Abstract
Neurogenesis contributes to poststroke recovery. Long noncoding RNAs (lncRNAs) participate in the regulation of stem cell self-renewal and differentiation. However, the role of lncRNAs in stroke-induced neurogenesis remains unknown. In this study, we found that H19 was the most highly upregulated lncRNA in neural stem cells (NSCs) of the subventricular zone (SVZ) of rats subjected to focal cerebral ischemia. Deletion of H19 suppressed cell proliferation, promoted cell death, and blocked NSC differentiation. RNA sequencing analysis revealed that genes deregulated by H19 knockdown were those that are involved in transcription, apoptosis, proliferation, cell cycle, and response to hypoxia. H19 knockdown significantly increased the transcription of cell cycle-related genes including p27, whereas overexpression of H19 substantially reduced expression of these genes through the interaction with chromatin remodeling proteins EZH2 and SUZ12. Moreover, H19 regulated neurogenesis-related miRNAs. Inactivation of H19 in NSCs of ischemic rats attenuated spontaneous functional recovery after stroke. Collectively, our data provide novel insights into the epigenetic regulation of lncRNAs in stroke-induced neurogenesis.
Keywords: epigenetics, H19, long noncoding RNA, neurogenesis, stroke
1 |. INTRODUCTION
Stroke is a leading cause of disability worldwide with limited treatment options.1 Neural stem cells (NSCs) are located in the subventricular zone (SVZ) of the lateral ventricle and the subgranular zone (SGZ) of the dentate gyrus (DG) in the hippocampus.2 NSCs from the SVZ continually generate neuroblasts and oligodendrocyte progenitor cells. The neuroblasts migrate to the olfactory bulb where they differentiate into interneurons. Neurogenesis contributes to limited spontaneous recovery after stroke.3,4 Elucidating the underlying molecular mechanisms in poststroke neurogenesis could provide new therapies to amplify endogenous neurogenesis and to improve neurological function during stroke recovery.
Long noncoding RNAs (lncRNAs) are a class of regulatory RNAs that are longer than 200 nucleotides, lack protein-coding potential, and are implicated in mediating physiologic and pathologic processes in mammals.5,6 LncRNAs mediate gene expression via chromatin modification, translational and posttranslational regulation.7 Recent studies show that lncRNAs express in a lineage-specific manner, suggesting that lncRNAs may have a function in particular cell types, including cells in the brain.8,9 Wang et al. reported that the lncRNA expression profiling was altered in the sera of stroke patients and the ischemic brains of experimental rodents.10 Moreover, the roles of lncRNAs in ischemic stroke have been recently studied in different brain cell types, including neurons, glial cells, and vascular cells. Among them, LncRNA-N1LR, SNHG1, C2dat1, and MALAT1 have been shown to facilitate neuroprotection after stroke, whereas TUG1, GAS5, FosDT, and MEG3 exacerbate ischemic brain injury.11–18 SNHG12, MEG3, and MALAT1 enhance stroke-induced angiogenesis.19–21 The functions of lncRNAs in neurogenesis are just beginning to be appreciated.22,23
H19, encoding a polyadenylated lncRNA of ~2.6 kb, is located on 11p15.5 in humans and chromosome 7 in mice.24 H19 is the earliest defined maternally imprinted lncRNA and it is involved in diverse biological processes and pathology as a multifunctional regulatory transcript.25–27 Hypermethylation or hypomethylation of H19 during development induces human genetic syndrome, for example, Beckwith-Wiedemann syndrome and Silver-Russel syndrome with clinical features of asymmetry and growth retardation.28 In adults, H19 maintains the character of hematopoietic stem cells and pluripotent stem cells and promotes skeletal muscle differentiation and regeneration.27,29,30 Also, H19 regulates adipogenesis of bone marrow cells and embryonic stem cell differentiation.31,32 Recently, six single-nucleotide polymorphisms (SNPs) of H19 were detected in ischemic stroke patients, whereas a preclinical study demonstrated that H19 is involved in acute stroke-induced neuroinflammation.10 These data suggest a critical role of H19 in the progression of stroke.
In the present study, we analyzed the lncRNA profiles alteration in the NSCs before and after stroke, investigated the role of H19 in mediating stroke-induced neurogenesis, and demonstrated that lncRNA H19 in the NSCs mediate stroke-augmented neurogenesis by potentially affecting chromatin remodeling proteins.
2 |. MATERIALS AND METHODS
2.1 |. Animals
All experimental procedures in this study were carried out according to the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee (IACUC) of Henry Ford Hospital.
2.2 |. Middle cerebral artery occlusion (MCAO)
Male Wistar rats (~10 weeks, purchased from Jackson Laboratory) were anesthetized with 2% isoflurane. Exposed the common carotid artery (CCA), external carotid artery (ECA), and internal carotid artery (ICA) on the right side and gently insert a nylon filament (4 ± 0) from the ECA into the lumen of the ICA.
2.3 |. Neural stem cell culture
Neural stem cells (NSCs) were isolated from nonischemic rats and ischemic rats on day 7 after MCAO when stroke-increased neurogenesis reaches a peak, as previously described.33,34 Briefly, the NSCs localized to the lateral walls of the lateral ventricles of the SVZ in sagittally dissected brain slices were isolated and digested in 0.05% trypsin (MilliporeSigma) at 37°C for 1 hour and the digestion was stopped with an equal volume of 0.1% Trypsin inhibitor (MilliporeSigma). For NSC proliferation, NSCs were cultured in a proliferation medium which consisted of Dulbecco’s modified Eagle’s medium (DMEM)/F-12 medium containing 1% B27 (Life Technologies), epidermal growth factor (EGF, 20 ng/mL, R&D System, USA), and basic fibroblast growth factor (bFGF, 20 ng/mL, R&D System). The formation of neurospheres was measured and characterized.35 For NSC differentiation, the primary neurospheres were mechanically dissociated and seeded as single cells. The cells were incubated in the differentiation medium which consisted of DMEM/F-12 medium and 2% fetal bovine serum but without the growth factors. Passages 2 to 5 of the primary NSCs were used in the following experiments.
2.4 |. LncRNA array analysis
Total RNA was isolated from NSCs using TRIzol reagent and quantified using the NanoDrop ND-1000. Then, each RNA sample was amplified and transcribed into cDNA. The labeled cDNAs were then hybridized onto Rat LncRNA Array v2.0 (Arraystar). The arrays were scanned using the Agilent Scanner G2505C.
2.5 |. mRNA-sequencing analysis
Approximate 2 μg total RNA from each sample was used to prepare the sequencing. Agilent 2100 Bioanalyzer was used to qualify the completed libraries, and RNA content was quantified by real-time PCR absolute quantification method. The differentially expressed genes and transcripts were filtered using R package Ballgown. The novel genes were predicted by comparing to the reference annotation using StringTie and Ballgown.
2.6 |. Gene ontology (GO)
Gene ontology was used to describe gene and gene product properties. The ontology covers biological process, cellular component, and molecular function. The P-value denotes the significance of GO terms enrichment in the deregulated genes (a P-value ≤.05 is considered significant).
2.7 |. LncRNA-protein-coding gene coexpression analysis
The coexpression network for lncRNAs and the associated protein-coding genes was constructed.36 For each pair of genes, Pearson correlation coefficients (PCCs) between the normalized data of provided lncRNAs and all coding gene expression were calculated. The coding genes with abs (PCC) ≥0.9 and FDR ≤0.05 were selected.
2.8 |. H19 knockdown with clustered regularly interspaced short palindromic repeat-associated system (CRISPR) associated 9 (Cas9) genome editing
CAG-mKate2-Cas9 expression plasmid DNA, trans-activating crRNA (tracrRNA), and single-guide RNAs (sgRNA nontargeting control [sgRNA-NC] and sgRNA-H19) were purchased from Dharmacon (Lafayette, Colorado). Four sgRNAs containing seed sequence targeting H19 were synthesized by Dharmacon. For each transfection, NSCs (~106) were electroporated in electroporation buffer containing 10 μg CAG-mKate-Cas9 expression plasmid DNA using nucleofector electroporation with program A33 (Amaxa Biosystems, Germany). The cells were incubated for 48 hours and transfected with a sgRNA-tracrRNA mixture using the same electroporation program. After an additional 48 hours, cells were used for the following procedure.
2.9 |. Production of adeno-associated virus carrying H19 construction and delivery
Guide sequences and guide RNA scaffold were subcloned into the corresponding sites of pAV-FH (Vigene Biosciences, Maryland) containing the adeno-associated virus (AAV) vector backbone with Cre recombinase. Mouse Ascl1 clone (MPRM39894, GeneCopoeia) was subcloned into the pAV-FH plasmid. Infectious recombinant AAV vector particles were generated in HEK293T cells by a cross-packaging approach whereby the vector genome was packaged into AAV serotype-9. Viral stocks were obtained through PEG precipitation and CsCl2 gradient centrifugation and stored at −80°C until use. The physical titer of recombinant AAVs was quantified using real-time PCR against a standard curve of a plasmid containing vector genome (vg).
For in vitro infection, NSCs were incubated with AAV-containing media (104 vg per cell) for 12 hours and an equal volume of media was added to the cultures. Seven days after the infection, NSCs were lysed and gene expression was tested.
For in vivo ablation of H19, floxed Cas9 knockin mice were used. Briefly, AAV-H19 gRNA (2 μL, 2 × 1013 vg/mL) was injected into the ipsilateral SVZ and hippocampus of transgenic mice. Mice that received AAV-GFP injection were used as a control. One week later, all the animals were subjected to MCAO surgery. Three weeks after MCAO, mice were euthanized and their brains were processed for neurogenesis analysis according to our published protocol.34
2.10 |. RNA immunoprecipitation (RIP)
RIP was performed using Magna RIP kit (MilliporeSigma). One hundred microliters of NSCs lysates were incubated with magnetic protein A/G beads precoated with SUZ12 or EZH2 antibody (Abcam) at 4°C overnight. Digested the protein with Proteinase K and isolated the immunoprecipitated RNA for quantitative RT-PCR (qRT-PCR) analysis. The presence of the binding targets was detected using the respective primers.
2.11 |. Chromatin Immunoprecipitation (ChIP) coupled with quantitative PCR
NSCs were cross-linked with 1% (v/v) formaldehyde for 10 minutes and sonicated until DNA was sheared to the lengths of 200 ~ 1000 bps. Chromatin extracts were immunoprecipitated with H3K27me3 antibody (Abcam) on Protein A Agarose/Salmon Sperm DNA. The ChIP DNA was isolated and the enrichment of H3K27me3 in the promoter of p27 was analyzed using real-time PCR and SYBR Green probes.
2.12 |. Chromatin isolation by RNA purification (ChIRP)
ChIRP was performed using Magna ChIRP RNA Interactome Kit (MilliporeSigma) following the manufacturer’s protocol. Biotinylated H19 probes were designed by Biosearch Technologies (Petaluma) and labeled as odd and even groups.
2.13 |. Statistical analysis
Data are presented as mean ± SE of the mean (SEM). The GraphPad Prism software (GraphPad Software Inc, California) was used for statistical analysis. Nonparametric one-way analysis of variance (ANOVA) followed by Tukey’s post hoc test was performed for multiple groups comparisons. Two-tailed Student’s t test was performed for two-group comparisons. The ImageJ software was used to quantify the band intensity in Western blot images. Quantification was performed from at least three independent experiments. A value of P < .05 was considered significant.
3 |. RESULTS
3.1 |. The alteration of lncRNAs and mRNAs profiles in adult neural stem cells after stroke
We first performed lncRNA profile analysis in NSCs isolated from nonischemic and ischemic rats. In total, 6101 lncRNAs were detected. Among the significantly and differentially expressed transcripts, 155 lncRNAs were downregulated (<0.33-fold), 86 lncRNAs were upregulated (>1.5-fold) after stroke (Figure 1A). The top three elevated lncRNAs in ischemic NSCs were lnc-NR_027324 (H19, 20-fold), lnc-EF094477 (6-fold), and lnc-BC090003 (6-fold), while top three decreased ones were lnc-M64384 (−5-fold), lnc-MRAK013682 (−4-fold), and lnc-MRAK051099 (−4-fold). mRNA-sequencing analysis showed a total of 11 591 mRNAs were detected in adult NSCs. 409 were significantly upregulated (>1.5-fold) and 255 were downregulated (<0.33-fold) in ischemic NSCs compared with nonischemic NSCs (Figure S1). GO analysis demonstrated that the stroke-induced upregulated mRNAs in NSCs were correlated with chemokine signaling and cell cycle pathways (Figure 1B), whereas the downregulated mRNAs were associated with calcium signaling, glutathione metabolism, and lysosome pathways (Figure 1C).
3.2 |. Networks of lncRNAs and protein-coding genes in NSCs
LncRNAs regulate the expression or function of genes in cis.37 In ischemic NSCs, four upregulated and two downregulated lncRNAs were positively correlated with the genes in cis, but no negative correlation was detected. Gene coexpression networks were built according to PCCs between lncRNA and mRNA expression. The network consists of upregulated lncRNAs showed 1679 positive matched lncRNA-mRNA correlations (Figure S2A). The network consists of downregulated lncRNAs identified 1944 negative matched lncRNA-mRNA correlations (Figure S2B). Gene set enrichment analysis (GESA) was performed to predict the functions of coexpressed genes. The coexpressed genes positively modulated by lncRNAs were related to cognition, synapse plasticity, neuron development, mitochondrion, calcium channel regulator, neurotransmitter signal, etc. The coexpressed genes negatively regulated by lncRNAs were associated with cytokine production, DNA replication, cell adhesion, NF-κB signaling, metabolic process, T cell process, and so on.
3.3 |. Stroke upregulates lncRNA H19 expression in NSCs
H19 (NR_027324) was the most upregulated lncRNA after stroke. H19 expression was significantly increased by 19- and 9-fold in ischemic SVZ and DG, compared with the nonischemic tissues (Figure 2A). In situ hybridization (ISH) further confirmed that H19 expression was enhanced in ischemic SVZ (Figure 2B). In contrast, the content of H19 was not increased in cortex and striatum (Figure 2A). Using fluorescence-activated cell sorting (FACS), we isolated Ascl1 positive NSCs from ischemic Ascl1-CreERT2;tomato reporter mice and DCX positive neuroblasts from and DCX-CreERT2;tomato reporter mice. QRT-PCR analysis revealed that H19 was mainly expressed in Ascl1+ NSCs rather than DCX+ neuroblasts (Figure 2C).
3.4 |. Ablation of H19 suppresses the proliferation and neuronal differentiation of NSCs in vitro
We knocked down H19 in ischemic NSCs using CRISPR-Cas9 genome editing, and examined the alteration in the self-renewal and multipotency of ischemic NSCs (Figure 2D). Among the four sgRNAs designed to pair H19 (sgH19), sgH19-1 caused a 70% reduction in H19 expression compared with nontargeting control sgRNA (sgRNA-NC) in ischemic NSCs (Figure 2D–F). Immunostaining analysis demonstrated that the H19 knockdown significantly decreased the proliferation rate and increased the apoptosis of ischemic NSCs, as assayed by the percentage of BrdU+ cell (Figure 2G,H) and Caspase-3/7 activity (Figure 2J), compared with sgRNA-NC treated cells. Moreover, H19 knockdown significantly reduced the percentage of Tuj1+ neurons, NG2+ oligodendrocyte progenitor cells (OPCs), and GFAP+ astrocytes in ischemic NSCs (Figure 2G–I).
3.5 |. Effects of H19 on the key genes in the proliferation and survival of NSCs
To elucidate the effects of H19 in the regulation of NSCs growth and differentiation, RNA-seq analysis was performed to examine transcriptome alteration after H19 knockdown. Compared with ischemic NSCs treated with sgRNA-NC, 356 genes were upregulated (>2-fold), whereas 228 genes were downregulated (<0.5-fold) in H19 knockdown cells. GO analysis revealed that upregulated genes were closely related to oxidative response, NF-κβ signaling, cell cycle, translation, proliferation, apoptosis, response to hypoxia, and transcription (Figure 3A). Many of these upregulated genes located both in nucleus and cytoplasm (Figure 3B), and this result was further confirmed using ISH analysis (Figure 3C). Furthermore, qRT-PCR analysis revealed that H19 content was enhanced in both cytoplasmic and nuclear RNA extractions of ischemic NSCs (Figure 3D).
We then examined the impact of cytoplasmic and nuclear H19 on gene transcription. Western blot analysis revealed that the expression of cell cycle-related genes, as well as apoptosis-related genes, was increased upon H19 depletion (Figure 3E,F). These genes have been shown to inhibit NSC proliferation and survival.38 In contrast, H19 overexpression (H19+/+) in NSCs by AAV9-H19 cDNA infection led to the inverse expression pattern of the genes (Figure 3E,F). These data suggest that H19 mediates the transcriptional program that plays critical roles in regulating cell cycles and programmed cell death.
3.6 |. H19 improves repressive H3K27me3 histone modification
Using ChIP coupled with qPCR, we found that repressive H3K27me3 histone modification was highly enriched in the bp −2000 to −1000 from the transcription start site of p27 in ischemic NSCs (Figure 4A). Furthermore, knockdown of H19 directly reduced the enrichment of the H3K27me3 in the promoter of the p27 gene (Figure 4B), leading to the upregulation of p27 expression in NSCs (Figure 3E,F). These results indicate that repressive H3K27me3 histone modification in neurogenesis-related gene transcription is involved in the regulation of H19 in poststroke neurogenesis.
3.7 |. H19 interacts with SUZ12 and EZH2 chromatin remodeling factors
Polycomb repressive complex 2 (PRC2) induces gene silencing by methylation of histone H3 on K27 via its subunit EZH2.39 We next performed RIP using antibodies against SUZ12 and EZH2 and observed a significant enrichment of H19 to chromatin proteins SUZ12 and EZH2 in NSCs after stroke (Figure 4C). Using ChIRP-Western blot, SUZ12, EZH2, H3K27me3 were observed significantly higher in the purified H19 regulatory complexes isolated from ischemic NSCs compared with those from nonischemic NSCs (Figure 4D). Also, stroke significantly increased SUZ12 and EZH2 in NSCs derived from SVZ and SGZ, whereas histone active marker H3K4me3 exhibited a significant decrease (Figure 4E,F). These data suggest that H19 promotes SUZ12 and EZH2 recruitment after stroke.
3.8 |. H19 regulates its encoding miR-675 expression in NSCs
H19 is the primary precursor of miR-675.40 We found the expression of miR-675-5p and −3p was significantly increased after stroke (Figure 5A), which was positively correlated with the upregulation of H19. Furthermore, when the H19 expression was manipulated, the content of miR-675-3p and miR-675-5p showed an identical change trend (Figure 5B). Meanwhile, we observed that the expression of insulin-like growth factor-1 receptor (IGF1R) was significantly reduced in H19 knockdown NSCs (Figure 5C,D), indicating IGF1R was not a direct target gene of H19/miR-675. Further analysis revealed that TGF-β1, a target gene of miR-675, showed an inverse expression pattern of miR-675 (Figure 5C,D). These data suggest that H19/miR-675/TGF-β1 participates in the regulation of poststroke neurogenesis.
3.9 |. Argonaute 2 is indispensable for H19-regulated miRNAs
Argonaute 2 (Ago2) is a key RNA binding protein in miRNA processing and function.41 We assessed whether Ago2 was required in the miRNA regulation of H19. NSCs were isolated from transgenic mice with conditional ablation of Ago2 in Ascl1 lineage cells. Western blot analysis showed that Ago2 expression was suppressed compared with wild-type mice (Figure 5F,G). QRT-PCR analysis showed that the ablation of Ago2 in Ascl1 lineage cells significantly reduced levels of H19 and encoding miR-675 compared with NSCs isolated from wild-type mice (Figure 5E). Meanwhile, TGF-β1 was significantly increased in Ago2-KO NSCs (Figure 5F,G). To determine the mechanism underlying Ago2-regulated H19 expression, we blocked the on-going transcription using actinomycin D (AMD) and observed that Ago2-KO enhanced H19 degradation (Figure 5H).
We and others previously demonstrated that miRNAs contribute to stroke-increased neurogenesis.35,42 However, whether deregulated lncRNAs affect miRNAs that mediate stroke-increased neurogenesis remains unknown. When H19 was knocked down in ischemic NSCs by sgH19, miR-146a, miR-17-92 cluster, miR-21, miR-124a, and miR-23 were substantially suppressed (Figure 5I). Besides, H19 knockdown did not significantly alter Ago2 expression, but substantially reduced the expression of Dicer in NSCs (Figure 5J,K).
3.10 |. H19 inactivation in neural stem cells attenuates motor and cognitive function after stroke
To assess the impact of H19 on poststroke functional outcome, we selectively knocked down H19 in NSCs by injection of AAV-H19 gRNA into the ipsilateral SVZ of floxed Cas9 knockin mice. Compared with ischemic mice that received AAV-GFP, ischemic mice treated with AAV-H19 gRNA exhibited significant impairments of neurological function at 14 and 21 days after MCAO assayed by an array of behavioral tests (Figure 6A,B). Moreover, ischemic mice with H19 knockdown in NSCs significantly diminished their sociability and novel object recognition compared with the mice treated with AAV-GFP (Figure 6C,D). RT-PCR analysis showed the downregulation of H19 both in the SVZ and in the hippocampus (Figure S3). Double-immunofluorescent staining analysis demonstrated that knockdown of H19 in NSCs significantly reduced the numbers of GFP+/BrdU+ proliferating progenitor cells and GFP+/DCX+ neuroblasts in SVZ and DG of the hippocampus (Figure 6E–L), as well as reduced GFP+/calretinin+ newly born neurons in DG (Figure 6M,N).
4 |. DISCUSSION
In this study, we for the first time provide a comprehensive lncRNA profile in NSCs and analyze the association of lncRNAs with corresponding protein-coding genes after stroke. Ischemic stroke robustly upregulated lncRNA H19 in NSCs. The deletion of H19 suppressed the proliferation and survival and blocked the neuronal differentiation of NSCs, leading to an exacerbation of motor and cognitive deficits after stroke. H19 physically interacted with and regulated levels of enriched SUZ12 and EZH2 chromatin proteins that catalyzed H3K27 trimethylation on p27. Besides, H19 regulated neurogenesis-related miRNAs in the NSCs. These data indicate that H19 contributes to stroke-increased neurogenesis.
The roles of lncRNAs in neurogenesis have been recently unveiled.43 We systematically analyzed transcriptome profile alteration in ischemic and nonischemic NSCs in attempting to identify lncRNAs that are potentially involved in the regulation of neurogenesis. Six lncRNAs were discovered positively associated with their adjacent genes (Table. 1). Stat1 is reported to mediate neurogenesis when activated by interferon-γ.44,45 Lamp2 is a lysosome marker gene and participates in ischemic neuronal cell damage.46 GFAP is a widely used marker for NSCs.35,47 Rt1.aa, an MHC class antigen, was increased after stroke and correlated with lncRNA NR_002597. The function of Zfhx2 and Cyld in adult neurogenesis has not been documented in adult NSCs after stroke, but both genes are involved in cell proliferation and differentiation.48,49 PCC analysis revealed numerous integrative networks of deregulated lncRNAs with distant genes. Further investigation is needed to elucidate mechanisms underlying the effects of lncRNA poststroke.
TABLE 1.
LncRNAs | Fold changes | Associated Gene name | Associated_protein_name | Fold changes | Relationship |
---|---|---|---|---|---|
MRAK088874 | 2.49 | Stat1 | signal transducer and activator of transcription | 4.41 | sense_exon_overlap |
MRAK080234 | 2.19 | Lamp2 | lysosomal-associated membrane protein 2 | 2.71 | sense_intron_overlap |
EF094477 | 6.38 | GFAP | glial fibrillary acidic protein | 3.57 | sense_exon_overlap |
NR_002597 | 2.29 | Rt1.aa | MHC class I RT1.Aa alpha-chain | 3.76 | sense_intron_overlap |
MRAK138051 | −1.75 | Zfhx2 | zinc finger homeobox 2 | −3.36 | sense_exon_overlap |
AY383658 | −2.09 | Cyld | ubiquitin carboxyl-terminal hydrolase CYLD | −2.23 | sense_exon_overlap |
The present study showed that H19 was among the most upregulated lncRNAs in ischemic NSCs and H19 knockdown in NSCs led to suppressed proliferation and neuronal differentiation, which was associated with the induction of cognitive deficits. Using a transgenic ablation of doublecortin-expressing cells, with doublecortin as a marker of neuroblasts, and nestin transgenic mice, recent studies have demonstrated that ablation of newly generated neuroblasts or NSCs significantly suppresses the neurogenesis in the SVZ and the SGZ of the hippocampus, leading to the impairment of cognition.50,51 Moreover, the ablation of newly generated neuroblasts and NSCs in the ischemic brain reduces spontaneous functional recovery.52 It is reasonable to assume that H19 may improve neurological outcomes after stroke.
SUZ12 and EZH2 are polycomb repressive complex 2 (PRC2) core proteins that catalyze the di/trimethylation of H3K27.53,54 We observed that upregulated H19 was physically associated with SUZ12 and EZH2 in NSCs. The absence of H19 markedly reduced levels of EZH2 and H3K27me3 at the promoters of cell cycle-related genes. Also, we demonstrated that H19 not only directly regulates its encoding miRNAs but also indirectly impacts its nonneighboring miRNAs, which is consistent with previous studies showing that lncRNAs can function as molecular decoys of miRNAs.6 Meanwhile, we found that ablation of Ago2 in Ascl1 lineage NSCs reduces the expression of H19 and its stability and related miRNAs. In comparison, knockdown of H19 has no significant effect on Ago2 expression, suggesting Ago2 is required for H19-mediated miRNA modulation but not vice versa. Interestingly, we revealed that knockdown of H19 reduces Dicer expression, another key factor in the miRNA biogenesis, which provides new insight into the role of lncRNA in miRNA biogenesis. Nevertheless, it remains to be determined how H19 regulates the transcription or translation of the Dicer gene in NSCs. Our novel observations suggest that H19 regulates stroke-induced neurogenesis by altering the epigenetic states to modulate the transcriptional silencing of genes as well as by the interaction with noncoding miRNAs, which underscore molecular mechanisms underlying the effect of H19 on adult neurogenesis.
Recently, it was reported that H19 is involved in the neuroinflammation and autophagy at the acute stage of stroke, indicating a detrimental effect of H19 on stroke.10,55 The differences between our study and the prior reports may lie in the stages poststroke that were focused on. In this study, we mainly investigated the differential role of H19 at the neurorestorative stage. GESA analysis revealed that H19 was correlated with immune responses signaling pathways, such as Toll-like receptors (TLR). In the later stage poststroke, the immune response can promote NSCs proliferation and neuroblast migration.56,57
Adult neurogenesis declines in aging rodents and primates. During aging, NSCs and their progenitors display reduced proliferation and new neuron generation, which contribute to age-related cognitive impairment and reduced neuroplasticity for brain repair.58,59 Compared with young animals, aged animals subjected to stroke showed more severe impairment of neurogenesis and worse neurological recovery, suggesting that age affects the stroke-induced neurogenesis and neurological outcomes. Dysregulation of lncRNAs is associated with age-associated pathologies and related neurodegenerative disorders.60,61 It remains unknown if lncRNAs will contribute to the stroke-induced neurogenesis in aging rodents. Further studies to investigate the function of H19 in neurogenesis using aging animals are warranted.
5 |. CONCLUSION
Therapies designed to boost neurogenesis following stroke offers a potentially effective approach to treatment aimed at improving recovery. Our study may set the groundwork for the development of a lncRNA-based therapy for improvement of neurological function after stroke.
Supplementary Material
Significance statement.
Adult neurogenesis contributes to neurological function. Elucidating the underlying molecular mechanisms in post-stroke neurogenesis could provide new therapies to amplify endogenous neurogenesis and to improve neurological function during stroke recovery. The present study for the first time reveals that stroke substantially changes lncRNA profiles and lncRNA-mRNA coexpression networks in the neural stem cells. Furthermore, it demonstrates that lncRNA H19 mediates stroke-augmented neurogenesis by recruiting chromatin remodeling proteins and regulating microRNA expression to modulate neurogenesis-related transcription. Our results provide new insights into the epigenetic control of adult neurogenesis after cerebral ischemia.
ACKNOWLEDGMENTS
We thank Julie Landschoot-Ward, Qinge Lu, and Sutapa Santra for the immunostaining. This study was supported by American Heart/Stroke Association Grant (18IPA34170331); U.S. National Institutes of Health (NIH) National Institute of Diabetes and Digestive and Kidney Diseases Grant RO1 (DK102861); and NIH National Institute of Neurological Disorders and Stroke Grants RO1 (NS075156) and RO1 (NS088656).
Funding information
American Heart Association, Grant/Award Number: 18IPA34170331; NIH National Institute of Diabetes and Digestive and Kidney Diseases, Grant/Award Number: DK102861; NIH National Institute of Neurological Disorders and Stroke, Grant/Award Number: NS075156; NIH National Institute of Neurological Disorders and Stroke Grants, Grant/Award Number: NS088656
Footnotes
CONFLICT OF INTEREST
The authors declared no potential conflicts of interest.
SUPPORTING INFORMATION
Additional supporting information may be found online in the Supporting Information section at the end of this article.
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available on request from the corresponding author.
REFERENCES
- 1.Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart disease and stroke statistics—2017 update: a report from the American Heart Association. Circulation. 2017;135(10):e146–e603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Doetsch F A niche for adult neural stem cells. Curr Opin Genet Dev 2003;13(5):543–550. [DOI] [PubMed] [Google Scholar]
- 3.Jin K, Minami M, Lan JQ, et al. Neurogenesis in dentate subgranular zone and rostral subventricular zone after focal cerebral ischemia in the rat. Proc Natl Acad Sci USA 2001;98(8):4710–4715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jin K, Wang X, Xie L, et al. Evidence for stroke-induced neurogenesis in the human brain. Proc Natl Acad Sci USA 2006;103(35):13198–13202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ulitsky I, Bartel DP. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154(1):26–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43(6):904–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009;10(3):155–159. [DOI] [PubMed] [Google Scholar]
- 8.Mercer TR, Dinger ME, Sunkin SM, Mehler MF, Mattick JS. Specific expression of long noncoding RNAs in the mouse brain. Proc Natl Acad Sci USA 2008;105(2):716–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dong X, Chen K, Cuevas-Diaz Duran R, et al. Comprehensive identification of long non-coding RNAs in purified cell types from the brain reveals functional LncRNA in OPC fate determination. PLoS Genet 2015;11(12):e1005669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wang J, Zhao H, Fan Z, et al. Long noncoding RNA H19 promotes neuroinflammation in ischemic stroke by driving histone deacetylase 1-dependent M1 microglial polarization. Stroke. 2017;48(8):2211–2221. [DOI] [PubMed] [Google Scholar]
- 11.Wu Z, Wu P, Zuo X, et al. LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation. Mol Neurobiol 2017;54(10):7670–7685. [DOI] [PubMed] [Google Scholar]
- 12.Zhang L, Luo X, Chen F, et al. LncRNA SNHG1 regulates cerebrovascular pathologies as a competing endogenous RNA through HIF-1alpha/VEGF signaling in ischemic stroke. J Cell Biochem 2018;119 (7):5460–5472. [DOI] [PubMed] [Google Scholar]
- 13.Xu Q, Deng F, Xing Z, et al. Long non-coding RNA C2dat1 regulates CaMKIIδ expression to promote neuronal survival through the NF-κB signaling pathway following cerebral ischemia. Cell Death Dis 2016;7: e2173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Qiao Y, Peng C, Li J, Wu D, Wang X. LncRNA MALAT1 is neuro-protective in a rat model of spinal cord ischemia-reperfusion injury through miR-204 regulation. Curr Neurovasc Res 2018;15(3):211–219. [DOI] [PubMed] [Google Scholar]
- 15.Chen S, Wang M, Yang H, et al. LncRNA TUG1 sponges microRNA-9 to promote neurons apoptosis by up-regulated Bcl2l11 under ischemia. Biochem Biophys Res Commun 2017;485(1):167–173. [DOI] [PubMed] [Google Scholar]
- 16.Chen F, Zhang L, Wang E, Zhang C, Li X. LncRNA GAS5 regulates ischemic stroke as a competing endogenous RNA for miR-137 to regulate the Notch1 signaling pathway. Biochem Biophys Res Commun 2018;496(1):184–190. [DOI] [PubMed] [Google Scholar]
- 17.Mehta SL, Kim T, Vemuganti R. Long noncoding RNA FosDT promotes ischemic brain injury by interacting with REST-associated chromatin-modifying proteins. J Neurosci 2015;35(50):16443–16449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yan H, Yuan J, Gao L, Rao J, Hu J. Long noncoding RNA MEG3 activation of p53 mediates ischemic neuronal death in stroke. Neuroscience. 2016;337:191–199. [DOI] [PubMed] [Google Scholar]
- 19.Ruan W, Wang P, Feng S, Xue Y, Li Y. Long non-coding RNA small nucleolar RNA host gene 12 (SNHG12) promotes cell proliferation and migration by upregulating angiomotin gene expression in human osteosarcoma cells. Tumor Biol 2016;37(3):4065–4073. [DOI] [PubMed] [Google Scholar]
- 20.Gordon FE, Nutt CL, Cheunsuchon P, et al. Increased expression of angiogenic genes in the brains of mouse meg3-null embryos. Endocrinology. 2010;151(6):2443–2452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Michalik KM, You X, Manavski Y, et al. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ Res 2014;114(9):1389–1397. [DOI] [PubMed] [Google Scholar]
- 22.Ramos AD, Andersen RE, Liu SJ, et al. The long noncoding RNA Pnky regulates neuronal differentiation of embryonic and postnatal neural stem cells. Cell Stem Cell. 2015;16(4):439–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ng SY, Bogu GK, Soh BS, Stanton LW. The long noncoding RNA RMST interacts with SOX2 to regulate neurogenesis. Mol Cell. 2013; 51(3):349–359. [DOI] [PubMed] [Google Scholar]
- 24.Monnier P, Martinet C, Pontis J, Stancheva I, Ait-Si-Ali S, Dandolo L. H19 lncRNA controls gene expression of the imprinted gene network by recruiting MBD1. Proc Natl Acad Sci USA 2013;110(51):20693–20698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gabory A, Ripoche MA, Le Digarcher A, et al. H19 acts as a trans regulator of the imprinted gene network controlling growth in mice. Development. 2009;136(20):3413–3421. [DOI] [PubMed] [Google Scholar]
- 26.Ghazal S, McKinnon B, Zhou J, et al. H19 lncRNA alters stromal cell growth via IGF signaling in the endometrium of women with endometriosis. EMBO Mol Med 2015;7(8):996–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dey BK, Pfeifer K, Dutta A. The H19 long noncoding RNA gives rise to microRNAs miR-675–3p and miR-675–5p to promote skeletal muscle differentiation and regeneration. Genes Dev 2014;28(5):491–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Jacob KJ, Robinson WP, Lefebvre L. Beckwith-Wiedemann and Silver-Russell syndromes: opposite developmental imbalances in imprinted regulators of placental function and embryonic growth. Clin Genet 2013;84(4):326–334. [DOI] [PubMed] [Google Scholar]
- 29.Ratajczak MZ. Igf2-H19, an imprinted tandem Yin-Yanggene and its emerging role in development, proliferation of pluripotent stem cells, senescence and cancerogenesis. J Stem Cell Res Ther 2012;2(4). pii: 108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tabano S, Colapietro P, Cetin I, et al. Epigenetic modulation of the IGF2/H19 imprinted domain in human embryonic and extraembryonic compartments and its possible role in fetal growth restriction. Epigenetics. 2010;5(4):313–324. [DOI] [PubMed] [Google Scholar]
- 31.Huang Y, Zheng Y, Jin C, Li X, Jia L, Li W. Long non-coding RNA H19 inhibits adipocyte differentiation of bone marrow mesenchymal stem cells through epigenetic modulation of histone deacetylases. Sci Rep 2016;6:28897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Poirier F, Chan CT, Timmons PM, Robertson EJ, Evans MJ, Rigby PW. The murine H19 gene is activated during embryonic stem cell differentiation in vitro and at the time of implantation in the developing embryo. Development. 1991;113(4):1105–1114. [DOI] [PubMed] [Google Scholar]
- 33.Zhang RL, Zhang ZG, Zhang L, Chopp M. Proliferation and differentiation of progenitor cells in the cortex and the subventricular zone in the adult rat after focal cerebral ischemia. Neuroscience. 2001; 105(1):33–41. [DOI] [PubMed] [Google Scholar]
- 34.Zhang R, Zhang Z, Wang L, et al. Activated neural stem cells contribute to stroke-induced neurogenesis and neuroblast migration toward the infarct boundary in adult rats. J Cereb Blood Flow Metab 2004;24 (4):441–448. [DOI] [PubMed] [Google Scholar]
- 35.Liu XS, Chopp M, Zhang RL, et al. MicroRNA profiling in subventricular zone after stroke: MiR-124a regulates proliferation of neural progenitor cells through Notch signaling pathway. PLoS One. 2011;6(8):e23461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 2005;102(43):15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Orom UA, Derrien T, Beringer M, et al. Long noncoding RNAs with enhancer-like function in human cells. Cell. 2010;143(1):46–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ramasamy S, Narayanan G, Sankaran S, Yu YH, Ahmed S. Neural stem cell survival factors. Arch Biochem Biophys 2013;534(1–2):71–87. [DOI] [PubMed] [Google Scholar]
- 39.Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D. Histone methyltransferase activity associated with a human multiprotein complex containing the enhancer of zeste protein. Genes Dev 2002;16(22):2893–2905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Keniry A, Oxley D, Monnier P, et al. The H19 lincRNA is a developmental reservoir of miR-675 that suppresses growth and Igf1r. Nat Cell Biol 2012;14(7):659–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hock J, Meister G. The Argonaute protein family. Genome Biol 2008; 9(2):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu XS, Fan BY, Pan WL, et al. Identification of miRNomes associated with adult neurogenesis after stroke using Argonaute 2-based RNA sequencing. RNA Biol 2017;14(5):488–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Deng B, Cheng X, Li H, Qin J, Tian M, Jin G. Microarray expression profiling in the denervated hippocampus identifies long noncoding RNAs functionally involved in neurogenesis. BMC Mol Biol 2017;18 (1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Borsini A, Cattaneo A, Malpighi C, et al. Interferon-alpha reduces human hippocampal neurogenesis and increases apoptosis via activation of distinct STAT1-dependent mechanisms. Int J Neuropsychopharmacol 2018;21(2):187–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pereira L, Medina R, Baena M, et al. IFN gamma regulates proliferation and neuronal differentiation by STAT1 in adult SVZ niche. Front Cell Neurosci 2015;9:270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rothaug M, Stroobants S, Schweizer M, et al. LAMP-2 deficiency leads to hippocampal dysfunction but normal clearance of neuronal substrates of chaperone-mediated autophagy in a mouse model for Danon disease. Acta Neuropathol Commun 2015;3:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pan WL, Chopp M, Fan B, et al. Ablation of the microRNA-17–92 cluster in neural stem cells diminishes adult hippocampal neurogenesis and cognitive function. FASEB J 2019;33(4):5257–5267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Komine Y, Takao K, Miyakawa T, Yamamori T. Behavioral abnormalities observed in Zfhx2-deficient mice. PLoS One. 2012;7 (12):e53114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tauriello DV, Haegebarth A, Kuper I, et al. Loss of the tumor suppressor CYLD enhances Wnt/beta-catenin signaling through K63-linked ubiquitination of Dvl. Mol Cell. 2010;37(5):607–619. [DOI] [PubMed] [Google Scholar]
- 50.Jin K, Wang X, Xie L, Mao XO, Greenberg DA. Transgenic ablation of doublecortin-expressing cells suppresses adult neurogenesis and worsens stroke outcome in mice. Proc Natl Acad Sci USA 2010;107 (17):7993–7998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Curtis MA, Kam M, Nannmark U, et al. Human neuroblasts migrate to the olfactory bulb via a lateral ventricular extension. Science. 2007; 315(5816):1243–1249. [DOI] [PubMed] [Google Scholar]
- 52.Sun F, Wang X, Mao X, Xie L, Jin K. Ablation of neurogenesis attenuates recovery of motor function after focal cerebral ischemia in middle-aged mice. PLoS One. 2012;7(10):e46326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Portoso M, Ragazzini R, Brencic Z, et al. PRC2 is dispensable for HOTAIR-mediated transcriptional repression. EMBO J 2017;36(8): 981–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett 2013;333(2): 213–221. [DOI] [PubMed] [Google Scholar]
- 55.Wang J, Cao B, Han D, Sun M, Feng J. Long non-coding RNA H19 induces cerebral ischemia reperfusion injury via activation of autophagy. Aging Dis 2017;8(1):71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Thored P, Heldmann U, Gomes-Leal W, et al. Long-term accumulation of microglia with proneurogenic phenotype concomitant with persistent neurogenesis in adult subventricular zone after stroke. Glia 2009;57(8):835–849. [DOI] [PubMed] [Google Scholar]
- 57.Yan YP, Sailor KA, Lang BT, Park SW, Vemuganti R, Dempsey RJ. Monocyte chemoattractant protein-1 plays a critical role in neuroblast migration after focal cerebral ischemia. J Cereb Blood Flow Metab 2007;27(6):1213–1224. [DOI] [PubMed] [Google Scholar]
- 58.Darsalia V, Heldmann U, Lindvall O, Kokaia Z. Stroke-induced neurogenesis in aged brain. Stroke. 2005;36(8):1790–1795. [DOI] [PubMed] [Google Scholar]
- 59.Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD, Sonntag WE, Riddle DR. Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience. 2001;107(4):603–613. [DOI] [PubMed] [Google Scholar]
- 60.Wan P, Su W, Zhuo Y. The role of long noncoding RNAs in neurodegenerative diseases. Mol Neurobiol 2017;54(3):2012–2021. [DOI] [PubMed] [Google Scholar]
- 61.Wei CW, Luo T, Zou SS, Wu AS. The role of long noncoding RNAs in central nervous system and neurodegenerative diseases. Front Behav Neurosci 2018;12:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support the findings of this study are available on request from the corresponding author.