phosphorylation are examined along the transcriptional process and as a function of physiological context including cell stress and early development ´ ´ ´ Correspondence to O Bensaude, Genetique Moleculaire, ´ UMR 8541 CNRS, Ecole Normale Superieure, 46 rue d’Ulm, 75230 Paris Cedex 05, France Fax: + 33 44323941, Tel.: + 33 44323410, E-mail: bensaude@biologie.ens.fr Abbreviations: CDK, cyclin-dependent kinase; CTD, C-terminal domain (of Rpb1); DSIF, DRB sensitivity inducing factor; HIV-LTR, long-terminal repeats of the human immunodeficiency virus; FCP1, TFIIF-dependent CTD phosphatase; NELF, negative elongation factor; MAPK, mitogen-activated protein kinase; Rpb1, largest subunit of RNAP II; RNAP II, RNA polymerase II; SCP, small CTD phosphatase; TFIIx, x type general class II gene transcription factor (Received 12 June 2003, revised 13 August 2003, accepted 15 August 2003) Keywords: RNA polymerase II; CTD-phosphorylation; CTD-kinase; CTD-phosphatase; transcription; mRNA processing Multiple RNAP II phosphoisoforms Two distinct fractions of RNAP II were purified from mammalian cells in early studies and designated RNAP IIA and RNAP IIO [20] They differ in the phosphorylation status of their largest subunit (Rpb1), respectively, designated as IIa and IIo forms [13] The CTD is hypophosphorylated in the IIa form and multiphosphorylated in the IIo form It is noteworthy that some preparations of RNAP II contain a third fraction, RNAP IIB, which results from an Ó FEBS 2003 3860 B Palancade and O Bensaude (Eur J Biochem 270) Fig Sequence of the human CTD Selfalignment of human RNAP II largest subunit sequence from amino acids 1593–1970 (last residue before the stop codon) (GeneBank accession number NM_000937) Note that serine at position in the heptad motif is not conserved in the C-terminal section of the CTD Serines and 5, the major phosphorylated residues are in red Non-conserved amino acid residues are in blue in vitro proteolysis of the CTD thus generating the IIb form of Rpb1 The CTD is phosphorylated mainly on serine residues, although phosphothreonine and phosphotyrosine have been detected to a minor extent [21,22] CTD phosphorylation provokes a remarkable shift in electrophoretic migration equivalent to an apparent molecular weight increase from 210 to 240 kDa in vertebrates [23] The number of phosphates in the mammalian IIo form has been evaluated as close to 50 residues, corresponding to one phosphate per repeat on average [24] Phosphorylated forms that migrate between the IIa and IIo forms have also been described An IIi (intermediary) form develops during Herpes simplex virus infection [25,26] An IIm (MAP kinase-dependent) form is generated during serum-stimulation of quiescent cells or during osmotic and oxidative stress [27,28] An IIe (embryonic) form is found in nontranscribing early embryos from vertebrates [28,29] and insects [30] Hence SDS/PAGE analysis unravels the diversity in phosphorylated forms of RNAP II Phosphorylation-dependent epitopes within the CTD Due to the elevated number of phosphate acceptors and to the repetition of the consensus motif, it is a formidable task to determine precisely which serine residues are phosphorylated in a particular repeat Recently, a set of monoclonal antibodies became a powerful tool to investigate phosphorylated residues within the CTD (Table 1) These antibodies have been characterized by Western blot using yeast CTD fusion proteins in which serines at positions or in all 26 heptads had been replaced either by alanine or by glutamate [31] Some of these antibodies react preferentially with the unphosphorylated CTD whereas recognition by others requires phosphorylated serines at specific positions It is worth mentioning that monoclonal antibodies H5 and H14 are often referred to as specific for phosphoserines and 5, respectively Table Phosphorylation dependence of anti-CTD monoclonal antibodies Characterization of CTD phosphoepitopes: (1) using a set of yeast CTD fusion proteins mutated or not [31]; (2) using a set of synthetic peptides phosphorylated or not ([34]; A Albert, S Lavoie and M Vincent, personal communication); (3) using purified RNAP II phosphorylated or not by MAP kinase [27] Name Epitope Method Reference 8WG16 V15 V6 MARA3 B3 H14 H5 CC3 MPM2 Unphosphorylated Ser2 Unphosphorylated Unphosphorylated Ser2 Ser2-P or Ser5-P Ser2-P or Ser5-P Ser5-P Ser2-P Ser2-P Ser2-P and Ser5-P (1) (3) (3) (1) (1) (1) (1) (1) (2) [123] [27] [27] [31] [124] [110] [110] [125] [37] (2) (2) (2) (2) Limitations in epitope data interpretation The specificity of monoclonal antibodies should be handled cautiously For instance, depending on investigators, the 8WG16 antibody binds to the unphosphorylated CTD exclusively, or to both phosphorylated and unphosphorylated CTD indistinctly [31–33] In addition, although ELISA assays confirmed that H5 and H14 antibodies distinguished peptides phosphorylated on serines or at low antigen concentration, this specificity was lost at high antigen concentrations [34] Moreover, a weaker binding with peptides phosphorylated simultaneously on serines and suggests that phosphorylation of adjacent sites might impair recognition Furthermore, little is known about the consequences of amino acid substitution occurring at nonphosphorylated positions on degenerated repeats as found in the C-terminal half of the mammalian CTD Finally, some of these antibodies such as H5, CC3 and MPM2 have been shown to cross-react with proteins other than the RNAP II largest subunit [35–37] Ó FEBS 2003 RNA polymerase II CTD phosphorylation (Eur J Biochem 270) 3861 Despite these restrictions in establishing a correspondence between serine or serine phosphorylation and H5 or H14 recognition, these antibodies provide useful tools with which to characterize the phosphorylation of the CTD As will be seen below, H5 and H14 reactivities unravel variations in the phosphorylation sites and may reflect differential action of CTD-kinases and CTD-phosphatases Moreover, H5 reactivity might be used as a marker for transcribing RNAP II molecules Amino acid residues targeted by CTD-kinases in vitro CTD-kinases belong to three subclasses: cyclin-dependent kinases, mitogen-activated protein kinases and others (see Table and [15,17,18]) Two methods have been used to determine phosphorylation site preference for a given CTDkinase The first method uses synthetic peptides consisting in one or more CTD heptads with the phosphorylable serine, threonine or tyrosine replaced by alanine [38,39] Serine phosphorylation is preferred for CDK7 [39–41], CDK8 [42,43], CDK9 [38] and MAPK [39] Kinases such as CDK1 and CDK2 phosphorylate both serine and serine [41,44] Efficiencies of the different kinases are influenced by the position of the targeted repeats within the CTD [38,45] and by the neighboring residue’s identity [38,39,43] Slightly different conclusions were reached when determining phosphoepitopes that were generated on purified RNAP II For example, CDK7 (TFIIH) generates H14 epitope (serine 5) as predicted [46,47] but also the H5 one [48] and the CC3 one [45], which are specific for phosphoserine [31] Furthermore, CDK8 generates both H14 and H5 reactivity upon long phosphorylation times [46] In discrepancy with peptide-driven conclusions, CDK9 generates H5 reactivity [47,49,50] One might consider that genuine RNAP II CTD is a complex array of degenerated repeats differing from the peptides used in biochemical studies Transcription factors influence phosphorylation site preference CTD kinases phosphorylate RNAP II within large supramolecular complexes that may influence the phosphorylation reaction course Recruitment of CDK7 in a DNA-bound transcription preinitiation complex increases its efficiency towards RNAP II CTD [51] and TFIIE enforces its preference for serine [48] Transcriptional regulators contribute to recruit CTD kinases Activation of HIV-LTR transcription by the viral transactivator Tat provides a paradigm of such a phenomenon [52,53] In the presence of Tat, CDK9 phosphorylates serine (H14) in addition to serine (H5) [50] Several cellular activators also recruit CDK9 to enhance genespecific transcription [54–57] In contrast, the glucocorticoid receptor bound to its hormone represses nuclear factorkappa B activation and this effect correlates with a decreased phosphorylation of serine (H5) on target genes [58] Amino acid residues targeted by CTD-phosphatases In contrast to the large number of characterized CTDkinases, few CTD-phosphatases have been identified (Table and references therein); most studies concern the TFIIF-dependent CTD phosphatase (FCP1) [59] Recently, three small FCP1 related proteins, SCP1/NLI-IF, SCP2/ OS4 and SCP3/HYA22, have been identified as also being CTD phosphatases [60] In addition, the contribution of type protein phosphatase (PP1) has been discussed in two reports [61,62] and the SSU72 transcription/processing factor has been hypothesized as a phosphotyrosine CTD phosphatase [63,64] In contrast to CTD-kinases that accept a wide range of substrates, the substrate specificity of FCP1 phosphatase is strictly restricted to the CTD within RNA polymerase II core enzyme [59] The site preference of CTD phosphatases was determined using various RNAP IIO isozymes: FCP1 dephosphorylates RNAP IIO purified from mammalian cells [65] or RNAP IIO generated in vitro by phosphorylation of RNAP IIA with purified kinases such as CDK7, CDK9, CDK1 or MAPK [66] Consistently, FCP1 phosphatase removes both H5 and H14 phosphoepitopes from mammalian RNAP IIO [66] Therefore, in higher eukaryotes, phosphates on serine (H5) and serine (H14) appear equally removed by FCP1 phosphatase In contrast, it has been proposed that fission yeast FCP1 specifically target serine [34] However, the Schizosaccharomyces pombe FCP1 assays were performed using peptides and highly acidic nonphysiological conditions Saccharomyces cerevisiae experiments involving FCP1 inactivation followed by chromatin immunoprecipitation (see below) suggested the occurrence of a distinct serine 5-specific phosphatase [34] Interestingly, in contrast to FCP1, the small FCP1-related CTD phosphatase, SCP1, might preferentially dephosphorylate phosphoserine as CDK7-phosphorylated RNAP IIO is its preferred substrate [60] Changes in RNAP II phosphorylation along gene transcription Initial studies established that RNAP II is hypophosphorylated when recruited to a promoter and then hyperphosphorylated during messenger RNA synthesis [67–70] Serine phosphorylation by the mediator complex subunit Srb10 (CDK8 ortholog) precludes this recruitment [42] An elegant study using step by step in vitro transcription in mammalian nuclear extracts has indicated that serine (H14) is phosphorylated first in the initiation complex (likely by CDK7) and that serine (H5) is phosphorylated by CDK9 upon entry into elongation [49] When engaged in transcription elongation complexes, RNAP II becomes a poor substrate for FCP1, suggesting that conformational changes or protecting factors affect the efficiency of the phosphatase [71,72] In support of the latter hypothesis, proteins that bind the phosphorylated CTD inhibit FCP1 activity (B Palancade, N F Marshall, O Bensaude, M Dahmus & M.-F Dubois, unpublished observation) Cellular DNA can be cross-linked to proteins in its vicinity and then immunoprecipitated with antibodies directed against these proteins The presence of specific DNA sequences in the immunoprecipitated material is probed by polymerase chain reaction (PCR) amplification Such chromatin immunoprecipitation (CHIP) assays have helped to map the evolution of CTD phosphorylation along a transcribed gene Antibodies directed to the hypo- and the Cyclin Cyclin H Ccl1 Cyclin C Srb11 CDK7 (MO15) Kin28 CDK8 Srb10 MAT1 Tfb3? Other partners Ctk2 Bur2 c-abl Others DNA-PK Mitogen-activated protein kinases ERK 1–2 Mpk1 Ctk1 Bur1 (Sgv1) Ku70 Ku86 Ctk3 Ser ND ND Tyr DNA-PK Ser Ser CTDK-I Ser 5* Mediator Ser 5 5 ND ND H14 H5 H14 H5 (H14 ?) H14 (H5 ?) H14 (H5 ?) H5 H14 ND Function in transcription Repression of initiation [21] [151] [39] [39,149] [27] ? ? ? [38,49,50, Entry into processive elongation 139,140,141] Tat-dependent HIV-1 transcription [145–147] Elongation? [148] Elongation? [42,43,46, 134–136] Repression of transcription during mitosis ? [44] Tat-dependent HIV-1 transcription ? [39–41,46–48, Initiation, recruitment of 130] capping enzymes [41,127] Peptides Phosphoepitopes Reference Site preference Ser Ser Ser Ser TFIIH or Ser CAK MPF Complex CDK9 (PITALRE) Cyclin T1, T2, K 7SK snRNA P-TEFb MAQ1 (HEXIM1) Cyclin E CDK2 Cyclin-dependent kinases CDK1 (cdc2) Cyclin A, B Kinase Associated factor DNA-PK, PI3-kinase, several nuclear proteins [153] p53, chromatin components [152] c-fos, c-jun, transcription factors [150] ND ND Spt5 [36,142] Rb [143], MyoD [144] CDK1, CDK2 [131] p53 [132], RARa [156], TBP, TFIIE, TFIIF [133] Cyclin H [107] Gcn4, Msn2 [137], Gal4 [138] Structural proteins, cell cycle regulators [128] Cell cycle regulators [129] Other substrates Table CTD-kinases and their features Usual mammalian protein names are indicated in bold characters, other nomenclatures are in parenthesis and S cerevisiae orthologues are in italic Ctk1 and Bur1 have both been proposed as budding yeast CDK9 counterparts Site preferences have been deduced from in vitro phosphorylation of synthetic peptides, or from phosphoepitopes generated on the CTD ´ ` ´ ` MPF, maturation-promoting factor; CAK, CDK-activating kinase; RARa, retinoic acid receptor; MAT1, Ômenage-a-troisÕ; MAQ1, Ômenage-a-quatreÕ [126] *, Although CDK9 preferentially phosphorylates serine on peptides, it generates phosphoserine (H5 epitope) in vitro and in vivo ND, not determined; ?, unknown 3862 B Palancade and O Bensaude (Eur J Biochem 270) Ó FEBS 2003 Ó FEBS 2003 RNA polymerase II CTD phosphorylation (Eur J Biochem 270) 3863 Table The CTD-phosphatases and their features The site preference has been determined by CTD-phosphatase assays in vitro Previous nomenclature of SCPs is given in parenthesis NLI-IF, nuclear LIM interactor–interacting factor; OS4, osteosarcoma gene 4; HYA22, human orthologue of S pombe YA22 locus; ND, not determined; ?, unknown Phosphatase Regulatory protein TFIIF-dependent phosphatases FCP1 RAP74 (TFIIF) Site preference References Function in transcription Other substrates Ser (H5) [59,66] Recycling of RNAP II after termination Regulation of elongation RNAP II mobilization CTD dephosphorylation after initiation? ? Developmentally regulated transcription? ? ? TFIIB Ser (H14) SCP1 (NLI-IF) RAP74 (TFIIF) Ser [60] SCP2 (OS4) RAP74 (TFIIF) ND [60,154] SCP3 (HYA22) ? ND [60] Phosphoserine/phosphothreonine protein phospshatases (PPP) PP1 At least seven different Ser (H5) [62] regulatory subunits Ser (H14) [155] hyperphosphorylated CTD of Drosophila RNAP II demonstrated that RNAP II molecules pause at the 5¢ ends of heat shock genes in an unphosphorylated state [73] In mammalian cells, phosphoserine (H14) is concentrated near the promoter while serine phosphorylation (H5) is observed throughout the gene [74] In yeast, serine is phosphorylated at initiation in a CDK7/Kin28-dependent manner [75,76] The Ctk1-dependent serine (H5) phosphorylation develops next at the beginning of elongation and decrease in immunoprecipitation suggested that serine was dephosphorylated at this stage [34,76] Alternatively, the latter finding might be interpreted as a loss of phosphoserine recognition when both serines and are phosphorylated Taken together, the chromatin immunoprecipitation experiments demonstrate that distinct residues are phosphorylated on the CTD during the progression of RNAP II along the genes in vivo (Fig 2) Interestingly, an alanine scanning analysis of the CTD in yeast RNAP II had suggested that phosphorylation on both serine and serine position in the repeats was required for viability but a search for suppressor mutations indicated that these positions differed functionally [77,78] Changes in CTD phosphorylation facilitate pre-mRNA processing The CTD mediates the coupling between transcription and pre-mRNA processing The phosphorylated CTD recruits the pre-mRNA processing machinery [79] Serine phosphorylation of the CTD by CDK7/Kin28 is sufficient to recruit and stimulate capping enzymes upon initiation [75,76,80] The capping enzyme may in turn, assist the recruitment of CDK9 as in S pombe, the CDK9 ortholog binds Pct1, a subunit of the capping apparatus [81] Consistently, inactivation of capping enzyme in Caenorhabditis elegans results in a loss of serine (H5) phosphorylation [82] Capping might be a prerequisite to elongate transcription and contribute to a transcriptional checkpoint: Tat-dependent HIV-1 transcription? ? ? ? Transcription factors, splicing factors, cell cycle regulators [155] only capped mRNAs would be transcribed efficiently The phosphorylated CTD next recruits SR proteins involved in splicing [83–85] followed by the cleavage-polyadenylation factors [86] The serine 2-kinase Ctk1 is required for 3¢-end processing in S cerevisiae [87] Consistently, the Pcf11p cleavage-polyadenylation factor preferentially associates to the CTD phosphorylated on serine rather than serine [88] Hence, transcription-dependent changes in CTD phosphorylation sites might be required to coordinate sequential recruitment of the processing machinery In addition to phosphorylation, degeneracy of the CTD adds a further level of complexity (Fig 1) Indeed, the N-terminal half of the CTD only stimulates capping whereas the C-terminal half supports capping as well as splicing and 3¢-end processing [89,90] Different repeats in the CTD may serve distinct functions Changes in CTD phosphorylation determine the recruitment of chromatin modification enzymes Recently, CTD phosphorylation has been shown to be involved in histone methylation control in budding yeast Indeed, phosphorylation of RNAP II mediates the recruitment of histone H3 methyltransferases from the SET family [79,91] Serine 5-phosphorylated RNAP II contributes to the association of the Set1/COMPASS methyltransferase complex onto the 5¢-portion of actively transcribed genes shortly after initiation, thereby enhancing histone H3 K4 trimethylation and providing a molecular mark of recent transcriptional activity [92,93] Subsequently, the Set2 histone H3 K39 methyltransferase is recruited to the elongation complex through direct binding to the CTD phosphorylated by Ctk1 on serines at position [94,95] Thus, distinct phosphorylated RNAP II isoforms contribute to chromatin imprinting Specific CTD phosphorylation patterns therefore determine and coordinate the interactions with protein partners such as transcription factors, histone-modifying complexes or mRNA-processing enzymes 3864 B Palancade and O Bensaude (Eur J Biochem 270) Ó FEBS 2003 Fig CTD phosphorylation and the transcription cycle (A) Recycling The ÔfreeÕ RNAP II core enzyme is not phosphorylated on the CTD It may assemble with coactivators such as the mediator complex (Med) thus forming a holoenzyme Premature CTD phosphorylation by CDK8 prevents the assembly of RNAP II on the promotor (B) Preinitiation The unphosphorylated RNAP II core or holoenzyme assemble onto the promoter sequences with general transcription factors such as TFIID, TFIIA, TFIIB, TFIIE, TFIIF and TFIIH thus forming a preinititiation complex of transcription The CTD is phosphorylated on serines at position (5P) by the CDK7 subunit of TFIIH (C) Initiation Transcription begins The phosphorylated CTD recruits the capping enzymes and the nascent transcript is capped at its 5¢ end (D) Elongation Phosphorylation of the CTD on serines at position (2P) by the CDK9 subunit of the positive transcription elongation factor (P-TEFb) is required to remove to block opposed by the DSIF/NELF factors and elongate transcription (E) RNA processing and transcription termination The phosphorylated CTD recruits the splicing machinery to remove introns and, finally recruits the cleavage and polyadenylation factors that cleave the transcript and add a polyadenylic tail at its 3¢ end This step signals transcription to terminate and RNAP II falls off its DNA template The resulting mRNA is then exported to the cytoplasm (F) To be recycled for another transcription round, RNAP II is dephosphorylated by the FCP1 CTD phosphatase Global changes in RNAP II phosphorylation The IIa and IIo RNAP II phosphoisoforms coexist in a dynamic equilibrium in yeast [33], insect [96], amphibian [28] and mammalian cells [97] Importantly, this dynamic exchange does not require transcription as it is also observed in a nontranscribing cell system such as the amphibian egg [98] Addition of purified phosphorylated RNAP IIO to a crude egg extract demonstrated a half turnover of CTD phosphates of 2–4 In this system, the antagonistic action of FCP1 and of ERK type MAP kinase accounts for the distribution between phosphorylated and unphosphorylated RNAP II forms [98,99] In transcribing cells, the CTD phosphorylation dynamic is unraveled upon CTD kinase/phosphatase inactivation In S cerevisiae, inactivation of either Kin28 (the CDK7 orthologue) or Ccl1, its cyclin partner, results in a rapid decrease in the amount of CTD-phosphorylated RNAP II [33,100] Conversely, Fcp1 inactivation leads to an increase in the amount of phosphorylated RNAP II [32] Selective gene inactivation in mammalian cells is less straightforward but, fortunately, numerous chemical inhibitors are available instead Kinase inhibitors have generally limited specificity but this problem might be circumvented by using a panel of compounds belonging to distinct chemical families and by comparing efficiencies in vitro and in vivo This procedure has been used to suggest that CDK9 is the major CTD kinase generating the bulk IIo form in interphasic mammalian cells [36,97,101] RNAP II hyperphosphorylation increases following treatments such as exposure to a-amanitin [102] or actinomycin D [101], or upon UV irradiation [103] As this increase is suppressed by CDK9 inhibitors [101], it is suggested to result from an enhanced activity of CDK9 provoked by the transcriptional arrest [104,105] Similarly, Ctk1 is required for serine phosphorylation during the diauxic shift or following DNA damage in S cerevisiae [106] Ó FEBS 2003 RNA polymerase II CTD phosphorylation (Eur J Biochem 270) 3865 However, the effect of kinase inhibition on CTD phosphorylation may be indirect For instance, CDK8 phosphorylates cyclin H thereby inactivating CDK7 [107] In turn, TFIIH inhibits the autophosphorylation of CDK9, which enhances its kinase activity [47] Therefore, a body of arguments is required to strengthen the interpretation For instance, the identification of ERK1/2 MAP kinase as the major CTD kinase generating the IIm forms relies on several arguments [27,98,99]: (a) the appearance of the IIm form correlates with MAP kinase activation; (b) it is insensitive to CDK9 inhibitors but suppressed in the presence of inhibitors of MAP kinase activation; (c) the immunogenicity of the IIm form matches that of RNAP II phosphorylated in vitro by MAP kinase Serine phosphorylation (H5) as a landmark of transcribing RNAP II Immunofluorescence analysis of polytene chromosomes revealed that hyperphosphorylated RNAP II is associated with actively transcribed sequences such as Drosophila ecdysone-induced puffs [108] Interestingly, this study also suggested that transcribing RNAP II is less phosphorylated under heat-shock conditions Although the H14 monoclonal antibody stains diffusely nuclei of interphasic mammalian cells, H5 reveals punctuated nuclear structures that match transcription foci [109] During mitosis, at metaphase, both H14 and H5 epitopes are excluded from chromatin [110] However, H14 staining is diffuse whereas H5 decorates mitotic granules (MIGs) which are speckle-like structures containing pre-mRNA processing material [111] At telophase, H14 immunostaining develops in daughter nuclei simultaneously with recruitment of the general transcription machinery Nuclear H5 staining develops at early G1 phase, in coincidence with the onset of transcription In many metazoans, early embryonic development following fertilization is characterized by global repression of zygotic transcription Gene expression is initiated abruptly at the so-called mid-blastula transition (MBT) in amphibians, or zygotic genome activation (ZGA) in mammals This period is marked by important CTD phosphorylation changes that have been documented in nematodes, flies [30,112], fishes [113], amphibians [28] and mammals [29] Before ZGA or MBT, the RNAP II CTD does not react with H5, which is consistent with the absence of transcription at this period H5 reactivity has therefore been proposed as a landmark of zygotic gene activation in early embryos [28] In nematode embryos, a mixed diffuse/punctuated H14 staining develops in nuclei of somatic cells throughout embryogenesis whereas transcription-inactive germline cells show only two discrete foci [112] In contrast, H5 staining appears in conjunction with ZGA at the four-cell stage and remains restricted to the transcriptionally active somatic cells, except for mutants that disrupt the asymmetric divisions of germline cells [114] In contrast to H14, the H5 phosphoepitope is absent in embryos when transcription is globally impaired following RNA interference inactivation of essential components of the transcription machinery such as CDK9 [115] or TBP-like factor (TLF) [116,117] Both H14 and H5 staining are decreased upon CDK7 [118], RGR-1 mediator subunit [119], TFIIB or TAFIIs [120] inactivation Similar data are available for vertebrate embryos H5-reactive RNAP II is absent from zebrafish embryos inactivated for TBP [121], and both H5 and H14 staining are attenuated in mouse embryos following inactivation of MAT1, the CDK7 partner [122] These findings are consistent with an H14 staining attributed in part to initiating polymerases (productive or abortive) and an H5 staining of elongating polymerases Conclusion Determining the sites phosphorylated in vivo within RNAP II CTD is a formidable task due to the repetitive nature of this domain However, phosphospecific antibodies such as H5 and H14 have been successfully used in a wide range of experiments (Table 1) Using these tools, qualitative changes in the CTD phosphorylation pattern have been found to occur during the course of transcription or in response to physiological stimuli These changes might contribute to synchronize the successive steps of nuclear mRNA metabolism (Fig 2) Many data point out that recognition of the CTD by the H5 antibody might be specific for RNAP II molecules engaged in mRNA synthesis It is expected that an expanded use of other anti-CTD antibodies will contribute to improve our knowledge of CTD phosphorylation Deciphering this ÔCTD codeÕ may help our global understanding of gene expression mechanisms [58] Acknowledgements We are much indebted to Keith Blackwell and Marie-Francoise Dubois ¸ for critical reading of the manuscript We thank Michael E Dahmus, ă Michel Vincent and members of their lab for sharing unpublished information This work was supported by grants from the Association pour la Recherche sur le Cancer (ARC 6250), the Ligue Nationale ´ Contre le Cancer (Comite de Paris) and the Agence Nationale de Recherche sur le SIDA References Corden, J.L (1993) RNA polymerase II transcription cycles Curr Opin Genet Dev 3, 213–218 Meininghaus, M., Chapman, R.D & Eick, D (2000) Conditional expression of RNA polylmerase II in mammalian cells Deletion of the carboxy-terminal domain of the large subunit affects early steps in transcription J Biol Chem 275, 24375–24382 Meisels, E., Gileadis, O & Corden, J.L (1995) Partial truncation of the yeast RNA polymerase II carboxy-terminal domain preferentially reduces expression of glycolytic genes J Biol Chem 270, 31255–31261 Scafe, C., Chao, D., Lopes, J., Hirsch, J.P., Henry, S & Young, R.A (1990) RNA polymerase II C-terminal repeat influences response to transcriptional enhancer signals Nature 347, 491– 494 Gerber, H.P., Hagmann, M., Seipel, K., Georgiev, O., West, M.A.L., Litingung, Y., Schaffner, W & Corden, J.L (1995) RNA polymerase II C-terminal domain required for enhancerdriven transcription Nature 374, 660–662 Misteli, T (2000) Cell biology of transcription and pre-mRNA splicing: nuclear architecture meets nuclear function J Cell Sci 113, 1841–1849 Proudfoot, N.J., Furger, A & Dye, M.J (2002) Integrating mRNA processing with transcription Cell 108, 501–512 Ó FEBS 2003 3866 B Palancade and O Bensaude (Eur J Biochem 270) Howe, K.J (2002) RNA polymerase II conducts a symphony of pre-mRNA processing activities Biochim Biophys Acta 1577, 308–324 Corden, J.L (1990) Tails of RNA polymerase II Trends Biochem Sci 15, 383–387 10 Corden, J.L & Ingles, C.J (1992) Carboxy-terminal domain of the largest subunit of RNA polymerase II In Transcriptional Regulation (McKnight, S.L & Yamamoto, K.R., eds), pp 81– 128, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY ´ 11 Bensaude, O., Bonnet, F., Casse, C., Dubois, M.-F., Nguyen, V.-T & Palancade, B (1999) Regulated phosphorylation of the RNA polymerase II C-terminal domain (CTD) Biochem Cell Biol 77, 1–7 12 Kelly, W.G., Dahmus, M.E & Hart, G.W (1993) RNA polymerase II is a glycoprotein: modification of the COOH-terminal domain by O-GlcNAc J.Biol Chem 268, 10416–10424 13 Dahmus, M.E (1996) Reversible phosphorylation of the C-terminal domain of RNA polymerase II J Biol Chem 271, 19009– 19012 14 Riedl, T & Egly, J.M (2000) Phosphorylation in transcription: the CTD and more Gene Expr 9, 3–13 15 Oelgeschlager, T (2002) Regulation of RNA polymerase II activity by CTD phosphorylation and cell cycle control J Cell Physiol 190, 160–169 16 Kobor, M.S & Greenblatt, J (2002) Regulation of transcription elongation by phosphorylation Biochim Biophys Acta 1577, 261–275 17 Majello, B & Napolitano, G (2001) Control of RNA polymerase II activity by dedicated CTD kinases and phosphatases Front Biosci 6, D1358–D1368 18 Prelich, G (2002) RNA polymerase II carboxy-terminal domain kinases: emerging clues to their function Eukaryot Cell 1, 153– 162 19 Bentley, D (2002) The mRNA assembly line: transcription and processing machines in the same factory Curr Opin Cell Biol 14, 336–342 20 Schwartz, L.B & Roeder, R.G (1975) Purification and subunit structure of deoxyribonucleic acid-dependent ribonucleic acid polymerase II from the mouse plasmacytoma, MOPC 315 J Biol Chem 250, 3221–3228 21 Baskaran, R., Dahmus, M.E & Wang, J.Y.J (1993) Tyrosine phosphorylation of mammalian RNA polymerase II carboxylterminal domain Proc Natl Acad Sci USA 90, 11167–11171 22 Zhang, J & Corden, J.L (1991) Identification of phosphorylation sites in the repetitive CTD of the mouse RNA polymerase II largest subunit J Biol Chem 266, 2290–2296 23 Dahmus, M.E & Kedinger, C (1983) Transcription of adenovirus-2 major late promoter inhibited by monoclonal antibody directed against RNA polymerases II0 and IIA J Biol Chem 258, 2303–2307 24 Payne, J.M & Dahmus, M.E (1993) Partial purification and characterization of two distinct protein kinases that differentially phosphorylate the carboxy-terminal domain of RNA polymerase subunit IIa J Biol Chem 268, 80–87 25 Jenkins, H.L & Spencer, C.A (2001) RNA polymerase II holoenzyme modifications accompany transcription reprogramming in herpes simplex virus type 1-infected cells J Virol 75, 9872–9884 26 Rice, S.A., Long, M.C., Lam, V & Spencer, C.A (1994) RNA polymerase II is aberrantly phosphorylated and localized to viral replication compartments following Herpes Simplex virus infection J Virol 68, 988–1001 27 Bonnet, F., Vigneron, M., Bensaude, O & Dubois, M.-F (1999) Transcription-independent phosphorylation of the RNA 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 polymerase II carboxy-terminal domain (CTD) recquiring ERK kinase activity Nucleic Acids Res 27, 4399–4404 Palancade, B., Bellier, S., Almouzni, G & Bensaude, O (2001) Incomplete RNA polymerase II phosphorylation in Xenopus laevis early embryos J Cell Sci 114, 2483–2489 Bellier, S., Chastant, S., Adenot, P., Vincent, M., Renard, J.-P & Bensaude, O (1997) Nuclear translocation and carboxyl-terminal domain phosphorylation of RNA polymerase II delineate the two phases of zygotic gene activation in mammalian embryos EMBO J 16, 6250–6262 Leclerc, V., Raisin, S & Leopold, P (2000) Dominant-negative mutants reveal a role for the Cdk7 kinase at the mid-blastula transition in Drosophila embryos EMBO J 19, 1567–1575 Patturajan, M., Schulte, R.J., Sefton, B.M., Berezney, R., Vincent, M., Bensaude, O., Warren, S.L & Corden, J.L (1998) Growth-related changes in phosphorylation of yeast RNA polymerase II J Biol Chem 273, 4689–4694 Kobor, M.S., Archambault, J., Lester, W., Holstege, F.C., Gileadi, O., Jansma, D.B., Jennings, E.G., Kouyoumdjian, F., Davidson, A.R., Young, R.A & Greenblatt, J (1999) An unusual eukaryotic protein phosphatase required for transcription by RNA polymerase II and CTD dephosphorylation in S cerevisiae Mol Cell 4, 55–62 Valay, J.-G., Simon, M., Dubois, M.-F., Bensaude, O., Facca, C & Faye, G (1995) The KIN28 gene is required both for RNA polymerase II mediated transcription and phosphorylation of the Rpb1p CTD J Mol Biol 249, 535–544 Cho, E.J., Kobor, M.S., Kim, M., Greenblatt, J & Buratowski, S (2001) Opposing effects of Ctk1 kinase and Fcp1 phosphatase at Ser2 of the RNA polymerase II C-terminal domain Genes Dev 15, 3319–3329 Doyle, O., Corden, J.L., Murphy, C & Gall, J.G (2002) The distribution of RNA polymerase II largest subunit (RPB1) in the Xenopus germinal vesicle J Struct Biol 140, 154–166 Lavoie, S.B., Albert, A.L., Handa, H., Vincent, M & Bensaude, O (2001) The peptidyl-prolyl isomerase Pin1 interacts with hSpt5 phosphorylated by Cdk9 J Mol Biol 312, 675–685 Albert, A., Lavoie, S & Vincent, M (1999) A hyperphosphorylated form of RNA polymerase II is the major interphase antigen of the phosphoprotein antibody MPM-2 and interacts with the peptidyl-prolyl isomerase Pin1 J Cell Sci 112, 2493–2500 Ramanathan, Y., Rajpara, S.M., Reza, S.M., Lees, E., Shuman, S., Mathews, M.B & Pe’ery, T (2001) Three RNA polymerase II carboxyl-terminal domain kinases display distinct substrate preferences J Biol Chem 276, 10913–10920 Trigon, S., Serizawa, H., Conaway, J.W., Conaway, R.C., Jackson, S.P & Morange, M (1998) Characterization of the residues phosphorylated in vitro by different C-terminal domain kinases J Biol Chem 273, 6769–6775 Roy, R., Adamczewski, J.P., Seroz, T., Vermeulen, W., Tassan, J.-P., Schaeffer, L., Nigg, E.A., Hoeijmakers, J.H.J & Egly, J.-M (1994) The MO15 cell cycle kinase is associated with the TFIIH transcription-DNA repair factor Cell 79, 1093–1101 Gebara, M.M., Sayre, M.H & Corden, J.L (1997) Phosphorylation of the carboxy-terminal repeat domain in RNA polymerase II by cyclin-dependent kinases is sufficient to inhibit transcription J Cell Biochem 64, 390–402 Hengartner, C.J., Myer, V.E., Liao, S.M., Wilson, C.J., Koh, S.S & Young, R.A (1998) Temporal regulation of RNA polymerase II by Srb10 and Kin28 cyclin-dependent kinases Mol Cell 2, 43–53 Rickert, P., Corden, J.L & Lees, E (1999) Cyclin C/CDK8 and cyclin H/CDK7/p36 are biochemically distinct CTD kinases Oncogene 18, 1093–1102 Ó FEBS 2003 RNA polymerase II CTD phosphorylation (Eur J Biochem 270) 3867 44 Deng, L., Ammosova, T., Pumfery, A., Kashanchi, F & Nekhai, S (2002) HIV)1 Tat interaction with RNA polymerase II CTD and a dynamic association with CDK2 induce CTD phosphorylation and transcription from HIV-1 promoter J Biol Chem 277, 33922–33929 45 Dubois, M.-F., Vincent, M., Adamczewski, J., Egly, J.M & Bensaude, O (1997) Heat shock inactivation of the TFIIHassociated kinase and change in the phosphorylation sites on RNA polymerase II largest subunit Nucleic Acids Res 25, 694–700 46 Sun, X., Zhang, Y., Cho, H., Rickert, P., Lees, E., Lane, W & Reinberg, D (1998) NAT, a human complex containing Srb polypeptides that functions as a negative regulator of activated transcription Mol Cell 2, 213–222 47 Zhou, M., Nekhai, S., Bharucha, D.C., Kumar, A., Ge, H., Price, D.H., Egly, J.M & Brady, J.N (2001) TFIIH inhibits CDK9 phosphorylation during human immunodeficiency virus type transcription J Biol Chem 276, 44633–44640 48 Yamamoto, S., Watanabe, Y., van der Spek, P.J., Watanabe, T., Fujimoto, H., Hanaoka, F & Ohkuma, Y (2001) Studies of nematode TFIIE function reveal a link between Ser-5 phosphorylation of RNA polymerase II and the transition from transcription initiation to elongation Mol Cell Biol 21, 1–15 49 Kim, Y.K., Bourgeois, C.F., Isel, C., Churcher, M.J & Karn, J (2002) Phosphorylation of the RNA polymerase II carboxylterminal domain by CDK9 is directly responsible for human immunodeficiency virus type Tat-activated transcriptional elongation Mol Cell Biol 22, 4622–4637 50 Zhou, M., Halanski, M.A., Radonovich, M.F., Kashanki, F., Peng, J., Price, D.H & Brady, J.N (2000) Tat modifies the activity of CDK9 to phosphorylate serine of the RNA polymerase II carboxy-terminal domain during immunodeficiency virus type transcription Mol Cell Biol 20, 5077–5086 51 Rossignol, M., Kolb-Cheynel, I & Egly, J.-M (1997) Substrate specificity of the cdk-activating kinase (CAK) is altered upon association with TFIIH EMBO J 16, 1628–1637 52 Mancebo, H.S., Lee, G., Flygare, J., Tomassini, J., Luu, P., Zhu, Y., Peng, J., Blau, C., Hazuda, D., Price, D & Flores, O (1997) P-TEFb kinase is required for HIV Tat transcriptional activation in vivo and in vitro Genes Dev 11, 2633–2644 53 Cujec, T., Okamoto, H., Fujinaga, K., Meyer, J., Chamberlin, H., Morgan, D.O & Peterlin, B.M (1997) The HIV transactivator TAT binds to the CDK-activating kinase and activates the phosphorylation of the carboxy-terminal domain of RNA polymerase II Genes Dev 11, 2645–2657 54 Kanazawa, S., Okamoto, T & Peterlin, B.M (2000) Tat competes with CIITA for the binding to P-TEFb and blocks the expression of MHC class II genes in HIV infection Immunity 12, 61–70 55 Lee, D.K., Duan, H.O & Chang, C (2001) Androgen receptor interacts with the positive elongation factor P-TEFb and enhances the efficiency of transcriptional elongation J Biol Chem 276, 9978–9984 56 Eberhardy, S.R & Farnham, P.J (2001) c-Myc mediates activation of the cad promoter via a post-RNA polymerase II recruitment mechanism J Biol Chem 276, 48562–48571 57 Barboric, M., Nissen, R.M., Kanazawa, S., Jabrane-Ferrat, N & Peterlin, B.M (2001) NF-kappaB binds P-TEFb to stimulate transcriptional elongation by RNA polymerase II Mol Cell 8, 327–337 58 Nissen, R.M & Yamamoto, K.R (2000) The glucocorticoid receptor inhibits NFkB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain Genes Dev 14, 2314–2329 59 Lin, P.S., Marshall, N.F & Dahmus, M.E (2002) CTD phosphatase: role in RNA polymerase II cycling and the regulation of 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 transcript elongation Prog Nucleic Acid Res Mol Biol 72, 333–365 Yeo, M., Lin, P.S., Dahmus, M.E & Gill, G.N (2003) A novel RNA polymerase II C-terminal domain phosphatase that preferentially dephosphorylates serine J Biol Chem 278, 26078–26085 Bharucha, D.C., Zhou, M., Nekhai, S., Brady, J.N., Shukla, R.R & Kumar, A (2002) A protein phosphatase from human T cells augments tat transactivation of the human immunodeficiency virus type long-terminal repeat Virology 296, 6–16 Washington, K., Ammosova, T., Beullens, M., Jerebtsova, M., Kumar, A., Bollen, M & Nekhai, S (2002) Protein phosphatase1 dephosphorylates the C-terminal domain of RNA polymeraseII J Biol Chem 15, 15 Ganem, C., Devaux, F., Torchet, C., Jacq, C., QuevillonCheruel, S., Labesse, G., Facca, C & Faye, G (2003) Ssu72 is a phosphatase essential for transcription termination of snoRNAs and specific mRNAs in yeast EMBO J 22, 1–11 Meinhart, A., Silberzahn, T & Cramer, P (2003) The mRNA transcription/processing factor Ssu72 is a potential tyrosine phosphatase J Biol Chem 18, 15917–15921 Cho, H., Kim, T.K., Mancebo, H., Lane, W.S., Flores, O & Reinberg, D (1999) A protein phosphatase functions to recycle RNA polymerase II Genes Dev 13, 1540–1552 Lin, P.S., Dubois, M.-F & Dahmus, M.E (2002) TFIIF-associating carboxyl-terminal domain phosphatase dephosphorylates phosphoserines and of RNA polymerase II J Biol Chem 277, 45949–45956 Cadena, D.L & Dahmus, M.E (1987) Messenger RNA synthesis in mammalian cells is catalyzed by the phosphorylated form of RNA polymerase II J Biol Chem 262, 12468–12474 Payne, J.M., Laybourn, P.J & Dahmus, M.E (1989) The transition of RNA polymerase II from initiation to elongation is associated with phosphorylation of the carboxyl-terminal domain of subunit IIa J Biol Chem 264, 19621–19629 Lu, H., Flores, O., Weinmann, R & Reinberg, D (1991) The nonphosphorylated form of RNA polymerase II preferentially associates with the preinitiation complex Proc Natl Acad Sci USA 88, 10004–10008 Chesnut, J.D., Stephens, J.H & Dahmus, M.E (1992) The interaction of RNA polymerase II with the adenovirus-2 major late promoter precluded by phosphorylation of the C-terminal domain of subunit IIa J Biol Chem 267, 10500–10506 Lehman, A.L & Dahmus, M.E (2000) The sensitivity of RNA polymerase II in elongation complexes to C-terminal domain phosphatase J Biol Chem 275, 14923–14932 Marshall, N.F & Dahmus, M.E (2000) C-terminal domain phosphatase sensitivity of RNA polymerase II in early elongation complexes on the HIV-1 and adenovirus major late templates J Biol Chem 275, 32430–32437 O’Brien, T., Hardin, S., Greenleaf, A & Lis, J.T (1994) Phosphorylation of RNA polymerase II C-terminal domain and transcriptional elongation Nature 370, 75–77 Cheng, C & Sharp, P.A (2003) RNA polymerase II accumulation in the promoter-proximal region of the dihydrofolate reductase and gamma-actin genes Mol Cell Biol 23, 1961–1967 Komarnitsky, P., Cho, E.J & Buratowski, S (2000) Different phosphorylated forms of RNA polymerase II and associated mRNA processing factors during transcription Genes Dev 14, 2452–2460 Schroeder, S.C., Schwer, B., Shuman, S & Bentley, D (2000) Dynamic association of capping enzymes with transcribing RNA polymerase II Genes Dev 14, 2435–2440 West, M.L & Corden, J.L (1995) Construction and analysis of yeast RNA polymerase II CTD deletion and substitution mutations Genetics 140, 1223–1233 Ó FEBS 2003 3868 B Palancade and O Bensaude (Eur J Biochem 270) 78 Yuryev, A & Corden, J.L (1996) Suppression analysis reveals a functional difference between the serines in positions two and five in the consensus sequence of the C-terminal domain of yeast RNA polymerase II Genetics 143, 661–671 79 Carty, S.M & Greenleaf, A.L (2002) Hyperphosphorylated C-terminal repeat domain-associating proteins in the nuclear proteome link transcription to DNA/chromatin modification and RNA processing Mol Cell Proteomics 1, 598–610 80 Ho, C.K & Shuman, S (1999) Distinct roles for CTD Ser-2 and Ser-5 phosphorylation in the recruitment and allosteric activation of mammalian capping enzyme Mol Cell 3, 405–411 81 Pei, Y., Schwer, B & Shuman, S (2003) Interactions between fission yeast Cdk9, its cyclin partner Pch1, and mRNA capping enzyme Pct1 suggest an elongation checkpoint for mRNA quality control J Biol Chem 278, 7180–7188 82 Takagi, T., Walker, A.K., Sawa, C., Diehn, F., Takase, Y., Blackwell, T.K & Buratowski, S (2003) The Caenorhabditis elegans mRNA 5¢-capping enzyme: in vitro and in vivo characterization J Biol Chem 278, 14174–14184 83 Greenleaf, A.L (1993) A positive addition to a negative tail’s tale Proc Natl Acad Sci USA 90, 10896–10897 84 Patturajan, M., Wei, X., Berezney, R & Corden, J.L (1998) A nuclear matrix protein interacts with the phosphorylated C-terminal domain of RNA polymerase II Mol Cell Biol 18, 2406–2415 85 Kim, E.L., Bregman, D.B & Warren, S.L (1997) Splicing factors associate with hyperphosphorylated RNA polymerase II in the absence of pre-mRNA J Cell Biol 136, 19–28 86 McCracken, S., Fong, N., Yankulov, K., Ballantyne, S., Pan, G., Greenblatt, J., Patterson, S.D., Wickens, M & Bentley, D.L (1997) The C-terminal domain of RNA polymerase II couples mRNA processing to transcription Nature 385, 357–361 87 Skaar, D.A & Greenleaf, A.L (2002) The RNA polymerase II CTD kinase CTDK-I affects pre-mRNA-3¢ cleavage/polyadenylation through the processing component Pti1p Mol Cell 10, 1429–1439 88 Licatalosi, D.D., Geiger, G., Minet, M., Schroeder, S., Cilli, K., McNeil, J.B & Bentley, D.L (2002) Functional interactions of yeast pre-mRNA-3¢ end processing factors with RNA polymerase II Mol Cell 9, 1101–1111 89 Ryan, K., Murthy, K.G., Kaneko, S & Manley, J.L (2002) Requirements of the RNA polymerase II C-terminal domain for reconstituting pre-mRNA-3¢ cleavage Mol Cell Biol 22, 1684– 1692 90 Fong, N & Bentley, D.L (2001) Capping, splicing, and 3¢ processing are independently stimulated by RNA polymerase II: different functions for different segments of the CTD Genes Dev 15, 1783–1795 91 Hampsey, M & Reinberg, D (2003) Tails of intrigue: phosphorylation of RNA polymerase II mediates histone methylation Cell 113, 429–432 92 Ng, H.H., Robert, F., Young, R.A & Struhl, K (2003) Targeted recruitment of Set1 histone methylase by elongating Pol II provides a localized mark and memory of recent transcriptional activity Mol Cell 11, 709–719 93 Krogan, N.J., Dover, J., Wod, A., Schneider, J., Heidt, J., Boateng, M.A., Dean, K., Ryan, O.W., Golshani, A., Johnston, M., Greenblatt, J.F & Shilatifard, A (2003) The Paf1 complex is required for histone H3 methylation by COMPASS and Dot1p: linking transcriptional elongation to histone methylation Mol Cell 11, 721–729 94 Li, J., Moazed, D & Gygi, S.P (2002) Association of the histone methyltransferase Set2 with RNA polymerase II plays a role in transcription elongation J Biol Chem 277, 49383–49388 95 Xiao, T., Hall, H., Kizer, K.O., Shibata, Y., Hall, M.C., Borchers, C.H & Strahl, B.D (2003) Phosphorylation of RNA 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 polymerase II CTD regulates H3 methylation in yeast Genes Dev 17, 654–663 Venetianer, A., Dubois, M.-F., Nguyen, V.T., Seo, S.J., Bellier, S & Bensaude, O (1995) Phosphorylation state of RNA polymerase II C-terminal domain (CTD) in heat-shocked cells: possible involvement of the stress activated MAP kinases Eur J Biochem 233, 83–92 Dubois, M.F., Nguyen, V.T., Bellier, S & Bensaude, O (1994) Inhibitors of transcription such as 5,6-dichloro-1-b-D-ribofuranosyl benzimidazole (DRB) and isoquinoline sulfonamide derivatives (H-8 and H-7*), promote the dephosphorylation of the C-terminal domain (CTD) of RNA polymerase II largest subunit J Biol Chem 269, 13331–13336 Palancade, B., Dubois, M.F., Dahmus, M.E & Bensaude, O (2001) Transcription-independent RNA polymerase II dephosphorylation by the FCP1 CTD-phosphatase in Xenopus laevis early embryos Mol Cell Biol 21, 6359–6368 Bellier, S., Dubois, M.-F., Nishida, E., Almouzni, G & Bensaude, O (1997) Phosphorylation of RNA polymerase II largest subunit during Xenopus laevis oocyte maturation Mol Cell Biol 17, 1434–1440 Valay, J.G., Dubois, M.-F., Bensaude, O & Faye, G (1996) Ccl1, a cyclin associated with protein kinase Kin28 controls the phosphorylation of RNA polymerase II largest subunit and mRNA transcription C.R Acad Sci Paris 319, 183–190 ´ Casse, C., Giannoni, F., Nguyen, V.T., Dubois, M.-F & Bensaude, O (1999) The transcriptional inhibitors, actinomycin D and a-amanitin, activate the HIV-1 promoter and favor phosphorylation of the RNA polymerase II C-terminal domain J Biol Chem 274, 16097–16106 Nguyen, V.T., Giannoni, F., Dubois, M.-F., Seo, S.-J., Vigneron, ´ M., Kedinger, C & Bensaude, O (1996) In vivo degradation of RNA polymerase II largest subunit triggered by a-amanitin Nucleic Acids Res 24, 2924–2929 Bregman, D.B., Halaban, R., Van Gool, A.J., Henning, K.A., Friedberg, E.C & Warren, S.L (1996) UV-induced ubiquitination of RNA polymerase II: a novel modification deficient in Cockayne syndrome cells Proc Natl Acad Sci USA 93, 11586–11590 Nguyen, V.T., Kiss, T., Michels, A.A & Bensaude, O (2001) 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes Nature 414, 322–325 Yang, Z., Zhu, Q., Luo, K & Zhou, Q (2001) The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription Nature 414, 317–322 Ostapenko, D & Solomon, M.J (2003) Budding yeast CTDK-I is required for DNA damage-induced transcription Eukaryot Cell 2, 274–283 Akoulitchev, S., Chuikov, S & Reinberg, D (2000) TFIIH is negatively regulated by cdk8-containing mediator complexes Nature 407, 102–106 Weeks, J.R., Hardin, S.E., Shen, J., Lee, J.M & Greenleaf, A (1993) Locus-specific variation in phosphorylation state of RNA polymerase II in vivo: correlations with gene activity and transcript processing Genes Dev 7, 2329–2344 Zeng, C., Kim, E., Warren, S.L & Berget, S.M (1997) Dynamic relocation of transcription and splicing factors dependent upon transcriptional activity EMBO J 16, 1401–1412 Bregman, D.B.L., van der Zee, S & Warren, S.L (1995) Transcription-dependent redistribution of the large subunit of RNA polymerase II to discrete nuclear domains J Cell Biol 129, 287–298 Prasanth, K.V., Sacco-Bubulya, P.A., Prasanth, S.G & Spector, D.L (2003) Sequential entry of components of gene expression machinery into daughter nuclei Mol Biol Cell 14, 1043–1057 Ó FEBS 2003 RNA polymerase II CTD phosphorylation (Eur J Biochem 270) 3869 112 Seydoux, G & Dunn, M.A (1997) Transcriptionally-repressed germ cells lack a subpopulation of phosphorylated RNA polymerase II in early embryos of C elegans and D melanogaster Development 124, 2191–2201 113 Knaut, H., Pelegri, F.K.B., Schwarz, H & Nusslein-Volhard, C (2000) Zebrafish vasa RNA but not its protein is a component of the germ plasm and segregates asymmetrically before germline specification J Cell Biol 149, 875–888 114 Tenenhaus, C., Schubert, C & Seydoux, G (1998) Genetic requirements for PIE-1 localization and inhibition of gene expression in the embryonic germ lineage of Caenorhabditis elegans Dev Biol 200, 212–224 115 Shim, E.Y., Walker, A.K., Shi, Y & Blackwell, T.K (2002) CDK-9/cyclin T (P-TEFb) is required in two postinitiation pathways for transcription in the C elegans embryo Genes Dev 16, 2135–2146 116 Dantonel, J.C., Quintin, S., Lakatos, L., Labouesse, M & Tora, L (2000) TBP-like factor is required for embryonic RNA polymerase II transcription in C elegans Mol Cell 6, 715–722 117 Kaltenbach, L., Horner, M.A., Rothman, J.H., Mango, S.E & Sharp, P.A (2000) The TBP-like factor CeTLF is required to activate RNA polymerase II transcription during C elegans embryogenesis Mol Cell 6, 705–713 118 Wallenfang, M.R & Seydoux, G (2002) cdk-7 is required for mRNA transcription and cell cycle progression in Caenorhabditis elegans embryos Proc Natl Acad Sci USA 99, 5527–5532 119 Shim, E.Y., Walker, A.K & Blackwell, T.K (2002) Broad requirement for the mediator subunit RGR-1 for transcription in the C elegans embryo J Biol Chem 277, 30413–30416 120 Walker, A.K., Rothman, J.H., Shi, Y & Blackwell, T.K (2001) Distinct requirements for C.elegans TAFIIs in early embryonic transcription EMBO J 20, 5269–5279 121 Muller, F., Lakatos, L., Dantonel, J., Strahle, U & Tora, L (2001) TBP is not universally required for zygotic RNA polymerase II transcription in zebrafish Curr Biol 11, 282–287 122 Rossi, D.J., Londesborough, A., Korsisaari, N., Pihlak, A., Lehtonen, E., Henkemeyer, M & Makela, T.P (2001) Inability to enter S phase and defective RNA polymerase II CTD phosphorylation in mice lacking Mat1 EMBO J 20, 2844– 2856 123 Thompson, N.E., Steinberg, T.H., Aronson, D.B & Burgess, R.R (1989) Inhibition of in vivo and in vitro transcription by monoclonal antibodies prepared against wheat germ RNA polymerase II that react with the heptapeptide repeat of eukaryotic RNA polymerase II J Biol Chem 264, 11511– 11520 124 Mortillaro, M.J., Blencowe, B.J., Wei, X., Du Nakayasu, H.L., Warren, S.L., Sharp, P.A & Berezney, R (1996) A hyperphosphorylated form of the large subunit of RNA polymerase II is associated with splicing complexes and the nuclear matrix Proc Natl Acad Sci USA 93, 8253–8257 125 Vincent, M., Lauriault, P., Dubois, M.-F., Lavoie, S., Bensaude, O & Chabot, B (1996) The nuclear matrix protein p255 is a highly phosphorylated form of RNA polymerase II largest subunit which associates with the spliceosome Nucleic Acids Res 24, 4649–4652 126 Michels, A.A., Nguyen, V.T., Fraldi, A., Labas, V., Edwards, M., Bonnet, F., Lania, L & Bensaude, O (2003) MAQ1 ´ ` (Ômenage a quatreÕ) and 7SK RNA interact with CDK9/CyclinT complexes in a transcription-dependent manner Mol Cell Biol 23, 4859–4869 127 Cisek, L.J & Corden, J.L (1989) Phosphorylation of RNA polymerase by the murine homologue of the cell-cycle control protein cdc2 Nature 339, 679–684 128 Nigg, E.A (1995) Cyclin-dependent protein kinases: key regulators of the eukaryotic cell cycle Bioessays 17, 471–480 129 Zhao, J., Dynlacht, B., Imai, T., Hori, T & Harlow, E (1998) Expression of NPAT, a novel substrate of cyclin E-CDK2, promotes S-phase entry Genes Dev 12, 456–461 130 Svejstrup, J.Q., Vichi, P & Egly, J.-M (1996) The multiple roles of transcription/repair factor TFIIH Trends Biochem Sci 21, 346–350 131 Fisher, R.P & Morgan, D.O (1994) A novel cyclin associates with MO15/CDK7 to form the CDK-activating kinase Cell 78, 713–724 132 Ko, L.J., Shieh, S.Y., Chen, X., Jayaraman, L., Tamai, K., Taya, Y., Prives, C & Pan, Z.Q (1997) p53 is phosphorylated by CDK7-cyclin H in a p36-MAT1 dependent manner Mol Cell Biol 17, 7220–7229 133 Ohkuma, Y & Roeder, R.G (1994) Regulation of TFIIH ATPase and kinase activities by TFIIE during active initiation complex formation Nature 368, 160–163 134 Liao, S.-M., Zhang, J., Jeffery, D.A., Koleske, A.J., Thompson, C.M., Chao, D.M., Viljoen, M., van Vuuren, H.J.J & Young, R.A (1995) A kinase-cyclin pair in the RNA polymerase II holoenzyme Nature 374, 193–196 ´ 135 Leclerc, V., Tassan, J.P., O’Farrell, P.H., Nigg, E.A & Leopold, P (1996) Drosophila cdk8, a kinase partner of cyclin C that interacts with the large subunit of RNA polymerase II Mol Biol Cell 7, 505–513 136 Rickert, P., Seghezzi, W., Shanahan, F., Cho, H & Lees, E (1996) Cyclin C/CDK8 is a novel CTD kinase associated with RNA polymerase II Oncogene 12, 2631–2640 137 Chi, Y., Huddleston, M.J., Zhang, X., Young, R.A., Annan, R.S., Carr, S.A & Deshaies, R.J (2001) Negative regulation of Gcn4 and Msn2 transcription factors by Srb10 cyclin-dependent kinase Genes Dev 15, 1078–1092 138 Hirst, M., Kobor, M.S., Kuriakose, N., Greenblatt, J & Sadowski, I (1999) GAL4 is regulated by the RNA polymerase II holoenzyme-associated cyclin-dependent kinase SRB10/CDK8 Mol Cell 3, 673–678 139 Zhu, Y., Pe’ery, T., Peng, J., Ramanathan, Y., Marshall, N., Marshall, T., Amendt, B., Mathews, M.B & Price, D.H (1997) Transcription elongation factor P-TEFb is required for HIV-1 Tat transactivation in vitro Genes Dev 11, 2622–2632 140 Price, D.H (2000) P-TEFb, a cyclin-dependent kinase controlling elongation by RNA polymerase II Mol Cell Biol 20, 2629–2634 141 Ramanathan, Y., Reza, S.M., Young, T.M., Mathews, M.B & Pe’ery, T (1999) Human and rodent transcription elongation factor P–TEFb: interactions with human immunodeficiency virus type Tat and carboxy-terminal domain substrate J Virol 73, 5448–5458 142 Kim, J.B & Sharp, P.A (2001) Positive transcription elongation factor b phosphorylates hSPT5 and RNA polymerase II carboxyterminal domain independently of cyclin-dependent kinase activating kinase J Biol Chem 276, 12317–12323 143 Simone, C., Bagella, L., Bellan, C & Giordano, A (2002) Physical interaction between pRb and cdk9/cyclin T2 complex Oncogene 21, 4158–4165 144 Simone, C., Stiegler, P., Bagella, L., Pucci, B., Bellan, C., De Falco, G., De Luca, A., Guanti, G., Puri, P.L & Giordano, A (2002) Activation of MyoD-dependent transcription by CDK9/ cyclin T2 Oncogene 21, 4137–4148 145 Lee, J.M & Greenleaf, A.L (1989) A protein kinase that phosphorylates the C-terminal repeat domain of the largest subunit of RNA polymerase II Proc Natl Acad Sci USA 86, 3624–3628 146 Sterner, D.E., Lee, J.M., Hardin, S.E & Greenleaf, A.L (1995) The yeast carboxyl-terminal repeat domain kinase CTDK-I is a divergent cyclin-cyclin-dependent kinase complex Mol Cell Biol 15, 5716–5724 147 Patturajan, M., Conrad, N.K., Bregman, D.B & Corden, J.L (1999) Yeast carboxyl-terminal domain kinase I positively and Ó FEBS 2003 3870 B Palancade and O Bensaude (Eur J Biochem 270) 148 149 150 151 negatively regulates RNA polymerase II carboxyl-terminal domain phosphorylation J Biol Chem 274, 27823–27828 Murray, S., Udupa, R., Yao, S., Hartzog, G & Prelich, G (2001) Phosphorylation of the RNA polymerase II carboxy-terminal domain by the Bur1 cyclin-dependent kinase Mol Cell Biol 21, 4089–4096 ` Dubois, M.-F., Nguyen, V.T., Dahmus, M.E., Pages, G., ´ Pouyssegur, J & Bensaude, O (1994) Enhanced phosphorylation of the C-terminal domain of RNA polymerase II upon serum stimulation of quiescent cells: possible involvement of MAP kinases EMBO J 13, 4787–4797 Treisman, R (1996) Regulation of transcription by MAP kinase cascades Curr Opin Cell Biol 8, 205–215 Dvir, A., Peterson, S.R., Knuth, M.W., Lu, H & Dynan, W.S (1992) Ku antigen is the regulatory component of a template- 152 153 154 155 156 associated protein kinase that phosphorylates RNA polymerase II Proc Natl Acad Sci USA 89, 11920–11924 Smith, G.C & Jackson, S.P (1999) The DNA-dependent protein kinase Genes Dev 13, 916–934 Van Etten, R.A (1999) Cycling, stressed-out and nervous: cellular functions of c-abl Trends Cell Biol 9, 179–186 Zohn, I.E & Brivanlou, A.H (2001) Expression cloning of Xenopus Os4, an evolutionarily conserved gene, which induces mesoderm and dorsal axis Dev Biol 239, 118–131 Bollen, M & Beullens, M (2002) Signaling by protein phosphatases in the nucleus Trends Cell Biol 12, 138–145 Keriel, A., Stary, A., Sarasin, A., Rochette-Egly, C & Egly, J.M (2002) XPD mutations prevent TFIH-dependent transactivation by nuclear receptors and phosphorylation of RARa Cell 109, 125–135 ... Yeast carboxyl-terminal domain kinase I positively and Ó FEBS 2003 3870 B Palancade and O Bensaude (Eur J Biochem 270) 148 149 150 151 negatively regulates RNA polymerase II carboxyl-terminal domain. .. recycled for another transcription round, RNAP II is dephosphorylated by the FCP1 CTD phosphatase Global changes in RNAP II phosphorylation The IIa and IIo RNAP II phosphoisoforms coexist in a dynamic... (DRB) and isoquinoline sulfonamide derivatives (H-8 and H-7*), promote the dephosphorylation of the C-terminal domain (CTD) of RNA polymerase II largest subunit J Biol Chem 269, 13331–13336 Palancade,

Ngày đăng: 17/03/2014, 10:20