Functional Analysis of lncRNAs

(Nakagawa Group)

To investigate the physiological significance of the acquisition of species-specific nuclear abundant lncRNAs, we have created knockout (KO) mice that lack Neat1, Gomafu, and Malat1 expression. Phenotypic analyses of these mice have revealed that none of these abundant nuclear lncRNAs are essential for viability, but all of them played important biological roles when the animals were placed in a specific conditions/environments.

Neat1 - an architectural lncRNA that supports female reproduction

Neat1 is an lncRNA that serves as an architectural component of the nuclear body paraspeckle (reviewed in Nakagawa and Hirose, 2012). Neat1 is highly expressed in the corpus luteum in the female ovary (Nakagawa et al., 2011), and the fertility of Neat1 KO mice was severely decreased due to the lack of corpus luteum formation and the consequent decrease in levels of serum progesterone, a steroid hormone essential for the establishment and maintenance of pregnancy (Nakagawa et al., 2014).

Interestingly, only half of the copulation events lead to conception failure, and even the same female mouse that failed to become pregnant can become pregnant from a subsequent copulation. These observations suggested that Neat1 and nuclear body paraspeckle is stochastically required for the formation of the pregnant corpus luteum under certain environments, of which the particular condition still remains unknown.

(A) Neat1 expression in mouse tissues. Note that Neat1 is expressed in only small populations of cells in various tissues, whereas strong and uniform expression is detected in the corpus luteum. 
(B) Fertility of female mice is severely decreased in the Neat1 KO mice. (C) Expression levels of genes that regulate formation of a pregnant corpus luteum are dramatically downregulated in the corpus luteum in Neat1 KO mice.

Gomafu - a neuronal lncRNA controls animal behavior

Gomafu is a neuron-specific lncRNA that forms unique nuclear bodies (Sone et al., 2007). Gomafu is strongly expressed in neurons, and expression is controlled by neuronal activities. The Gomafu KO mice exhibit marginal hyper-locomotive activities in a series of behavior tests, whereas anxiety-related behavior or learning and memory are not significantly affected (Ip et al., 2016).

(A) Expression of Gomafu in the adult mouse brain revealed by in situ hybridization. The transcripts strongly accumulate in neuronal nuclei. Mt, mitral cells, CA1, hippocampus CA1 neurons, V, pyramidal neurons in layer V of the cortex. (B) Total distances the animals traveled in the open field test. WT and KO mice injected with saline or methamphetamine (MAP) for 5 consecutive days 60 minutes after the beginning of the behavior test. Gomafu KO mice (▲) exhibit strikingly enhanced responses to MAP compared to the WT animals (△).

Identification of hnRNP U as the chromosomal localization factor of Xist

Xist is the master regulator of X chromosome inactivation, an epigenetic mechanism that silences one of the two X chromosomes in mammalian females. Notably, Xist accumulate on the entire inactive X chromosome (Xi), resulting in the recruitment of chromatin-modifying complexes and subsequent heterochromatin formation. While basic molecular processes leading to the formation of Xi have been well described, it has long remained elusive how Xist RNA transcripts associate with the chromosome. To identify the chromosomal localization factor of Xist, we hypothesized that a certain RNA-binding protein tethers the transcripts to the chromosome. Thus, we prepared a custom siRNA library designed against abundant RNA-binding proteins and examined if the localization of Xist RNA was affected upon knockdown of each gene. We found that the siRNA-mediated knockdown of hnRNP U leads to dissociation of Xist RNA into the nucleoplasm. This effect was rescued by introduction of full-length hnRNP U, but not the deletion mutant molecules of hnRNP U lacking either its DNA- or RNA-binding domains, suggesting that hnRNP U regulates the chromosomal localization of Xist RNA through its bipartite nucleic-acid binding domains. We also found that hnRNP U is required for the formation of an inactive X chromosome, suggesting that hnRNP U-mediated chromosomal localization of Xist RNA is an essential step during X chromosome inactivation.

(A) Xist (green) and hnRNP U (magenta) in Neuro2a cells. Note that Xist RNA is diffusely localized in the nucleus of the hnRNP U upon knockdown of this protein by siRNA.  (B) Schematic drawing of the role of hnRNP U in Xist RNA localization of the Xi. hnRNP U interacts with X-chromosome and Xist RNA through the SAF and RGG domain, respectively, and recruit chromatin modifying complex such as PRC2 to induce inactivation of the chromosome. 

Gomafu regulates splicing kinetics via association with SF1

To obtain insight into the molecular function of Gomafu, we tried to identify protein that interact with the noncoding RNA. We initially searched sequence motif conserved between chicken, mouse, and human Gomafu, and found that all of the three genes contained a tandem repeat of UACUAAC, which is the essential and conserved intron branch point sequence. We then identified a general splicing factor SF1 as a Gomafu binding protein through affinity purification using the repeat sequence as a bait, and subsequent in vitro splicing assays revealed that the Gomafu repeat cause a distinct delay in the formation of the early spliceosome when using a IgM intron as a substrate. Notably, this effect was not observed with an intron substrate from adenovirus, suggesting that Gomafu regulates kinetics of splicing reaction of a subset of introns. Taken together, we propose that the Gomafu RNA regulates splicing efficiency by changing the local concentration of splicing factors within the nucleus (Tsuiji et al. 2011).

(A) Schematic representation of the UACUAAC repeats in Gomafu homologues and the sequences of the Gomafu repeats used for affinity purification experiments. Bold letters indicate the critical residues for branch point recognition that were mutated in the control. (B) Affinity purification of proteins associated with the Gomafu repeat RNA. SF1 was specifically precipitated by the Gomafu UACUAAC repeat. (C) Analysis of splicing complex formation. Splicing complexes were separated on a native 2% agarose gel. Formation of complex B was significantly retarded in the presence of the Gomafu repeat oligonucleotides. 

Formation of a novel nuclear body by Gomafu-associating proteins

To identify factor that can regulate localization or stability of Gomafu, we screened custom siRNA library designed against abundant RNA binding proteins. We found knockdown of Celf3, previously identified as a member of CUGBP1 (CUG repeat binding protein 1) protein family, leads to marked decrease of Gomafu expression. Cross-link immunoprecipitation analysis using antibodies raised against Celf3 confirmed specific interaction between Gomafu and Celf3. Interestingly, Celf3 formed novel nuclear bodies (CS bodies) in a neuroblastoma cell line Neuro2A, which localization was coincided with SF1, another Gomafu-interacting protein. On the other hand, Gomafu did not accumulate in the CS bodies but separately distributed throught the nucleus. We thus speculate that Gomafu indirectly modulate the function of RNA binding proteins in CS bodies by sequestering them in separate regions in the nucleus.

(A) In situ hybridization of Gomafu in cells depleted with Celf3. Knockdown of Celf3 (Celf3 KD) leads to marked reduction of Gomafu. (B) Immunofluorescent detection of Celf3 (green) and SF1 (magenta). Note that Celf3 and SF1 are enriched in the same nuclear bodies. (C) Models of molecular mechanisms of Gomafu. Gomafu form RNA-protein complex containing splicing factors SF1 and Celf3. These splicing factors assemble on transcription site of certain RNA to form CS bodies. Gomafu assumingly sequester Celf3 and SF1 and control their dynamics in the nucleus. 

Analyses of molecular functions of clade-specific noncoding RNA 4.5SH

As described above, lnRNAs are more species-specific compared to protein-coding mRNAs. We have recently performed a series of functional analyses on 4.5SH, an lncRNA specific to the Myodonta clade (a group of rodents with small sizes and short lives) and highly homologous to the retrotransposon SINE B1. 4.5SH is localized in the nuclear compartment called nuclear speckles, which contain various types of splicing factors as well as pre- and processed mRNAs. We speculated that nuclear 4.5SH may display base pairing with target poly-A (+) mRNAs that contain an antisense insertion of SINE B1, which is highly homologous to 4.5SH. We also hypothesized that the formation of inter-molecular double-stranded RNA (dsRNA) structures lead to decreased gene expression via nuclear retention, as has been proposed for intra-molecular dsRNA structures. We found that 4.5SH inhibits the nuclear export of mRNAs containing an antisense insertion of the retrotransposon SINE B1 and thereby downregulates the expression of these mRNAs. Considering that the emergence of 4.5SH cluster genes occurred later than the expansion of the retrotransposon SINE B1, we propose that the emergence of the 4.5SH gene cluster in specific rodent species rapidly changed the cryptic genetic variations into phenotypic variations via regulation of the expression of genes containing antisense insertions of SINE B1s.

(A) Simultaneous detection of 4.5SH and nuclear speckle marker Srsf1. Insets show the higher-magnification image. Note the clear co-localization of the two signals. (B) Model for the molecular mechanism of the nuclear retention of asB1 genes. 4.5SH forms a complex with asB1 transcripts and prevents nuclear export of the mRNA via a certain dsRNA-binding protein Factor X. Upon knockdown of 4.5SH, the asB1 transcripts are released into the cytoplasm. (C) Speculation of gene expression changes during the evolution of the Myodonta clade. Initially, retrotransposition of SINE B1 generated a number of protein-coding genes that contain asB1 in the untranslated region. At this point, the insertions do not dramatically affect the expression of the host genes, resulting in the generation of silent genetic variations. The subsequent emergence of 4.5SH induced batch downregulation of asB1 genes, resulting in phenotypic diversity. Many of the gene expression changes might be deleterious for the survival of the individuals and were removed upon natural selection, whereas certain combinations of gene downregulation might be advantageous and have delineated the course of the Myodonta clade evolution. 

Super-resolution Observation of Nuclear Bodies

Groups of mammalian-specific lncRNAs abundantly accumulate in the nucleus, forming non-membranous nuclear bodies. The sizes of these RNA-containing nuclear bodies are usually on a submicron scale, making it difficult to observe fine structures using optical microscopy due to the diffraction limitation (δ ≈ 200nm), which is given by the wavelength of the detection light (λ) and the numerical aperture (NA) of the objective lens (δ=0.61λ/NA). However, the emergence of super-resolution microscopy allows us to readily observe these structures at submicron levels beyond the diffraction limit.

We have recently found that a super-resolution microscope, called the Structured Illumination Microscope (SIM), is extremely useful for fine structural analyses of nuclear bodies such as paraspeckles built on lncRNAs (Mito et al., 2016; Okada and Nakagawa, 2015). For example, the diameter of the paraspeckle is usually 200–300 nm, which is a size similar to the diffraction limit of visible light. Indeed, paraspeckle components are co-localized to the paraspeckles, and we cannot discriminate if they occupy distinct positions in the paraspeckles using conventional confocal microscopy. However, SIM observation clearly revealed the core-shell structure of paraspeckles with the 5' and 3' regions of Neat1 located at the surface of the paraspeckles and the middle region embedded in the core. Paraspeckle proteins also displayed distinct distributions: Sfpq and Nono were localized to the core of the paraspeckles, whereas TDP43 was observed mainly at the surface of the paraspeckles. These observations suggested that the paraspeckles are not random aggregates of the proteins and the noncoding RNA but rather that these components are regularly arranged, forming characteristic core-shell structures.