Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Available online at www.sciencedirect.

com

Exploring new roles for RNA-binding proteins in


epigenetic and gene regulation☆
Pedro Avila-Lopez 1,2 and Shannon M Lauberth 1,2,* ]]
]]]]]]
]]

A significant portion of the human proteome comprises RNA- metabolism as RBPs regulate the entire life cycle of
binding proteins (RBPs) that play fundamental roles in RNA. RBPs form functional interactions with RNA
numerous biological processes. In the last decade, there has during transcription, splicing, polyadenylation, sub­
been a staggering increase in RBP identification and cellular localization, translation, and decay [6]. Notably,
classification, which has fueled interest in the evolving roles of an increasingly supported paradigm is that RNAs reg­
RBPs and RBP-driven molecular mechanisms. Here, we focus ulate their own transcription by interacting and reg­
on recent insights into RBP-dependent regulation of the ulating sequence-specific DNA-binding transcription
epigenetic and transcriptional landscape. We describe factors (TFs) and chromatin-regulatory proteins [7–22].
advances in methodologies that define the RNA-protein TFs consist of two primary protein domains required to
interactome and machine-learning algorithms that are regulate gene expression that include a DNA-binding
streamlining RBP discovery and predicting new RNA-binding domain (DBD) that directs TF binding to cis-regulatory
regions. Finally, we present how RBP dysregulation leads to elements and effector domains that facilitate TF inter­
alterations in tumor-promoting gene expression and discuss actions with cofactors [23–25]. Similarly, epigenetic
the potential for targeting these RBPs for the development of regulators often engage with chromatin through reader
new cancer therapeutics. domains that facilitate recognition of specific histone
posttranslational modifications [26–28]. However, in­
Addresses creasing evidence finds that TFs and epigenetic reg­
1
Simpson Querrey Institute for Epigenetics, Feinberg School of ulators also bind RNA and often through domains other
Medicine, Northwestern University, Chicago, IL 60611, USA
2 than the well-known RNA-binding domains (RBDs)
Department of Biochemistry and Molecular Genetics, Feinberg School
of Medicine, Northwestern University, Chicago, IL 60611, USA that include the RNA recognition motif, K-homology
domain, double-strand RNA-binding domain, and zinc
Corresponding author: Lauberth, Shannon M finger domain [9,29–31]. Thus, the mechanisms under­
(shannon.lauberth@northwestern.edu) lying the recruitment/stabilization and functions of TFs
*
Twitter account: @LauberthLab
and chromatin regulators at specific genomic regions
Current Opinion in Genetics & Development 2024, 84:102136
require additional consideration for how RNA may be
involved. Also, since initial RBP annotations were lar­
This review comes from a themed issue on Cancer Genomics
gely based on the presence of conserved RBR domain
Edited by Shannon M. Lauberth and Ali Shilatifard structure and function [30], the breadth of RBPs that
exist, particularly those devoid of typical RBRs, has not
yet been fully realized. The recent and significant ad­
vances in experimental technologies, including machine-
Available online xxxx
learning-based algorithms, promise to unveil new RBPs
https://doi.org/10.1016/j.gde.2023.102136 and RBRs. Thus, a comprehensive identification of
0959–437X/© 2023 Published by Elsevier Ltd. RBPs and their functional consequences will provide
new insights into the regulation of gene expression
during normal development and disease and pro­
vides potential new targets for developing RBP-based
therapeutics.

Numerous reviews have detailed roles for RBPs in reg­


Introduction ulating posttranscriptional regulatory networks in normal
Cumulative studies have raised the current census of development and disease [32–37]. In this review, we
RNA-binding proteins (RBPs) encoded for by the highlight recent technologies advancing the discovery of
human genome to ∼2500 proteins [1–5]. The growing diverse cellular RBPs and RBRs. We discuss the finding
number of RBPs has important implications for RNA that TFs and epigenetic regulators are among the


Given the role as Guest Editor, Shannon M. Lauberth had no involvement in the peer review of the article and has no access to information
regarding its peer-review. Full responsibility for the editorial process of this article was delegated to Ali Shilatifard.

www.sciencedirect.com Current Opinion in Genetics & Development 2024, 84:102136


2

Figure 1
Cancer Genomics

Current Opinion in Genetics & Development 2024, 84:102136


Current Opinion in Genetics and Development

Novel methodologies for investigating the RNA-binding capacity of RBPs. (a) UV cross-linking together with mass spectrometry-based profiling (RBR-ID) allows for the detection of RBPs and their
RBRs [43]. (b) Deep-learning models (HydRA, RNApred, catRAPID, RBPPred, and TriPepSVM) are proving effective in predicting noncanonical RBPs and RBRs [46–50]. (c) The KIN-CLIP approach
reveals the kinetics of RBP binding to RNA at endogenous gene loci [51]. (d) Split-pool recognition of interaction by tag extension (RNA & DNA SPRITE) enables simultaneous mapping of RNA–RNA,
RNA–DNA, and DNA–DNA contacts to comprehensively map RNA and DNA spatial organization [53]. Abbreviations: UV, ultraviolet; 4-SU, 4-thiouridine; PPI, protein–protein interactions; AI, artificial
intelligence.

www.sciencedirect.com
RNA binding proteins in cancer Avila-Lopez and Lauberth 3

growing list of RBPs and that RNA functions should be an approach is the kinetic cross-linking and im­
considered when defining mechanisms underlying munoprecipitation (KIN-CLIP) technique (Figure 1c)
chromatin and gene regulation. Finally, we discuss im­ [51]. KIN-CLIP is a time-resolved laser cross-linking
plications for RBPs in the dysregulation of gene ex­ approach that involves using a pulsed femtosecond (fs)
pression in cancer and the recent exploratory efforts for UV laser that can rapidly photo-cross-link proteins to
identifying molecules that probe RBP functions and RNA relative to the association and dissociation rate
exhibit potential as novel cancer therapeutics. constants for the RNA-protein interactions [51]. Thus,
the power of measuring RBP–RNA-binding kinetics
with KIN-CLIP is the ability to distinguish between the
Identification of RNA-binding proteins that effects of cross-linking and RBP–RNA interactions at
interface with chromatin and transcriptional individual binding sites in cells [51]. Determining the
control kinetic landscape of RBPs in cells provides important
Cataloging additional RBPs across different tissues, de­ information about RBP–RNA interactions at en­
velopmental stages, and disease states is an important but dogenous RBP-binding sites under different cellular
also challenging effort. In Figure 1, we highlight a few conditions. Finally, imaging technologies are at the
recent methods that have streamlined RBP identification forefront of advancing our understanding of the sub­
and characterization. RBP identification has largely cellular distribution and localization patterns of RBPs.
stemmed from proteomic profiling analyses following affi­ For example, the RBP Image database is a resource that
nity purification of polyadenylated RNAs [2,38–41] and provides microscopy imaging data and curated pattern
noncoding RNAs [42–44] and by employing organic phase- descriptions for the subcellular localization of hundreds
separation-based methods [3,45]. RBR identification tools, of RBPs [52]. Moreover, RNA & DNA SPRITE (RD-
including RNA-binding region identification (RBR-ID) SPRITE) (Figure 1d) provides comprehensive mapping
(Figure 1a), have defined RBRs with great precision using of the spatial organization of RNA and DNA in the
mass spectrometric detection of RNA-cross-linked peptide nucleus [53]. RD-SPRITE identifies higher-order RNA-
mass shifts following photoactivatable ribonucleoside-spe­ chromatin hubs through split-and-pool barcoding of all
cific UVA cross-linking with 4-thiouridine [7,43]. RBR DNA and RNA contained within cross-linked RNA,
mapping provides a powerful platform for prioritizing RBR DNA, and protein complexes. The DNA and RNA se­
mutant design for exploring the functional significance of quencing reads with identical barcodes are merged into a
RBP–RNA interactions. RBR-ID also recently provided cluster that enables identification of simultaneous asso­
the unexpected realization that approximately half of the ciations of DNA and RNA molecules. This approach has
TFs identified in the dataset bind RNA and in­ provided functional insights into the role that RNA in­
clude GATA1, GATA2, and RUNX1, MYC, and MAX [7]. cluding low-abundance RNA molecules play in reg­
This finding supports an emerging paradigm that the role ulating the diffusion of other RNAs and proteins into
of RNA should be considered when defining gene-reg­ three-dimensional (3D) structures [53]. Taken together,
ulatory mechanisms. Increasing developments in machine- these various resources are available for guiding func­
learning technologies are also proving powerful in ex­ tional investigations that will undeniably advance our
pediting progress toward RBP identification by increasing understanding of how RNA through RBP interactions
the predictive power of proteome-wide RNA-binding ca­ regulates gene control in normal development and
pacity. One such example is the hybrid ensemble classifier human diseases including cancer.
for RBPs (HydRA) (Figure 1b) [46]. HydRA is a novel
machine-learning-based algorithm that integrates deep-
learning algorithms, such as convolutional neural network, RNA-binding proteins functions associated
transformer, and support vector machines to predict RBPs with epigenetic and transcriptional regulation
and RBRs [46]. HydRA predictions have revealed 1487 While TFs and epigenetic regulators are among the
previously unclassified human RBPs and 76 new RBRs growing number of noncanonical RBPs, it remains to be
[46]. Other examples of RBP classification tools that are determined whether RNA regulates their chromatin and
dependent on machine-learning models that utilize protein gene-regulatory functions or leads to other functional
sequence information include RNApred [47], catRAPID consequences. Here, we focus on current studies that
signature [48], RNA-binding protein predictor (RBPPred) demonstrate the importance of RNA through RBP in­
[49], and TriPepSVM [50]. Taken together, these various teractions in contributing to multifaceted roles in reg­
technological advances are supporting the growing interest ulating gene expression as highlighted in Figure 2.
in understanding how RNA alters the functions of an in­ Specifically, we discuss the role of RBP–RNA interac­
creasing number of RBPs. tions in regulating the following: (i) TF and epigenetic
regulator recruitment/stabilization on chromatin, (ii) re­
Recent technological developments also make it feasible lease of paused RNA polymerase II (RNAPII) into
to measure the kinetics of RBP binding to and dis­ productive elongation, (iii) altering chromatin state and
sociation from their cellular RNAs. An example of such accessibility, (iv) shaping 3D nuclear organization, and

www.sciencedirect.com Current Opinion in Genetics & Development 2024, 84:102136


4 Cancer Genomics

Figure 2

Current Opinion in Genetics and Development

Multifaceted roles of RBPs in gene regulation. (a) RNA fine-tunes gene expression by contributing to the recruitment/stabilization of TFs, RNAPII, and
chromatin regulators [7–11,14,18,20,21]. (b) RNA-dependent recruitment/stabilization of TFs and cofactors on chromatin facilitates the release of
paused RNAPII into productive elongation [11,17]. (c) RNA regulates epigenetic regulators binding chromatin, which in turn leads to changes in the
PTMs of histones and nucleosome remodeling [9,10,13,15,16,19,20,22,29,58]. (d) RNA interactions with CTCF inhibit CTCF binding to low-affinity
binding sites across the genome, which in turn alters the 3D organization of the genome and leads to alterations in the expression of genes involved in
development and differentiation [59–62]. (e) RNA concentrates RBPs in phase condensates to inhibit PUM proteins from leading to defects in mitosis
[65]. Abbreviations: CTD, C-terminal domain; me3, trimethylation.

(v) regulation of RBP activities through phase-separation arginine-rich motif (ARM) [7]. Specifically, the ARM-like
condensate formation. domains were found to be more highly enriched in TFs
relative to other proteins within the proteome and are lo­
An increasing number of studies have revealed that RNA cated adjacent to TF DBDs [7]. RNA-dependent recruit­
contributes to both transcriptional repression and activation ment/stabilization of TFs and cofactors on chromatin
through the dynamic regulation of TF and cofactor chro­ promotes activation through enhanced RNAPII recruit­
matin engagement [7–11,14,18,20,21]. A recent study ment and/or activity. Specifically, several RBPs, including
found that TFs interact with RNA through RBDs that FUS [17] and ribogenesis factor WDR43 [11], have been
resemble the HIV trans-activator of transcription (Tat) shown through RNA interactions to facilitate release of

Current Opinion in Genetics & Development 2024, 84:102136 www.sciencedirect.com


RNA binding proteins in cancer Avila-Lopez and Lauberth 5

paused RNAPII into productive elongation. Also, RNAs RNAs been shown to employ phase separation as a
have been shown to interface with chromatin reader do­ means of controlling RBP activities. For example, RNAs
mains to augment their recruitment/stabilization on chro­ facilitate the sequestration of factors in condensates.
matin. For example, enhancer-derived RNAs (eRNAs) This sequestration function is demonstrated for the long
directly bind to the tandem bromodomains (BDs) of bro­ noncoding NORAD that maintains genomic stability by
modomain and extra-terminal motif (BET) protein BRD4, sequestering PUMILIO (PUM) proteins in phase-sepa­
which increases BRD4 and RNAPII recruitment to aug­ rated condensates to inhibit their roles that otherwise
ment enhancer- and tumor-promoting gene activation [8]. lead to aberrant mitosis [65]. Another example includes
Similarly, RNA interacts with the BDs of PBRM1, a sub­ RNA interactions with the paraspeckle protein PSPC1
unit of the PBAF chromatin remodeling complex, which that induces formation of transcription condensates that
facilitates PBAF chromatin access and PBRM1-mediated tightly control the phosphorylation and RNA-induced
cell growth effects [14]. Another well-known example is release of RNAPII into productive elongation [69].
RNA interactions with polycomb-repressive complex 2 Thus, further consideration of how RNA impacts the
(PRC2), which has been shown to associate with numerous binding specificity, affinity, and nuclear organization of
primary messenger RNAs (mRNAs) and long noncoding TFs and cofactors will further our understanding of the
RNAs [54–57]. Yet, the mechanisms underlying RNA comprehensive mechanisms underlying gene regulation.
regulation of PRC2 have remained disparate. For example,
RNA has been shown to enhance PRC2 chromatin
binding, increase deposition of H3K27me3, and drive re­ Emerging identification of RNA-binding
pression of specific genes involved in cardiomyocyte dif­ proteins involved in altering gene expression
ferentiation [13,29]. On the flip side of the coin, PRC2 can in human cancers
be removed from active chromatin by transferring from Numerous RBPs have been shown to dysregulate post­
chromatin to RNA upon transcriptional activation, which transcriptional events in cancer, which leads to rapid
prevents PRC2 deposition of H3K27me3 on genes changes in protein expression levels that promote cancer
[9,10,13,15,16,19,20,22]. The role of RNA in competitively cell growth and progression [32,70]. The increasing
inhibiting access to active gene loci to protect from tran­ identification of TFs that bind RNA suggests additional
scriptional repression has also been demonstrated for the opportunity for defining new gene-regulatory mechan­
ATP-dependent nucleosome remodeler and transcriptional isms important in cancer. Consistent with RBPs involved
inhibitor Mi-2/CHD4 [58]. In addition to RNA regulating in gene control impacting tumorigenesis is the im­
chromatin engagement, several studies have also unveiled balance in RBP expression levels identified by publicly
that RNA impacts the enzymatic activity of chromatin- available databases, including the Cancer Genome Atlas
modifying enzymes [12,19]. For example, a recent struc­ (TCGA) and GTEx [71,72]. Among the differentially
tural study revealed that RNA inhibits PRC2 enzymatic expressed RBPs that contribute to altered chromatin and
activity by facilitating PRC2 dimerization, which in turn gene control in cancer are the well-known epigenetic
sequesters PRC2’s catalytic subunit [19]. In addition, and transcriptional regulators, including TET2 [73–75],
eRNAs have been shown to reduce CBP’s autoinhibition MYCN [76–78], MEL18 [79,80], NANOG [81–84],
by preventing CBP’s basic activation loop from blocking BRD4 [8,85,86], and PRC2 [79,87]. While these factors
CBP’s active site [12]. RNA has also been shown to shape are known to play critical roles in tumorigenesis, their
3D nuclear organization by modulating the genome-wide relationship with RNAs has not been fully elucidated.
binding profile of CCCTC-binding factor (CTCF) [59–62]. Thus, there is an imminent need for defining the re­
Specifically, RNA promotes CTCF self-association, which lative contributions of RNA in altering the epigenetic
in turn correlates with CTCF chromatin binding and DNA and transcriptional functions of these and the numerous
loop formation [60,62]. Interestingly, RNA–CTCF inter­ other factors recently identified [7]. Such studies will
actions lead to the formation of distinct classes of chro­ undeniably advance our understanding of the dysregu­
matin loops, which suggests an RNA-dependent lation of tumor-promoting gene networks and the de­
mechanism for establishing cell-specific CTCF loops [62]. velopment of new targets with therapeutic implications.
In addition, the long noncoding RNA Jpx has been shown
to inhibit ectopic CTCF binding to chromatin, which can In addition to dysregulated RBP expression, RBPs are
otherwise lead to alterations in anchor site usage and a also enriched for mutations in various cancer types [88].
downregulation of Jpx target gene loci [59]. An assessment of the mutational landscape of thousands
of RBPs across cancer genomes revealed that RBPs un­
RNA contributions to transcriptional regulation also in­ dergo frequent frameshift and in-frame deletions and
volve its role in regulating the formation of membrane­ missense and nonsense mutations [88]. Moreover, hun­
less subcellular compartments, known as biomolecular dreds of candidate driver RBPs associated with func­
condensates that promote the regulation of biochemical tional mutations in cancer were identified using
activities inside cells [53,63–67]. While RNAs and RBPs OncodriveFM [89]. This approach computes the func­
can undergo cophase separation [68], only recently have tional impact of nonsynonymous mutations in genes

www.sciencedirect.com Current Opinion in Genetics & Development 2024, 84:102136


6 Cancer Genomics

across a list of tumor samples and their impact on protein enzyme-active sites to directly examine enzyme func­
function as an indication of positive selection and de­ tion within complex proteomes [101,102]. In addition,
tection of candidate driver genes [89]. Taken together, proteomic platforms that integrate size-exclusion chro­
mutational analysis and the prediction of candidate dri­ matography and high-throughput screening of electro­
vers is informing altered RBP functions in cancer [90] philic compound libraries have accelerated the discovery
and suggests the importance of defining mechanisms of small molecules that can impact protein complexes in
underlying RNA-dependent functions of RBPs in cancer human cells [103]. Importantly, these analyses have re­
initiation and progression. Consistent with mutations sulted in identifying effective chemical probes that
altering RBP function are mutations in splicing factors, target protein domains extending well beyond enzyme-
SF3B1, U2AF1, and PCBP1, that are linked to sig­ active sites [103]. An example of small compounds co-
nificant splicing defects associated with cancer pheno­ opting RBPs that regulate gene expression is the recent
types in multiple cancer types [91]. Recently, an analysis finding that compounds engaging RBP NONO suppress
of publicly available databases, including ClinVar [92] pro-tumorigenic transcriptional networks and impair
and HGMD [93] that archive somatic and germline cancer cell proliferation [104]. This finding, together
variants associated with cancer phenotypes, revealed with the growing number of RBPs in the cell, suggests
hundreds of pathogenic mutations localized to TF-ARM that advances in chemical proteomics will undeniably
RBRs [7]. The significance of the TF-ARM mutations aid in the design of effective chemical probes targeting
was examined through an evaluation of the R269C mu­ RBPs. Moreover, RNA and RBP contributions to phase
tation in the ARM domain of the estrogen receptor 1 separation that in turn impacts RBP functions may also
(ESR1), which is a mutation identified in a subset of inform the design of prospective therapeutics. Con­
patients with pancreatic cancer [94]. Interestingly, this sistent is the finding that numerous transcriptional reg­
mutation localized to the ARM domain disrupts ulators have been linked to the dysregulation of phase
ESR1–RNA interactions [7], which suggests that cancer- separation, including UTX [105], MLL4 [106], WASP
associated mutations in the RBR regions of TFs can [107], AKAP95 [108], and TAZ [109]. Also consistent is
disrupt RBP–RNA interactions. Similarly, the glyci­ the growing evidence that condensate dysregulation is
ne–arginine-rich domain that serves as a conserved RBR linked to pathogenic mutations that map to condensate-
domain in RBPs has been shown to be associated with promoting protein features including protein in­
several diseases including neurological diseases and trinsically disordered regions (IDRs) [110]. With IDRs
cancer [95,96]. Interestingly, chromatin reader BDs of contributing to the arrangement of RNA-binding mod­
the BET family members, that serve as targets for ules within an RBP, which impacts RNA-binding affinity
pharmacological agents, also dual purpose as readers of and specificity, targeting molecular condensates formed
histone PTMs and RNA [8]. Similar to the arginine-rich by aberrant RBP–RNA interactions for removal may
domain in TFs, the BDs of all BET family members leverage new cancer treatments [111,112].
consist of a basic amino acid patch that surrounds the
acetyl-binding pocket and supports interactions with In conclusion, while a growing number of chromatin-
acetylated histones [97,98]. Thus, it is possible that and transcription-regulatory proteins have been de­
these positively charged patches may facilitate BD-RNA monstrated to bind RNA, the functional implications
interactions. Supporting this potential mechanism is the remain largely unexplored. This calls for future studies
finding that positively charged patches facilitate BRDT focused on examining the functional relationships be­
BD1 [99] and BRG1 BD [100] interactions with DNA. tween RBPs and their RNAs and the mechanisms that
With the growing number of RBPs and RBRs, further ensue because these RNA interactions are formed. The
mechanistic studies that directly manipulate the RBR link between RBPs and gene control is consistent with
are needed to discern the direct (causal) functions of entering a new era of gene regulation that will largely
RNA–RBP interactions in the dysregulation of gene comprise exploring and highlighting new roles for RBPs
expression networks in various cancers. and to assess the actions of RBPs in cancer. Moreover,
the identified nexus between dysregulated RBP pro­
RNA-binding proteins as therapeutic targets duction and function promises to elicit new therapeutic
for cancer targets for the tuning of gene expression programs that
There is a growing interest in generating chemical shift disease-related networks.
probes, small-molecule ligands that can inform RBP
function and enable the discovery of pharmacological
agents. Chemical probe design and development Data
have benefited from robust methodologies, including
activity-based protein profiling (ABPP). ABPP utilizes No data were used for the research described in the ar­
small activity-based probes designed to bind or modify ticle.

Current Opinion in Genetics & Development 2024, 84:102136 www.sciencedirect.com


RNA binding proteins in cancer Avila-Lopez and Lauberth 7

Declaration of Competing Interest of PRC2 with RNA or chromatin is mutually antagonistic.


Genome Res 2016, 26:896-907.

The authors declare that they have no known competing 11. Bi X, Xu Y, Li T, Li X, Li W, Shao W, Wang K, Zhan G, Wu Z, Liu W,
et al.: RNA targets ribogenesis factor WDR43 to chromatin for
financial interests or personal relationships that could transcription and pluripotency control. Mol Cell 2019,
have appeared to influence the work reported in this 75:102-116 e109.
paper. 12. Bose DA, Donahue G, Reinberg D, Shiekhattar R, Bonasio R,
Berger SL: RNA binding to CBP stimulates histone acetylation
and transcription. Cell 2017, 168:135-149 e122.
Acknowledgements
Research in the Lauberth lab is supported by a Grant from the NIH/ 13. Cifuentes-Rojas C, Hernandez AJ, Sarma K, Lee JT: Regulatory
National Institute of General Medical Sciences (R35 GM128900), the interactions between RNA and polycomb repressive complex
Robert H. Lurie Comprehensive Cancer Center, and Harold E Eisenberg 2. Mol Cell 2014, 55:171-185.
Foundation. 14. De Silva SM, Dhiman A, Sood S, Mercedes KF, Simmons WJ,
Henen MA, Vogeli B, Dykhuizen EC, Musselman CA: PBRM1
bromodomains associate with RNA to facilitate chromatin
References and recommended reading association. Nucleic Acids Res 2023, 51:3631-3649.
Papers of particular interest, published within the period of review, have
15. Kaneko S, Son J, Bonasio R, Shen SS, Reinberg D: Nascent RNA
been highlighted as: interaction keeps PRC2 activity poised and in check. Genes
Dev 2014, 28:1983-1988.
•• of special interest
•• of outstanding interest 16. Kaneko S, Son J, Shen SS, Reinberg D, Bonasio R: PRC2 binds
active promoters and contacts nascent RNAs in embryonic
1. Baltz AG, Munschauer M, Schwanhausser B, Vasile A, Murakawa stem cells. Nat Struct Mol Biol 2013, 20:1258-1264.
Y, Schueler M, Youngs N, Penfold-Brown D, Drew K, Milek M, 17. Schwartz JC, Ebmeier CC, Podell ER, Heimiller J, Taatjes DJ,
et al.: The mRNA-bound proteome and its global occupancy Cech TR: FUS binds the CTD of RNA polymerase II and
profile on protein-coding transcripts. Mol Cell 2012, regulates its phosphorylation at Ser2. Genes Dev 2012,
46:674-690. 26:2690-2695.
2. Castello A, Fischer B, Eichelbaum K, Horos R, Beckmann BM, 18. Sigova AA, Abraham BJ, Ji X, Molinie B, Hannett NM, Guo YE,
Strein C, Davey NE, Humphreys DT, Preiss T, Steinmetz LM, et al.: Jangi M, Giallourakis CC, Sharp PA, Young RA: Transcription
Insights into RNA biology from an atlas of mammalian mRNA- factor trapping by RNA in gene regulatory elements. Science
binding proteins. Cell 2012, 149:1393-1406. 2015, 350:978-981.
3. Queiroz RML, Smith T, Villanueva E, Marti-Solano M, Monti M,
19. Song J, Gooding AR, Hemphill WO, Love BD, Robertson A, Yao L,
Pizzinga M, Mirea DM, Ramakrishna M, Harvey RF, Dezi V, et al.:
Zon LI, North TE, Kasinath V, Cech TR: Structural basis for
Comprehensive identification of RNA-protein interactions in
inactivation of PRC2 by G-quadruplex RNA. Science 2023,
any organism using orthogonal organic phase separation
381:1331-1337.
(OOPS). Nat Biotechnol 2019, 37:169-178.
20. Wang X, Paucek RD, Gooding AR, Brown ZZ, Ge EJ, Muir TW,
4. Trendel J, Schwarzl T, Horos R, Prakash A, Bateman A, Hentze
Cech TR: Molecular analysis of PRC2 recruitment to DNA in
MW, Krijgsveld J: The human RNA-binding proteome and its
chromatin and its inhibition by RNA. Nat Struct Mol Biol 2017,
dynamics during translational arrest. Cell 2019, 176:391-403
24:1028-1038.
e319.
5. Urdaneta EC, Vieira-Vieira CH, Hick T, Wessels HH, Figini D, 21. Wei C, Xiao R, Chen L, Cui H, Zhou Y, Xue Y, Hu J, Zhou B,
Moschall R, Medenbach J, Ohler U, Granneman S, Selbach M, Tsutsui T, Qiu J, et al.: RBFox2 binds nascent RNA to globally
et al.: Purification of cross-linked RNA-protein complexes by regulate polycomb complex 2 targeting in mammalian
phenol-toluol extraction. Nat Commun 2019, 10:990. genomes. Mol Cell 2016, 62:982.

6. Hentze MW, Castello A, Schwarzl T, Preiss T: A brave new world 22. Zhang Q, McKenzie NJ, Warneford-Thomson R, Gail EH, Flanigan
of RNA-binding proteins. Nat Rev Mol Cell Biol 2018, SF, Owen BM, Lauman R, Levina V, Garcia BA, Schittenhelm RB,
19:327-341. et al.: RNA exploits an exposed regulatory site to inhibit the
enzymatic activity of PRC2. Nat Struct Mol Biol 2019,
7. Oksuz O, Henninger JE, Warneford-Thomson R, Zheng MM, Erb 26:237-247.
•• H, Vancura A, Overholt KJ, Hawken SW, Banani SF, Lauman R,
et al.: Transcription factors interact with RNA to regulate 23. Frankel AD, Kim PS: Modular structure of transcription factors:
genes, e13, Mol Cell 2023, 83:2449-2463, https://doi.org/10. implications for gene regulation. Cell 1991, 65:717-719.
1016/j.molcel.2023.06.012 Epub 2023 Jul 3. 24. Lambert SA, Jolma A, Campitelli LF, Das PK, Yin Y, Albu M, Chen
n this study, the authors utilize an RNA-protein crosslinking assay (RBR- X, Taipale J, Hughes TR, Weirauch MT: The human transcription
ID) for high-resolution mapping of RBPs and RNA binding regions. The factors. Cell 2018, 172:650-665.
proteomic analyses performed in this study revealed that many se­
quence-specific TFs bind RNA. The TF RNA binding interface was 25. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM:
mapped to a non-canonical RNA binding region referred to as an ARM A census of human transcription factors: function, expression
that is highly conserved and was shown by the authors to be required for and evolution. Nat Rev Genet 2009, 10:252-263.
TF function during embryonic development. Moreover, the authors re­
vealed that TF-RNA interactions are important for increasing TF binding 26. Vann KR, Klein BJ, Kutateladze TG: Mechanistic similarities in
to DNA, which in turn contributes to fine-tuning gene regulation. recognition of histone tails and DNA by epigenetic readers.
Curr Opin Struct Biol 2021, 71:1-6.
8. Rahnamoun H, Lee J, Sun Z, Lu H, Ramsey KM, Komives EA,
Lauberth SM: RNAs interact with BRD4 to promote enhanced 27. Musselman CA, Lalonde ME, Cote J, Kutateladze TG: Perceiving
chromatin engagement and transcription activation. Nat the epigenetic landscape through histone readers. Nat Struct
Struct Mol Biol 2018, 25:687-697. Mol Biol 2012, 19:1218-1227.
9. Beltran M, Tavares M, Justin N, Khandelwal G, Ambrose J, Foster 28. Andrews FH, Strahl BD, Kutateladze TG: Insights into newly
BM, Worlock KB, Tvardovskiy A, Kunzelmann S, Herrero J, et al.: discovered marks and readers of epigenetic information. Nat
G-tract RNA removes Polycomb repressive complex 2 from Chem Biol 2016, 12:662-668.
genes. Nat Struct Mol Biol 2019, 26:899-909.
29. Long Y, Hwang T, Gooding AR, Goodrich KJ, Rinn JL, Cech TR:
10. Beltran M, Yates CM, Skalska L, Dawson M, Reis FP, Viiri K, RNA is essential for PRC2 chromatin occupancy and function
Fisher CL, Sibley CR, Foster BM, Bartke T, et al.: The interaction in human pluripotent stem cells. Nat Genet 2020, 52:931-938.

www.sciencedirect.com Current Opinion in Genetics & Development 2024, 84:102136


8 Cancer Genomics

30. Lunde BM, Moore C, Varani G: RNA-binding proteins: modular 48. Livi CM, Klus P, Delli Ponti R, Tartaglia GG: catRAPID signature:
design for efficient function. Nat Rev Mol Cell Biol 2007, identification of ribonucleoproteins and RNA-binding regions.
8:479-490. Bioinformatics 2016, 32:773-775.
31. Maris C, Dominguez C, Allain FH: The RNA recognition motif, a 49. Zhang X, Liu S: RBPPred: predicting RNA-binding proteins
plastic RNA-binding platform to regulate post-transcriptional from sequence using SVM. Bioinformatics 2017, 33:854-862.
gene expression. FEBS J 2005, 272:2118-2131.
50. Bressin A, Schulte-Sasse R, Figini D, Urdaneta EC, Beckmann
32. Pereira B, Billaud M, Almeida R: RNA-binding proteins in BM, Marsico A: TriPepSVM: de novo prediction of RNA-binding
cancer: old players and new actors. Trends Cancer 2017, proteins based on short amino acid motifs. Nucleic Acids Res
3:506-528. 2019, 47:4406-4417.
33. Choi PS, Thomas-Tikhonenko A: RNA-binding proteins of 51. Sharma D, Zagore LL, Brister MM, Ye X, Crespo-Hernandez CE,
COSMIC importance in cancer. J Clin Investig (18) 2021, Licatalosi DD, Jankowsky E: The kinetic landscape of an RNA-
131:e151627. binding protein in cells. Nature 2021, 591:152-156.
34. Shi DL, Cheng XN, Saquet A, Grifone R: Emerging roles of RNA- 52. Benoit Bouvrette LP, Wang X, Boulais J, Kong J, Syed EU, Blue
binding proteins in inner ear hair cell development and SM, Zhan L, Olson S, Stanton R, Wei X, et al.: RBP Image
regeneration. Int J Mol Sci (20) 2022, 23:12393. Database: a resource for the systematic characterization of
35. Corley M, Burns MC, Yeo GW: How RNA-binding proteins the subcellular distribution properties of human RNA binding
interact with RNA: molecules and mechanisms. Mol Cell 2020, proteins. Nucleic Acids Res 2023, 51:D1549-D1557.
78:9-29. 53. Quinodoz SA, Jachowicz JW, Bhat P, Ollikainen N, Banerjee AK,
36. Gebauer F, Schwarzl T, Valcarcel J, Hentze MW: RNA-binding • Goronzy IN, Blanco MR, Chovanec P, Chow A, Markaki Y, et al.:
proteins in human genetic disease. Nat Rev Genet 2021, RNA promotes the formation of spatial compartments in the
22:185-198. nucleus. Cell 2021, 184:5775-5790 e5730.
Importantly, this study provides new insights into RNA involvement in
37. Nussbacher JK, Tabet R, Yeo GW, Lagier-Tourenne C: the formation of molecular condensates. The authors describe an im­
Disruption of RNA metabolism in neurological diseases and proved method that they refer to as RNA & DNA SPRITE (RD-SPRITE),
emerging therapeutic interventions. Neuron 2019, which enables the simultaneous mapping of RNA-RNA, RNA-DNA, and
102:294-320. DNA-DNA contacts in the cell. Using this powerful methodology, the
authors unveil that nascent RNA, even that which is produced at low
38. Beckmann BM, Horos R, Fischer B, Castello A, Eichelbaum K, levels can drive and organize functional compartments comprised of
Alleaume AM, Schwarzl T, Curk T, Foehr S, Huber W, et al.: The regulatory factors within the nucleus to regulate gene expression.
RNA-binding proteomes from yeast to man harbour
conserved enigmRBPs. Nat Commun 2015, 6:10127. 54. Yan J, Dutta B, Hee YT, Chng WJ: Towards understanding of
PRC2 binding to RNA. RNA Biol 2019, 16:176-184.
39. Conrad T, Albrecht AS, de Melo Costa VR, Sauer S, Meierhofer D,
Orom UA: Serial interactome capture of the human cell 55. Zhao J, Ohsumi TK, Kung JT, Ogawa Y, Grau DJ, Sarma K, Song
nucleus. Nat Commun 2016, 7:11212. JJ, Kingston RE, Borowsky M, Lee JT: Genome-wide
identification of polycomb-associated RNAs by RIP-seq. Mol
40. Kwon SC, Yi H, Eichelbaum K, Fohr S, Fischer B, You KT, Castello Cell 2010, 40:939-953.
A, Krijgsveld J, Hentze MW, Kim VN: The RNA-binding protein
repertoire of embryonic stem cells. Nat Struct Mol Biol 2013, 56. Davidovich C, Zheng L, Goodrich KJ, Cech TR: Promiscuous
20:1122-1130. RNA binding by Polycomb repressive complex 2. Nat Struct
Mol Biol 2013, 20:1250-1257.
41. Perez-Perri JI, Rogell B, Schwarzl T, Stein F, Zhou Y, Rettel M,
Brosig A, Hentze MW: Discovery of RNA-binding proteins and 57. Rosenberg M, Blum R, Kesner B, Aeby E, Garant JM, Szanto A,
characterization of their dynamic responses by enhanced Lee JT: Motif-driven interactions between RNA and PRC2 are
RNA interactome capture. Nat Commun 2018, 9:4408. rheostats that regulate transcription elongation. Nat Struct
42. Bao X, Guo X, Yin M, Tariq M, Lai Y, Kanwal S, Zhou J, Li N, Lv Y, Mol Biol 2021, 28:103-117.
Pulido-Quetglas C, et al.: Capturing the interactome of newly 58. Ullah I, Tholken C, Zhong Y, John M, Rossbach O, Lenz J,
transcribed RNA. Nat Methods 2018, 15:213-220. Gossringer M, Nist A, Albert L, Stiewe T, et al.: RNA inhibits dMi-
43. He C, Sidoli S, Warneford-Thomson R, Tatomer DC, Wilusz JE, 2/CHD4 chromatin binding and nucleosome remodeling. Cell
Garcia BA, Bonasio R: High-resolution mapping of RNA- Rep 2022, 39:110895.
binding regions in the nuclear proteome of embryonic stem
59. Oh HJ, Aguilar R, Kesner B, Lee HG, Kriz AJ, Chu HP, Lee JT: Jpx
cells. Mol Cell 2016, 64:416-430. • RNA regulates CTCF anchor site selection and formation of
44. Huang R, Han M, Meng L, Chen X: Capture and identification of chromosome loops. Cell 2021, 184:6157-6173 e6124.
RNA-binding proteins by using click Chemistry-assisted RNA- This study provides new functional insights into a chromatin archi­
interactome Capture (CARIC) strategy. J Vis Exp 2018, 140, tectural role for the Jpx RNA that has been classically known for its role
https://doi.org/10.3791/58580 e58580. in controlling X-inactivation. The authors report that the Jpx RNA reg­
ulates CTCF binding on a global scale by releasing CTCF from DNA at
45. Caudron-Herger M, Wassmer E, Nasa I, Schultz AS, Seiler J, low-affinity CTCF binding sites. This regulation of CTCF by Jpx has
Kettenbach AN, Diederichs S: Identification, quantification and important implications for the displacement of chromosome loops and
bioinformatic analysis of RNA-dependent proteins by RNase the formation of new loops that are associated with the downregulation
treatment and density gradient ultracentrifugation using of genes involved in development and differentiation.
R-DeeP. Nat Protoc 2020, 15:1338-1370.
60. Saldana-Meyer R, Rodriguez-Hernaez J, Escobar T, Nishana M,
46. Jin W, Brannan KW, Kapeli K, Park SS, Tan HQ, Gosztyla ML, Jacome-Lopez K, Nora EP, Bruneau BG, Tsirigos A, Furlan-
• Mujumdar M, Ahdout J, Henroid B, Rothamel K, et al.: HydRA: Magaril M, Skok J, et al.: RNA interactions are essential for
deep-learning models for predicting RNA-binding capacity CTCF-mediated genome organization. Mol Cell 2019,
from protein interaction association context and protein 76:412-422 e415.
sequence. Mol Cell 2023, 83:2595-2611 e2511.
In this study, the authors demonstrate the power of implementing a ma­ 61. Saldana-Meyer R, Gonzalez-Buendia E, Guerrero G, Narendra V,
chine-learning-based algorithm referred to as hybrid ensemble classifier Bonasio R, Recillas-Targa F, Reinberg D: CTCF regulates the
(HydRA). By incorporating both intermolecular protein context and se­ human p53 gene through direct interaction with its natural
quence-level information, this algorithm guides predictions of unexpected antisense transcript, Wrap53. Genes Dev 2014, 28:723-734.
RBPs and the regions that are involved in RBP-RNA interactions.
62. Hansen AS, Hsieh TS, Cattoglio C, Pustova I, Saldana-Meyer R,
47. Kumar M, Gromiha MM, Raghava GP: SVM based prediction of Reinberg D, Darzacq X, Tjian R: Distinct classes of chromatin
RNA-binding proteins using binding residues and loops revealed by deletion of an RNA-binding region in CTCF.
evolutionary information. J Mol Recognit 2011, 24:303-313. Mol Cell 2019, 76:395-411 e313.

Current Opinion in Genetics & Development 2024, 84:102136 www.sciencedirect.com


RNA binding proteins in cancer Avila-Lopez and Lauberth 9

63. Weber AP: SC: evidence for and against liquid-liquid phase 81. Gong S, Li Q, Jeter CR, Fan Q, Tang DG, Liu B: Regulation of
separation in the nucleus. Noncoding RNA (4) 2019, 5:50. NANOG in cancer cells. Mol Carcinog 2015, 54:679-687.
64. Pandya-Jones A, Markaki Y, Serizay J, Chitiashvili T, Mancia Leon 82. Jeter CR, Badeaux M, Choy G, Chandra D, Patrawala L, Liu C,
WR, Damianov A, Chronis C, Papp B, Chen CK, McKee R, et al.: A Calhoun-Davis T, Zaehres H, Daley GQ, Tang DG: Functional
protein assembly mediates Xist localization and gene evidence that the self-renewal gene NANOG regulates human
silencing. Nature 2020, 587:145-151. tumor development. Stem Cells 2009, 27:993-1005.

65. Elguindy MM, Mendell JT: NORAD-induced Pumilio phase 83. Lin T, Ding YQ, Li JM: Overexpression of Nanog protein is
separation is required for genome stability. Nature 2021, associated with poor prognosis in gastric adenocarcinoma.
595:303-308. Med Oncol 2012, 29:878-885.

66. Nair SJ, Yang L, Meluzzi D, Oh S, Yang F, Friedman MJ, Wang S, 84. Meng HM, Zheng P, Wang XY, Liu C, Sui HM, Wu SJ, Zhou J,
•• Suter T, Alshareedah I, Gamliel A, et al.: Phase separation of Ding YQ, Li J: Over-expression of Nanog predicts tumor
ligand-activated enhancers licenses cooperative progression and poor prognosis in colorectal cancer. Cancer
chromosomal enhancer assembly. Nat Struct Mol Biol 2019, Biol Ther 2010, 9:295-302.
26:193-203.
85. Du Z, Song X, Yan F, Wang J, Zhao Y, Liu S: Genome-wide
Here, the authors provide one of the first lines of evidence that RNAs
transcriptional analysis of BRD4-regulated genes and
drive phase separation to facilitate regulatory control over RBP activ­
pathways in human glioma U251 cells. Int J Oncol 2018,
ities. By focusing on lncRNA NORAD that is activated by DNA damage,
52:1415-1426.
the authors reveal mechanistic insights into NORAD regulation of
Pumilio RBPs, PUM1, and PUM2 in maintaining genome stability. 86. French CA: Small-molecule targeting of BET proteins in
Specifically, the authors reveal that NORAD effectively competes with cancer. Adv Cancer Res 2016, 131:21-58.
other cellular RNAs for interactions with PUM proteins by forming bio­
molecular condensates. This RNA-dependent mechanism sequesters 87. Laugesen A, Hojfeldt JW, Helin K: Role of the Polycomb
and inhibits PUM proteins, which has important implications for genome Repressive Complex 2 (PRC2) in transcriptional regulation
maintenance. and cancer. Cold Spring Harb Perspect Med (9) 2016, 6:a026575.
67. Markaki Y, Chong JG, Wang Y, Jacobson EC, Luong C, Tan SYX, 88. Neelamraju Y, Gonzalez-Perez A, Bhat-Nakshatri P, Nakshatri H,
Jachowicz JW, Strehle M, Maestrini D, Banerjee AK, et al.: Xist Janga SC: Mutational landscape of RNA-binding proteins in
nucleates local protein gradients to propagate silencing human cancers. RNA Biol 2018, 15:115-129.
across the X chromosome. Cell 2021, 184:6212.
89. Gonzalez-Perez A, Lopez-Bigas N: Functional impact bias
68. Lin Y, Protter DS, Rosen MK, Parker R: Formation and reveals cancer drivers. Nucleic Acids Res 2012, 40:e169.
maturation of phase-separated liquid droplets by RNA-
binding proteins. Mol Cell 2015, 60:208-219. 90. Castello A, Fischer B, Hentze MW, Preiss T: RNA-binding
proteins in Mendelian disease. Trends Genet 2013, 29:318-327.
69. Shao W, Bi X, Pan Y, Gao B, Wu J, Yin Y, Liu Z, Peng M, Zhang W,
Jiang X, et al.: Phase separation of RNA-binding protein 91. Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M,
promotes polymerase binding and transcription. Nat Chem Zhang Q, McMichael JF, Wyczalkowski MA, et al.: Mutational
Biol 2022, 18:70-80. landscape and significance across 12 major cancer types.
Nature 2013, 502:333-339.
70. Kang D, Lee Y, Lee JS: RNA-binding proteins in cancer:
92. Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla
functional and therapeutic perspectives. Cancers (9) 2020,
S, Gu B, Hart J, Hoffman D, Jang W, et al.: ClinVar: improving
12:2699.
access to variant interpretations and supporting evidence.
71. Chang K, Creighton CJ, Davis C, Donehower L, Drummond J, Nucleic Acids Res 2018, 46:D1062-D1067.
Wheeler D, Ally A, Balasundaram M, Birol I, Butterfield YSN, et al.: 93. Stenson PD, Mort M, Ball EV, Chapman M, Evans K, Azevedo L,
The cancer genome Atlas Pan-Cancer analysis project. Nat Hayden M, Heywood S, Millar DS, Phillips AD, et al.: The Human
Genet 2013, 45:1113-1120. Gene Mutation Database (HGMD((R))): optimizing its use in a
72. Consortium GT: The Genotype-Tissue Expression (GTEx) clinical diagnostic or research setting. Hum Genet 2020,
project. Nat Genet 2013, 45:580-585. 139:1197-1207.
94. Boldes T, Merenbakh-Lamin K, Journo S, Shachar E, Lipson D,
73. Bensberg M, Rundquist O, Selimovic A, Lagerwall C, Benson M,
Yeheskel A, Pasmanik-Chor M, Rubinek T, Wolf I: R269C variant
Gustafsson M, Vogt H, Lentini A, Nestor CE: TET2 as a tumor
of ESR1: high prevalence and differential function in a subset
suppressor and therapeutic target in T-cell acute
of pancreatic cancers. BMC Cancer 2020, 20:531.
lymphoblastic leukemia. Proc Natl Acad Sci USA (34) 2021,
118:e2110758118. 95. Doron-Mandel E, Koppel I, Abraham O, Rishal I, Smith TP,
Buchanan CN, Sahoo PK, Kadlec J, Oses-Prieto JA, Kawaguchi
74. Lio CJ, Yuita H, Rao A: Dysregulation of the TET family of R, et al.: The glycine arginine-rich domain of the RNA-binding
epigenetic regulators in lymphoid and myeloid malignancies. protein nucleolin regulates its subcellular localization. EMBO
Blood 2019, 134:1487-1497. J 2021, 40:e107158.
75. Wang L, Ozark PA, Smith ER, Zhao Z, Marshall SA, Rendleman 96. Jarvelin AI, Noerenberg M, Davis I, Castello A: The new (dis)
EJ, Piunti A, Ryan C, Whelan AL, Helmin KA, et al.: TET2 order in RNA regulation. Cell Commun Signal 2016, 14:9.
coactivates gene expression through demethylation of
enhancers. Sci Adv 2018, 4:eaau6986. 97. Filippakopoulos P, Picaud S, Mangos M, Keates T, Lambert JP,
Barsyte-Lovejoy D, Felletar I, Volkmer R, Muller S, Pawson T,
76. Huang M, Weiss WA: Neuroblastoma and MYCN. Cold Spring et al.: Histone recognition and large-scale structural analysis
Harb Perspect Med 2013, 3:a014415. of the human bromodomain family. Cell 2012, 149:214-231.
77. Rickman DS, Schulte JH, Eilers M: The expanding world of N- 98. Fujisawa T, Filippakopoulos P: Functions of bromodomain-
MYC-driven tumors. Cancer Discov 2018, 8:150-163. containing proteins and their roles in homeostasis and
cancer. Nat Rev Mol Cell Biol 2017, 18:246-262.
78. Liu R, Shi P, Wang Z, Yuan C, Cui H: Molecular mechanisms of
MYCN dysregulation in cancers. Front Oncol 2020, 10:625332. 99. Miller TC, Simon B, Rybin V, Grotsch H, Curtet S, Khochbin S,
Carlomagno T, Muller CW: A bromodomain-DNA interaction
79. Parreno V, Martinez AM, Cavalli G: Mechanisms of Polycomb facilitates acetylation-dependent bivalent nucleosome
group protein function in cancer. Cell Res 2022, 32:231-253. recognition by the BET protein BRDT. Nat Commun 2016,
7:13855.
80. Piunti A, Shilatifard A: The roles of Polycomb repressive
complexes in mammalian development and cancer. Nat Rev 100. Morrison EA, Sanchez JC, Ronan JL, Farrell DP, Varzavand K,
Mol Cell Biol 2021, 22:326-345. Johnson JK, Gu BX, Crabtree GR, Musselman CA: DNA binding

www.sciencedirect.com Current Opinion in Genetics & Development 2024, 84:102136


10 Cancer Genomics

drives the association of BRG1/hBRM bromodomains with 105. Shi B, Li W, Song Y, Wang Z, Ju R, Ulman A, Hu J, Palomba F,
nucleosomes. Nat Commun 2017, 8:16080. Zhao Y, Le JP, et al.: UTX condensation underlies its tumour-
suppressive activity. Nature 2021, 597:726-731.
101. Liu Y, Patricelli MP, Cravatt BF: Activity-based protein profiling:
the serine hydrolases. Proc Natl Acad Sci USA 1999, 106. Fasciani A, D’Annunzio S, Poli V, Fagnocchi L, Beyes S, Michelatti
96:14694-14699. D, Corazza F, Antonelli L, Gregoretti F, Oliva G, et al.: MLL4-
associated condensates counterbalance Polycomb-mediated
102. Speers AE, Cravatt BF: Activity-Based Protein Profiling (ABPP) nuclear mechanical stress in Kabuki syndrome. Nat Genet
and Click Chemistry (CC)-ABPP by MudPIT mass 2020, 52:1397-1411.
spectrometry. Curr Protoc Chem Biol 2009, 1:29-41.
103. Lazear MR, Remsberg JR, Jaeger MG, Rothamel K, Her HL, 107. Yuan B, Zhou X, Suzuki K, Ramos-Mandujano G, Wang M,
• DeMeester KE, Njomen E, Hogg SJ, Rahman J, Whitby LR, Tehseen M, Cortes-Medina LV, Moresco JJ, Dunn S, Hernandez-
Benitez R, et al.: Wiskott-Aldrich syndrome protein forms
et al.: Proteomic discovery of chemical probes that perturb
protein complexes in human cells. Mol Cell 2023, nuclear condensates and regulates alternative splicing. Nat
83:1725-1742 e1712. Commun 2022, 13:3646.
In this study, the authors describe a novel screening platform used for the 108. Li W, Hu J, Shi B, Palomba F, Digman MA, Gratton E, Jiang H:
discovery of chemical probes with therapeutic relevance. The functional Biophysical properties of AKAP95 protein condensates regulate
screen that is described in their study is focused on disrupting native splicing and tumorigenesis. Nat Cell Biol 2020, 22:960-972.
protein complexes. Specifically, through a combination of size exclusion
chromatography and cysteine-directed ABPP, the authors reveal the ef­ 109. Lu Y, Wu T, Gutman O, Lu H, Zhou Q, Henis YI, Luo K: Phase
fects of electrophilic compounds on altering native protein complexes. This separation of TAZ compartmentalizes the transcription
function-based screening platform has great potential for expanding the machinery to promote gene expression. Nat Cell Biol 2020,
types of proteins that can be targeted and will undeniably aid in the dis­ 22:453-464.
covery of new chemical probes for the ‘ undruggable ’ proteome.
110. Banani SF, Afeyan LK, Hawken SW, Henninger JE, Dall’Agnese A,
104. Kathman SG, Koo SJ, Lindsey GL, Her HL, Blue SM, Li H, Jaensch Clark VE, Platt JM, Oksuz O, Hannett NM, Sagi I, et al.: Genetic
•• S, Remsberg JR, Ahn K, Yeo GW, et al.: Remodeling oncogenic variation associated with condensate dysregulation in
transcriptomes by small molecules targeting NONO. Nat disease. Dev Cell 2022, 57:1776-1788 e1778.
Chem Biol 2023, 19:825-836.
This study performed activity-based profiling analysis of a collection of 111. Chandra B, Michmerhuizen NL, Shirnekhi HK, Tripathi S, Pioso
electrophilic compounds that resulted in the identification of effective com­ BJ, Baggett DW, Mitrea DM, Iacobucci I, White MR, Chen J, et al.:
pounds that suppress the expression of oncogenic TF, androgen receptor. Phase separation mediates NUP98 fusion oncoprotein
The target of the AR-inhibiting compounds was identified as the RBP, leukemic transformation. Cancer Discov 2022, 12:1152-1169.
NONO. Further mechanistic insights revealed that the NONO ligands lead to
alterations in the transcriptome of cancer cells by stabilizing NONO-mRNA 112. Boija A, Klein IA, Young RA: Biomolecular condensates and
interactions that lead to aberrant accumulation of NONO in perinuclear cancer. Cancer Cell 2021, 39:174-192.
bodies.

Current Opinion in Genetics & Development 2024, 84:102136 www.sciencedirect.com

You might also like