リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「tRIP‐seq reveals repression of premature polyadenylation by co‐transcriptional FUS‐U1 snRNP assembly」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

tRIP‐seq reveals repression of premature polyadenylation by co‐transcriptional FUS‐U1 snRNP assembly

Masuda, Akio Kawachi, Toshihiko Takeda, Jun‐ichi Ohkawara, Bisei Ito, Mikako Ohno, Kinji 名古屋大学

2020.05.06

概要

RNA processing occurs co‐transcriptionally through the dynamic recruitment of RNA processing factors to RNA polymerase II (RNAPII). However, transcriptome‐wide identification of protein–RNA interactions specifically assembled on transcribing RNAPII is challenging. Here, we develop the targeted RNA immunoprecipitation sequencing (tRIP‐seq) method that detects protein–RNA interaction sites in thousands of cells. The high sensitivity of tRIP‐seq enables identification of protein–RNA interactions at functional subcellular levels. Application of tRIP‐seq to the FUS‐RNA complex in the RNAPII machinery reveals that FUS binds upstream of alternative polyadenylation (APA) sites of nascent RNA bound to RNAPII, which retards RNAPII and suppresses the recognition of the polyadenylation signal by CPSF. Further tRIP‐seq analyses demonstrate that the repression of APA is achieved by a complex composed of FUS and U1 snRNP on RNAPII, but not by either one alone. Moreover, our analysis reveals that FUS mutations in familial amyotrophic lateral sclerosis (ALS) that impair the FUS‐U1 snRNP interaction aberrantly activate the APA sites. tRIP‐seq provides new insights into the regulatory mechanism of co‐transcriptional RNA processing by RNA processing factors.

この論文で使われている画像

参考文献

1.

Gerstberger S, Hafner M, Tuschl T (2014) A census of human RNA-binding proteins. Nat

Rev Genet 15: 829-45

2.

Bentley DL (2014) Coupling mRNA processing with transcription in time and space. Nat

Rev Genet 15: 163-75

3.

Harlen KM, Churchman LS (2017) The code and beyond: transcription regulation by the

RNA polymerase II carboxy-terminal domain. Nat Rev Mol Cell Biol 18: 263-273

4.

Nojima T, Rebelo K, Gomes T, Grosso AR, Proudfoot NJ, Carmo-Fonseca M (2018) RNA

Polymerase II Phosphorylated on CTD Serine 5 Interacts with the Spliceosome during Cotranscriptional Splicing. Molecular cell 72: 369-379 e4

5.

Masuda A, Takeda J, Ohno K (2016) FUS-mediated regulation of alternative RNA

processing in neurons: insights from global transcriptome analysis. Wiley Interdiscip Rev RNA 7:

330-40

6.

Zhou Z, Licklider LJ, Gygi SP, Reed R (2002) Comprehensive proteomic analysis of the

human spliceosome. Nature 419: 182-5

7.

Rappsilber J, Ryder U, Lamond AI, Mann M (2002) Large-scale proteomic analysis of the

human spliceosome. Genome Res 12: 1231-45

8.

Tan AY, Manley JL (2009) The TET family of proteins: functions and roles in disease. J

Mol Cell Biol 1: 82-92

9.

Ratti A, Buratti E (2016) Physiological functions and pathobiology of TDP-43 and

FUS/TLS proteins. J Neurochem 138 Suppl 1: 95-111

10.

Ballarino M, Jobert L, Dembele D, de la Grange P, Auboeuf D, Tora L (2013) TAF15 is

important for cellular proliferation and regulates the expression of a subset of cell cycle genes

through miRNAs. Oncogene 32: 4646-55

11.

Sohn EJ, Park J, Kang SI, Wu YP (2012) Accumulation of pre-let-7g and downregulation

of mature let-7g with the depletion of EWS. Biochem Biophys Res Commun 426: 89-93

12.

Chi B, O'Connell JD, Yamazaki T, Gangopadhyay J, Gygi SP, Reed R (2018) Interactome

analyses revealed that the U1 snRNP machinery overlaps extensively with the RNAP II machinery

and contains multiple ALS/SMA-causative proteins. Sci Rep 8: 8755

13.

Schwartz JC, Ebmeier CC, Podell ER, Heimiller J, Taatjes DJ, Cech TR (2012) FUS binds

the CTD of RNA polymerase II and regulates its phosphorylation at Ser2. Genes and Development

26: 2690-5

14.

Bertolotti A, Melot T, Acker J, Vigneron M, Delattre O, Tora L (1998) EWS, but not EWSFLI-1, is associated with both TFIID and RNA polymerase II: interactions between two members of

31

the TET family, EWS and hTAFII68, and subunits of TFIID and RNA polymerase II complexes.

Mol Cell Biol 18: 1489-97

15.

Kwon I, Kato M, Xiang S, Wu L, Theodoropoulos P, Mirzaei H, Han T, Xie S, Corden JL,

McKnight SL (2013) Phosphorylation-regulated binding of RNA polymerase II to fibrous polymers

of low-complexity domains. Cell 155: 1049-60

16.

Pomeranz Krummel DA, Oubridge C, Leung AK, Li J, Nagai K (2009) Crystal structure of

human spliceosomal U1 snRNP at 5.5 A resolution. Nature 458: 475-80

17.

Wahl MC, Will CL, Luhrmann R (2009) The spliceosome: design principles of a dynamic

RNP machine. Cell 136: 701-18

18.

Almada AE, Wu X, Kriz AJ, Burge CB, Sharp PA (2013) Promoter directionality is

controlled by U1 snRNP and polyadenylation signals. Nature 499: 360-3

19.

Kaida D, Berg MG, Younis I, Kasim M, Singh LN, Wan L, Dreyfuss G (2010) U1 snRNP

protects pre-mRNAs from premature cleavage and polyadenylation. Nature 468: 664-8

20.

Berg MG, Singh LN, Younis I, Liu Q, Pinto AM, Kaida D, Zhang Z, Cho S, Sherrill-Mix S,

Wan L, et al. (2012) U1 snRNP determines mRNA length and regulates isoform expression. Cell

150: 53-64

21.

Oh JM, Di C, Venters CC, Guo J, Arai C, So BR, Pinto AM, Zhang Z, Wan L, Younis I, et

al. (2017) U1 snRNP telescripting regulates a size-function-stratified human genome. Nat Struct

Mol Biol 24: 993-999

22.

So BR, Di C, Cai Z, Venters CC, Guo J, Oh JM, Arai C, Dreyfuss G (2019) A Complex of

U1 snRNP with Cleavage and Polyadenylation Factors Controls Telescripting, Regulating mRNA

Transcription in Human Cells. Molecular cell

23.

Venters CC, Oh JM, Di C, So BR, Dreyfuss G (2019) U1 snRNP Telescripting:

Suppression of Premature Transcription Termination in Introns as a New Layer of Gene Regulation.

Cold Spring Harb Perspect Biol 11

24.

Yamazaki T, Chen S, Yu Y, Yan B, Haertlein TC, Carrasco MA, Tapia JC, Zhai B, Das R,

Lalancette-Hebert M, et al. (2012) FUS-SMN protein interactions link the motor neuron diseases

ALS and SMA. Cell Rep 2: 799-806

25.

Sun S, Ling SC, Qiu J, Albuquerque CP, Zhou Y, Tokunaga S, Li H, Qiu H, Bui A, Yeo

GW, et al. (2015) ALS-causative mutations in FUS/TLS confer gain and loss of function by altered

association with SMN and U1-snRNP. Nature communications 6: 6171

26.

Ishigaki S, Masuda A, Fujioka Y, Iguchi Y, Katsuno M, Shibata A, Urano F, Sobue G,

Ohno K (2012) Position-dependent FUS-RNA interactions regulate alternative splicing events and

transcriptions. Scientific reports 2: 529

32

27.

Masuda A, Takeda J, Okuno T, Okamoto T, Ohkawara B, Ito M, Ishigaki S, Sobue G, Ohno

K (2015) Position-specific binding of FUS to nascent RNA regulates mRNA length. Genes Dev 29:

1045-57

28.

Van Nostrand EL, Pratt GA, Shishkin AA, Gelboin-Burkhart C, Fang MY, Sundararaman

B, Blue SM, Nguyen TB, Surka C, Elkins K, et al. (2016) Robust transcriptome-wide discovery of

RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat Methods 13: 508-14

29.

Yeo GW, Coufal NG, Liang TY, Peng GE, Fu XD, Gage FH (2009) An RNA code for the

FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat Struct

Mol Biol 16: 130-7

30.

Huppertz I, Attig J, D'Ambrogio A, Easton LE, Sibley CR, Sugimoto Y, Tajnik M, Konig J,

Ule J (2014) iCLIP: protein-RNA interactions at nucleotide resolution. Methods 65: 274-87

31.

Takeda JI, Masuda A, Ohno K (2017) Six GU-rich (6GUR) FUS-binding motifs detected

by normalization of CLIP-seq by Nascent-seq. Gene 618: 57-64

32.

Sasagawa Y, Nikaido I, Hayashi T, Danno H, Uno KD, Imai T, Ueda HR (2013) QuartzSeq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic

gene-expression heterogeneity. Genome Biol 14: R31

33.

Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR (2012)

Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop

codons. Cell 149: 1635-46

34.

Zhao BS, Roundtree IA, He C (2017) Post-transcriptional gene regulation by mRNA

modifications. Nat Rev Mol Cell Biol 18: 31-42

35.

Naftelberg S, Schor IE, Ast G, Kornblihtt AR (2015) Regulation of alternative splicing

through coupling with transcription and chromatin structure. Annu Rev Biochem 84: 165-98

36.

Porrua O, Libri D (2015) Transcription termination and the control of the transcriptome:

why, where and how to stop. Nat Rev Mol Cell Biol 16: 190-202

37.

Schonemann L, Kuhn U, Martin G, Schafer P, Gruber AR, Keller W, Zavolan M, Wahle E

(2014) Reconstitution of CPSF active in polyadenylation: recognition of the polyadenylation signal

by WDR33. Genes Dev 28: 2381-93

38.

Engreitz JM, Sirokman K, McDonel P, Shishkin AA, Surka C, Russell P, Grossman SR,

Chow AY, Guttman M, Lander ES (2014) RNA-RNA interactions enable specific targeting of

noncoding RNAs to nascent Pre-mRNAs and chromatin sites. Cell 159: 188-199

39.

Kondo Y, Oubridge C, van Roon AM, Nagai K (2015) Crystal structure of human U1

snRNP, a small nuclear ribonucleoprotein particle, reveals the mechanism of 5' splice site

recognition. eLife 4

33

40.

Yu Y, Reed R (2015) FUS functions in coupling transcription to splicing by mediating an

interaction between RNAP II and U1 snRNP. Proc Natl Acad Sci U S A 112: 8608-13

41.

Derti A, Garrett-Engele P, Macisaac KD, Stevens RC, Sriram S, Chen R, Rohl CA, Johnson

JM, Babak T (2012) A quantitative atlas of polyadenylation in five mammals. Genome Research

22: 1173-83

42.

Kapeli K, Martinez FJ, Yeo GW (2017) Genetic mutations in RNA-binding proteins and

their roles in ALS. Hum Genet 136: 1193-1214

43.

Yu Y, Chi B, Xia W, Gangopadhyay J, Yamazaki T, Winkelbauer-Hurt ME, Yin S, Eliasse

Y, Adams E, Shaw CE, et al. (2015) U1 snRNP is mislocalized in ALS patient fibroblasts bearing

NLS mutations in FUS and is required for motor neuron outgrowth in zebrafish. Nucleic Acids Res

43: 3208-18

44.

De Santis R, Santini L, Colantoni A, Peruzzi G, de Turris V, Alfano V, Bozzoni I, Rosa A

(2017) FUS Mutant Human Motoneurons Display Altered Transcriptome and microRNA Pathways

with Implications for ALS Pathogenesis. Stem Cell Reports 9: 1450-1462

45.

Ha KCH, Blencowe BJ, Morris Q (2018) QAPA: a new method for the systematic analysis

of alternative polyadenylation from RNA-seq data. Genome Biol 19: 45

46.

Lee FCY, Ule J (2018) Advances in CLIP Technologies for Studies of Protein-RNA

Interactions. Molecular cell 69: 354-369

47.

Burke KA, Janke AM, Rhine CL, Fawzi NL (2015) Residue-by-Residue View of In Vitro

FUS Granules that Bind the C-Terminal Domain of RNA Polymerase II. Molecular cell 60: 231-41

48.

Boehning M, Dugast-Darzacq C, Rankovic M, Hansen AS, Yu T, Marie-Nelly H,

McSwiggen DT, Kokic G, Dailey GM, Cramer P, et al. (2018) RNA polymerase II clustering

through carboxy-terminal domain phase separation. Nat Struct Mol Biol

49.

Kwiatkowski TJ, Jr., Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis

A, Gilchrist J, Kasarskis EJ, Munsat T, et al. (2009) Mutations in the FUS/TLS gene on

chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323: 1205-8

50.

Masuda A, Andersen HS, Doktor TK, Okamoto T, Ito M, Andresen BS, Ohno K (2012)

CUGBP1 and MBNL1 preferentially bind to 3' UTRs and facilitate mRNA decay. Sci Rep 2: 209

51.

Dunn JJ (1976) RNase III cleavage of single-stranded RNA. Effect of ionic strength on the

fideltiy of cleavage. Journal of Biological Chemistry 251: 3807-14

52.

Martin M (2011) Cutadapt Removes Adapter Sequences From High-Throughput

Sequencing Reads. EMBnet journal 17: 10-12

53.

Bao W, Kojima KK, Kohany O (2015) Repbase Update, a database of repetitive elements

in eukaryotic genomes. Mob DNA 6: 11

34

54.

Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M,

Gingeras TR (2013) STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29: 15-21

55.

Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers

RM, Brown M, Li W, et al. (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9:

R137

56.

Schneider TD, Stephens RM (1990) Sequence logos: a new way to display consensus

sequences. Nucleic Acids Res 18: 6097-100

57.

Machanick P, Bailey TL (2011) MEME-ChIP: motif analysis of large DNA datasets.

Bioinformatics 27: 1696-7

58.

Shen L, Shao N, Liu X, Nestler E (2014) ngs.plot: Quick mining and visualization of nextgeneration sequencing data by integrating genomic databases. BMC Genomics 15: 284

59.

Fagerland MW, Sandvik L (2009) Performance of five two-sample location tests for

skewed distributions with unequal variances. Contemp Clin Trials 30: 490-6

35

Figure Legends

36

Figure 1. Identification of protein-RNA interactions by tRIP-seq

A. Schematic representation of Terminator 5′-phosphate-dependent exonuclease (TEX) treatment

of immunoprecipitated RNA. TEX digestion stops at the antibody-tethered nucleotide, while TEX

eliminates the non-specific RNA and linkers remaining on the beads. Green circle represents m6A

nucleotide.

B. Distribution of RBFOX2-eCLIP reads and RBFOX2-tRIP reads on chr10: 75,544,000–

75,635,000 (GRCh37/hg19).

C. Distributions of FUS-HITS-CLIP reads, FUS-tRIP reads, and ContAb-tRIP reads on chr15:

26,165,000–28,520,000 (GRCm38/mm10). The number of N2A cells is indicated.

D. Correlation of read densities constituting MACS-defined peaks between RBFOX2-tRIP and

RBFOX2-eCLIP. Scatter plot indicates RPM of tRIP-seq and eCLIP constituting each peak.

E. Correlation in read densities constituting MACS-defined peaks between FUS-tRIP and FUSHITS-CLIP. Scatter plot indicates RPM of tRIP-seq and HITS-CLIP constituting each peak.

F. The RBFOX2 motif identified by MEME in the peaks of RBFOX2-tRIP.

G. The FUS motif identified by MEME in the peaks of FUS-tRIP of 1 × 108 cells.

H. Distributions of FUS-tRIP reads (upper panel) and ContAb-tRIP reads (lower panel) mapped to

the relative positions of all coding genes in mouse. The ngs.plot tool [58] was used to calculate the

average RPM for a gene structure. The average RPM at each position was normalized based on the

total RPM mapped to each gene. The standard error of normalized RPM is shown as a semitransparent shade around the average curve. TSS, transcriptional start site; TTS, transcriptional

termination site.

37

38

Figure 2. Identification of protein-RNA interactions in the RNAPII machinery by tRIP-seq

A. Schematic representation of immunoprecipitation of protein-RNA complexes engaged to

RNAPII shown in (B). With UV-crosslinking of protein-RNA interactions (UV+) or without (UV-),

the chromatin fraction is isolated from the cells and treated with (Pre-RNase+) or without RNase A

(Pre-RNase-). RNAPII, shown in gray, is 1st immunoprecipitated. The immunoprecipitants are

treated with RNase A to release a protein, shown in green, from the protein-RNA-RNAPII complex.

B. RNAPII-RBP-RNA complex was immunoprecipitated by anti-RNAPII antibody or control

antibody (cont), and subjected to immunoblotting. The input lanes contained 0.4% of lysates used in

the immunoprecipitation experiments.

C. Generation of tRIP-seq libraries of RNAPII-RBP-RNA complexes. After UV crosslinking,

protein-RNA complexes were immunoprecipitated with anti-RNAPII (1st IP). RNA of the isolated

RNAPII-RBP-RNA complexes was partially digested with RNase on beads, and RBP-RNA

complexes dissociated from the bead were further immunoprecipitated with anti-FUS or anti-U1C

antibody bound to the other beads (2nd IP). The immunoprecipitated FUS-RNA or U1C-RNA

complex was treated with TEX, and then subjected to tRIP-seq library generation. The bottom

panels show PCR-amplification of tRIP-seq libraries generated from RNAPII-FUS-RNA complex

(left panel) and RNAPII-U1C-RNA complex (right panel).

D. Read distributions of Chr-FUS-tRIP, U1C-FUS-tRIP, and RNAPII-FUS-tRIP mapped to the

relative positions of all coding genes in mouse. The ngs.plot tool [58] was used to calculate the

average RPM for a gene structure. The average RPM at each position was normalized for the total

RPM mapped to each gene. The standard error of normalized RPM is shown as a semi-transparent

shade around the average curve. Arrowhead indicates a peak before the transcription termination

site (TTS) in RNAPII-FUS-tRIP.

39

E. Read distributions of RNAPII-FUS-tRIP (purple lines) and Chr-FUS-tRIP (black lines) around

the APA sites repressed (bold lines) or activated (thin lines) by FUS. The p-values for the

differences between RNAPII-FUS-tRIP and Chr-FUS-tRIP around FUS-repressed APA sites are

indicated by circles. An arrowhead indicates a peak upstream to APA sites in RNAPII-FUS-tRIP.

F. Read distributions of RNAPII-CPSF160-tRIPs generated from Fus-silenced cells (siFus, pink

line) and those of control siRNA-treated cells (siCont, green line). The p-values for the differences

between siFus and siCont are indicated by circles. An arrowhead indicates a peak before APA sites

in RNAPII-CPSF160-tRIP of siFus-treated cells.

40

Figure 3. Identification of protein-RNA interactions in the RNAPII machinery by tRIP-seq to

investigate FUS-dependent enhancement of U1 snRNP-RNA interactions around alternative

splice sites

A. Read distributions of Chr-FUS-tRIP mapped around constitutive splice sites (CS sites, gray

dotted line) and alternative splice sites (AS sites, black solid line). Positions of CS sites and AS

sites are according to the ENSEMBL annotations on GRCm38/mm10. 3SS, 3′ splice site; 5SS, 5′

splice site.

B. Read distributions of RNAPII-FUS-tRIP (RNAPII-FUS, purple line), U1C-FUS-tRIP (U1CFUS, blue line), and Chr-FUS-tRIP (Chr-FUS, black line) around AS sites. The p-values for the

41

differences between RNAPII-FUS and Chr-FUS are indicated by circles.

C. Read distributions of Chr-U1C-tRIPs generated from Fus-silenced cells (siFus, pink line) and

those of control siRNA-treated cells (siCont, green line). Arrowhead indicates a peak at the 5′ splice

site, which disappears upon Fus-silencing.

D. Read distributions of RNAPII-U1C-tRIPs generated from Fus-silenced cells (siFus, pink line)

and control siRNA-treated cells (siCont, green line).

Data information: C and D. Left panels show read distributions of tRIP-seqs mapped to the

relative positions of all coding genes in mouse. The standard error of mean is shown as a semitransparent shade around the average curve. Right panels show read distributions of tRIP-seqs

mapped around AS sites. The p-values for the differences between siFus and siCont are indicated by

circles.

42

Figure 4. FUS-U1 snRNP complex on RNAPII suppresses APA

A and B. PolyA-seq was performed using N2A cells to examine the effect of downregulation of

FUS and U1 snRNP on polyA site usage. Cells were treated with siRNA against Fus (siFus) and/or

U1 antisense morpholino (U1AS). Control cells (Cont) were treated with both control siRNA and

control antisense morpholino. A. Correlation of changes in polyA site usage between U1-inhibition

43

and Fus-silencing. Fold change in the RPM of a polyA site between siFus and Cont (siFus/Cont)

was plotted against that between U1AS and Cont (U1AS/Cont). Pearson’s correlation coefficient (r)

and the slope of the regression line are shown. B. Correlation of changes in polyA site usage

between U1-inhibition alone and the combination of Fus-silencing and U1-inhibition. Fold change

in the RPM of a polyA site between siFus+U1AS and Cont (siFus+U1AS/Cont) was plotted against

that between U1AS and Cont (U1AS/Cont). Pearson’s correlation coefficient (r) and the slope of

the regression line are shown. A slope of 1.0 indicates that Fus-silencing has no additive effect on

U1-inhibition.

C. Read distributions of RNAPII-FUS-tRIP (left) and RNAPII-U1C-tRIP (right) around APA sites

repressed [the first quadrant in (A)] or activated [the third quadrant in (A)] by both FUS and U1

snRNP. The p-values for the differences between the repressed and the activated APA sites are

indicated by circles. Arrowheads indicate noticeable peaks of RNAPII-FUS and RNAPII-U1C

upstream to the repressed APA sites.

D. Effects of Fus-silencing (pink line) and U1-inhibition (yellow line) on read distributions of

RNAPII-FUS-tRIP (left) and RNAPII-U1C-tRIP (right) around APA sites repressed by both FUS

and U1 snRNP [the first quadrant in (A)]. The p-values for the differences between contAS and

U1AS (left panel), as well as those between siCont and siFus (right panel), are indicated by circles.

E. Read distributions of U1A-FUS-tRIP (left) and U1C-FUS-tRIP (right) around APA sites

repressed [the first quadrant in (A)] or activated [the third quadrant in (A)] by both FUS and U1

snRNP. The p-values for the differences between repressed and activated APA sites are indicated

by circles. Arrowheads indicate noticeable peaks of U1A/U1C-FUS upstream to the repressed APA

sites.

44

Figure 5. A proposed model for the repression of APA by a complex composed of FUS and U1

snRNP.

FUS interacts with U1 snRNP, and binds upstream to APA to repress transcription termination and

polyadenylation, which allows further elongation of nascent RNA.

45

Figure 6. The ALS mutation in FUS, R495X, aberrantly activates APA sites, which are

normally repressed by FUS-U1 snRNP complex

A. Localization of FUS in R495X_N2A cells (R495X), and wild-type N2A cells (WT). Cells were

fractionated to separate nucleus and cytoplasm. Equal proportion of cell extracts from nuclear and

cytoplasmic fractions were immunoblotted with indicated antibodies.

B. Interactions between FUS and U1 snRNP in the R495X_N2A cells (R495X) and wild-type N2A

cells (WT). Total cell lysates were immunoprecipitated with anti-U1-70K antibody or a control

antibody (cont), and immunoblotted with indicated antibodies.

C. Effects of the R495X mutation on four categories of polyA sites. Four categories of polyA sites

are comprised of (1) FUS-repressed (rep) /U1-repressed sites, (2) FUS-activated (act) /U1-repressed

sites, (3) FUS-repressed/U1-activated sites, and (4) FUS-activated/U1-activated sites, according to

Fig 4A. Fold change of each polyA site usage by the R495X mutation (R495X/WT) is shown in

violin plots. Bars indicate the median and 95% confidential intervals for each category. *** p <

0.001, **** p < 0.0001, based on the Steel-Dwass test for comparing all 6 pairs of categories.

46

D. Change in polyA site usage by the R495X mutation. Fold change of polyA site usage by the

R495X mutation (R495X/WT) on the relative positions of all mouse coding genes is plotted. The

average is shown in blue line. The standard error of the mean is shown in semi-transparent blue

shade.

47

Expanded View Figure Legends

Figure EV1. Schematic

representation of the tRIP workflow

RNA binding protein (RBP)-RNA complex is immunoprecipitated with an antibody against the

RBP. Then, the immunoprecipitated RNA is partially digested with RNase on beads. After stringent

washes, a 64-nt linker is ligated to the 3’ end of the RNA fragment, and immunoprecipitants are

treated with TEX to eliminate non-specifically remaining RNA and free linkers. The TEX digests

immunoprecipitated RNA up to the RBP-tethered site. The immunoprecipitants are then treated

48

with proteinase K, followed by column purification of RNA. After the first-strand synthesis with

reverse transcription, a polyA tail is added to the 3’ end of the first-strand cDNA. After the secondstrand synthesis with the tagging primer, the double-strand cDNAs are PCR-amplified and

subjected to high-throughput sequencing analysis.

49

Figure EV2. Read distributions of m6A-tRIP and PTBP1-tRIP

A. The enrichment of m6A motif (GGAC) around the 5′ ends of mapped reads obtained from m6AtRIP and m6A-MeRIP (RNA-seq of conventional RNA immunoprecipitation using At-m6A

antibody).

B. Correlation of read densities constituting MACS-defined peaks between m6A-MeRIP and m6AtRIP. Scatter plot indicates reads per million mapped reads (RPM) of tRIP and MeRIP constituting

each peak. Pearson’s correlation coefficient (r) is indicated.

C. The CU-rich PTBP1 motifs (upper panels) and the m6A motifs (lower panels) identified by

MEME in the peaks of PTBP1-tRIPs and m6A-tRIPs, respectively, performed using indicated

number of C2C12 cells.

D and E. Distributions of PTBP1-tRIP reads (D) and m6A-tRIP reads (E) mapped to the relative

positions of all mouse coding genes. The ngs.plot tool [58] was used to calculate average RPM on a

50

gene structure. Shown are tRIP-seqs of indicated number of C2C12 cells. The standard error of

average RPM is shown as a semi-transparent shade around the average curve.

51

Figure EV3. The recruitment of FUS upstream to polyA sites slows down transcription and

inhibits the recognition of polyA signal by CPSF

A. Read distributions of Chr-RNAP II-tRIPs of Fus-silenced cells (siFus, pink line) and control

siRNA-treated cells (siCont, green line) around APA sites repressed (left graph) or activated (right

graph) by FUS. The p-values for the differences between siFus and siCont are indicated by circles.

An arrowhead indicates a peak upstream to the repressed APA sites detected in Chr-RNAP II-tRIP

of siCont-treated cells.

52

B. Expanded view of read distributions of RNAP II-FUS-tRIP (Fig 2E, bold purple line), RNAP IItRIP of siCont-treated cells (Fig EV3A, green line), and RNAP II-CPSF160-tRIP of Fus-silenced

cells (Fig 2F, pink line) around FUS-repressed APA sites. The bottom graph shows frequency of the

AAUAAAA polyadenylation signal (blue line). Arrowheads indicate noticeable peaks detected in

the respective tRIP-seqs.

C. Read distributions of Chr-CPSF160-tRIPs of Fus-silenced cells (siFus, pink line) and control

siRNA-treated cells (siCont, green line) around APA sites repressed (left graph) or activated (right

graph) by FUS. The p-values for the differences between siFus and siCont are indicated by circles.

An arrowhead indicates a peak upstream to the repressed APA sites detected in Chr-CPSF160-tRIP

of Fus-silenced cells.

D. A proposed model for FUS-dependent repression of APA. The recruitment of FUS upstream to

polyA sites slows down transcription by RNAP II and inhibits the recognition of polyA signal by

CPSF, which suppresses subsequent APA and keeps elongation by RNAP II.

53

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る