Abstract
Actin is a highly conserved protein in mammals. The actin dynamics is regulated by actin-binding proteins and actin-related proteins. Nuclear actin and these regulatory proteins participate in multiple nuclear processes, including chromosome architecture organization, chromatin remodeling, transcription machinery regulation, and DNA repair. It is well known that the dysfunctions of these processes contribute to the development of cancer. Moreover, emerging evidence has shown that the deregulated actin dynamics is also related to cancer. This chapter discusses how the deregulation of nuclear actin dynamics contributes to tumorigenesis via such various nuclear events.
Keywords: nuclear actin, chromosome architecture, chromatin remodeling, BAF complex, INO80 complex, transcription machinery, RNA polymerase, DNA repair, gene expression, cancer
1. Introduction
Actin is a highly conserved protein family in mammals. It participates in multiple cellular processes, including muscle contraction, cell motility, cell division, organelle movement, material transportation, signal transduction, cell junction establishment, and cell shape maintenance [1–3]. The actin family is classified into three types of isoforms in humans: α-actins (ACTA1, ACTA2, and ACTC1), β-actins (ACTB and ACTBL2), and γ-actins (ATCG1 and ACTG2) (Table 1) [2]. They share 93% amino acid identity, with slight length variations at N-terminus [2, 4].
Table 1.
Actin genes.
| Gene symbol | Gene name | Expressing cells |
|---|---|---|
| ACTA1 | Actin alpha 1, skeletal muscle | Skeletal muscle cells |
| ACTA2 | Actin alpha 2, smooth muscle | Vascular smooth muscle cells |
| ACTB | Actin beta | Ubiquitous in non-muscle cells |
| ACTBL2 | Actin beta like 2 | Ubiquitous in non-muscle cells |
| ACTC1 | Actin alpha cardiac muscle 1 | Cardiac muscle cells |
| ACTG1 | Actin gamma 1 | Ubiquitous in non-muscle cells |
| ACTG2 | Actin gamma 2, smooth muscle | Enteric smooth muscle cells |
1.1. Actin dynamics in the cytoplasm
In the cytoplasm, actin exists as either a globular monomer (G-actin) or a filamentous polymer (F-actin) [1]. The G-actin structure is divided into two lobes by a deep cleft in the middle (Figure 1A). The upper cleft between subdomains 2 and 4 is the binding site for ATP, ADP, and cations. The lower cleft between subdomains 1 and 3 is the target site for actin-binding proteins (ABPs) [1, 4]. F-actin is a linear chain of G-actins [1]. It is the basic building structure of microfilaments with two ends, the (+) end (barbed end) and the (−) end (pointed end).
Figure 1. Cytoplasmic actin dynamics and nuclear actin visualization.

(A) Structure of ACTA1 (PDB: 4PKG). (B) The process of actin polymerization with three phases: the nucleation phase, the elongation phase, and the steady phase. (C) The treadmilling of F-actin. The rates of G-actin assembly and disassembly depend on the concentrations of free ATP-G-actin on both ends. When the concentration of free ATP-G-actin is between 0.12 µM and 0.6 µM, the (+) end is elongated, and the (−) end is shortened, demonstrating the treadmilling of F-actin. (D) The regulation of actin turnover by actin-binding proteins (ABPs). In the cofilin cycle, cofilin binds to the (−) end of F-actin containing ADP-actin, inducing them to fragment and thus enhancing depolymerization. In the profilin cycle, profilin binds ADP-G-actin and catalyzes the exchange of ADP for ATP. The ATP-G-actin-profilin complex delivers actin to the (+) end with dissociation and recycling of profilin. (E) Live-cell imaging of nuclear actin by stably expressing actin-chromobody-GFP-NLS. The colon epithelial cell line (CCD841CoN) shows high levels of nuclear F-actin. Mucinous colorectal cancer cell line (LS174T) exhibits the reduced nuclear F-actin.
In a physiological condition, G-actin and F-actin are under a dynamical equilibrium between polymerization and depolymerization (Figure 1B) [5]. At the initial polymerization phase, ATP-bound G-actins combine into an oligomer as a nucleus with actin-nucleating proteins, like formin and actin-related protein 2/3 (ARP2/3) complex (Table 2), under the existence of Mg2+, K+, or Na+. Then, the nucleus of actin polymerization rapidly increases in length at both ends. This elongation phase is powered by the hydrolysis of ATP-G-actin, which transforms to ADP-G-actin and releases inorganic phosphate (Pi). Finally, a steady conformational state between F-actin and G-actin is reached, with no further elongation of F-actin. Briefly, when ATP is bound to G-actin with the existence of cations, G-actin polymerizes into F-actin. This process is reversible when the free ATP-G-actin amount or the cation strength is low.
Table 2.
Actin dynamics-related protein families.
| Actin-nucleating proteins | Actin-binding proteins [6, 28] | Actin-related proteins [28, 251] | Capping proteins [7] | Capping protein regulators [8] |
|---|---|---|---|---|
| ARP2/3 complex [252] | Cofilin [253] | ACTL6A | ADD1 | CARMIL1 |
| Formin [254] | Gelsolin [255] | ACTL6B | ADD2 | CARMIL2 |
| Spire [256] | Profilin [257] | ACTL7A | CAPG | CARMIL3 |
| Thymosin beta-4 [258] | ACTL7B | CAPZA1 | CD2AP | |
| ACTL8 | CAPZA2 | CRACD [10] | ||
| ACTL9 | CAPZA3 | IQANK1 | ||
| ACTL10 | CAPZB | MTPN | ||
| ACTRT1 | PLEKHO1 | |||
| ACTRT2 | RCSD1 | |||
| ACTRT3 | SH3KBP1 | |||
| ACTR1A | WASHC2A | |||
| ACTR1B | WASHC2C | |||
| ACTR2 | ||||
| ACTR3 | ||||
| ACTR3B | ||||
| ACTR3C | ||||
| ACTR5 | ||||
| ACTR6 | ||||
| ACTR8 | ||||
| ACTR10 |
The polymerization process that the (+) end grows whereas the (−) end loses subunits is called treadmilling, which drives the intracellular movement of F-actin (Figure 1C). Treadmilling is accelerated by ABPs (Table 2), such as cofilin and profilin (Figure 1D) [6]. Cofilin binds to ADP-G-actin at the (−) end to enhance its dissociation from the chain. Profilin binds to free ADP-G-actin and catalyzes the exchange of ADP for ATP, delivering ATP-G-actin back to the (+) end.
The actin dynamics are further regulated by capping proteins (CPs) (Table 2) binding to the (+) end, and tropomodulin, which binds to the (−) end, inhibiting the uncontrolled polymerization and depolymerization [7]. As a new actin polymerization regulator (Table 2), CRACD (capping protein-inhibiting regulator of actin dynamics) has been recently identified as a tumor suppressor in colorectal cancer, indicating the implication of deregulated actin dynamics in cancer [8–10].
1.2. Actin visualization
To visualize actin-related events, several actin-detecting probes have been generated by fusing the fluorophore or fluorescent protein to actin (actin-GFP), actin antibody (actin-chromobody), actin-modulating drugs (phalloidin, SiR-actin, and SPY-actin), or ABPs (F-tractin, Lifeact, and UtrCH) (Table 3) [11, 12]. Unlike live-cell imaging, optimal fixation conditions are crucial to represent physiologically relevant actin dynamics. Fixation by methanol, ethanol, or acetone destroys the native quaternary structure of F-actin, which creates dotted artifacts in fixed cells [13, 14]. Paraformaldehyde has been validated as the best fixation solution to retain actin structure [15, 16]. Furthermore, to avoid ethanol during the dehydration and hydration of paraffin-embedded tissues, cryo-section is the preferred choice to stain F-action by phalloidin in tissue samples [17].
Table 3.
Actin-detecting probes.
| Probe | Description | Mechanisms | Pitfalls |
|---|---|---|---|
| Actin-chromobody [259] | A DNA plasmid encoding an anti-actin VHH nanobody fused to a fluorescent protein. | Fluorescent-labeled nanobody labels endogenous actin by antigen-antibody reaction. | High background when transient transfected into cells. Signal quenching after cell fixation. |
| Actin-GFP [260] | A DNA plasmid encoding human actin fused to GFP. | Ectopic expression of fluorescent-labeled actin into cells. | Interfere with the physiological actin dynamics. |
| F-tractin [261] | A DNA plasmid encoding N-terminus 10–52 AA peptides of rat inositol-trisphosphate 3-kinase (Itpk) [262]. | Itpk has a F-actin specifically binding domain at the N-terminus 1–66 AA region [263]. | Inhibit ABPs binding to F-actin. |
| Lifeact [264] | A DNA plasmid encoding N-terminus 1–17 AA peptides of yeast ABP140 [265]. | Lifeact binds to G-actin with an affinity 10-fold higher than F-actin. | Alter F-actin organization [266]. |
| Phalloidin [267] | Bicyclic heptapeptide from death cap mushroom. | Phalloidin specifically binds at the interface between subunits of F-actin, locking the F-actin structure and preventing depolymerization. | Prevent F-actin depolymerization. Cytotoxicity. |
| SiR-actin [268] | Silicon-rhodamine (SiR) conjugated to desbromo-desmethyl-jasplakinolide. | SiR is a fluorophore [269]. Jasplakinolide binds at the interface of G-actin oligomers at the nucleation phase [270, 271]. | Enhance F-actin polymerization. Cytotoxicity. |
| SPY-actin | Improved version of the SiR-actin by utilizing SPY dyes instead of SiR. | ||
| UtrCH [272] | A DNA plasmid encoding N-terminus 1–261 AA peptides of human utrophin. | The N-terminus of utrophin has calponin-homology (CH) domains, which specifically binds to F-actin [273]. | Alter F-actin organization. |
The presence of nuclear actin was first described in the calf thymus cells in 1963 [18]. However, due to the lack of nucleus-permeable actin probes, the existence of nuclear actin has been in debate for decades [19]. Initially, nuclear actin was considered to be an artifact from cytoplasmic actin contamination [20]. Thanks to the development of microscopy and actin-detecting constructs fused with nuclear localization signal (NLS), convincing evidence of nuclear actin have been introduced [21–24]. For example, the endogenous nuclear F-actin and G-actin are detectable by using the actin-chromobody-GFP-NLS in the normal colon epithelial cells and mucinous colorectal cancer cells (Figure 1E).
1.3. Nuclear actin dynamics
Actin protein is constantly and rapidly shuttled into and out of the nucleus via the nuclear pore complex (NPC) to maintain the actin balance between the cytoplasm and the nucleus [25]. By active transport, G-actin is imported into the nucleus by importin-9 (IPO9) in a complex with cofilin and exported out by exportin-6 (XPO6) coupled with profilin [26]. ABPs and actin-related proteins (ARPs) also exist in the nucleus (Table 2) [27–29]. Additionally, SUMOylation, a type of post-translational modification, of actin interferes with the actin-XPO6 interaction to retain actin in the nucleus [30].
In this chapter, we focus on the roles of nuclear actin in various biological processes, including chromosome architecture organization, chromatin remodeling, transcription machinery regulation, and DNA repair, in the aspect of cancer [27, 31–34].
2. Nuclear Actin and Chromosome Architecture
2.1. The hierarchy of chromosome architecture
The two-meter length of mammalian DNA is organized into a highly condensed chromosome at a supreme level of hierarchy [35]. In the classical model, chromatin is described as an alternation of euchromatin and heterochromatin. Euchromatin is loose and transcriptionally active with enrichments of specific histone modifications (H3K4me3, H3K36me3, and H3K79me3), mainly located in the nuclear interior [36]. Heterochromatin is dense and transcriptionally repressed, marked by repressive histone modifications (H3K9me2, H3K9me3, and H3K27me3), located at the nuclear periphery [36, 37].
Based on the genome-wide chromosome conformation capture sequencing (Hi-C-seq) [38], the chromosome architectural hierarchy is divided into the nucleosomes, chromatin fibers, chromatin loops, topologically associating domains (TADs), chromosome compartments, and chromosome territories in a decreasing resolution (Figure 2) [39–42].
Figure 2. The hierarchy of chromosome architecture.

DNA chain (<10 nm) wraps around the histone octamer to form the nucleosome (10–30 nm). The beads-on-a-string arrays of nucleosomes coil into chromatin fibers (30 nm). The liner chromatin fibers loop out to form the chromatin loops (30–100 nm). Topologically adjacent and preferentially interacting chromatin loops construct into topologically associating domains (TADs) (100–500 nm). Topologically interacting TADs form the chromosome compartments (500–1000 nm). Chromosome territories (1000–2000 nm) are the discrete space for each chromosome in the nucleus.
Nucleosomes (10–30 nm):
Nucleosome is the fundamental unit of chromatin, containing a histone octamer (two copies of each H2A, H2B, H3, H4) wrapped with 147 bp DNA [43]. Adjacent nucleosomes are connected by the linker DNA associated with the linker histone protein (H1 or H5) to form a beads-on-a-string array at a diameter of 11 nm [43].
Chromatin fibers (30 nm):
The beads-on-a-string arrays coil into a 30 nm diameter helical structure known as the chromatin fiber under the shape of solenoid or zig-zag [44].
Chromatin loops (30–100 nm):
It is suggested that, on the linear chromatin fiber, the distal enhancers physically bind to the promoters of target genes at spatial proximity to initiate the transcription by looping out to form the chromatin loops [45, 46]. This process is mediated by anchoring several proteins, including transcription factors (TFs), RNA polymerase II, CCCTC-binding factor (CTCF), cohesin, and mediator [47]. These loops form the active chromatin hub (ACH) spanned by CTCF-CTCF homodimer with point-to-point interactions between loci [46]. The well-appreciated example of ACH is that the long-range cis-regulatory elements of hemoglobin subunit beta (HBB) interact strongly and facilitate transcription by forming the chromatin loops in erythroid cells [48]. The loops that are not spanned by CTCFs are called ordinary domains [41]. The formation of chromatin loops is regulated by the loop extrusion process [49]. In this process, the cohesin complex binds to a chromatin fiber and reels it to form the loops [50]. With the increasing density of loops, more advanced structures like TADs are formed.
TADs (100–500 nm):
TAD is a chromatin region formed by bunches of topologically adjacent and preferentially interacting chromatin loops and ordinary domains [51]. TADs are separated by boundaries that are formed by CTCFs and cohesins [42]. Inside of a specific TAD or between similar TADs, chromatin loops interact with each other more frequently than sequences in the adjacent non-TAD regions [39, 42]. TADs were first identified as sub-chromosomal domains in the 1980s, and validated by Hi-C-seq, appearing as individual triangles on the heatmap [40, 52]. They can be further subdivided into smaller ones called subTADs when increasing the resolution [53].
Chromosome compartments (500–1000 nm):
The chromosome is mainly compartmented by TADs [39]. It can be defined as A (euchromatic) or B (heterochromatic) by the principal component analysis of Hi-C-seq [54]. A is in a transcriptionally active state, while B is in a repressed state. Chromosome compartments can switch between each other in a cell-type-specific manner [55].
Chromosome territories (1000–2000 nm):
Chromosome territory is the discrete space each chromosome occupies in the nucleus [56].
Recently, a new technique named targeted chromatin capture (T2C) combining a simulation method [57, 58] proposed a new model of chromosome architectural hierarchy that the chromatin quasi-fibers (in a length of 80–120 nm) fold into stable chromatin loops and cluster into aggregate/rosette-like sub-chromosomal domains [52, 57, 59].
2.2. Organizing chromosome architecture by nuclear actin
Accumulating evidence suggests that nuclear actin plays a vital role in organizing chromosome architecture. For example, knock-out (KO) of the Actb gene encoding mouse β-actin upregulates the intensity of heterochromatin in the nuclear interior [60]. In humans, a higher proportion of heterochromatin was observed at the mitotic exit when nuclear F-actin polymerization was inhibited [61]. Furthermore, the impaired nuclear F-actin increases the degree but reduces the dynamics of chromatin compaction in the postmitotic nucleus, whereas the enhanced nuclear F-actin does reversely [61, 62]. The underlying mechanisms of how nuclear actin modulates chromosome architecture are described below.
Actin and nuclear lamina:
Nuclear lamina affects chromosome architecture [46, 63]. Heterochromatin binds to the nuclear lamina at the nuclear periphery to form the region called lamina-associated domains (LADs) [63], whereas euchromatin loops out into the nucleus interior. When cells differentiate, constitutive LADs remain attached to the lamina, whereas facultative LADs become detached, and the genes they contain become actively transcribed. After mitosis, LADs relocate to the nuclear periphery. It was reported that the perturbation of perinuclear actin can deform the nuclear lamina integrity and consequently alter the heterochromatin localization and enhance chromatin condensation [64, 65]. This deformation may be via the linker of nucleoskeleton and cytoskeleton (LINC) complex, which couples the cytoplasmic actin with the nuclear lamina, and does the mechanotransduction [66]. Though the interactions between nuclear actin and nuclear lamina have not been elucidated, the chromosome architecture might likely be organized by nuclear actin via nuclear lamina based on the close connection between cytoplasmic and nuclear actin.
Nuclear actin and chromatin remodeling (for nuclear actin-related chromatin remodeling, see next section):
The Hi-C-seq and assay for transposase-accessible chromatin sequencing (ATAC-seq) of mouse embryonic fibroblasts showed that the deficiency of the BAF chromatin remodeling complexes induced by Actb KO is related to the transitions of chromosome compartments [67]. CTCF binds to the BRK (brm and kis proteins from fly) domain of SMARCA4, the core subunit of BAF complexes [68–70]. The cohesin occupancy at enhancers is also severely perturbed upon SMARCA4 depletion [71, 72]. Therefore, it is plausible that nuclear actin-related chromatin remodeling complexes might stabilize the chromosome compartments by CTCF or cohesin.
2.3. Chromosome architecture and cancers
Accumulating evidence demonstrates that distinct chromosome architecture is associated with cancer [73–82]. The computational model shows that chromosome architecture shapes the landscape of somatic copy-number alterations in cancer [76]. The chromosome decompaction caused by the loss of linker histone H1 leads to the activations of cell stemness-related genes and enhances lymphoma growth [80]. The dynamic changes of chromosome compartments caused by estrogen stimulation create active open chromatins enriched with cancer invasion signaling activities and promote estrogen receptor (ER)-positive breast cancer [82]. Interestingly, large-scale sequencing identified a new chromosome compartment restraining the malignant progression of colorectal cancers [77].
3. Nuclear Actin and Chromatin Remodeling
In the transcriptionally repressed state, DNA is inaccessible as a packaged nucleosome array. Chromatin accessibility refers to the degree to which chromatin-binding factors, such as TFs, RNA polymerase II, or architectural proteins (CTCF and cohesin), physically bind to the open chromatin to initiate the transcription [83]. Chromatin remodeling complexes modulate chromatin accessibility by sliding, inserting, or ejecting the nucleosomal core with the energy of ATP hydrolysis [84]. There are four chromatin remodeling families in mammals (Table 4): the BAF family (canonical BAF complex [cBAF], polybromo-associated BAF complex [pBAF], and non-canonical BAF complex [ncBAF]), the CHD family (CHD1–2 complexes as subfamily I, CHD3–5 complexes as subfamily II, and CHD6–9 complexes as subfamily III), the INO80 family (INO80, SRCAP, and TRRAP complexes), and the ISWI family (ACF, CHRAC, NoRC, NURF, RSF, and WICH complexes) [85–89]. Among those chromatin remodeling families, BAF and INO80 families contain both ACTB and ARPs (ACTL6A, ACTL6B, ACTR5, ACTR6, and ACTR8) as core components [90–92].
Table 4.
Subunits of mammalian BAF, CHD, INO80 and ISWI chromatin remodeling complexes [85, 87–92, 112, 114, 115, 274–281].
| BAF family | CHD family | INO80 family | ISWI family | |||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| cBAF | pBAF | ncBAF | Subfamily I (CHD1–2) | Subfamily II (CHD3–5) | Subfamily III (CHD6–9) | INO80 | SRCAP | TRRAP | ACF | CHRAC | NoRC | NURF | RSF | WICH |
| SMARCB1 | SMARCB1 | SMARCC1 | CHD1 | CDK2AP1 | ASH2L | ACTB | ACTB | ACTB | BAZ1A | BAZ1A | BAZ2A | BAP18 | RSF1 | BAZ1B |
| SMARCC1 | SMARCC1 | SMARCC2 | CHD2 | CHD3 | CHD6 | ACTL6A | ACTL6A | ACTL6A | SMARCA5 | CHRAC1 | SMARCA5 | BPTF | SMARCA5 | SMARCA5 |
| SMARCC2 | SMARCC2 | SMARCD1 | CHD4 | CHD7 | ACTR5 | ACTR6 | ANP32E | POLE3 | HMGXB4 | |||||
| SMARCD1 | SMARCD1 | SMARCD2 | CHD5 | CHD8 | ACTR8 | DMAP1 | BRD8 | SMARCA5 | RBBP4 | |||||
| SMARCD2 | SMARCD2 | SMARCD3 | CSNK2A1 | CHD9 | INO80 | H2AZ1 | DMAP1 | RBBP7 | ||||||
| SMARCD3 | SMARCD3 | BICRA | GATAD2A | PARP1 | INO80B | H2B | EP400 | SMARCA1 | ||||||
| SMARCE1 | SMARCE1 | BICRAL | GATAD2B | RBBP5 | INO80C | RUVBL1 | EPC1 | |||||||
| ACTB | ACTB | ACTB | HDAC1 | WDR5 | INO80D | RUVBL2 | EPC2 | |||||||
| ACTL6A | ACTL6A | ACTL6A | HDAC2 | INO80E | SRCAP | ING3 | ||||||||
| ACTL6B | ACTL6B | ACTL6B | MBD2 | MCRS1 | VPS72 | KAT5 | ||||||||
| BCL7A | BCL7A | BCL7A | MBD3 | NFRKB | YEATS4 | MEAF6 | ||||||||
| BCL7B | BCL7B | BCL7B | MTA1 | RUVBL1 | ZNHIT1 | MORF4L1 | ||||||||
| BCL7C | BCL7C | BCL7C | MTA2 | RUVBL2 | MORF4L2 | |||||||||
| SMARCA2 | SMARCA2 | SMARCA2 | MTA3 | TFPT | MRGBP | |||||||||
| SMARCA4 | SMARCA4 | SMARCA4 | RBBP4 | UCHL5 | RUVBL1 | |||||||||
| SS18 | BRD7 | SS18 | RBBP7 | YY1 | RUVBL2 | |||||||||
| SS18L1 | PBRM1 | SS18L1 | ZBTB7A | YY1AP1 | TRRAP | |||||||||
| ARID1A | ARID2 | BRD9 | VPS72 | |||||||||||
| ARID1B | PHF10 | YEATS4 | ||||||||||||
| DPF1 | ||||||||||||||
| DPF2 | ||||||||||||||
| DPF3 | ||||||||||||||
This section focuses on the roles of nuclear actin and ARPs in modulating the BAF and INO80 chromatin remodeling families in cancer.
3.1. Nuclear actin in the chromatin remodeling complexes
In yeast, the subdomains 1–2 and 3–4 of Act1 (yeast homolog of actin) are twisted 6° by the interaction with both Arp4 (yeast homolog of ACTL6A) and the helicase-SANT-associated (HSA) domain of Snf2 (yeast homolog of SMARCA4) [93]. This spatial twist hinders the cleft of Act1 from binding with latrunculin, an actin polymerization inhibiting toxin, and masks the (+) end of Act1 to prevent the actin polymerization [93, 94]. The Act1 is also observed unable to bind with profilin, which might be due to its embedding into the Baf complex [93]. In contrast, these findings were not observed in the mammalian BAF complex, which needs further investigation [95]. In the yeast Ino80 complex, the (+) end of Act1 is also masked, making it unable to interact with profilin, while the subdomain 2 of Act1 is still accessible to DNase I [96].
3.2. Nuclear actin and cBAF complex
The human cBAF complex consists of three modules (Figure 3A): ATPase, ARP, and base modules (Figure 2) [88, 91]. The residues at 521–1647 amino acids (AAs) of SMARCA4 form the ATPase module, which grabs the nucleosome. The HSA domain (residues at 446–520 AAs) of SMARCA4 binds to the heterodimer constructed by ACTB and ACTL6A to form the ARP module, which maintains the rigid structure of HSA to couple the motions of the ATPase and base modules during chromatin remodeling. The pre-HSA region (residues at 350–445 AAs) of SMARCA4 is anchored into the base module, in which the SMARCB1 packs against the bottom of the nucleosome.
Figure 3. Human canonical BAF and INO80 chromatin remodeling complexes [282].

(A) Cryogenic electron microscopy (Cryo-EM) structure of human cBAF complex binding with the nucleosome (PDB: 6LTJ) [91]. (B) Cryo-EM structure of human INO80 complex binding with the nucleosome (PDB: 6HTS) [90].
When the nucleosome is recruited, the cBAF complex sandwiches it to provide a structural basis for chromatin remodeling [91]. Upon ATP hydrolysis, the ATPase module is positioned to engage with the nucleosome and translocate DNA. The DNA translocation creates DNA tensions to eject nucleosomes or peels the DNA off from the adjacent nucleosomes, which creates nucleosome-depleted regions for chromatin-binding factors to access and bind.
In mice, the genetic deletion of the subdomain of Actl6b, an ARP component of neuron-specific BAF complex, induces impairments of phosphorylation of synaptic cofilin, memory, and synaptic potentiation [97]. These neurological impairments are rescued by restoring the nuclear actin dynamics using a phosphomimetic mutant of cofilin, an ABP accelerating actin depolymerization [97]. This study demonstrates the functional interaction between nuclear actin dynamics and the BAF complex.
3.3. BAF family and cancers
The genes encoding the subunits of the BAF family are highly mutated in many human cancers [98]. In animal models, the loss-of-function of subunits contributes to in vivo tumorigenesis, implying the overall roles of the BAF complex as tumor suppressors. For instance, the genetic mutations (loss-of-function) in the SMARCC1 gene are frequently observed in colon and ovarian cancer [99]. The truncated mutations of the SMARCE1 gene are enriched in clear cell meningioma [100]. The mutation at the splicing site of the BCL7A gene interferes with its binding to the BAF complex in the diffuse large B-cell lymphoma [101]. In lung adenocarcinoma, the loss of SMARCA4 promotes the malignant transformation of the CCSP (club cell secretory protein)-positive cells with early metastasis [102]. As a cBAF-specific subunit, ARID1A loss initiates transdifferentiation from ER-dependent luminal cells to ER-independent basal-like cells in breast cancer. This cellular plasticity is mediated by the reconfigured BAF complex activating the luminal lineage-determining TFs, including ESR1, FOXA1, and GATA3 [103]. As a pBAF-specific subunit, PBRM1 loss reduces the binding of SMARCA4 to the IFNGR2 promoter, decreasing the expression of downstream target genes and enhancing the resistance to immune checkpoint blockade in renal cancer [104].
In contrast, some subunits of the BAF complexes appear to play oncogenic roles. For example, the ACTL6A gene is amplified, and its encoded protein interacts with TP53 to promote cell proliferation and cancer cell stemness in head and neck squamous cell carcinoma [105]. As a ncBAF-specific subunit, BRD9 induces the binding of the ncBAF complex to the enhancers of the cancer-related gene, STAT5A, promoting leukemia cell survival [106]. Recently, protein structure-based assessment of the SMARCA4 showed the different impacts of various mutations in the SMARCA4 gene on the chromatin remodeling activity [107–111]. In addition to the genetic alterations in the BAF complexes, the direct impacts of nuclear actin dynamics on the chromatin remodeling complex need further investigation.
3.4. Nuclear actin and INO80 complex
The human INO80 complex is constructed on the frame of INO80 (Figure 3B) [90, 112]. The N-terminus domain (residues at 1–267 AAs) of INO80 recruits metazoan-specific subunits, including INO80D, INO80E, MCRS1, NFRKB, TFPT, and UCHL5 [113]. The HSA domain (residues at 273–404 AAs) of INO80 directly binds with ACTR8 followed by ACTB, ACTL6A, YY1, and YY1AP1 to form the subcomplex 1 [114, 115]. The residues at 487–1556 AAs of INO80 contact RUVBL1-RUVBL2 hexamer to form the subcomplex 2, which serves as an ATPase. The architecture of subcomplex 2 is relatively rigid because the wheel-like insert domain (residues at 835–1083 AAs) of INO80 is inserted into the barrel-like RUVBL1-RUVBL2 hexamer to restrain its conformation. The subcomplex 2 accommodates a tail-like structure constructed by ACTR5, INO80B, and INO80C to form the subcomplex 2 plus.
When the nucleosome is recruited, the ATPase of the INO80 complex grabs the nucleosome against the ACTR5-INO80C heterodimer to provide a structural basis for chromatin remodeling [90]. Upon ATP hydrolysis, the ATPase pumps DNA towards the nucleosome dyad, unwrapping DNA from the nucleosome surface. When the ACTR5-INO80C heterodimer slips, the DNA wrap is pushed forward and slides across the nucleosome surface, releasing open DNA. In yeast, the requirement of Arp5-Ies6 (yeast homologs of ACTR5-INO80C) heterodimer for chromatin remodeling activity was also observed[116].
The yeast Ino80 complex has two different nucleosome binding states switched by the subcomplex 1 (Act1-Arp4-Arp8 module) [117, 118]. In state I, Arp8 (yeast homolog of ACTR8) grabs the linker DNA of the nucleosome via its Insert 2A (residues at 301–390 AAs) domain [119–121]. In state II, Arp8 folds toward the Tip49A-Tip49B (yeast homologs of RUVBL1-RUVBL2) hexamer to wrap the exposed histone surface of the nucleosome and moves Act1 and Arp8 toward the nucleosome to build direct contacts. Furthermore, act1–2 (an A58T substitution in ACT1) compromises the function of the yeast Ino80 complex, with a significant reduction of ATPase activity, nucleosome binding affinity, and chromatin remodeling activity [96].
Consequently, the gene transcription can also be affected by the disruption of nuclear actin dynamics via the INO80 complex. Additionally, ACTR5 KO-induced dysfunction of the INO80 complex impairs the opening of the cis-regulatory region of heme oxygenase 1 (HMOX1), resulting in a deficiency of transcriptional activator binding [122]. These studies suggest the crucial roles of nuclear actin and ARPs in INO80 complex-mediated gene regulation.
3.5. INO80 family and cancers
It has been well documented that the INO80 family regulates transcription, DNA replication, DNA repair and catalyzes the exchange of H2AZ1-H2B heterodimers with free H2A-H2B in biological processes [123]. In non-small cell lung cancer and melanoma, the INO80 complex is highly correlated with H3K4me1 and H3K27ac histone modifications and enhances the assembly of enhancers to activate the cancer-related genes [124–126]. ATAC-seq elucidates that this enhancer-mediated oncogenic transcription is due to the increased nucleosome occupancy configured by the INO80 complex [126]. During DNA replication in prostate cancer cells, the chromatin remodeling driven by the INO80 complex resolves the R-loop, a DNA-RNA hybrid structure for transcription, and reduces the R-loop-induced DNA damage in cancer cells [127].
The high expression of SRCAP is found in colon cancer [128]. In prostate cancer, SRCAP knockdown decreases the expression of prostate-specific antigen KLK3 by reducing the binding of H2AZ1 to its enhancers [129].
TRRAP shields mutant TP53 protein against the degradation machinery via its HEAT repeat region (residues at 1050–1158 AAs) in lymphoma [130]. It also maintains the cell stemness derived from glioblastoma multiforme by transactivating the self-renewal-related gene, Cyclin A2 (CCNA2). In contrast, its silencing decreases the tumorigenicity of cancer stem cells both in vitro and in vivo due to the reduction of H3 acetylation and H3K4me3 [131]. Similarly, TRAAP knockdown suppresses the expression of stemness-associated markers, including NANOG, POU5F1, and SOX2 in ovarian cancer [132]. Once TRRAP or KAT5, a subunit of the TRRAP complex, is depleted, hepatocellular carcinoma cells become senescent and get arrested at the G2/M phase [133]. Interestingly, the report based on clinical samples and the survival data of breast cancer patients shows that lower expression of TRRAP is observed in tumors compared to normal tissues, and higher TRRAP expression indicates smaller tumor size with better overall survival [134].
Due to the potential oncogenic roles of the INO80 family in cancer, specific disruption of INO80-associated ARPs (ACTR5, ACTR6, or ACTR8) might be a plausible option for cancer prevention or treatment.
4. Nuclear Actin and Transcription Machinery
4.1. Nuclear actin and MRTFs
In 1984, Egly et al. found that nuclear actin stimulates RNA polymerase II-mediated transcription in vitro [135]. Similarly, Scheer et al. demonstrated the involvement of nuclear actin in modulating transcription in salamander oocytes [136]. Later, the vital role of nuclear actin in the initiation and elongation of transcription in eukaryotes was unveiled [137]. After these milestone studies on the potential role of nuclear actin in transcription, more researchers began to study the function of nuclear actin [135, 136]. To date, accumulating evidence suggests that in combination with TFs, ABPs, or transcription complexes, nuclear actin modulates gene expression from transcription initiation to transcription elongation [138].
Among the actin-mediated transcriptions, the regulation of myocardin-related transcription factor A (MRTFA) is one of the most well-demonstrated examples (Figure 4A) [139]. MRTFA is a transcription co-activator of serum response factor (SRF), which regulates the expression of muscle-specific, immediate-early, and cytoskeletal genes in response to changes in G-actin levels [140–142]. The N-terminus of MRTFA contains a domain with three RPEL (G-actin-binding sites) motifs that operate as a G-actin sensor, regulating both subcellular localization and nuclear activity of MRTFA [143–145]. G-actin can interact with the RPEL domains directly [146]. Within the RPEL domain, a bipartite NLS is embedded, preventing the nuclear import of the pentameric actin complex [147, 148]. In unstimulated conditions, when G-actin levels are relatively high, MRTFA is predominantly localized in the cytoplasm due to efficient actin-dependent and exportin 1 (XPO1)-mediated nuclear export of MRTFA. This is because G-actin binding occludes the bipartite NLS in the RPEL domain [143, 148, 149]. However, upon serum stimulation that activates transient nuclear actin polymerization, G-actin is transformed into F-actin, which results in the release and nuclear accumulation of MRTFA by increased nuclear import, and decreased XPO1-dependent nuclear export, followed by the subsequent activation of SRF target gene transcription [21, 62, 150].
Figure 4. Nuclear actin functions in the nucleus.

(A) G-actin inhibits the MRTFA/SRF-mediated transcription. Nuclear actin polymerization releases MRTFA from G-actin to activate SRF-mediated transcription upon serum stimulation. (B) Actin is involved in transcription. Actin and ABPs are associated with RNA polymerases I/II/III, and interacts with P-TEFb, snRNPs, and hnRNPs, regulating transcription initiation and elongation. (C) The association of nuclear actin with pre-mRNA splicing and processing. The molecular mechanism of actin-controlled pre-mRNA splicing is unclear. (D) Nuclear actin modulates Wnt signaling-mediated transcription via direct interaction with Wnt signaling-related components, including β-catenin, CRACD, and APC (not shown). (E, F) When double strand of DNA breaks in the nucleus, nuclear F-actin participates in the non-homologous end joining (NHEJ) and homologous recombination (HR) repair pathways.
Additionally, nuclear actin polymerization and MRTFA/SRF-mediated transcription are also regulated by other factors, including F-actin-monooxygenase MICAL2 [151], cyclic-AMP(cAMP) signaling [152], and Ras association domain-containing protein 1 isoform A (RASSF1A) [153]. MICAL2 regulates nuclear actin through redox modification that decreases G-actin levels inside the nucleus. This, in turn, increases MRTFA accumulation in the nucleus and MRTFA/SRF-dependent gene transcription [151]. Recently, McNeill et al. showed that elevated c-AMP increases nuclear G-actin levels in vascular smooth muscle cells, suppressing cell proliferation and migration by inhibiting MRTFA/SRF and YAP/TAZ-TEAD-dependent gene expression [152]. RASSF1A is a tumor suppressor frequently epigenetically suppressed in tumors and forms a complex with XPO6 and RAN GTPase, promoting the XPO6-mediated nuclear export of actin, thus regulating transcription via MRTFA/SRF [153]. Interestingly, this pathway is deregulated in cancer cells, which leads to the accumulation of nuclear G-actin and suppression of MRTFA/SRF-mediated transcription [153]. Besides, there is another MRTF named MRTFB which can also stimulate SRF-dependent transcription. In contrast to MRTFA, MRTFB has a different tissue distribution and relatively weak affinities with SRF [154]. Kuwahara et al. found that in NIH3T3 cells, MRTFB also undergoes nuclear translocation react to Rho signaling and nuclear actin polymerization, even though it is slightly less responsive than MRTFA upon serum stimulation [155]. In human aortic endothelial cells, Hayashi et al. showed that the nuclear localization of MRTFA and MRTFB is affected by actin dynamics involved in gene expression [156].
Together, MRTF/SRF transcriptional activity is regulated by signal-induced nuclear actin polymerization and depolymerization cycle and G-actin binding to the RPEL domain of MRTFA [21, 157, 158]. It is noteworthy that many cytoskeletal genes, including actin, are also regulated by MRTF/SRF, suggesting that the actin-MRTF-SRF signaling axis forms a feedback loop where actin dynamics regulates the transcriptional homeostasis of the cytoskeleton [158, 159].
4.2. Nuclear actin and RNA polymerases
In the eukaryotes, RNA polymerases (Pols) I, II, and III catalyze DNA-dependent RNA synthesis [160]. Pol I synthesizes ribosomal RNA (rRNA), and Pol II and Pol III synthesize mainly mRNA and tRNAs, respectively [160]. Several studies showed that nuclear actin directly binds to all three RNA Pols (Figure 4B) [161–164]. Later, the impacts of nuclear actin-associated RNA Pols on gene regulation were unveiled.
RNA Pol I:
As a molecular motor, nuclear F-actin interacts with the RNA Pol I complex together with nuclear myosin I (NM1) and is involved in the transcription of ribosomal RNA genes (rDNA) [165, 166]. Philimonenko et al. found that nuclear actin and NM1 are associated with rDNA, and microinjection of antibodies against actin or NM1 into the nuclei of cells decreased the Pol I-mediated transcription in vivo and in vitro [167]. Ye et al.’s study showed that drugs inhibiting actin polymerization or myosin function blocked Pol I-driven transcription in vivo and in vitro [168]. Meanwhile, actin mutants (S14C, G15S, and V159N) stabilizing F-actin tightly bind to Pol I and activate transcription [168, 169]. Conversely, a polymerization-deficient actin mutant does not interact with Pol I and fails to activate transcription [168]. Moreover, the association of nuclear actin and NM1 with Pol I is interrupted when ATP exists but is stabilized by ADP, and by anchoring NM1 to DNA and nuclear F-actin to RNA polymerase, the nuclear actomyosin complex serves as a motor that works with nuclear RNA polymerases to activate transcription [170, 171]. These studies suggest the crucial role of nuclear actin polymerization in Pol I-mediated transcription [168].
RNA Pol II:
In 1984, Scheer et al. showed that injection of actin antibodies into the nuclei of salamander oocytes inhibited the transcription [172]. Hofmann et al. found that actin is associated with actively transcribed genes and plays a pivotal role in the activation of transcription [161]. Later, in vivo and in vitro studies have recapitulated the requirement for actin in RNA Pol-mediated transcription activation, initiation, and elongation [159]. In 2019, to determine the nuclear actin interactome, Viita et al. employed two mass spectrometry (MS)-based techniques, affinity purification (AP)-MS and biotin identification (BioID)-MS [173]. The MS data identified nuclear actin as a component of the RNA Pol II pre-initiation complex [173]. Sokolova et al. performed chromatin immunoprecipitation sequencing (ChIP-seq) and genome-wide analysis in fly ovaries. For the first time, they showed that nuclear actin in physical conjunction with Pol II co-occupies the promoters associated with gene bodies of actively transcribed genes [174]. Furthermore, by using immunoprecipitation, immunofluorescence, and glutathione S-transferase (GST) pull-down assay, Qi et al. and Hu et al. showed that nuclear actin directly interacts with Pol II subunits POLR2E and POLR2G, as well as Pol III subunits POLR3C, POLR2F, and POLR2H [175, 176]. During the initiation and elongation of transcription, cyclin-dependent kinase 9 (CDK9), a subunit of positive transcription elongation factor b (P-TEFb) and RNA helicase A (RHA) are associated with G-actin in the nucleus, which physically links nuclear actin with Pol II [176, 177].
RNA Pol III:
Utilizing protein purification of Pol III from human IMR90 cells expressing a double-tagged Pol III subunit, Hu et al. observed that actin is co-purified with Pol III [175]. They showed that actin is stably associated with one or more of the POLR3C, POLR2F, and POLR2H subunits of Pol III via direct interaction, required for Pol III-mediated transcription in vitro [175]. Moreover, ChIP experiments showed that actin occupies the promoter of the U6 gene actively transcribed by Pol III [178, 179]. Additionally, the treatment of cells with methane methylsulfonate, an inhibitor of Pol III, released the transcription initiation complex from the U6 promoter and uncoupled the actin protein from the Pol III complex [178, 180]. Moreover, it was shown that the monomeric form of actin is essential for Pol III-driven transcription [181].
In addition to nuclear actin, the actin polymerization and depolymerization regulators were shown to be engaged in RNA Pols. For instance, the ARP2/3 complex and its activators N-WASP (neural Wiskott-Aldrich syndrome protein), WASF1 (WASP family member 1), WASH (WASP family homolog), and motor protein myosin are associated with nuclear RNA Pols and transcription processes [182–185]. Recently, using next-generation transcriptome sequencing and super-resolution microscopy, Wei et al. showed that the formation of RNA Pol II complex is facilitated by the N-WASP/ARP2/3-dependent polymerization of nuclear actin filaments [186]. Together, these lines of evidence suggest that nuclear actin and its regulatory proteins are physically and functionally associated with RNA polymerases-controlled transcriptional activation, initiation, and elongation.
4.3. Nuclear actin and pre-mRNA splicing
Actin was also detected in pre-messenger ribonucleoprotein (pre-mRNP) [187], implying the potential roles of nuclear actin in pre-mRNA processing (Figure 4C) [188, 189]. In 2019, the mass spectrometry-based nuclear actin interactome approaches not only revealed the association of actin with pre-initiation complex (PIC), transcription elongation but also pre-mRNA splicing and processing [173]. Viita et al. found that alterations in nuclear actin levels disturb alternative splicing of reporter gene constructs, suggesting that nuclear actin affects pre-mRNA splicing directly or indirectly, likely by affecting the transcription elongation rate [173]. Another study combining bioinformatics with protein binding analysis showed that nuclear actin interacts with the unique region of the pre-mRNA of the Epstein-Barr virus (EBV) latent membrane protein 2 (LMP2) [190]. Treatment of EBV-positive cells with drugs inhibiting actin polymerization showed a significant decrease of spliced isoform levels of the pre-mRNA, indicating the role of nuclear actin in modulating viral RNA splicing [190].
4.4. Other mechanisms
Ribonucleoproteins:
Besides the roles of nuclear actin in regulating transcriptional activation and mRNA splicing, nuclear actin is engaged in RNA processing and transportation [191]. Nuclear actin is also associated with small nuclear ribonucleoproteins (snRNPs) modulating mRNA processing and viral RNA nuclear export [191–193]. Moreover, nuclear actin binds to heterogeneous nuclear ribonucleoprotein (hnRNP) protein hrp65–2 in the Chironomus tentans cells and hnRNP U protein in the mammalian cells [189, 194, 195]. Sjölinder et al. showed that growing pre-mRNA recruits actin, hnRNP proteins, and chromatin remodeling complexes to actively transcribed genes for ongoing transcription [196].
Wnt signaling:
Wnt signaling is a highly conserved pathway orchestrating various cellular processes and is hyperactivated in many human cancers [197–199]. Recent studies suggested that nuclear actin modulates Wnt signaling-mediated transcription via direct interaction with Wnt signaling-related components, including β-catenin, CRACD, and APC (adenomatous polyposis coli) (Figure 4D) [10, 200–202]. In 2016, Yamazaki et al. found that nuclear F-actin colocalizes with β-catenin, increases the nuclear accumulation of β-catenin, and enhances the transcriptional β-catenin downstream targeting genes of the Wnt/β-catenin signaling pathway [202]. Jung et al. identified CRACD as a regulator stabilizing the cadherin-catenin-actin complex via capping protein inhibition in the nucleus. The frequent inactivation of CRACD in colorectal cancer inhibits actin polymerization, resulting in G-actin release and accumulation in the nucleus with Wnt signaling hyperactivation mucinous colorectal cancer in vivo [10].
As a protein destruction complex, APC binds to and induces the degradation of β-catenin [203, 204]. The APC gene is highly mutated in colorectal cancer, resulting in the hyperactivation of Wnt/β-catenin signaling [205]. In addition to the cytoplasmic APC, APC contains the NLS and is also located in the nucleus [206, 207]. In conjunction with an actin-nucleating protein, formin mDia1, the C-terminus ‘basic’ domain of APC protein nucleates the formation of actin filaments and stimulates actin filament assembly [201]. Baarlink et al. found that the mDia1 triggers the nuclear actin polymerization in response to serum stimulation [208]. Therefore, nuclear APC likely regulates nuclear actin dynamics via actin nucleation.
Emerging evidence demonstrated that nuclear actin and ABPs are physically and functionally associated with various proteins related to gene expression [159]. Given the distinct feature of nuclear actin in cancer cells, the impacts of deregulated nuclear actin dynamics on aberrant gene expression in tumorigenesis need further interrogation.
5. Nuclear Actin and DNA Repair
In the mid-1930s, Timoféeff-Ressovsky et al. found that ionizing and ultraviolet (UV) radiation induces DNA damage [209]. At the end of the 1940s, Kelner and Dulbecco et al. discovered the DNA repair mechanism in cells and bacteriophages using UV radiation [210, 211]. Genotoxic stress induces DNA damage, which includes disruption or addition to the nucleotide of the DNA or the breakage of one chain of the DNA or DNA double-strand break (DSB) [212, 213]. Unresolved DNA damage results in a variety of human disorders and cancers [214, 215].
5.1. DSB repair
DNA damage response includes repair and tolerance [213]. However, severe DNA damage such as DSBs should be repaired to avoid cell death [216]. DSBs may lead to chromosomal rearrangements, including deletion, translocation, and amplification, which can trigger the activation of oncogenes or the inactivation of tumor suppressors for tumorigenesis [215]. There are two major DSB repair mechanisms: non-homologous end joining (NHEJ) and homologous recombination (HR) (Figure 4E, F) [213]. NHEJ takes place in both dividing and non-dividing cells, whereas HR occurs only mainly in dividing cells during the late S-G2 phase because HR utilizes a homologous sister chromatid as a template [217, 218].
NHEJ is a fast and predominant DSB repair mechanism in mammalian cells [219]. NHEJ often results in the loss of genetic information at the site of the DSB [219]. At the beginning of NHEJ, the Ku (Ku70/Ku80) heterodimer recognizes and binds to the two DSB DNA ends directly, followed by recruitment of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) [220, 221]. Then, the two DNA-PKcs positioned at each DSB terminus align the two DNA ends together, activating processing factors such as Artemis, which generates overhangs at the DNA ends [222]. Finally, DNA polymerases fill in the gaps, followed by end-joining via X-ray repair cross complementing 4 (XRCC4)-DNA Ligase IV complex in collaboration with non-homologous end-joining factor 1 (NHEJ1) [223–225].
HR uses the sister chromatid as a template to repair DSB, which leads to a high-fidelity repair of DSBs [226]. HR mainly occurs in the late S-G2 phase and includes ssDNA overhang generation, strand invasion, homologous pairing, Holliday junction formation, DNA synthesis, branch migration, and Holliday junction resolution [227]. The key step of HR is initiated by recognition of the DSB by the MRN (MRE11-RAD50-NBS1) complex [228]. As a break sensor, the MRN complex is associated with DNA endonuclease RB-binding protein 8 (RBBP8) and recruits the serine-protein kinase ataxia telangiectasia mutated (ATM) to DSB sites, which result in the generation of 5′−3′ end resection and the 3′ ssDNA overhang [229–232]. Then, the ssDNA overhang is stabilized by replication protein A (RPA) binding, DNA repair protein 51 (RAD51) is loaded onto the ssDNA overhang [233]. Next, RAD52 is recruited to RPA, and the RAD52-RPA complex is replaced by the RAD51-BRCA2 complex [233, 234]. Then, RAD51-coated ssDNA promotes invasion of the template strand, which generates a Holliday junction [235]. Later, the DNA strand is synthesized by a polymerase using the sister strand as a template, followed by branch migration and subsequent resolution of the heteroduplex [236, 237]. Finally, the two broken DNA ends are rejoined by a DNA ligase [238].
5.2. Nuclear actin-mediated repair of DSBs
In the past decades, there has been increasing evidence showing the vital role of nuclear actin in the DSB repair process [159]. In 2012, Andrin et al. performed the pull-down assay with purified F-actin protein and found that F-actin binds to DSB repair proteins including Ku80, MRE11, and RAD51 in vitro, suggesting that actin polymerization may be engaged in DSB repair [239]. Later, utilizing actin probes, Belin et al. found that DNA damage induces the generation of long nuclear actin filaments, short nucleolus-associated filaments, and dense nuclear actin clusters in living cells [240]. In 2018, two independent studies demonstrated that nucleator ARP2/3 complex-mediated nuclear actin filament assembly is required for DSB repair in different cell lines [241, 242].
Nuclear F-actin participates in both NHEJ and HR repair pathways [239]. In the NHEJ pathway, depolymerization of endogenous nuclear actin alters the retention of Ku80 at DNA damage sites in human cells [239]. In the HR pathway, nuclear F-actin drives DSB dynamics in a somewhat different way between heterochromatin and euchromatin [243]. In fly and mouse cells, DSB detection and resection occur within the heterochromatin domain [244–246]. Firstly, the early DSB signaling, processing factor Mre11 and heterochromatin protein 1a (Hp1a) promote the recruitment of Arp2/3 and myosins to DSBs [241]. Secondly, Arp2/3 activation promotes actin polymerization and filament growth towards the nuclear periphery [247]. Thirdly, Smc5/6 blocks Rad51 recruitment and instead recruits Unc45 to activate nuclear myosins [247, 248]. Finally, the myosin-Smc5/6-chromatin repair complex travels along nuclear actin filaments and anchors DSBs to nuclear pores, where HR repair continues with Rad51 recruitment and strand invasion [241, 242, 248]. In fly and mouse cells, nuclear F-actin is detected by the live-cell imaging with nuclear F-actin marker chromobody and F-actin staining with phalloidin [241]. The live-cell imaging shows that re-localization of heterochromatic DSBs occurs by directly moving along a nuclear actin filament network assembled at the repair sites by Arp2/3 and extension toward the nuclear periphery [241, 247]. In euchromatin, Mre11 and resection promote the movement of DSB repair sites via Arp2/3-induced short nuclear actin polymers, which also travel with euchromatic repair sites [241, 249]. In response to DSB in human cells, enriched ARP2/3 and nuclear actin polymerization at the DSB repair sites facilitate focus clustering, DSB resection, DSB movement, and HR completion without myosins [241, 242].
Genomic instability, one of the hallmarks of cancer, is mainly due to the impaired DNA repair pathway [250]. Therefore, cancer-actin dynamics affect genomic instability via nuclear actin-mediated DNA repair.
6. Concluding Remarks
Actin is the most abundant protein in the cells [1]. The canonical features of actin include cytoplasmic localization, GTPase activity, and highly dynamic transition between polymerization and depolymerization [1–3]. These properties led to the intense investigation of actin’s roles in orchestrating the cytoskeleton, resulting in the seminal findings of actin cytoskeleton-mediated cell morphology, cell migration, and cell adhesion.
Evolved from these classical outlooks, accumulating evidence has unveiled that actin is also engaged in diverse nuclear processes. Beyond its roles as a structural protein composing the cytoskeleton, the nuclear actin is being highlighted in the context of chromosome architecture, chromatin remodeling complexes, transcription machinery, and DNA repair [27, 31–34, 67]. These actin-associated nuclear events were generally appreciated as the perspective of a similar mechanism of the cytosolic actin dynamics between polymerization and depolymerization. For instance, nuclear actin polymerization mechanically regulates chromosome architecture [67]. Additionally, the polymerized nuclear actin serves as a railroad facilitating the movement of DNA repair proteins [32]. Notwithstanding, it is noteworthy that monomeric and oligomeric nuclear actin also exists as a distinct structure, unlike the one in the cytoplasm. For example, monomeric nuclear actin in the chromatin remodeling complex displays a different structure from the cytoplasmic G-actin [93]. Besides such a conformational difference, the nuclear import and export of actin, ARPs, and their post-translational modifications may provide additional regulatory layers of nuclear actin dynamics [30]. Therefore, along with its role as a building block in the cytoplasm, both monomeric and oligomeric actin in the nucleus should be comprehensively appreciated as a critical component of various nuclear processes. Despite the current limitations in dissecting nuclear actin in vivo, the ongoing technical improvement in visualization and quantification is expected to unravel nuclear actin-associated biological events up-close.
The hallmarks of cancer include the loss of genomic/chromosomal integrity and aberrant gene expression, which are physically and functionally associated with nuclear actin. Thus, it is reasonable to assume that fine control of nuclear actin dynamics is a gatekeeper of tumorigenesis (Figure 5). Indeed, recent genome-wide studies showed that actin and actin regulatory proteins are genetically and epigenetically dysregulated in cancer [10, 105]. Therefore, further understanding of the pathophysiological impact of nuclear actin deregulation on chromosome architecture, chromatin remodeling complexes, transcription machinery, and DNA repair will lead to biomarker identification, therapy development, or biomarker-guided molecular targeting of cancer.
Figure 5. Nuclear actin, a gatekeeper of genomic integrity and gene expression.

In normal cells, the well-balanced nuclear actin dynamics plays crucial roles in modulating chromosome architecture, chromatin remodeling complexes, transcriptional machinery, and DNA repair, which maintains genomic integrity and orchestrates gene expression for tissue homeostasis. Conversely, dysregulated actin dynamics impairs the fine control of chromosomal architecture, chromatin remodeling, transcriptional machinery, and DNA repair. Consequently, genomic instability, inactivation of tumor suppressor genes, and hyperactivation of oncogenes contribute to tumorigenesis.
Acknowledgments
We apologize for not including all relevant studies in the field. This work was supported by grants to the Cancer Prevention and Research Institute of Texas (RP200315 to J.-I.P.) and the National Institutes of Health (R01 CA193297 and R03 CA256207 to J.-I.P.).
References
- 1.Dominguez R, Holmes KC. Actin structure and function. Annu Rev Biophys 2011;40:169–86. doi: 10.1146/annurev-biophys-042910-155359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Perrin BJ, Ervasti JM. The actin gene family: function follows isoform. Cytoskeleton (Hoboken) 2010;67(10):630–4. doi: 10.1002/cm.20475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pollard TD, Cooper JA. Actin, a central player in cell shape and movement. Science (New York, NY) 2009;326(5957):1208–12. doi: 10.1126/science.1175862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kühn S, Mannherz HG. Actin: Structure, Function, Dynamics, and Interactions with Bacterial Toxins. In: Mannherz HG, editor. The Actin Cytoskeleton and Bacterial Infection Cham: Springer International Publishing; 2017. p. 1–34. [DOI] [PubMed] [Google Scholar]
- 5.Carlsson AE. Actin dynamics: from nanoscale to microscale. Annu Rev Biophys 2010;39:91–110. doi: 10.1146/annurev.biophys.093008.131207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pollard TD. Actin and Actin-Binding Proteins. Cold Spring Harb Perspect Biol 2016;8(8). doi: 10.1101/cshperspect.a018226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wear MA, Cooper JA. Capping protein: new insights into mechanism and regulation. Trends Biochem Sci 2004;29(8):418–28. doi: 10.1016/j.tibs.2004.06.003. [DOI] [PubMed] [Google Scholar]
- 8.Edwards M, Zwolak A, Schafer DA, Sept D, Dominguez R, Cooper JA. Capping protein regulators fine-tune actin assembly dynamics. Nature reviews Molecular cell biology 2014;15(10):677–89. doi: 10.1038/nrm3869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Eng G, Braverman J, Yilmaz OH. CRAD as a cytoskeletal tumour suppressor. Nature cell biology 2018;20(11):1232–3. doi: 10.1038/s41556-018-0225-x. [DOI] [PubMed] [Google Scholar]
- 10.Jung YS, Wang W, Jun S, Zhang J, Srivastava M, Kim MJ, et al. Deregulation of CRAD-controlled cytoskeleton initiates mucinous colorectal cancer via beta-catenin. Nat Cell Biol 2018;20(11):1303–14. doi: 10.1038/s41556-018-0215-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Belin BJ, Goins LM, Mullins RD. Comparative analysis of tools for live cell imaging of actin network architecture. Bioarchitecture 2014;4(6):189–202. doi: 10.1080/19490992.2014.1047714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Melak M, Plessner M, Grosse R. Actin visualization at a glance. Journal of cell science 2017;130(3):525–30. doi: 10.1242/jcs.189068. [DOI] [PubMed] [Google Scholar]
- 13.DesMarais V, Eddy RJ, Sharma VP, Stone O, Condeelis JS. Optimizing leading edge F-actin labeling using multiple actin probes, fixation methods and imaging modalities. Biotechniques 2019;66(3):113–9. doi: 10.2144/btn-2018-0112. [DOI] [PubMed] [Google Scholar]
- 14.Loureiro SO, Heimfarth L, Reis K, Wild L, Andrade C, Guma FT, et al. Acute ethanol exposure disrupts actin cytoskeleton and generates reactive oxygen species in c6 cells. Toxicol In Vitro 2011;25(1):28–36. doi: 10.1016/j.tiv.2010.09.003. [DOI] [PubMed] [Google Scholar]
- 15.Danchenko M, Csaderova L, Fournier PE, Sekeyova Z. Optimized fixation of actin filaments for improved indirect immunofluorescence staining of rickettsiae. BMC Res Notes 2019;12(1):657. doi: 10.1186/s13104-019-4699-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Leyton-Puig D, Kedziora KM, Isogai T, van den Broek B, Jalink K, Innocenti M. PFA fixation enables artifact-free super-resolution imaging of the actin cytoskeleton and associated proteins. Biol Open 2016;5(7):1001–9. doi: 10.1242/bio.019570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lin G, Qiu X, Fandel TM, Albersen M, Wang Z, Lue TF, et al. Improved penile histology by phalloidin stain: circular and longitudinal cavernous smooth muscles, dual-endothelium arteries, and erectile dysfunction-associated changes. Urology 2011;78(4):970 e1–8. doi: 10.1016/j.urology.2011.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ohnishi T, Kawamura H, Yamamoto T. [Extraction of a Protein Resembling Actin from the Cell Nucleus of the Calf Thymus]. J Biochem 1963;54:298–300. doi: 10.1093/oxfordjournals.jbchem.a127789. [DOI] [PubMed] [Google Scholar]
- 19.Tsopoulidis N, Kaw S, Laketa V, Kutscheidt S, Baarlink C, Stolp B, et al. T cell receptor-triggered nuclear actin network formation drives CD4(+) T cell effector functions. Sci Immunol 2019;4(31). doi: 10.1126/sciimmunol.aav1987. [DOI] [PubMed] [Google Scholar]
- 20.Zahler S Nuclear actin in cancer biology. Int Rev Cell Mol Biol 2020;355:53–66. doi: 10.1016/bs.ircmb.2020.04.001. [DOI] [PubMed] [Google Scholar]
- 21.Baarlink C, Wang H, Grosse R. Nuclear actin network assembly by formins regulates the SRF coactivator MAL. Science 2013;340(6134):864–7. doi: 10.1126/science.1235038. [DOI] [PubMed] [Google Scholar]
- 22.Lamm N, Read MN, Nobis M, Van Ly D, Page SG, Masamsetti VP, et al. Nuclear F-actin counteracts nuclear deformation and promotes fork repair during replication stress. Nat Cell Biol 2020;22(12):1460–70. doi: 10.1038/s41556-020-00605-6. [DOI] [PubMed] [Google Scholar]
- 23.Serebryannyy LA, Parilla M, Annibale P, Cruz CM, Laster K, Gratton E, et al. Persistent nuclear actin filaments inhibit transcription by RNA polymerase II. Journal of cell science 2016;129(18):3412–25. doi: 10.1242/jcs.195867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wilkie AR, Lawler JL, Coen DM. A Role for Nuclear F-Actin Induction in Human Cytomegalovirus Nuclear Egress. Mbio 2016;7(4). doi: ARTN e01254 10.1128/mBio.01254-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kyheroinen S, Vartiainen MK. Nuclear actin dynamics in gene expression and genome organization. Semin Cell Dev Biol 2019. doi: 10.1016/j.semcdb.2019.10.012. [DOI] [PubMed]
- 26.Dopie J, Skarp KP, Rajakyla EK, Tanhuanpaa K, Vartiainen MK. Active maintenance of nuclear actin by importin 9 supports transcription. Proceedings of the National Academy of Sciences of the United States of America 2012;109(9):E544–52. doi: 10.1073/pnas.1118880109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen M, Shen X. Nuclear actin and actin-related proteins in chromatin dynamics. Current opinion in cell biology 2007;19(3):326–30. doi: 10.1016/j.ceb.2007.04.009. [DOI] [PubMed] [Google Scholar]
- 28.Kristo I, Bajusz I, Bajusz C, Borkuti P, Vilmos P. Actin, actin-binding proteins, and actin-related proteins in the nucleus. Histochem Cell Biol 2016;145(4):373–88. doi: 10.1007/s00418-015-1400-9. [DOI] [PubMed] [Google Scholar]
- 29.Oma Y, Harata M. Actin-related proteins localized in the nucleus: from discovery to novel roles in nuclear organization. Nucleus 2011;2(1):38–46. doi: 10.4161/nucl.2.1.14510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hofmann WA, Arduini A, Nicol SM, Camacho CJ, Lessard JL, Fuller-Pace FV, et al. SUMOylation of nuclear actin. The Journal of cell biology 2009;186(2):193–200. doi: 10.1083/jcb.200905016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Caridi CP, Plessner M, Grosse R, Chiolo I. Nuclear actin filaments in DNA repair dynamics. Nature cell biology 2019;21(9):1068–77. doi: 10.1038/s41556-019-0379-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hurst V, Shimada K, Gasser SM. Nuclear Actin and Actin-Binding Proteins in DNA Repair. Trends Cell Biol 2019;29(6):462–76. doi: 10.1016/j.tcb.2019.02.010. [DOI] [PubMed] [Google Scholar]
- 33.Olave IA, Reck-Peterson SL, Crabtree GR. Nuclear actin and actin-related proteins in chromatin remodeling. Annu Rev Biochem 2002;71:755–81. doi: 10.1146/annurev.biochem.71.110601.135507. [DOI] [PubMed] [Google Scholar]
- 34.Percipalle P, Visa N. Molecular functions of nuclear actin in transcription. J Cell Biol 2006;172(7):967–71. doi: 10.1083/jcb.200512083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gilbert N, Allan J. The many length scales of DNA packaging. Essays Biochem 2019;63(1):1–4. doi: 10.1042/EBC20190040. [DOI] [PubMed] [Google Scholar]
- 36.Bartova E, Krejci J, Harnicarova A, Galiova G, Kozubek S. Histone modifications and nuclear architecture: a review. J Histochem Cytochem 2008;56(8):711–21. doi: 10.1369/jhc.2008.951251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liu J, Ali M, Zhou Q. Establishment and evolution of heterochromatin. Ann N Y Acad Sci 2020;1476(1):59–77. doi: 10.1111/nyas.14303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Belaghzal H, Dekker J, Gibcus JH. Hi-C 2.0: An optimized Hi-C procedure for high-resolution genome-wide mapping of chromosome conformation. Methods 2017;123:56–65. doi: 10.1016/j.ymeth.2017.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bonev B, Cavalli G. Organization and function of the 3D genome. Nat Rev Genet 2016;17(11):661–78. doi: 10.1038/nrg.2016.112. [DOI] [PubMed] [Google Scholar]
- 40.Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 2012;485(7398):376–80. doi: 10.1038/nature11082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 2014;159(7):1665–80. doi: 10.1016/j.cell.2014.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rowley MJ, Corces VG. Organizational principles of 3D genome architecture. Nat Rev Genet 2018;19(12):789–800. doi: 10.1038/s41576-018-0060-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.McGinty RK, Tan S. Nucleosome structure and function. Chem Rev 2015;115(6):2255–73. doi: 10.1021/cr500373h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li G, Zhu P. Structure and organization of chromatin fiber in the nucleus. FEBS Lett 2015;589(20 Pt A):2893–904. doi: 10.1016/j.febslet.2015.04.023. [DOI] [PubMed] [Google Scholar]
- 45.Kolovos P, Knoch TA, Grosveld FG, Cook PR, Papantonis A. Enhancers and silencers: an integrated and simple model for their function. Epigenetics Chromatin 2012;5(1):1. doi: 10.1186/1756-8935-5-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pombo A, Dillon N. Three-dimensional genome architecture: players and mechanisms. Nature reviews Molecular cell biology 2015;16(4):245–57. doi: 10.1038/nrm3965. [DOI] [PubMed] [Google Scholar]
- 47.Davidson IF, Peters JM. Genome folding through loop extrusion by SMC complexes. Nature reviews Molecular cell biology 2021;22(7):445–64. doi: 10.1038/s41580-021-00349-7. [DOI] [PubMed] [Google Scholar]
- 48.Palstra RJ, Tolhuis B, Splinter E, Nijmeijer R, Grosveld F, de Laat W. The beta-globin nuclear compartment in development and erythroid differentiation. Nat Genet 2003;35(2):190–4. doi: 10.1038/ng1244. [DOI] [PubMed] [Google Scholar]
- 49.Cutts EE, Vannini A. Condensin complexes: understanding loop extrusion one conformational change at a time. Biochem Soc Trans 2020;48(5):2089–100. doi: 10.1042/BST20200241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Banigan EJ, van den Berg AA, Brandao HB, Marko JF, Mirny LA. Chromosome organization by one-sided and two-sided loop extrusion. Elife 2020;9. doi: 10.7554/eLife.53558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dekker J, Heard E. Structural and functional diversity of Topologically Associating Domains. FEBS Lett 2015;589(20 Pt A):2877–84. doi: 10.1016/j.febslet.2015.08.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.A. Knoch T. A Consistent Systems Mechanics Model of the 3D Architecture and Dynamics of Genomes Chromatin and Epigenetics. 2020. [Google Scholar]
- 53.Bak JH, Kim MH, Liu L, Hyeon C. A unified framework for inferring the multi-scale organization of chromatin domains from Hi-C. PLoS Comput Biol 2021;17(3):e1008834. doi: 10.1371/journal.pcbi.1008834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Falk M, Feodorova Y, Naumova N, Imakaev M, Lajoie BR, Leonhardt H, et al. Heterochromatin drives compartmentalization of inverted and conventional nuclei. Nature 2019;570(7761):395–9. doi: 10.1038/s41586-019-1275-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang S, Su JH, Beliveau BJ, Bintu B, Moffitt JR, Wu CT, et al. Spatial organization of chromatin domains and compartments in single chromosomes. Science (New York, NY) 2016;353(6299):598–602. doi: 10.1126/science.aaf8084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cremer T, Cremer M. Chromosome territories. Cold Spring Harb Perspect Biol 2010;2(3):a003889. doi: 10.1101/cshperspect.a003889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Knoch TA. Simulation of different three-dimensional polymer models of interphase chromosomes compared to experiments-an evaluation and review framework of the 3D genome organization. Semin Cell Dev Biol 2019;90:19–42. doi: 10.1016/j.semcdb.2018.07.012. [DOI] [PubMed] [Google Scholar]
- 58.Kolovos P, Brouwer RWW, Kockx CEM, Lesnussa M, Kepper N, Zuin J, et al. Investigation of the spatial structure and interactions of the genome at sub-kilobase-pair resolution using T2C. Nat Protoc 2018;13(3):459–77. doi: 10.1038/nprot.2017.132. [DOI] [PubMed] [Google Scholar]
- 59.Knoch TA, Wachsmuth M, Kepper N, Lesnussa M, Abuseiris A, Ali Imam AM, et al. The detailed 3D multi-loop aggregate/rosette chromatin architecture and functional dynamic organization of the human and mouse genomes. Epigenetics Chromatin 2016;9:58. doi: 10.1186/s13072-016-0089-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Xie X, Almuzzaini B, Drou N, Kremb S, Yousif A, Farrants AO, et al. beta-Actin-dependent global chromatin organization and gene expression programs control cellular identity. FASEB J 2018;32(3):1296–314. doi: 10.1096/fj.201700753R. [DOI] [PubMed] [Google Scholar]
- 61.Baarlink C, Plessner M, Sherrard A, Morita K, Misu S, Virant D, et al. A transient pool of nuclear F-actin at mitotic exit controls chromatin organization. Nature cell biology 2017;19(12):1389–99. doi: 10.1038/ncb3641. [DOI] [PubMed] [Google Scholar]
- 62.Wang Y, Sherrard A, Zhao B, Melak M, Trautwein J, Kleinschnitz EM, et al. GPCR-induced calcium transients trigger nuclear actin assembly for chromatin dynamics. Nat Commun 2019;10(1):5271. doi: 10.1038/s41467-019-13322-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.van Steensel B, Belmont AS. Lamina-Associated Domains: Links with Chromosome Architecture, Heterochromatin, and Gene Repression. Cell 2017;169(5):780–91. doi: 10.1016/j.cell.2017.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ramdas NM, Shivashankar GV. Cytoskeletal control of nuclear morphology and chromatin organization. J Mol Biol 2015;427(3):695–706. doi: 10.1016/j.jmb.2014.09.008. [DOI] [PubMed] [Google Scholar]
- 65.Toh KC, Ramdas NM, Shivashankar GV. Actin cytoskeleton differentially alters the dynamics of lamin A, HP1alpha and H2B core histone proteins to remodel chromatin condensation state in living cells. Integr Biol (Camb) 2015;7(10):1309–17. doi: 10.1039/c5ib00027k. [DOI] [PubMed] [Google Scholar]
- 66.Jahed Z, Soheilypour M, Peyro M, Mofrad MR. The LINC and NPC relationship - it’s complicated! Journal of cell science 2016;129(17):3219–29. doi: 10.1242/jcs.184184. [DOI] [PubMed] [Google Scholar]
- 67.Mahmood SR, Xie X, Hosny El Said N, Venit T, Gunsalus KC, Percipalle P. beta-actin dependent chromatin remodeling mediates compartment level changes in 3D genome architecture. Nat Commun 2021;12(1):5240. doi: 10.1038/s41467-021-25596-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Allen MD, Bycroft M, Zinzalla G. Structure of the BRK domain of the SWI/SNF chromatin remodeling complex subunit BRG1 reveals a potential role in protein-protein interactions. Protein Sci 2020;29(4):1047–53. doi: 10.1002/pro.3820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Daubresse G, Deuring R, Moore L, Papoulas O, Zakrajsek I, Waldrip WR, et al. The Drosophila kismet gene is related to chromatin-remodeling factors and is required for both segmentation and segment identity. Development 1999;126(6):1175–87. [DOI] [PubMed] [Google Scholar]
- 70.Valletta M, Russo R, Baglivo I, Russo V, Ragucci S, Sandomenico A, et al. Exploring the Interaction between the SWI/SNF Chromatin Remodeling Complex and the Zinc Finger Factor CTCF. Int J Mol Sci 2020;21(23). doi: 10.3390/ijms21238950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rhodes J, Mazza D, Nasmyth K, Uphoff S. Scc2/Nipbl hops between chromosomal cohesin rings after loading. Elife 2017;6. doi: 10.7554/eLife.30000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhu Y, Denholtz M, Lu H, Murre C. Calcium signaling instructs NIPBL recruitment at active enhancers and promoters via distinct mechanisms to reconstruct genome compartmentalization. Genes Dev 2021;35(1–2):65–81. doi: 10.1101/gad.343475.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Barutcu AR, Hong D, Lajoie BR, McCord RP, van Wijnen AJ, Lian JB, et al. RUNX1 contributes to higher-order chromatin organization and gene regulation in breast cancer cells. Biochim Biophys Acta 2016;1859(11):1389–97. doi: 10.1016/j.bbagrm.2016.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Feng Y, Pauklin S. Revisiting 3D chromatin architecture in cancer development and progression. Nucleic Acids Res 2020;48(19):10632–47. doi: 10.1093/nar/gkaa747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Feng Y, Liu X, Pauklin S. 3D chromatin architecture and epigenetic regulation in cancer stem cells. Protein Cell 2021;12(6):440–54. doi: 10.1007/s13238-020-00819-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fudenberg G, Getz G, Meyerson M, Mirny LA. High order chromatin architecture shapes the landscape of chromosomal alterations in cancer. Nat Biotechnol 2011;29(12):1109–13. doi: 10.1038/nbt.2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Johnstone SE, Reyes A, Qi Y, Adriaens C, Hegazi E, Pelka K, et al. Large-Scale Topological Changes Restrain Malignant Progression in Colorectal Cancer. Cell 2020;182(6):1474–89 e23. doi: 10.1016/j.cell.2020.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Senigagliesi B, Penzo C, Severino LU, Maraspini R, Petrosino S, Morales-Navarrete H, et al. The High Mobility Group A1 (HMGA1) Chromatin Architectural Factor Modulates Nuclear Stiffness in Breast Cancer Cells. Int J Mol Sci 2019;20(11). doi: 10.3390/ijms20112733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yun J, Song SH, Kim HP, Han SW, Yi EC, Kim TY. Dynamic cohesin-mediated chromatin architecture controls epithelial-mesenchymal plasticity in cancer. EMBO Rep 2016;17(9):1343–59. doi: 10.15252/embr.201541852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yusufova N, Kloetgen A, Teater M, Osunsade A, Camarillo JM, Chin CR, et al. Histone H1 loss drives lymphoma by disrupting 3D chromatin architecture. Nature 2021;589(7841):299–305. doi: 10.1038/s41586-020-3017-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zeitz MJ, Ay F, Heidmann JD, Lerner PL, Noble WS, Steelman BN, et al. Genomic interaction profiles in breast cancer reveal altered chromatin architecture. PLoS One 2013;8(9):e73974. doi: 10.1371/journal.pone.0073974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhou Y, Gerrard DL, Wang J, Li T, Yang Y, Fritz AJ, et al. Temporal dynamic reorganization of 3D chromatin architecture in hormone-induced breast cancer and endocrine resistance. Nature communications 2019;10(1):1522. doi: 10.1038/s41467-019-09320-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Klemm SL, Shipony Z, Greenleaf WJ. Chromatin accessibility and the regulatory epigenome. Nat Rev Genet 2019;20(4):207–20. doi: 10.1038/s41576-018-0089-8. [DOI] [PubMed] [Google Scholar]
- 84.Narlikar GJ, Sundaramoorthy R, Owen-Hughes T. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell 2013;154(3):490–503. doi: 10.1016/j.cell.2013.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bartholomew B ISWI chromatin remodeling: one primary actor or a coordinated effort? Curr Opin Struct Biol 2014;24:150–5. doi: 10.1016/j.sbi.2014.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gerhold CB, Gasser SM. INO80 and SWR complexes: relating structure to function in chromatin remodeling. Trends in cell biology 2014;24(11):619–31. doi: 10.1016/j.tcb.2014.06.004. [DOI] [PubMed] [Google Scholar]
- 87.Hota SK, Bruneau BG. ATP-dependent chromatin remodeling during mammalian development. Development 2016;143(16):2882–97. doi: 10.1242/dev.128892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mashtalir N, D’Avino AR, Michel BC, Luo J, Pan J, Otto JE, et al. Modular Organization and Assembly of SWI/SNF Family Chromatin Remodeling Complexes. Cell 2018;175(5):1272–88 e20. doi: 10.1016/j.cell.2018.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sims JK, Wade PA. SnapShot: Chromatin remodeling: CHD. Cell 2011;144(4):626–e1. doi: 10.1016/j.cell.2011.02.019. [DOI] [PubMed] [Google Scholar]
- 90.Ayala R, Willhoft O, Aramayo RJ, Wilkinson M, McCormack EA, Ocloo L, et al. Structure and regulation of the human INO80-nucleosome complex. Nature 2018;556(7701):391–5. doi: 10.1038/s41586-018-0021-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.He S, Wu Z, Tian Y, Yu Z, Yu J, Wang X, et al. Structure of nucleosome-bound human BAF complex. Science (New York, NY) 2020;367(6480):875–81. doi: 10.1126/science.aaz9761. [DOI] [PubMed] [Google Scholar]
- 92.Mashtalir N, Suzuki H, Farrell DP, Sankar A, Luo J, Filipovski M, et al. A Structural Model of the Endogenous Human BAF Complex Informs Disease Mechanisms. Cell 2020;183(3):802–17 e24. doi: 10.1016/j.cell.2020.09.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cao T, Sun L, Jiang Y, Huang S, Wang J, Chen Z. Crystal structure of a nuclear actin ternary complex. Proceedings of the National Academy of Sciences of the United States of America 2016;113(32):8985–90. doi: 10.1073/pnas.1602818113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Spector I, Shochet NR, Kashman Y, Groweiss A. Latrunculins: novel marine toxins that disrupt microfilament organization in cultured cells. Science (New York, NY) 1983;219(4584):493–5. doi: 10.1126/science.6681676. [DOI] [PubMed] [Google Scholar]
- 95.Zhao K, Wang W, Rando OJ, Xue Y, Swiderek K, Kuo A, et al. Rapid and phosphoinositol-dependent binding of the SWI/SNF-like BAF complex to chromatin after T lymphocyte receptor signaling. Cell 1998;95(5):625–36. doi: 10.1016/s0092-8674(00)81633-5. [DOI] [PubMed] [Google Scholar]
- 96.Kapoor P, Chen M, Winkler DD, Luger K, Shen X. Evidence for monomeric actin function in INO80 chromatin remodeling. Nat Struct Mol Biol 2013;20(4):426–32. doi: 10.1038/nsmb.2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Vogel Ciernia A, Kramar EA, Matheos DP, Havekes R, Hemstedt TJ, Magnan CN, et al. Mutation of neuron-specific chromatin remodeling subunit BAF53b: rescue of plasticity and memory by manipulating actin remodeling. Learn Mem 2017;24(5):199–209. doi: 10.1101/lm.044602.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mittal P, Roberts CWM. The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol 2020;17(7):435–48. doi: 10.1038/s41571-020-0357-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.DelBove J, Rosson G, Strobeck M, Chen J, Archer TK, Wang W, et al. Identification of a core member of the SWI/SNF complex, BAF155/SMARCC1, as a human tumor suppressor gene. Epigenetics 2011;6(12):1444–53. doi: 10.4161/epi.6.12.18492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Smith MJ, Wallace AJ, Bennett C, Hasselblatt M, Elert-Dobkowska E, Evans LT, et al. Germline SMARCE1 mutations predispose to both spinal and cranial clear cell meningiomas. J Pathol 2014;234(4):436–40. doi: 10.1002/path.4427. [DOI] [PubMed] [Google Scholar]
- 101.Balinas-Gavira C, Rodriguez MI, Andrades A, Cuadros M, Alvarez-Perez JC, Alvarez-Prado AF, et al. Frequent mutations in the amino-terminal domain of BCL7A impair its tumor suppressor role in DLBCL. Leukemia 2020;34(10):2722–35. doi: 10.1038/s41375-020-0919-5. [DOI] [PubMed] [Google Scholar]
- 102.Concepcion CP, Ma S, LaFave LM, Bhutkar A, Liu M, DeAngelo LP, et al. SMARCA4 inactivation promotes lineage-specific transformation and early metastatic features in the lung. Cancer Discov 2021. doi: 10.1158/2159-8290.CD-21-0248. [DOI] [PMC free article] [PubMed]
- 103.Xu G, Chhangawala S, Cocco E, Razavi P, Cai Y, Otto JE, et al. ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat Genet 2020;52(2):198–207. doi: 10.1038/s41588-019-0554-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Liu XD, Kong W, Peterson CB, McGrail DJ, Hoang A, Zhang X, et al. PBRM1 loss defines a nonimmunogenic tumor phenotype associated with checkpoint inhibitor resistance in renal carcinoma. Nature communications 2020;11(1):2135. doi: 10.1038/s41467-020-15959-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Saladi SV, Ross K, Karaayvaz M, Tata PR, Mou H, Rajagopal J, et al. ACTL6A Is Co-Amplified with p63 in Squamous Cell Carcinoma to Drive YAP Activation, Regenerative Proliferation, and Poor Prognosis. Cancer Cell 2017;31(1):35–49. doi: 10.1016/j.ccell.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Del Gaudio N, Di Costanzo A, Liu NQ, Conte L, Migliaccio A, Vermeulen M, et al. BRD9 binds cell type-specific chromatin regions regulating leukemic cell survival via STAT5 inhibition. Cell Death Dis 2019;10(5):338. doi: 10.1038/s41419-019-1570-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Fernando TM, Piskol R, Bainer R, Sokol ES, Trabucco SE, Zhang Q, et al. Functional characterization of SMARCA4 variants identified by targeted exome-sequencing of 131,668 cancer patients. Nature communications 2020;11(1):5551. doi: 10.1038/s41467-020-19402-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hodges HC, Stanton BZ, Cermakova K, Chang CY, Miller EL, Kirkland JG, et al. Dominant-negative SMARCA4 mutants alter the accessibility landscape of tissue-unrestricted enhancers. Nat Struct Mol Biol 2018;25(1):61–72. doi: 10.1038/s41594-017-0007-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hoffmeister H, Fuchs A, Strobl L, Sprenger F, Grobner-Ferreira R, Michaelis S, et al. Elucidation of the functional roles of the Q and I motifs in the human chromatin-remodeling enzyme BRG1. The Journal of biological chemistry 2019;294(9):3294–310. doi: 10.1074/jbc.RA118.005685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Schick S, Grosche S, Kohl KE, Drpic D, Jaeger MG, Marella NC, et al. Acute BAF perturbation causes immediate changes in chromatin accessibility. Nat Genet 2021;53(3):269–78. doi: 10.1038/s41588-021-00777-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Schoenfeld AJ, Bandlamudi C, Lavery JA, Montecalvo J, Namakydoust A, Rizvi H, et al. The Genomic Landscape of SMARCA4 Alterations and Associations with Outcomes in Patients with Lung Cancer. Clin Cancer Res 2020;26(21):5701–8. doi: 10.1158/1078-0432.CCR-20-1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Aramayo RJ, Willhoft O, Ayala R, Bythell-Douglas R, Wigley DB, Zhang X. Cryo-EM structures of the human INO80 chromatin-remodeling complex. Nat Struct Mol Biol 2018;25(1):37–44. doi: 10.1038/s41594-017-0003-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chen L, Conaway RC, Conaway JW. Multiple modes of regulation of the human Ino80 SNF2 ATPase by subunits of the INO80 chromatin-remodeling complex. Proceedings of the National Academy of Sciences of the United States of America 2013;110(51):20497–502. doi: 10.1073/pnas.1317092110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Guo DC, Duan XY, Regalado ES, Mellor-Crummey L, Kwartler CS, Kim D, et al. Loss-of-Function Mutations in YY1AP1 Lead to Grange Syndrome and a Fibromuscular Dysplasia-Like Vascular Disease. Am J Hum Genet 2017;100(1):21–30. doi: 10.1016/j.ajhg.2016.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wu S, Shi Y, Mulligan P, Gay F, Landry J, Liu H, et al. A YY1-INO80 complex regulates genomic stability through homologous recombination-based repair. Nat Struct Mol Biol 2007;14(12):1165–72. doi: 10.1038/nsmb1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yao W, Beckwith SL, Zheng T, Young T, Dinh VT, Ranjan A, et al. Assembly of the Arp5 (Actin-related Protein) Subunit Involved in Distinct INO80 Chromatin Remodeling Activities. The Journal of biological chemistry 2015;290(42):25700–9. doi: 10.1074/jbc.M115.674887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wu J, Lao Y, Li B. Nuclear actin switch of the INO80 remodeler. J Mol Cell Biol 2019;11(5):343–4. doi: 10.1093/jmcb/mjy083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhang X, Wang X, Zhang Z, Cai G. Structure and functional interactions of INO80 actin/Arp module. J Mol Cell Biol 2019;11(5):345–55. doi: 10.1093/jmcb/mjy062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Knoll KR, Eustermann S, Niebauer V, Oberbeckmann E, Stoehr G, Schall K, et al. The nuclear actin-containing Arp8 module is a linker DNA sensor driving INO80 chromatin remodeling. Nat Struct Mol Biol 2018;25(9):823–32. doi: 10.1038/s41594-018-0115-8. [DOI] [PubMed] [Google Scholar]
- 120.Saravanan M, Wuerges J, Bose D, McCormack EA, Cook NJ, Zhang X, et al. Interactions between the nucleosome histone core and Arp8 in the INO80 chromatin remodeling complex. Proceedings of the National Academy of Sciences of the United States of America 2012;109(51):20883–8. doi: 10.1073/pnas.1214735109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Udugama M, Sabri A, Bartholomew B. The INO80 ATP-dependent chromatin remodeling complex is a nucleosome spacing factor. Mol Cell Biol 2011;31(4):662–73. doi: 10.1128/MCB.01035-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Takahashi Y, Murakami H, Akiyama Y, Katoh Y, Oma Y, Nishijima H, et al. Actin Family Proteins in the Human INO80 Chromatin Remodeling Complex Exhibit Functional Roles in the Induction of Heme Oxygenase-1 with Hemin. Front Genet 2017;8:17. doi: 10.3389/fgene.2017.00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Morrison AJ, Shen X. Chromatin remodelling beyond transcription: the INO80 and SWR1 complexes. Nature reviews Molecular cell biology 2009;10(6):373–84. doi: 10.1038/nrm2693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Roadmap Epigenomics C, Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, et al. Integrative analysis of 111 reference human epigenomes. Nature 2015;518(7539):317–30. doi: 10.1038/nature14248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zhang S, Zhou B, Wang L, Li P, Bennett BD, Snyder R, et al. INO80 is required for oncogenic transcription and tumor growth in non-small cell lung cancer. Oncogene 2017;36(10):1430–9. doi: 10.1038/onc.2016.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhou B, Wang L, Zhang S, Bennett BD, He F, Zhang Y, et al. INO80 governs superenhancer-mediated oncogenic transcription and tumor growth in melanoma. Genes Dev 2016;30(12):1440–53. doi: 10.1101/gad.277178.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Prendergast L, McClurg UL, Hristova R, Berlinguer-Palmini R, Greener S, Veitch K, et al. Resolution of R-loops by INO80 promotes DNA replication and maintains cancer cell proliferation and viability. Nature communications 2020;11(1):4534. doi: 10.1038/s41467-020-18306-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Moon SW, Mo HY, Choi EJ, Yoo NJ, Lee SH. Cancer-related SRCAP and TPR mutations in colon cancers. Pathology, research and practice 2021;217:153292. doi: 10.1016/j.prp.2020.153292. [DOI] [PubMed] [Google Scholar]
- 129.Slupianek A, Yerrum S, Safadi FF, Monroy MA. The chromatin remodeling factor SRCAP modulates expression of prostate specific antigen and cellular proliferation in prostate cancer cells. J Cell Physiol 2010;224(2):369–75. doi: 10.1002/jcp.22132. [DOI] [PubMed] [Google Scholar]
- 130.Jethwa A, Słabicki M, Hüllein J, Jentzsch M, Dalal V, Rabe S, et al. TRRAP is essential for regulating the accumulation of mutant and wild-type p53 in lymphoma. Blood 2018;131(25):2789–802. doi: 10.1182/blood-2017-09-806679. [DOI] [PubMed] [Google Scholar]
- 131.Wurdak H, Zhu S, Romero A, Lorger M, Watson J, Chiang CY, et al. An RNAi screen identifies TRRAP as a regulator of brain tumor-initiating cell differentiation. Cell Stem Cell 2010;6(1):37–47. doi: 10.1016/j.stem.2009.11.002. [DOI] [PubMed] [Google Scholar]
- 132.Kang KT, Kwon YW, Kim DK, Lee SI, Kim K-H, Suh D-S, et al. TRRAP stimulates the tumorigenic potential of ovarian cancer stem cells. BMB Reports 2018;51(10):514–9. doi: 10.5483/BMBRep.2018.51.10.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kwan SY, Sheel A, Song CQ, Zhang XO, Jiang T, Dang H, et al. Depletion of TRRAP Induces p53-Independent Senescence in Liver Cancer by Down-Regulating Mitotic Genes. Hepatology 2020;71(1):275–90. doi: 10.1002/hep.30807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wang J, Shan M, Liu T, Shi Q, Zhong Z, Wei W, et al. Analysis of TRRAP as a Potential Molecular Marker and Therapeutic Target for Breast Cancer. J Breast Cancer 2016;19(1):61–7. doi: 10.4048/jbc.2016.19.1.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Egly JM, Miyamoto NG, Moncollin V, Chambon P. Is Actin a Transcription Initiation-Factor for Rna Polymerase-B. Embo J 1984;3(10):2363–71. doi: DOI 10.1002/j.1460-2075.1984.tb02141.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Scheer U, Hinssen H, Franke WW, Jockusch BM. Microinjection of actin-binding proteins and actin antibodies demonstrates involvement of nuclear actin in transcription of lampbrush chromosomes. Cell 1984;39(1):111–22. doi: 10.1016/0092-8674(84)90196-x. [DOI] [PubMed] [Google Scholar]
- 137.Percipalle P Co-transcriptional nuclear actin dynamics. Nucleus 2013;4(1):43–52. doi: 10.4161/nucl.22798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kyheroinen S, Vartiainen MK. Nuclear actin dynamics in gene expression and genome organization. Seminars in Cell & Developmental Biology 2020;102:105–12. doi: 10.1016/j.semcdb.2019.10.012. [DOI] [PubMed] [Google Scholar]
- 139.Miralles F, Posern G, Zaromytidou A-I, Treisman R. Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell 2003;113(3):329–42. [DOI] [PubMed] [Google Scholar]
- 140.Cen B, Selvaraj A, Prywes R. Myocardin/MKL family of SRF coactivators: Key regulators of immediate early and muscle specific gene expression. J Cell Biochem 2004;93(1):74–82. doi: 10.1002/jcb.20199. [DOI] [PubMed] [Google Scholar]
- 141.Han Z, Li XM, Wu J, Olson EN. A myocardin-related transcription factor regulates activity of serum response factor in Drosophila. P Natl Acad Sci USA 2004;101(34):12567–72. doi: DOI 10.1073/pnas.0405085101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Posern G, Treisman R. Actin’ together: serum response factor, its cofactors and the link to signal transduction. Trends Cell Biol 2006;16(11):588–96. doi: 10.1016/j.tcb.2006.09.008. [DOI] [PubMed] [Google Scholar]
- 143.Mouilleron S, Langer CA, Guettler S, McDonald NQ, Treisman R. Structure of a Pentavalent G-Actin.MRTF-A Complex Reveals How G-Actin Controls Nucleocytoplasmic Shuttling of a Transcriptional Coactivator. Science Signaling 2011;4(177). doi: ARTN ra40 10.1126/scisignal.2001750. [DOI] [PubMed] [Google Scholar]
- 144.Panayiotou R, Miralles F, Pawlowski R, Diring J, Flynn HR, Skehel M, et al. Phosphorylation acts positively and negatively to regulate MRTF-A subcellular localisation and activity. Elife 2016;5. doi: ARTN e15460 10.7554/eLife.15460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Vartiainen MK, Guettler S, Larijani B, Treisman R. Nuclear actin regulates dynamic subcellular localization and activity of the SRF cofactor MAL. Science 2007;316(5832):1749–52. doi: 10.1126/science.1141084. [DOI] [PubMed] [Google Scholar]
- 146.Mouilleron S, Langer CA, Guettler S, McDonald NQ, Treisman R. Structure of a Pentavalent G-Actin•MRTF-A Complex Reveals How G-Actin Controls Nucleocytoplasmic Shuttling of a Transcriptional Coactivator. Science Signaling 2011;4(177):ra40–ra. doi: doi: 10.1126/scisignal.2001750. [DOI] [PubMed] [Google Scholar]
- 147.Hirano H, Matsuura Y. Sensing actin dynamics: structural basis for G-actin-sensitive nuclear import of MAL. Biochem Biophys Res Commun 2011;414(2):373–8. doi: 10.1016/j.bbrc.2011.09.079. [DOI] [PubMed] [Google Scholar]
- 148.Pawlowski R, Rajakyla EK, Vartiainen MK, Treisman R. An actin-regulated importin alpha/beta-dependent extended bipartite NLS directs nuclear import of MRTF-A. Embo J 2010;29(20):3448–58. doi: 10.1038/emboj.2010.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Vartiainen MK, Guettler S, Larijani B, Treisman R. Nuclear actin regulates dynamic subcellular localization and activity of the SRF cofactor MAL. Science (New York, NY) 2007;316(5832):1749–52. doi: 10.1126/science.1141084. [DOI] [PubMed] [Google Scholar]
- 150.Plessner M, Melak M, Chinchilla P, Baarlink C, Grosse R. Nuclear F-actin Formation and Reorganization upon Cell Spreading*♦. J Biol Chem 2015;290(18):11209–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lundquist MR, Storaska AJ, Liu T-C, Larsen SD, Evans T, Neubig RR, et al. Redox modification of nuclear actin by MICAL-2 regulates SRF signaling. Cell 2014;156(3):563–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.McNeill MC, Wray J, Sala-Newby GB, Hindmarch CC, Smith SA, Ebrahimighaei R, et al. Nuclear actin regulates cell proliferation and migration via inhibition of SRF and TEAD. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 2020;1867(7):118691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Chatzifrangkeskou M, Pefani DE, Eyres M, Vendrell I, Fischer R, Pankova D, et al. RASSF 1A is required for the maintenance of nuclear actin levels. The EMBO journal 2019;38(16):e101168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Wang D-Z, Li S, Hockemeyer D, Sutherland L, Wang Z, Schratt G, et al. Potentiation of serum response factor activity by a family of myocardin-related transcription factors. Proceedings of the National Academy of Sciences 2002;99(23):14855–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kuwahara K, Barrientos T, Pipes GT, Li S, Olson EN. Muscle-specific signaling mechanism that links actin dynamics to serum response factor. Mol Cell Biol 2005;25(8):3173–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Ki Hayashi, Murai T, Oikawa H, Masuda T, Kimura K, Muehlich S, et al. A novel inhibitory mechanism of MRTF-A/B on the ICAM-1 gene expression in vascular endothelial cells. Sci Rep-Uk 2015;5(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Panayiotou R, Miralles F, Pawlowski R, Diring J, Flynn HR, Skehel M, et al. Phosphorylation acts positively and negatively to regulate MRTF-A subcellular localisation and activity. Elife 2016;5:e15460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Posern G, Treisman R. Actin’together: serum response factor, its cofactors and the link to signal transduction. Trends in cell biology 2006;16(11):588–96. [DOI] [PubMed] [Google Scholar]
- 159.Ulferts S, Prajapati B, Grosse R, Vartiainen MK. Emerging Properties and Functions of Actin and Actin Filaments Inside the Nucleus. Cold Spring Harb Perspect Biol 2021;13(3). doi: 10.1101/cshperspect.a040121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kuhn C-D, Geiger SR, Baumli S, Gartmann M, Gerber J, Jennebach S, et al. Functional architecture of RNA polymerase I. Cell 2007;131(7):1260–72. [DOI] [PubMed] [Google Scholar]
- 161.Hofmann WA, Stojiljkovic L, Fuchsova B, Vargas GM, Mavrommatis E, Philimonenko V, et al. Actin is part of pre-initiation complexes and is necessary for transcription by RNA polymerase II. Nat Cell Biol 2004;6(11):1094–101. doi: 10.1038/ncb1182. [DOI] [PubMed] [Google Scholar]
- 162.Hu P, Wu S, Hernandez N. A role for beta-actin in RNA polymerase III transcription. Gene Dev 2004;18(24):3010–5. doi: 10.1101/gad.1250804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Philimonenko VV, Zhao J, Iben S, Dingova H, Kysela K, Kahle M, et al. Nuclear actin and myosin I are required for RNA polymerase I transcription. Nature Cell Biology 2004;6(12):1165–72. doi: 10.1038/ncb1190. [DOI] [PubMed] [Google Scholar]
- 164.Stojiljkovic L, Fan JL, Goodrich J, Raychaudhuri P, Lessard J, de Lanerolle P. Nuclear myosin I and beta-actin are required for transcription by RNA polymerase II. Mol Biol Cell 2001;12:353a–a. [Google Scholar]
- 165.Fomproix N, Percipalle P. An actin–myosin complex on actively transcribing genes. Experimental cell research 2004;294(1):140–8. [DOI] [PubMed] [Google Scholar]
- 166.Ye J, Zhao J, Hoffmann-Rohrer U, Grummt I. Nuclear myosin I acts in concert with polymeric actin to drive RNA polymerase I transcription. Gene Dev 2008;22(3):322–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Philimonenko VV, Zhao J, Iben S, Dingova H, Kysela K, Kahle M, et al. Nuclear actin and myosin I are required for RNA polymerase I transcription. Nat Cell Biol 2004;6(12):1165–72. doi: 10.1038/ncb1190. [DOI] [PubMed] [Google Scholar]
- 168.Ye J, Zhao J, Hoffmann-Rohrer U, Grummt I. Nuclear myosin I acts in concert with polymeric actin to drive RNA polymerase I transcription. Genes Dev 2008;22(3):322–30. doi: 10.1101/gad.455908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Posern G, Sotiropoulos A, Treisman R. Mutant actins demonstrate a role for unpolymerized actin in control of transcription by serum response factor. Mol Biol Cell 2002;13(12):4167–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Grummt I. Actin and myosin as transcription factors. Current opinion in genetics & development 2006;16(2):191–6. [DOI] [PubMed] [Google Scholar]
- 171.Percipalle P, Fomproix N, Cavellán E, Voit R, Reimer G, Krüger T, et al. The chromatin remodelling complex WSTF–SNF2h interacts with nuclear myosin 1 and has a role in RNA polymerase I transcription. Embo Rep 2006;7(5):525–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Scheer U, Hinssen H, Franke WW, Jockusch BM. Microinjection of actin-binding proteins and actin antibodies demonstrates involvement of nuclear actin in transcription of lampbrush chromosomes. Cell 1984;39(1):111–22. [DOI] [PubMed] [Google Scholar]
- 173.Viita T, Kyheroinen S, Prajapati B, Virtanen J, Frilander MJ, Varjosalo M, et al. Nuclear actin interactome analysis links actin to KAT14 histone acetyl transferase and mRNA splicing. J Cell Sci 2019;132(8). doi: 10.1242/jcs.226852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Sokolova M, Moore HM, Prajapati B, Dopie J, Meriläinen L, Honkanen M, et al. Nuclear actin is required for transcription during Drosophila oogenesis. Iscience 2018;9:63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Hu P, Wu S, Hernandez N. A role for beta-actin in RNA polymerase III transcription. Genes Dev 2004;18(24):3010–5. doi: 10.1101/gad.1250804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Qi T, Tang W, Wang L, Zhai L, Guo L, Zeng X. G-actin participates in RNA polymerase II-dependent transcription elongation by recruiting positive transcription elongation factor b (P-TEFb). J Biol Chem 2011;286(17):15171–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Tang W, You W, Shi F, Qi T, Wang L, Djouder Z, et al. RNA helicase A acts as a bridging factor linking nuclear β-actin with RNA polymerase II. Biochem J 2009;420(3):421–8. [DOI] [PubMed] [Google Scholar]
- 178.Pederson T As functional nuclear actin comes into view, is it globular, filamentous, or both? The Journal of cell biology 2008;180(6):1061–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Schmidt EE, Bondareva AA, Radke JR, Capecchi MR. Fundamental cellular processes do not require vertebrate-specific sequences within the TATA-binding protein. J Biol Chem 2003;278(8):6168–74. [DOI] [PubMed] [Google Scholar]
- 180.Johnston IM, Allison SJ, Morton JP, Schramm L, Scott PH, White RJ. CK2 forms a stable complex with TFIIIB and activates RNA polymerase III transcription in human cells. Mol Cell Biol 2002;22(11):3757–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Reiner R, Ben-Asouli Y, Krilovetzky I, Jarrous N. A role for the catalytic ribonucleoprotein RNase P in RNA polymerase III transcription. Gene Dev 2006;20(12):1621–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Miyamoto K, Teperek M, Yusa K, Allen GE, Bradshaw CR, Gurdon J. Nuclear Wave1 is required for reprogramming transcription in oocytes and for normal development. Science 2013;341(6149):1002–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wu X, Yoo Y, Okuhama NN, Tucker PW, Liu G, Guan J-L. Regulation of RNA-polymerase-II-dependent transcription by N-WASP and its nuclear-binding partners. Nature cell biology 2006;8(7):756–63. [DOI] [PubMed] [Google Scholar]
- 184.Xia P, Wang S, Huang G, Zhu P, Li M, Ye B, et al. WASH is required for the differentiation commitment of hematopoietic stem cells in a c-Myc–dependent mannerWASH is essential for LT-HSC differentiation. The Journal of experimental medicine 2014;211(10):2119–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Yoo Y, Wu X, Guan J-L. A novel role of the actin-nucleating Arp2/3 complex in the regulation of RNA polymerase II-dependent transcription. J Biol Chem 2007;282(10):7616–23. [DOI] [PubMed] [Google Scholar]
- 186.Wei M, Fan X, Ding M, Li R, Shao S, Hou Y, et al. Nuclear actin regulates inducible transcription by enhancing RNA polymerase II clustering. Sci Adv 2020;6(16):eaay6515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Brunel C, Lelay MN. Two-dimensional analysis of proteins associated with heterogenous nuclear RNA in various animal cell lines. Eur J Biochem 1979;99(2):273–83. doi: 10.1111/j.1432-1033.1979.tb13254.x. [DOI] [PubMed] [Google Scholar]
- 188.Obrdlik A, Kukalev A, Louvet E, Östlund Farrants A-K, Caputo L, Percipalle P. The histone acetyltransferase PCAF associates with actin and hnRNP U for RNA polymerase II transcription. Mol Cell Biol 2008;28(20):6342–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Percipalle P, Jonsson A, Nashchekin D, Karlsson C, Bergman T, Guialis A, et al. Nuclear actin is associated with a specific subset of hnRNP A/B-type proteins. Nucleic Acids Res 2002;30(8):1725–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Kumarasinghe N, Moss WN. Analysis of a structured intronic region of the LMP2 pre-mRNA from EBV reveals associations with human regulatory proteins and nuclear actin. Bmc Res Notes 2019;12(1):1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.PAGOULATOS GN, YANIV M. Proteins Bound to Heterogeneous Nuclear RNA of Siman‐Virus‐40‐Infected Cells. European journal of biochemistry 1978;91(1):1–10. [DOI] [PubMed] [Google Scholar]
- 192.Gounon P, Karsenti E. Involvement of contractile proteins in the changes in consistency of oocyte nucleoplasm of the newt Pleurodeles waltlii. The Journal of cell biology 1981;88(2):410–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.MAUNDRELL K, SCHERRER K. Characterization of pre‐messenger‐RNA‐containing nuclear ribonucleoprotein particles from avian erythroblasts. European journal of biochemistry 1979;99(2):225–38. [DOI] [PubMed] [Google Scholar]
- 194.Percipalle P, Fomproix N, Kylberg K, Miralles F, Björkroth B, Daneholt B, et al. An actin–ribonucleoprotein interaction is involved in transcription by RNA polymerase II. Proceedings of the National Academy of Sciences 2003;100(11):6475–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Kukalev A, Nord Y, Palmberg C, Bergman T, Percipalle P. Actin and hnRNP U cooperate for productive transcription by RNA polymerase II. Nat Struct Mol Biol 2005;12(3):238–44. [DOI] [PubMed] [Google Scholar]
- 196.Sjölinder M, Björk P, Söderberg E, Sabri N, Farrants A-KÖ, Visa N. The growing pre-mRNA recruits actin and chromatin-modifying factors to transcriptionally active genes. Gene Dev 2005;19(16):1871–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Fultang N, Peethambaran B. Wnt Signaling in Breast Cancer Oncogenesis, Development and Progression. Advances in Cancer Signal Transduction and Therapy Bentham Science Publishes 2020:1–28.
- 198.Jung YS, Park JI. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond beta-catenin and the destruction complex. Exp Mol Med 2020;52(2):183–91. doi: 10.1038/s12276-020-0380-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Nusse R, Clevers H. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell 2017;169(6):985–99. [DOI] [PubMed] [Google Scholar]
- 200.Daugherty RL, Serebryannyy L, Yemelyanov A, Flozak AS, Yu H-J, Kosak ST, et al. α-Catenin is an inhibitor of transcription. Proceedings of the National Academy of Sciences 2014;111(14):5260–5. doi: 10.1073/pnas.1308663111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Okada K, Bartolini F, Deaconescu AM, Moseley JB, Dogic Z, Grigorieff N, et al. Adenomatous polyposis coli protein nucleates actin assembly and synergizes with the formin mDia1. Journal of Cell Biology 2010;189(7):1087–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Yamazaki S, Yamamoto K, de Lanerolle P, Harata M. Nuclear F-actin enhances the transcriptional activity of β-catenin by increasing its nuclear localization and binding to chromatin. Histochem Cell Biol 2016;145(4):389–99. [DOI] [PubMed] [Google Scholar]
- 203.Fodde R, Smits R, Clevers H. APC, signal transduction and genetic instability in colorectal cancer. Nature reviews cancer 2001;1(1):55–67. [DOI] [PubMed] [Google Scholar]
- 204.Polakis P Wnt signaling and cancer. Gene Dev 2000;14(15):1837–51. [PubMed] [Google Scholar]
- 205.De la Roche M, Worm J, Bienz M. The function of BCL9 in Wnt/β-catenin signaling and colorectal cancer cells. BMC cancer 2008;8(1):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Neufeld KL. Nuclear APC. APC Proteins 2009:13–29. [DOI] [PMC free article] [PubMed]
- 207.Rosin-Arbesfeld R, Townsley F, Bienz M. The APC tumour suppressor has a nuclear export function. Nature 2000;406(6799):1009–12. [DOI] [PubMed] [Google Scholar]
- 208.Baarlink C, Wang H, Grosse R. Nuclear actin network assembly by formins regulates the SRF coactivator MAL. Science 2013;340(6134):864–7. [DOI] [PubMed] [Google Scholar]
- 209.Timoféeff-Ressovsky N, Zimmer K, Delbrück M. Über die Natur der Genmutation und der Genstruktur 1935.
- 210.Dulbecco R Reactivation of ultra-violet-inactivated bacteriophage by visible light. Nature 1949;163(4155):949–50. [DOI] [PubMed] [Google Scholar]
- 211.Kelner A Effect of visible light on the recovery of Streptomyces griseus conidia from ultra-violet irradiation injury. P Natl Acad Sci USA 1949;35(2):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Bernstein C, Prasad AR, Nfonsam V, Bernstein H. DNA damage, DNA repair and cancer. New Research Directions in DNA Repair 2013:413–65.
- 213.Friedberg EC. DNA damage and repair. Nature 2003;421(6921):436–40. [DOI] [PubMed] [Google Scholar]
- 214.Chatterjee N, Walker GC. Mechanisms of DNA damage, repair, and mutagenesis. Environmental and molecular mutagenesis 2017;58(5):235–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature 2009;461(7267):1071–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Friedberg EC, Walker GC, Siede W, Wood RD. DNA repair and mutagenesis American Society for Microbiology Press; 2005. [Google Scholar]
- 217.Li X, Heyer W-D. Homologous recombination in DNA repair and DNA damage tolerance. Cell research 2008;18(1):99–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Panier S, Boulton SJ. Double-strand break repair: 53BP1 comes into focus. Nat Rev Mol Cell Bio 2014;15(1):7–18. [DOI] [PubMed] [Google Scholar]
- 219.Chang HH, Pannunzio NR, Adachi N, Lieber MR. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat Rev Mol Cell Bio 2017;18(8):495–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Getts RC, Stamato TD. Absence of a Ku-like DNA end binding activity in the xrs double-strand DNA repair-deficient mutant. J Biol Chem 1994;269(23):15981–4. [PubMed] [Google Scholar]
- 221.Taccioli GE, Gottlieb TM, Blunt T, Priestley A, Demengeot J, Mizuta R, et al. Ku80: product of the XRCC5 gene and its role in DNA repair and V (D) J recombination. Science 1994;265(5177):1442–5. [DOI] [PubMed] [Google Scholar]
- 222.Ma Y, Pannicke U, Schwarz K, Lieber MR. Hairpin opening and overhang processing by an Artemis/DNA-dependent protein kinase complex in nonhomologous end joining and V (D) J recombination. Cell 2002;108(6):781–94. [DOI] [PubMed] [Google Scholar]
- 223.Ahnesorg P, Smith P, Jackson SP. XLF interacts with the XRCC4-DNA ligase IV complex to promote DNA nonhomologous end-joining. Cell 2006;124(2):301–13. [DOI] [PubMed] [Google Scholar]
- 224.Andrade P, Martín MJ, Juárez R, de Saro FL, Blanco L. Limited terminal transferase in human DNA polymerase μ defines the required balance between accuracy and efficiency in NHEJ. Proceedings of the National Academy of Sciences 2009;106(38):16203–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Capp J-P, Boudsocq F, Bertrand P, Laroche-Clary A, Pourquier P, Lopez BS, et al. The DNA polymerase λ is required for the repair of non-compatible DNA double strand breaks by NHEJ in mammalian cells. Nucleic Acids Res 2006;34(10):2998–3007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Johnson RD, Jasin M. Sister chromatid gene conversion is a prominent double‐strand break repair pathway in mammalian cells. The EMBO journal 2000;19(13):3398–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.O’Driscoll M, Jeggo PA. The role of double-strand break repair—insights from human genetics. Nature Reviews Genetics 2006;7(1):45–54. [DOI] [PubMed] [Google Scholar]
- 228.Stracker TH, Petrini JH. The MRE11 complex: starting from the ends. Nat Rev Mol Cell Bio 2011;12(2):90–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Huertas P DNA resection in eukaryotes: deciding how to fix the break. Nat Struct Mol Biol 2010;17(1):11–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Lavin M ATM and the Mre11 complex combine to recognize and signal DNA double-strand breaks. Oncogene 2007;26(56):7749–58. [DOI] [PubMed] [Google Scholar]
- 231.Lavin MF, Kozlov S. ATM activation and DNA damage response. Cell Cycle 2007;6(8):931–42. [DOI] [PubMed] [Google Scholar]
- 232.Sartori AA, Lukas C, Coates J, Mistrik M, Fu S, Bartek J, et al. Human CtIP promotes DNA end resection. Nature 2007;450(7169):509–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.McIlwraith MJ, Van Dyck E, Masson J-Y, Stasiak AZ, Stasiak A, West SC. Reconstitution of the strand invasion step of double-strand break repair using human Rad51 Rad52 and RPA proteins. Journal of molecular biology 2000;304(2):151–64. [DOI] [PubMed] [Google Scholar]
- 234.New JH, Sugiyama T, Zaitseva E, Kowalczykowski SC. Rad52 protein stimulates DNA strand exchange by Rad51 and replication protein A. Nature 1998;391(6665):407–10. [DOI] [PubMed] [Google Scholar]
- 235.Pellegrini L, David SY, Lo T, Anand S, Lee M, Blundell TL, et al. Insights into DNA recombination from the structure of a RAD51–BRCA2 complex. Nature 2002;420(6913):287–93. [DOI] [PubMed] [Google Scholar]
- 236.Kass EM, Jasin M. Collaboration and competition between DNA double-strand break repair pathways. FEBS letters 2010;584(17):3703–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.McIlwraith MJ, Vaisman A, Liu Y, Fanning E, Woodgate R, West SC. Human DNA polymerase η promotes DNA synthesis from strand invasion intermediates of homologous recombination. Molecular cell 2005;20(5):783–92. [DOI] [PubMed] [Google Scholar]
- 238.San Filippo J, Sung P, Klein H. Mechanism of eukaryotic homologous recombination. Annu Rev Biochem 2008;77:229–57. [DOI] [PubMed] [Google Scholar]
- 239.Andrin C, McDonald D, Attwood KM, Rodrigue A, Ghosh S, Mirzayans R, et al. A requirement for polymerized actin in DNA double-strand break repair. Nucleus 2012;3(4):384–95. [DOI] [PubMed] [Google Scholar]
- 240.Belin BJ, Lee T, Mullins RD. DNA damage induces nuclear actin filament assembly by Formin-2 and Spire-1/2 that promotes efficient DNA repair. Elife 2015;4:e07735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Caridi CP, D’Agostino C, Ryu T, Zapotoczny G, Delabaere L, Li X, et al. Nuclear F-actin and myosins drive relocalization of heterochromatic breaks. Nature 2018;559(7712):54–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Schrank BR, Aparicio T, Li Y, Chang W, Chait BT, Gundersen GG, et al. Nuclear ARP2/3 drives DNA break clustering for homology-directed repair. Nature 2018;559(7712):61–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Caridi CP, Plessner M, Grosse R, Chiolo I. Nuclear actin filaments in DNA repair dynamics. Nature Cell Biology 2019;21(9):1068–77. doi: 10.1038/s41556-019-0379-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Guenatri M, Bailly D, Maison C, Almouzni G. Mouse centric and pericentric satellite repeats form distinct functional heterochromatin. Journal of Cell Biology 2004;166(4):493–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Li Q, Tjong H, Li X, Gong K, Zhou XJ, Chiolo I, et al. The three-dimensional genome organization of Drosophila melanogaster through data integration. Genome biology 2017;18(1):1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Riddle NC, Minoda A, Kharchenko PV, Alekseyenko AA, Schwartz YB, Tolstorukov MY, et al. Plasticity in patterns of histone modifications and chromosomal proteins in Drosophila heterochromatin. Genome research 2011;21(2):147–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.See C, Arya D, Lin E, Chiolo I. Live cell imaging of nuclear actin filaments and heterochromatic repair foci in Drosophila and mouse cells. Methods Mol Biol Springer; 2021. p. 459–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Dialynas G, Delabaere L, Chiolo I. Arp2/3 and Unc45 maintain heterochromatin stability in Drosophila polytene chromosomes. Exp Biol Med 2019;244(15):1362–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Chiolo I, Tang J, Georgescu W, Costes SV. Nuclear dynamics of radiation-induced foci in euchromatin and heterochromatin. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 2013;750(1–2):56–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Zahler S Nuclear actin in cancer biology. Int Rev Cel Mol Bio 2020;355:53–66. doi: 10.1016/bs.ircmb.2020.04.001. [DOI] [PubMed] [Google Scholar]
- 251.Schafer DA, Schroer TA. Actin-related proteins. Annu Rev Cell Dev Biol 1999;15:341–63. doi: 10.1146/annurev.cellbio.15.1.341. [DOI] [PubMed] [Google Scholar]
- 252.Goley ED, Welch MD. The ARP2/3 complex: an actin nucleator comes of age. Nature reviews Molecular cell biology 2006;7(10):713–26. doi: 10.1038/nrm2026. [DOI] [PubMed] [Google Scholar]
- 253.Tanaka K, Takeda S, Mitsuoka K, Oda T, Kimura-Sakiyama C, Maeda Y, et al. Structural basis for cofilin binding and actin filament disassembly. Nature communications 2018;9(1):1860. doi: 10.1038/s41467-018-04290-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Breitsprecher D, Goode BL. Formins at a glance. Journal of cell science 2013;126(Pt 1):1–7. doi: 10.1242/jcs.107250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Sun HQ, Yamamoto M, Mejillano M, Yin HL. Gelsolin, a multifunctional actin regulatory protein. The Journal of biological chemistry 1999;274(47):33179–82. doi: 10.1074/jbc.274.47.33179. [DOI] [PubMed] [Google Scholar]
- 256.Manor U, Bartholomew S, Golani G, Christenson E, Kozlov M, Higgs H, et al. A mitochondria-anchored isoform of the actin-nucleating spire protein regulates mitochondrial division. Elife 2015;4. doi: 10.7554/eLife.08828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Krishnan K, Moens PDJ. Structure and functions of profilins. Biophys Rev 2009;1(2):71–81. doi: 10.1007/s12551-009-0010-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Goldstein AL, Hannappel E, Sosne G, Kleinman HK. Thymosin beta4: a multi-functional regenerative peptide. Basic properties and clinical applications. Expert Opin Biol Ther 2012;12(1):37–51. doi: 10.1517/14712598.2012.634793. [DOI] [PubMed] [Google Scholar]
- 259.Traenkle B, Rothbauer U. Under the Microscope: Single-Domain Antibodies for Live-Cell Imaging and Super-Resolution Microscopy. Front Immunol 2017;8:1030. doi: 10.3389/fimmu.2017.01030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Nagasaki A, S TK, Yumoto T, Imaizumi, Yamagishi A, Kim H, et al. The Position of the GFP Tag on Actin Affects the Filament Formation in Mammalian Cells. Cell Struct Funct 2017;42(2):131–40. doi: 10.1247/csf.17016. [DOI] [PubMed] [Google Scholar]
- 261.Lopata A, Hughes R, Tiede C, Heissler SM, Sellers JR, Knight PJ, et al. Affimer proteins for F-actin: novel affinity reagents that label F-actin in live and fixed cells. Scientific reports 2018;8(1):6572. doi: 10.1038/s41598-018-24953-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Xia HJ, Yang G. Inositol 1,4,5-trisphosphate 3-kinases: functions and regulations. Cell Res 2005;15(2):83–91. doi: 10.1038/sj.cr.7290270. [DOI] [PubMed] [Google Scholar]
- 263.Schell MJ, Erneux C, Irvine RF. Inositol 1,4,5-trisphosphate 3-kinase A associates with F-actin and dendritic spines via its N terminus. The Journal of biological chemistry 2001;276(40):37537–46. doi: 10.1074/jbc.M104101200. [DOI] [PubMed] [Google Scholar]
- 264.Riedl J, Crevenna AH, Kessenbrock K, Yu JH, Neukirchen D, Bista M, et al. Lifeact: a versatile marker to visualize F-actin. Nat Methods 2008;5(7):605–7. doi: 10.1038/nmeth.1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Asakura T, Sasaki T, Nagano F, Satoh A, Obaishi H, Nishioka H, et al. Isolation and characterization of a novel actin filament-binding protein from Saccharomyces cerevisiae. Oncogene 1998;16(1):121–30. doi: 10.1038/sj.onc.1201487. [DOI] [PubMed] [Google Scholar]
- 266.Flores LR, Keeling MC, Zhang X, Sliogeryte K, Gavara N. Lifeact-GFP alters F-actin organization, cellular morphology and biophysical behaviour. Scientific reports 2019;9(1):3241. doi: 10.1038/s41598-019-40092-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Coluccio LM, Tilney LG. Phalloidin enhances actin assembly by preventing monomer dissociation. The Journal of cell biology 1984;99(2):529–35. doi: 10.1083/jcb.99.2.529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Lukinavicius G, Reymond L, D’Este E, Masharina A, Gottfert F, Ta H, et al. Fluorogenic probes for live-cell imaging of the cytoskeleton. Nat Methods 2014;11(7):731–3. doi: 10.1038/nmeth.2972. [DOI] [PubMed] [Google Scholar]
- 269.Lukinavicius G, Umezawa K, Olivier N, Honigmann A, Yang G, Plass T, et al. A near-infrared fluorophore for live-cell super-resolution microscopy of cellular proteins. Nat Chem 2013;5(2):132–9. doi: 10.1038/nchem.1546. [DOI] [PubMed] [Google Scholar]
- 270.Bubb MR, Senderowicz AM, Sausville EA, Duncan KL, Korn ED. Jasplakinolide, a cytotoxic natural product, induces actin polymerization and competitively inhibits the binding of phalloidin to F-actin. Journal of Biological Chemistry 1994;269(21):14869–71. doi: 10.1016/s0021-9258(17)36545-6. [DOI] [PubMed] [Google Scholar]
- 271.Bubb MR, Spector I, Beyer BB, Fosen KM. Effects of Jasplakinolide on the Kinetics of Actin Polymerization. Journal of Biological Chemistry 2000;275(7):5163–70. doi: 10.1074/jbc.275.7.5163. [DOI] [PubMed] [Google Scholar]
- 272.Belin BJ, Cimini BA, Blackburn EH, Mullins RD. Visualization of actin filaments and monomers in somatic cell nuclei. Mol Biol Cell 2013;24(7):982–94. doi: 10.1091/mbc.E12-09-0685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Galkin VE, Orlova A, VanLoock MS, Rybakova IN, Ervasti JM, Egelman EH. The utrophin actin-binding domain binds F-actin in two different modes: implications for the spectrin superfamily of proteins. J Cell Biol 2002;157(2):243–51. doi: 10.1083/jcb.200111097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Allen HF, Wade PA, Kutateladze TG. The NuRD architecture. Cell Mol Life Sci 2013;70(19):3513–24. doi: 10.1007/s00018-012-1256-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Bao Y, Shen X. SnapShot: chromatin remodeling complexes. Cell 2007;129(3):632. doi: 10.1016/j.cell.2007.04.018. [DOI] [PubMed] [Google Scholar]
- 276.Bao Y, Shen X. SnapShot: Chromatin remodeling: INO80 and SWR1. Cell 2011;144(1):158–.e2. doi: 10.1016/j.cell.2010.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Feng Y, Tian Y, Wu Z, Xu Y. Cryo-EM structure of human SRCAP complex. Cell Res 2018;28(11):1121–3. doi: 10.1038/s41422-018-0102-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Kasten MM, Clapier CR, Cairns BR. SnapShot: Chromatin remodeling: SWI/SNF. Cell 2011;144(2):310.e1. doi: 10.1016/j.cell.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 279.Obri A, Ouararhni K, Papin C, Diebold ML, Padmanabhan K, Marek M, et al. ANP32E is a histone chaperone that removes H2A.Z from chromatin. Nature 2014;505(7485):648–53. doi: 10.1038/nature12922. [DOI] [PubMed] [Google Scholar]
- 280.Toto M, D’Angelo G, Corona DF. Regulation of ISWI chromatin remodelling activity. Chromosoma 2014;123(1–2):91–102. doi: 10.1007/s00412-013-0447-4. [DOI] [PubMed] [Google Scholar]
- 281.Yadon AN, Tsukiyama T. SnapShot: Chromatin remodeling: ISWI. Cell 2011;144(3):453–.e1. doi: 10.1016/j.cell.2011.01.019. [DOI] [PubMed] [Google Scholar]
- 282.Pettersen EF, Goddard TD, Huang CC, Meng EC, Couch GS, Croll TI, et al. UCSF ChimeraX: Structure visualization for researchers, educators, and developers. Protein Sci 2021;30(1):70–82. doi: 10.1002/pro.3943. [DOI] [PMC free article] [PubMed] [Google Scholar]
