Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 5.
Published in final edited form as: Curr Opin Genet Dev. 2019 Aug 5;55:91–99. doi: 10.1016/j.gde.2019.06.008

Genome Organization around Nuclear Speckles

Yu Chen a,b, Andrew S Belmont c
PMCID: PMC6759399  NIHMSID: NIHMS1536694  PMID: 31394307

Abstract

Higher eukaryotic cell nuclei are highly compartmentalized into bodies and structural assemblies of specialized funcions. Nuclear speckles/IGCs are one of the most prominent nuclear bodies, yet their functional significance remains largely unknown. Recent advances in sequence-based mapping of nuclear genome organization now provide genome-wide analysis of chromosome organization relative to nuclear speckles. Here we review older microscopy-based studies on a small number of genes with the new genomic mapping data suggesting a significant fraction of the genome is near deterministically positioned near nuclear speckles. Both microscopy and genomic based approaches support the concept of the nuclear speckle periphery as a major active chromosomal compartment which may play an important role in fine-tuning gene regulation.

Discovery and rediscoveries of nuclear speckles

The “nuclear speckle” name is now reserved for one particular nuclear body, ranging in size from ~0.3 −3 um and in number from several to tens per nucleus (Fig. 1), as reviewed previously [1,2]. After nucleoli, these nuclear speckles represent the next largest nuclear body compartment in typical mammalian nuclei and are found over a wide range of metazoan species from plants to animals. Cajal first discovered nuclear speckles, which he named “hyaline grumes”, using a modified silver-staining protocol [3] (Fig. 1a). Nuclear speckles were rediscovered by electron microscopy (EM) as “Interchromatin Granule Clusters” (IGCs)- clusters of ~20–25 nm RNP granules lying between regions of chromatin [4] (Fig. 1b). Nuclear speckles were rediscovered yet again through the localization of various snRNP components, splicing factors (i.e. “SC35 domains”), and poly(A)+ RNAs (“poly(A)+ islands”) [57] (Fig. 2a).

1. Variable Identification/Visualization of Nuclear Speckles.

1.

(a) Cajal identified nuclear speckles (handwritten “a”) using different histochemical stains, including silver staining; also shown are nucleoli (handwritten “b”). Original drawing at the Cajal Institute, CSIC, Madrid. Reprinted from Fig. 2a, Ref. [3]. (b) Energy loss EM visualization of nuclear speckles in Hela cells as Interchromatin Granule Clusters (green arrows) surrounded by chromatin (red arrows); energy loss signal (black) is proportional to presence of nucleic acid (phosphorus). Image courtesy of Michael Hendzel. (c) EM section showing nuclear speckles in detergent-extracted nucleus stained with heavy metals in CHO cells; a peripheral zone (large arrowheads), which appears distinct from the surrounding chromatin (narrow arrows), surrounds the granular core of the speckle. Negative image is shown, with heavy metal staining appearing white and low staining black. Scale bar= 0.5 μm. Reprinted from Fig. 5B, Ref. [8]. (d) lncRNA Malat1 (red) and U2 snRNA (green) concentrate at the speckle periphery surrounding SC35 (blue) speckle core as visualized by 3D SIM light microscopy; U2 snRNA also localizes to small foci outside of speckles. Diffraction-limited image (top left) is shown for comparison. Reprinted from Fig. 1A, Ref. [9]. (e) The SON speckle marker (left) shows a higher, more localized concentration over nuclear speckles than SR splicing factors such as ASF/SF2 (right) which stains a larger region surrounding speckles, regions connecting speckles, and other focal accumulations. Scale bar= 2 μm. Reprinted from Fig. S1A, Ref. [24].

2. Poly(A)+ RNA, Decondensed Chromatin, and Transcription Sites at the Nuclear Speckle Periphery.

2.

(a) Nuclear speckles were identified as “Poly(A)+ RNA islands”(red) and “SC35 domains” (green); the Poly(A)+ RNA signal overlaps but is also peripheral to the SC35 domain. Reprinted from Fig. 2A,E,F, Ref. [2]. (b) Serial-section EM reconstruction of detergent-extracted nucleus stained with heavy metals in CHO cells. Negative image is shown, with heavy metal staining appearing white and low staining black. Locally decondensed large-scale chromatin fibers (small arrowheads) frequently mapped to the Interchromatin Granule Cluster periphery (large arrowheads). Scale bar= 120 nm Reprinted from Fig. 8E, Ref. [8]. (c-e) Active sites of transcription and Ser5p- RNA pol2 staining foci surround nuclear speckle periphery. (d-e) 5 min EU pulse-labeling of transcription (d) or Ser5p-RNA pol 2 staining (e) (red) relative to nuclear speckle marker (SON) (green in (c) and white in (d)). 2x enlarged views corresponding to regions surrounding speckles (arrows). Reprinted and Modified from Fig. 5C&D, Ref. [24, with permission from Rockefeller University Press.

Nuclear speckles remain an operational definition dependent on the experimental detection method. While EM uniquely identifies IGCs, they likely miss a surrounding peripheral zone which together with the IGC define the actual nuclear speckle functional unit. EM of detergent extracted nuclei shows an amorphous zone of variable width, stained by heavy metals but distinctive from the surrounding chromatin (Fig. 1c, arrows), surrounding the IGC [8] (Fig. 1c, arrowheads). The long non-coding RNA (lncRNA) Malat-1 is highly enriched in nuclear speckles, with its distribution extending slightly outside the core of clustered granules into this peripheral zone with which active genes and their nascent transcripts may largely interact [9] (Fig. 1d). Many spliceosome and splicing factors are also enriched within and surrounding the IGC but are also found in a reticular pattern spreading throughout the nucleus and in Cajal bodies and other small foci [6] (Fig. 1d&e). Even potential scaffolding proteins (e.g. SON [10], Fig. 1e) and other splicing factor markers (pSC35, Figs. 1d,2a), which may be enriched 8–10-fold in the IGC core of the nuclear speckle, are present elsewhere in the nucleus and in a given cell type and species may not uniquely mark nuclear speckles. For instance, in mouse 2-cell embryos, immuno-EM showed localization of SC35 in IGCs but also in more numerous smaller foci [11]. Nearly all known nuclear speckle protein components are factors involved in transcription, post-transcriptional RNA processing, or nuclear RNA export and are therefore found to varying degrees as a diffusible nucleoplasmic pool and bound to active gene loci, nascent transcripts, pre-mRNAs, mRNAs, and/or other focal nuclear accumulations.

This has led to the common misconception that nuclear speckles form wherever there is strong transcription. In fact, only a subset of highly transcriptionally active endogenous genes or transgenes show a specific association with nuclear speckles [2,12,13].

Proposed nuclear speckle functions

To date, two main classes of models have been proposed for the possible physiological functions of nuclear speckles [1,2].

Model 1 suggests nuclear speckles are storage sites for RNA processing factors [1]. Proteomics of RNP granules purified from IGCs revealed hundreds of proteins involved in various stages of RNA processing and nuclear export [14], while EM shows no DNA or active transcription inside speckles. Additionally, after transcriptional or splicing inhibition, splicing factors accumulate within larger and rounder nuclear speckles. A newer and more nuanced variation of this model would be that nuclear speckles actively buffer nucleoplasmic levels of factors regulating aspects of transcription and RNA processing for homeostasis control (Yaron Shav-Tal, personal communication).

Model 2 instead suggests nuclear speckles are “gene expression hubs”, enhancing transcription, post-transcriptional splicing, and/or other RNA processing activities for a subset of genes through rapid recycling of components between speckles and genes [2]. Immuno-FISH revealed ~half of 20–25 active genes localize near the nuclear speckle periphery [2], while certain co-expressed genes co- localized at the nuclear speckle periphery with frequencies higher than expected by chance [1517]. Entire gene-rich chromosome bands localize near nuclear speckles at increased frequency [18]. Moreover, the pre-mRNA and/or mRNAs of a few of these active genes (COLA1, MyHC) overlap with and enter within nuclear speckles, while poly(A)+ RNA accumulates inside and at the periphery of IGCs [2,19,20] (Fig. 2a). Local decondensation of chromatin near nuclear speckles has been visualized by EM (Fig. 2b), and a concentration of active transcription surrounding speckles has been visualized by pulse-labeling and RNA pol2 staining [2124](Fig. 2ce). Splicing defects and inhibition of either splicing or nuclear export lead to accumulation of RNAs inside speckles [2528], while microinjected RNAs with functional splice sites and/or splicing enhancers localize and, after export inhibition, accumulate inside nuclear speckles [27,2931].

Despite healthy debate for over 20 years, little progress has been made in distinguishing between the above two models. Two main questions need to be answered to judge the significance of the data on which Model 2 is based. How prevalent are genes that position at high frequency near nuclear speckles? Given the higher density of both nuclear speckles and active transcription in the nuclear interior, what is the actual mean speckle distance that should be considered unusually “close” to a nuclear speckle?

A subset of the active genome organizes around speckles

Missing until recently have been genome-wide methods capable of mapping chromosome regions relative to nuclear speckles. Several methods measuring molecular proximity have failed to identify speckle-associated chromosome regions. The high compaction of chromatin observed near nuclear speckles (Fig. 1b, c, Fig. 2b) predicts that speckle-associated chromosome regions should be large domains, 100s of Kbp to Mbp in size. However, ChIP against speckle-enriched factors (SRSF2/SC35, SON) shows localized peaks over promoters and gene bodies of active genes [32,33]. Similarly, CHART (capture hybridization analysis of RNA targets) and RAP (RNA anti-sense purification) show MALAT1 focally enriched over transcriptionally active and spliced gene bodies, possibly via interaction with pre-mRNAs since the association is transcription-elongation dependent [34,35]. Molecular proximity methods are likely detecting the small fraction of marker proteins/RNAs interacting at nascent transcripts and active genes, independent of speckle localization but within cross-linking distance of the DNA. Conversely, they fail to detect DNA within microscopic but not molecular proximity of these same marker proteins/RNAs concentrated within speckles.

Recently, three orthogonal methods have been applied to identify nuclear speckle associated chromosome regions genome-wide [24,36,37] (Fig. 3). MARGI (Mapping of RNAGenome Interactions) is a ligation-based method that reads the sequences of RNA-DNA molecules linked by a bridging adaptor oligonucleotide and exploits the known concentration of particular RNAs (snRNAs, 7S RNA, MALAT1) within nuclear speckles and their periphery [37] (Fig. 3a). SPRITE (Split-Pool Recognition of Interactions by Tag Extension) identifies DNA and potentially RNA molecules found in the same molecular complexes isolated after sonication of nuclei [36] (Fig. 3b). The molecular interaction distances producing a MARGI signal are unknown. For SPRITE, complexes containing increasing numbers of fragments should probe longer interaction distances, but these actual microscopic distances are unknown. TSA (Tyramide Signal Amplification)-Seq instead uses a cytological staining method producing a diffusible signal to label chromosome regions proportionally to their distance, up to ~ 1 um, from nuclear speckles [24]. TSA-Seq maps distances over cytological size scales and a physical model derived from the diffusion equation accurately converts sequence read count enrichment into an actual physical distance (Fig. 3c).

3. Genomic assays for speckle localization and correlation with speckle localization measured by FISH.

3.

(a) MARGI uses an oligonucleotide adaptor to ligate RNA and DNA fragments (top left). Counts of ligations with nuclear-speckle associated RNAs (saRNAs) show peaks and valleys across chromosomes (bottom left). FISH measurements show an increased level of SC35 colocalization with probes selected from peaks (nsaPeak) versus valleys (non-nsaPeak). Adapted under Creative Commons Attribution License (CC BY) from Graphical Summary, Fig. 4A,D, Ref. [37], (b) SPRITE measures colocalization of DNA and RNA fragments in isolated, chromatin complexes (top left). Large complexes with many DNA fragments show unusually high numbers of inter-chromosome interactions with a small number of active chromosomal regions, defining an “active chromosomal hub”. The frequency of chromosome interactions with this active hub (red, y-axis) correlate with Malat1 RAP-RNA-Seq (black, y-axis) (bottom left). FISH of different probes showed a linear correlation in percentage of alleles mapping <0.5 μm from a nuclear speckle (y-axis) with this active hub contact frequency (x-axis) (right). Adapted from Graphical Summary, Figs. 5A, 6D, Ref.[36]. (c) TSA-Seq measures spreading of diffusible tyramide free-radical, generated by a peroxidase localized near nuclear speckles by SON immunostaining, producing biotin-DNA labeling proportional to speckle distance. DNA purification is followed by biotinylated-DNA pull-down and sequencing (top left). TSA-Seq score uses log2 ratio of observed versus average read count (y-axis, log2 ratio of fold-enrichment), with peaks predicting regions near speckles (bottom left). An exponential relationship was observed between probe TSA-Seq fold-enrichment and the average FISH-signal distance from a speckle (right). Adapted from Figs. 1A, 2A,B, 3A,B, Ref. [24] with permission from Rockefeller University Press.

MARGI detects peaks of 100kb-13 Mbp width essentially over all active chromosomal regions (the A Hi-C compartment) as enriched in speckle-associated RNAs. Resolution currently is limited by the relatively low read count of DNA fragments linked to nuclear speckle associated RNAs. It remains unclear whether MARGI, like RAP and CHART, is simply detecting the focal enrichment of Malat1 and snRNAs near active gene bodies or whether it is actually measuring those active genes near nuclear speckles. SPRITE complexes containing large numbers of DNA fragments show increased numbers of inter-chromosomal contacts. Two distinct subsets, or “hubs”, of chromosome regions with unusually high numbers of such inter-chromosomal contacts emerge from analysis of these large clusters: transcriptionally inactive regions and gene-rich, transcriptionally active regions. Regions within each hub contact each other at high frequency, but show low contacts with the opposite hub. Contact frequencies of 1 Mbp-size chromosome regions with this active hub correlated with Malat1 lncRNA localization measured by RAP-RNA-Seq [35], and showed a linear correlation with the fraction of alleles (20–80%) mapping within 0.5 um from a nuclear speckle by FISH [35]. TSA-Seq currently maps speckle proximity at high resolution, estimating mean distance to nuclear speckles with an accuracy of ~50 nm. Most active chromosomal regions localize closer to speckles than the median distance across the genome.

Strikingly, in human K562 cells TSA-Seq reveals Speckle-Associated Domains (SPADs) corresponding to ~5% of the genome showing deterministic positioning within 500 nm of nuclear speckles with near 100% frequencies (estimated mean distance < 0.32 um to speckle periphery). More generally, the ~20% of the genome mapping closest to nuclear speckles correlates closely to the A1 HI-C subcompartment, with the A2 Hi-C subcompartment mapping instead at intermediate mean distances to speckles. This 20% of the genome closest to speckles shows high gene density and contains ~70% of super-enhancers and the top 5% expressed genes. TSA-Seq suggests that as an ensemble-average, interphase chromosomes traverse from nuclear periphery to interior and back to periphery, with gene expression “hot zones” located at the apex of these chromosome trajectories into the nuclear interior. These gene expression hot zones are either near deterministically positioned close to nuclear speckles (Type I) or located at intermediate distances (Type II). Type I gene expression hot zones contain the most highly expressed genes and have the higher gene density, the greater fraction of housekeeping genes, and the greater enrichment of genes with low RNA polymerase pausing, which may be linked to the increased concentration of factors involved in transcriptional pause release and elongation in nuclear speckles [38,39].

How MARGI, SPRITE and TSA-Seq predictions compare awaits data generation from the identical cell lines. Whereas MARGI and SPRITE, through cross-linking to speckle-enriched RNAs (MARGI) or active chromosomal hubs (SPRITE), are conceptualized as measuring the fraction of alleles within close distance to speckles, TSA-Seq instead measures mean speckle distances. Both A1 and A2 Hi-C subcompartments are identified as speckle-interacting by MARGI and SPRITE, but only A1 is identified as close to speckles by TSA-Seq. An important question is whether chromosome regions showing a low contact frequency by SPRITE and an intermediate distance to speckles by TSA-Seq are positioned persistently away from speckles (Fig. 4a), or might show frequent speckle associations over a physiologically meaningful time frame (either through gene movements to speckles or speckle protrusions and/or movements to genes (Fig. 4b)), or are positioned near smaller speckles giving rise to a proportionally lower TSA-Seq signal (Fig. 4c).

4. Models for explaining variable speckle proximity/distance observed by genomic methods.

4.

Variable size peaks observed for SPRITE [36], TSA-Seq [24], and MARGI [37] have been interpreted differently. (a) Model 1: SON TSA-Seq identified two types of transcription hot-zones mapping near TSA-Seq local maxima: Type I (large peaks, correlated with A1 Hi-C subcompartment) positioned close, on average, to nuclear speckles and Type II peaks (smaller peaks, correlated with A2 Hi-C subcompartment), positioned at intermediate distances, on average, from speckles, and possibly interacting with an unknown nuclear body. (b) Model 2: Larger TSA-Seq (Type I), MARGI, and SPRITE peaks could correspond to a larger fraction of alleles localizing close to nuclear speckles, with smaller TSA-Seq (Type II), MARGI and SPRITE peaks showing a smaller fraction of alleles close to speckles. (c) Model 3: Larger (Type I) versus smaller (Type II) TSA-Seq peaks could correspond to association of chromosome region with larger versus smaller nuclear speckles, respectively. Further work is needed to determine which model best describes chromosome positioning relative to nuclear speckles.

Beyond correlation- is there a functional significance to gene association with speckles?

These new genomic methods demonstrate that the nuclear speckle colocalization observed years ago for ~50% of ~25 protein-coding genes now extends to a comparable fraction of ALL expressed protein-coding genes. This high prevalence of nuclear speckle association across the genome greatly revives interest in the possible functional significance of speckle association, though it still remains only a correlation that does not distinguish cause and effect. However, what is also new has been the demonstration of the existence of an active mechanism for moving genes towards nuclear speckles. Using the Hsp70 HSPA1A gene as a model system, live cell microscopy revealed an actin-dependent, directional movement of plasmid HSPA1A transgene arrays, over distances of up to 4–5 um towards nuclear speckles with an average velocity of ~ 1 um/min after heat-shock induction [40]. The existence of an active mechanism for moving these Hsp70 transgene arrays to nuclear speckles strongly suggests the existence of active mechanism for positioning endogenous chromosomal regions near nuclear speckles.

Moreover, live-cell imaging of Hsp70 transgenes demonstrates a strong likelihood of functional significance of speckle association by showing that nascent transcripts of these transgenes become visible only after, and not before, first contact of the transgene with a nuclear speckle. Using a plasmid transgene HSPA1A array expressing MS2-tagged HSPA1A transcripts, visible accumulations of nascent transcripts after heat shock were observed only 0 to several minutes following first contact with nuclear speckles [40]. Like most multi-copy plasmid transgene arrays, this HSPA1A transgene array is heterochromatic and shows greatly delayed and asynchronous heat-shock induced expression, raising questions of physiological significance. However, more recent work using a BAC Hsp70 transgene system that more closely recapitulates normal Hsp70 gene expression points to a model in which amplification of gene expression is linked to nuclear speckle association (IMN Kim et al, bioRxiv 604298; doi: https://doi.org/10.1101/604298). Several fold higher levels of nascent transcripts are seen after heat shock using smRNA FISH for speckle-associated versus non-associated endogenous Hsp70 alleles, while live-cell imaging of Hsp70 BAC transgenes shows association of the transgene with nuclear speckles precedes the appearance of a MS2-nascent HSPA1B transcript signal by up to several minutes, while separation from nuclear speckles precedes by several minutes downregulation and disappearance of these MS2 signals.

This transcriptional amplification phenomenon appears to require actual physical contact, if only transient, with the nuclear speckle. Nuclear speckles behave as liquid phase-separated bodies, with measured viscosities comparable to other liquid-phase separated RNP bodies [41]. Biophysical analysis suggests liquid condensates may greatly enhance rates of certain reactions by concentrating factors that favor the reaction [42]. Moreover, interactions among components of nuclear speckles may lead to a multi-layer organization concentrating certain components, including MALAT1 and snRNAs, to the speckle periphery in position to interact with active chromosomal regions [9].

Nuclear speckles have also been suggested as playing a distinct role in post-transcriptional splicing and nuclear export of RNAs and changes in expression of a many nuclear speckle proteins are linked to human disease states, particularly cancer, and altered alternative splicing of particular mRNAs [43,44]. Both unspliced RNAs and spliced RNAs without Exon Junction Complex (EJC) proteins accumulate within nuclear speckles, suggesting that speckles may act as a quality control for mRNAs prior to export [2531,45]. The liquid phase condensation behavior of nuclear speckles could enhance the functions of speckles to regulate RNA processing activities both by accelerating reactions within speckles and by buffering concentrations of speckle components in the nucleoplasm.

Conclusions and perspectives

Gene expression in the nucleus is regulated at many levels including transcription, splicing, mRNA modification and export. Two of these at least- transcription and splicing-already are known to be tightly coupled [46]. Components related to all of these levels of gene expression are concentrated in nuclear speckles, including for example transcription (p300/CBP, pTEF-beta), splicing (snRNPs, SR proteins), and RNA capping, poly-adenylation, and export factors. The recent genome-wide demonstration of the prominent localization of a significant fraction of active gene loci near nuclear speckles revives old models of nuclear speckles and their periphery acting as a central hub to coordinate multiple steps of gene expression. Genomic analysis also reveals a linear gradient of gene expression as a function of mean speckle distance [24]. Years ago, models were presented by which speckle protrusions or a transport network might link the contents of nuclear speckles with sites of active transcription and RNA processing [47,48]. More recently, live-cell imaging has revealed directional movements of transgenes towards nuclear speckles and dynamic changes in distances of transgenes from speckles due to the mobility of both. It is fascinating to speculate that the dynamics of gene loci, speckles, and interchromatin granules might lead to a merging of both the older “storage site” and “transcriptional hub” models of nuclear speckle function. Live-cell imaging technologies should allow future exploration of how nuclear speckle and gene dynamics may contribute to the fine-tuning of gene expression.

Acknowledgements:

We thank Michael Hendzel, University of Alberta, for his contribution of Fig. 1b, and Yaron Shav-Tal, Bar-Ilan University, for his ideas about nuclear speckles buffering nucleoplasmic levels of RNA processing factors and sharing his paper prior to publication. We apologize to colleagues whose work was not mentioned due to space limitations. Funding: ASB acknowledges support from National Institutes of Health R01 grant GM58460 and from its Common Fund 4D Nucleome Program (U54 DK107965).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Spector DL, Lamond AI: Nuclear speckles. Cold Spring Harb Perspect Biol 2011, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hall LL, Smith KP, Byron M, Lawrence JB: Molecular anatomy of a speckle. Anat Rec A Discov Mol Cell Evol Biol 2006, 288:664–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lafarga M, Casafont I, Bengoechea R, Tapia O, Berciano MT: Cajal’s contribution to the knowledge of the neuronal cell nucleus. Chromosoma 2009, 118:437–443. [DOI] [PubMed] [Google Scholar]
  • 4.Thiry M: Differential location of nucleic acids within interchromatin granule clusters. Eur J Cell Biol 1993, 62:259–269. [PubMed] [Google Scholar]
  • 5.Carter KC, Taneja KL, Lawrence JB: Discrete nuclear domains of poly(A) RNA and their relationship to the functional organization of the nucleus. J Cell Biol 1991, 115:1191–1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Spector DL, Fu X-D, Maniatis T: Associations between distinct pre-mRNA splicing components and the cell nucleus. EMBO J 1991, 10:3467–3481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Carter KC, Bowman D, Carrington W, Fogarty K, McNeil JA, Fay FS, Lawrence JB: A three-dimensional view of precursor messenger RNA metabolism within the mammalian nucleus. Science 1993, 259:1330–1335. [DOI] [PubMed] [Google Scholar]
  • 8.Belmont AS, Bruce K: Visualization of G1 chromosomes: a folded, twisted, supercoiled chromonema model of interphase chromatid structure. J Cell Biol 1994, 127:287–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Fei J, Jadaliha M, Harmon TS, Li ITS, Hua B, Hao Q, Holehouse AS, Reyer M, Sun Q, Freier SM, et al. : Quantitative analysis of multilayer organization of proteins and RNA in nuclear speckles at super resolution. J Cell Sci 2017, 130:4180–4192. [DOI] [PMC free article] [PubMed] [Google Scholar]; **. 3D SIM super-resolution light microscopy analysis reveals nuclear speckle core regions stained by SC35 and SON proteins with a more peripheral distribution of lncRNA Malat1 and snRNA. Simulations predicted this multi-layer arrangement of speckle components, and changes induced by perturbation, based on molecular interactions between RNA and protein components.
  • 10.Sharma A, Takata H, Shibahara K, Bubulya A, Bubulya PA: Son is essential for nuclear speckle organization and cell cycle progression. Mol Biol Cell 2010, 21:650–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bogolyubova I, Bogolyubov D, Parfenov V: Localization of poly(A)+ RNA and mRNA export factors in interchromatin granule clusters of two-cell mouse embryos. Cell Tissue Res 2009, 338:271–281. [DOI] [PubMed] [Google Scholar]
  • 12.Hu Y, Plutz M, Belmont AS: Hsp70 gene association with nuclear speckles is Hsp70 promoter specific. J Cell Biol 2010, 191:711–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hu Y, Kireev I, Plutz M, Ashourian N, Belmont AS: Large-scale chromatin structure of inducible genes: transcription on a condensed, linear template. J Cell Biol 2009, 185:87–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Saitoh N, Spahr CS, Patterson SD, Bubulya P, Neuwald AF, Spector DL: Proteomic analysis of interchromatin granule clusters. Mol Biol Cell 2004, 15:3876–3890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rieder D, Ploner C, Krogsdam AM, Stocker G, Fischer M, Scheideler M, Dani C, Amri E-Z, Müller WG, McNally JG, et al. : Co-expressed genes prepositioned in spatial neighborhoods stochastically associate with SC35 speckles and RNA polymerase II factories. Cell Mol Life Sci 2014, 71:1741–1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Brown JM, Leach J, Reittie JE, Atzberger A, Lee-Prudhoe J, Wood WG, Higgs DR, Iborra FJ, Buckle VJ: Coregulated human globin genes are frequently in spatial proximity when active. J Cell Biol 2006, 172:177–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Brown JM, Green J, das Neves RP, Wallace HAC, Smith AJH, Hughes J, Gray N, Taylor S, Wood WG, Higgs DR, et al. : Association between active genes occurs at nuclear speckles and is modulated by chromatin environment. J Cell Biol 2008, 182:1083–1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Shopland LS, Johnson CV, Byron M, McNeil J, Lawrence JB: Clustering of multiple specific genes and gene-rich R-bands around SC-35 domains: evidence for local euchromatic neighborhoods. J Cell Biol 2003, 162:981–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xing Y, Johnson CV, Moen PT, McNeil JA, Lawrence JB: Nonrandom gene organization: structural arrangements of specific pre-mRNA transcription and splicing with SC-35 domains. J Cell Biol 1995, 131:1635–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smith KP, Moen PT, Wydner KL, Coleman JR, Lawrence JB: Processing of endogenous pre-mRNAs in association with SC-35 domains is gene specific. J Cell Biol 1999, 144:617–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wei X, Somanathan S, Samarabandu J, Berezney R: Three-Dimensional Visualization of Transcription Sites and Their Association with Splicing Factor–Rich Nuclear Speckles. J Cell Biol 1999, 146:543–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fakan S: The functional architecture of the nucleus as analysed by ultrastructural cytochemistry. Histochem Cell Biol 2004, 122:83–93. [DOI] [PubMed] [Google Scholar]
  • 23.Xie SQ, Martin S, Guillot PV, Bentley DL, Pombo A: Splicing speckles are not reservoirs of RNA polymerase II, but contain an inactive form, phosphorylated on serine2 residues of the C-terminal domain. Mol Biol Cell 2006, 17:1723–1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chen Y, Zhang Y, Wang Y, Zhang L, Brinkman EK, Adam SA, Goldman R, Van Steensel B, Ma J, Belmont AS: Mapping 3D genome organization relative to nuclear compartments using TSA-Seq as a cytological ruler. J Cell Biol 2018, 217:4025–4048. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Discovery paper for TSA-Seq mapping of genome organization relative to different nuclear compartments. TSA-Seq adapts the Tyramide Signal Amplification cytological staining method to measure relative distances of DNA to the stained compartment. For nuclear speckle staining, TSA-Seq uses a physical model of diffusion to convert sequence read counts into a calibrated mean distance, measured in microns, with an accuracy of ~50 nm. TSA-Seq identifies ~5% of the genome positioned at near 100% frequency within ~0.5 μm of a speckle.
  • 25.Johnson C, Primorac D, McKinstry M, McNeil J, Rowe D, Lawrence JB: Tracking COL1A1 RNA in osteogenesis imperfecta. splice-defective transcripts initiate transport from the gene but are retained within the SC35 domain. J Cell Biol 2000, 150:417–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Carvalho T, Martins S, Rino J, Marinho S, Carmo-Fonseca M: Pharmacological inhibition of the spliceosome subunit SF3b triggers exon junction complex-independent nonsense-mediated decay. J Cell Sci 2017, 130:1519–1531. [DOI] [PubMed] [Google Scholar]
  • 27.Dias AP, Dufu K, Lei H, Reed R: A role for TREX components in the release of spliced mRNA from nuclear speckle domains. Nat Commun 2010, 1:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Girard C, Will CL, Peng J, Makarov EM, Kastner B, Lemm I, Urlaub H, Hartmuth K, Lührmann R: Post-transcriptional spliceosomes are retained in nuclear speckles until splicing completion. Nat Commun 2012, 3:994. [DOI] [PubMed] [Google Scholar]
  • 29.Melčák I, Melčáková Š, Kopsky V, Večeřová J, Raška I: Prespliceosomal assembly on microinjected precursor mRNA takes place in nuclear speckles. Mol Biol Cell 2001, 12:393–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Akef A, Zhang H, Masuda S, Palazzo AF: Trafficking of mRNAs containing ALREX-promoting elements through nuclear speckles. Nucleus 2013, 4:326–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang K, Wang L, Wang J, Chen S, Shi M, Cheng H: Intronless mRNAs transit through nuclear speckles to gain export competence. J Cell Biol 2018, 217:3912–3929. [DOI] [PMC free article] [PubMed] [Google Scholar]; Intriguing observation that exonic splicing enhancers in intronless mRNAs promote localization in nuclear speckles, based largely on observations of RNAs produced by microinjected DNA templates. Experimental results suggest mRNAs in nuclear speckles gain competence for nuclear export through enhanced recruitment of the TREX export complex. Curiously, however, RNA FISH against endogenous genes showed RNAs accumulating equally in all or most nuclear speckles. This contrasts with previously published studies, in some cases studying the same endogenous genes, showing RNA FISH signals accumulating near the gene transcription site, and, for certain genes, within just the adjacent nuclear speckle.
  • 32.Ji X, Zhou Y, Pandit S, Huang J, Li H, Lin CY, Xiao R, Burge CB, Fu XD: SR proteins collaborate with 7SK and promoter-associated nascent RNA to release paused polymerase. Cell 2013, 153:855–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kim J-H, Baddoo MC, Park EY, Stone JK, Park H, Butler TW, Huang G, Yan X, Pauli-Behn F, Myers RM, et al. : SON and Its Alternatively Spliced Isoforms Control MLL Complex-Mediated H3K4me3 and Transcription of Leukemia-Associated Genes. Mol Cell 2016, 61:859–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.West JA, Davis CP, Sunwoo H, Simon MD, Sadreyev RI, Wang PI, Tolstorukov MY, Kingston RE: The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol Cell 2014, 55:791–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Engreitz JM, Sirokman K, McDonel P, Shishkin AA, Surka C, Russell P, Grossman SR, Chow AY, Guttman M, Lander ES: RNA-RNA interactions enable specific targeting of noncoding RNAs to nascent Pre-mRNAs and chromatin sites. Cell 2014, 159:188–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Quinodoz SA, Ollikainen N, Tabak B, Palla A, Schmidt JM, Detmar E, Lai MM, Shishkin AA, Bhat P, Takei Y, et al. : Higher-Order Inter-chromosomal Hubs Shape 3D Genome Organization in the Nucleus. Cell 2018, 174:744–757.e24. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Discovery paper for SPRITE mapping of genome organization which uses bar-coding of DNA and RNA fragments localizing within the same cross-linked chromatin complex to produce a ligation-independent, multi-fragment mapping method to measure genome proximity. An “active hub” with increased inter-chromosomal contacts and showing increased nuclear speckle association is identified; frequencies of chromosome region interactions with this active hub correlate with the percentage of alleles of these regions observed close to nuclear speckles.
  • 37.Chen W, Yan Z, Li S, Huang N, Huang X, Zhang J, Zhong S: RNAs as Proximity-Labeling Media for Identifying Nuclear Speckle Positions Relative to the Genome. iScience 2018, 4:204–215. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This paper leverages the MARGI mapping of RNA-DNA ligation events to identify genomic regions that are in contact with speckle-associated RNAs known to be enriched at the nuclear speckle periphery. Chromosome regions enriched in these speckle-associated RNA-DNA contacts are shown to have increased colocalization with immunostaining of SC35, a nuclear speckle protein marker.
  • 38.Dow EC, Liu H, Rice AP: T-loop phosphorylated Cdk9 localizes to nuclear speckle domains which may serve as sites of active P-TEFb function and exchange between the Brd4 and 7SK/HEXIM1 regulatory complexes. J Cell Physiol 2010, 224:84–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Herrmann CH, Mancini MA: The Cdk9 and cyclin T subunits of TAK/P-TEFb localize to splicing factor-rich nuclear speckle regions. J Cell Sci 2001, 114:1491–1503. [DOI] [PubMed] [Google Scholar]
  • 40.Khanna N, Hu Y, Belmont AS: HSP70 transgene directed motion to nuclear speckles facilitates heat shock activation. Curr Biol 2014, 24:1138–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kim J, Han KY, Khanna N, Ha T, Belmont AS: Nuclear speckle fusion via long-range directional motion regulates speckle morphology after transcriptional inhibition. J Cell Sci 2019, 132:jcs226563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Stroberg W, Schnell S: Do Cellular Condensates Accelerate Biochemical Reactions? Lessons from Microdroplet Chemistry. Biophys J 2018, 115:3–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Galganski L, Urbanek MO, Krzyzosiak WJ: Nuclear speckles: molecular organization, biological function and role in disease. Nucleic Acids Res 2017, 45:10350–10368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Corkery DP, Holly AC, Lahsaee S, Dellaire G: Connecting the speckles: Splicing kinases and their role in tumorigenesis and treatment response. Nucleus 2015, 6:279–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hochberg-Laufer H, Schwed-Gross A, Neugebauer KM, Shav-Tal Y: Uncoupling of nucleocytoplasmic RNA export and localization during stress. Nucleic Acids Res 2019, 47:4778–4797. [DOI] [PMC free article] [PubMed] [Google Scholar]; * An adenosine analog, tubercidin, causes a stress response leading to loss of of a subset of nuclear speckle components from the speckles. Although mRNAs were spliced, exon junction complex and TREX recruitment to mRNAs were reduced and mRNA nuclear export was inhibited. mRNAs accumulated in nuclear speckles, suggesting nuclear speckles might serve as a quality control check for export-competent mRNAs prior to nuclear export.
  • 46.McCracken S, Fong N, Yankulov K, Ballantyne S, Pan G, Greenblatt J, Patterson SD, Wickens M, Bentley DL: The C-terminal domain of RNA polymerase II couples mRNA processing to transcription. Nature 1997, 385:357–361. [DOI] [PubMed] [Google Scholar]
  • 47.Misteli T, Cáceres JF, Spector DL: The dynamics of a pre-mRNA splicing factor in living cells. Nature 1997, 387:523–527. [DOI] [PubMed] [Google Scholar]
  • 48.Spector DL: Higher order nuclear organization: three-dimensional distribution of small nuclear ribonucleoprotein particles. Proc Natl Acad Sci U S A 1990, 87:147–51. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES