Haifang Wang's research while affiliated with Chinese Academy of Sciences and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (12)


Correction: Single-cell in vivo imaging of cellular circadian oscillators in zebrafish
  • Article
  • Full-text available

August 2021

·

26 Reads

PLOS Biology

PLOS Biology

Haifang Wang

·

·

Xingxing Li

·

[...]

·

[This corrects the article DOI: 10.1371/journal.pbio.3000435.].

Download
Share

Molecular changes in the neurogenic niche induced by ischemia. (A) Volcano plot displays changes in gene expression in monkey SVZa. Transcripts up-or down-regulated and those unchanged are in red or blue and gray. (B) Cellular architecture of SVZa on the striatal and the callosal side of the lateral ventricle. For details see Text. (C,D) Heat maps illustrating gene expression changes of 150 genes in EL, SEL, CSVZ, STR and PVSEL upon ischemia. The 36 genes implicated in stem cell biology are marked (*). Original data are on “monkey-niche.org.” Expression of 150 genes, deposited in the “monkey-niche” database (Supplementary Table S7) is shown for five regions of interest (Figure 1B). The genes in (C,D) are grouped according to their postischemic upregulation: Group 1 (markedly enhanced expression levels in all RoIs), Group 2 (predominantly upregulated in the RoIs on the striatal side of the ventricle), Group 3 (strongly induced in EL, SEL and CVSZ), Group 4 (strongly induced in EL, CSVZ and STR), Group 5 (strongly induced in EL and SEL), Group 6 (strongly induced in EL and STR), Group 7 (strongly induced in EL), Group 8 (strongly induced in STR), Group 9 (genes with low level of expression or not belonging to the other groups).
Bioinformatics analysis of the genes differentially induced after ischemia in monkey SVZa. (A,B) Pie charts showing the Gene Ontology (GO) categories for SVZa up-regulated genes (SVZa-DE-UP) and SVZa down-regulated genes (SVZa-DE-DOWN). Only those GO categories are shown for which there is enrichment relative to the total SVZa genes (all genes expressed in the SVZa). The SVZa-DE-UP and SVZa-DE-DOWN genes for each GO category are listed in Supplementary Table S4. (C) GSEA comparison for SVZa-DE-UP versus SVZa-DE-DOWN genes. Fisher’s exact test was applied to identify the sets that show difference between SVZa-DE-UP and SVZa-DE-DOWN genes. The genes that correspond to each category are listed in Supplementary Table S5. (D) Protein interaction network of the differentially expressed transcription factors. The red nodes represent SVZa-DE-UP genes, while the blue nodes represent SVZa-DE-DOWN genes. The thin lines indicate low interaction score (<0.4), while the thick lines indicate medium or high interaction score (≥0.4).
Upregulation of cell-type specific genes in SVZa after ischemia. (A) Relative to the total SVZa genes, the SVZa-DE-UP were enriched for the transcription profiles of astrocytic or oligodendrocytic markers (Zhang et al., 2014). Monkey SVZa-DE-DOWN genes were significantly enriched for endothelial genes. * Fisher’s exact test p-value < 0.05; ** Fisher’s exact test p-value < 0.01; *** Fisher’s exact test p-value < 0.001. (B,C) Comparative heat maps showing differential expression of genes – assessed by visual scoring -which are characteristic for the different cortical cell types listed on the left. Horizontal double arrows refer to ISH data shown in (D–F). (D–F) ISH data illustrating expression of selected genes that are characteristic for astrocytes (D, CLU; E, TNC) or myelinating oligodendrocytes (F, FEZ1). Black arrows point at PVSEL. Expression of each gene was quantified in EL, SEL, and CSVZ (right column) using the Celldetekt. A bar plot shows the expression value of the respective gene as determined by RNA Seq (number of copies normalized by total count derived from 3 control and 3 ischemic brains); p-values are as in Panel A. See Supplementary Table S1 for atlas coordinates of the sections shown in panels (D–F). Scale bar 100 μm.
Prevalence of transcripts characteristic for quiescent NSCs in ischemic SVZa. (A) Monkey total SVZa genes or SVZa-DE genes were compared with the gene datasets enriched in mouse SVZ progenitors (Beckervordersandforth et al., 2010; Codega et al., 2014; Llorens-Bobadilla et al., 2015; Dulken et al., 2017). The enrichment for each of the progenitor transcription profiles in monkey SVZa-DE genes was statistically analyzed. Monkey genes that correspond to each progenitor type are listed in Supplementary Tables S9–S12. * Fisher’s exact test p-value < 0.05; ** Fisher’s exact test p-value < 0.01; *** Fisher’s exact test p-value < 0.001. (B,C) Comparative heat maps of 40 genes from the “monkey-niche.org” database which are found in the datasets of Codega et al., 2014; Dulken et al., 2017; Llorens-Bobadilla et al., 2015. Horizontal double arrows refer to ISH data shown in (D–K). (D–K) ISH data, Celldetekt-based quantification of ISH and RPKM based quantification revealed a region-specific strong post-ischemic enhancement in the EL (D, GPR37L1; E, GNAO1) or in the SEL (F, FAM107A; G, PHKG1). Other genes (H–K) were enhanced in EL, SEL and CSVZ (MLC1, GJA1, ATF7, and NACC2). Gene expression was quantified (right column) using Celldetekt and transcriptomic data as described in Figure 3. See Supplementary Table S1 for atlas coordinates of the sections shown in panels (D–K). Scale bar 100 μm.
Post-ischemic increase of expression of genes of NOTCH, BMP and WNT pathways. (A–C) NOTCH pathway: NOTCH1, RBPJ and LFNG. Note the strong post-ischemic enhancement of these three genes in the EL. (D–F) BMP pathway: BMPR1, NEO1 and CXXC5. Note the post-ischemic enhancement of the genes in the EL. (G–I) WNT pathway: BCAN, TNR and WLS. Note the post-ischemic enhancement of the genes in EL, SEL and for BCAN also in the CSVZ. Gene expression was quantified (right column) using Celldetekt and transcriptomic data as described in Figure 3. See Supplementary Table S1 for atlas coordinates of the sections shown in panels (A–I). Scale bar 100 μm.

+2

Transcriptome Response and Spatial Pattern of Gene Expression in the Primate Subventricular Zone Neurogenic Niche After Cerebral Ischemia

December 2020

·

205 Reads

·

7 Citations

The main stem cell niche for neurogenesis in the adult mammalian brain is the subventricular zone (SVZ) that extends along the cerebral lateral ventricles. We aimed at characterizing the initial molecular responses of the macaque monkey SVZ to transient, global cerebral ischemia. We microdissected tissue lining the anterior horn of the lateral ventricle (SVZa) from 7 day post-ischemic and sham-operated monkeys. Transcriptomics shows that in ischemic SVZa, 541 genes were upregulated and 488 genes were down-regulated. The transcription data encompassing the upregulated genes revealed a profile typical for quiescent stem cells and astrocytes. In the primate brain the SVZ is morphologically subdivided in distinct and separate ependymal and subependymal regions. The subependymal contains predominantly neural stem cells (NSC) and differentiated progenitors. To determine in which SVZa region ischemia had evoked transcriptional upregulation, sections through control and ischemic SVZa were analyzed by high-throughput in situ hybridization for a total of 150 upregulated genes shown in the www.monkey-niche.org image database. The majority of the differentially expressed genes mapped to the subependymal layers on the striatal or callosal aspect of the SVZa. Moreover, a substantial number of upregulated genes was expressed in the ependymal layer, implicating a contribution of the ependyma to stem cell biology. The transcriptome analysis yielded several novel gene markers for primate SVZa including the apelin receptor that is strongly expressed in the primate SVZa niche upon ischemic insult.


Fig 1. Construction of in vivo circadian reporter in zebrafish. (a) The upper graph shows the schematic of nr1d1:VNP construct design. The lower graph shows a magnified view of the nr1d1 promoter sequence used for driving the circadian expression of VNP. The putative RRE (Nr1d1/2 binding site), E-box (Bmal1/Clock binding site), Crx, Otx5, and Crx/Otx5 binding sites were indicated by red oval, blue rectangle, light green oval, yellow rectangle, and dark green oval, respectively. (b) Plot of real-time PCR results of nr1d1 and Venus-pest expression. Each time point has 2 replicates, and each replicate is the pool of 7-10 fish. The dots show the original value, while the solid lines and the bars
Fig 2. Characterization of nr1d1:VNP expressing cells. (a) t-SNE visualization of brain cell clusters. The clusters were annotated by comparing to the adult zebrafish scRNA-seq data. (b) Violin plot demonstrated the clusters with enriched expression of nr1d1:VNP. Y axis denotes the normalized expression value. Note the highest levels in the pineal gland. (c) Fluorescence images show the co-expression of
Single-cell in vivo imaging of cellular circadian oscillators in zebrafish

March 2020

·

206 Reads

·

18 Citations

PLOS Biology

PLOS Biology

The circadian clock is a cell-autonomous time-keeping mechanism established gradually during embryonic development. Here, we generated a transgenic zebrafish line carrying a destabilized fluorescent protein driven by the promoter of a core clock gene, nr1d1, to report in vivo circadian rhythm at the single-cell level. By time-lapse imaging of this fish line and 3D reconstruction, we observed the sequential initiation of the reporter expression starting at photoreceptors in the pineal gland, then spreading to the cells in other brain regions at the single-cell level. Even within the pineal gland, we found heterogeneous onset of nr1d1 expression, in which each cell undergoes circadian oscillation superimposed over a cell type–specific developmental trajectory. Furthermore, we found that single-cell expression of nr1d1 showed synchronous circadian oscillation under a light–dark (LD) cycle. Remarkably, single-cell oscillations were dramatically dampened rather than desynchronized in animals raised under constant darkness, while the developmental trend still persists. It suggests that light exposure in early zebrafish embryos has significant effect on cellular circadian oscillations.


TIP60/KAT5 is required for neuronal viability in hippocampal CA1

November 2019

·

195 Reads

·

16 Citations

Scientific Reports

Aberrant histone acetylation contributes to age-dependent cognitive decline and neurodegenerative diseases. We analyze the function of lysine acetyltransferase TIP60/KAT5 in neurons of the hippocampus using an inducible mouse model. TIP60-deficiency in the adult forebrain leads within days to extensive transcriptional dysfunction characterized by the presence of a neurodegeneration-related signature in CA1. Cell cycle- and immunity-related genes are upregulated while learning- and neuronal plasticity-related genes are downregulated. The dysregulated genes seen under TIP60-deficiency overlap with those in the well-characterized CK-p25 neurodegeneration model. We found that H4K12 is hypoacetylated at the transcriptional start sites of those genes whose expression is dampened in TIP60-deficient mice. Transcriptional dysregulation is followed over a period of weeks by activation of Caspase 3 and fragmentation of β-actin in CA1 neurites, eventually leading to severe neuronal loss. TIP60-deficient mice also develop mild memory impairment. These phenotypes point to a central role of TIP60 in transcriptional networks that are critical for neuronal viability.


Fig. 4. Circadian dynamics of pineal gland at higher temporal resolution. (a) Experimental design to examine the developmental dynamics of nr1d1:VNP expression at higher temporal resolution. (b) Fluorescence images illustrate the tracing of one nr1d1:VNP example cell every 2 hours. (c) Single-cell tracing results of all 117 nr1d1:VNP-positive cells in two zebrafish pineal glands (2 fish). The empty dots represent the original fluorescence signals, while the solid line represents the smoothed curve fitted by the cosine functions. The example cell in (b) is highlighted by a green square. (d) Circadian phase distributions of the 117 nr1d1:VNP-positive cells. (e) t-SNE visualization of the clustering result of the 117 nr1d1:VNP-positive cells. (f) Raw fluorescence intensity traces of the two types of nr1d1:VNP-positive cells in (e). (g & h) The Comparison of the baseline expression (g) and relative circadian amplitude (h) between the two types of cells. The colors of the boxes corresponds to (e). Two-tailed Student's t-test was applied to calculate the levels of significance between the two types of cells. *** represents P<0.001. (i & j) Scatter plot demonstrated the relationship
Fig. 5. Light-Dark cycle is essential to synchronous onset of nr1d1 oscillation. (a) Experimental design to investigate the effect of light on the onset of circadian clock development. (b) Expression patterns of all cells under DD condition during development (four fish). Each thin line represents one cell. Each thick line represents the loess-smoothed curve of all cells in each individual fish. The shaded area shows the 95% confidence level of the smoothed curve. (c) Comparison of oscillating coefficient (B) between LD and DD cells. Two-tailed Student's t-test was applied to calculate the levels of significance between the two types of cells. *** represents P<0.001. (d) Comparison of developmental coefficient (A) between LD and DD cells. Two-tailed Student's t-test was applied to calculate the levels of significance between the two types of cells. *** represents P<0.001. (e) Experimental design to examine the single-cell circadian clocks under DD after transferred from LD (LD_DD). (f) Expression patterns of LD_LD (three fish) and LD_DD cells (three fish) from 6.0 to 7.5dpf. Each thin line represents one cell and each dot represents raw fluorescence intensity. Thick lines represent the loess-smoothed curves for all the LD_DD cells in red and LD_LD cells in blue respectively. The shaded areas show the 95% confidence level of the smooth curve. (g) Log2-transformed dusk-dawn ratios of VNP fluorescence intensities of all the cells in (f). Each data point represents one cell and the colors correspond to different fish. The red dash line represents y=0. n.s. represents no significant difference in duskdawn ratios between different time points (two-tailed ANOVA test). (h) Experimental design to examine the expression pattern of DD cells at higher temporal resolution. (i) Expression patterns of LD (two fish) and DD cells (three fish) across one day at 2-h resolution. Each thin line represents one cell and each dot represents normalized fluorescence intensity by dividing minimal value of each cell. Thick lines represent the loess-smoothed curves for all the LD in blue and DD cells in red respectively. The
Single-cell in vivo imaging reveals light-initiated circadian oscillators in zebrafish

July 2019

·

62 Reads

Circadian clock is a cell-autonomous time-keeping mechanism established gradually during embryonic development. Here we generated a transgenic zebrafish line carrying a destabilized fluorescent protein driven by the promoter of a core clock gene, nr1d1, to report in vivo circadian rhythm at the single-cell level. By time-lapse imaging of this fish line, we observed the sequential initiation of the reporter expression starting at photoreceptors in pineal gland then spreading to cells in other brain regions. Even within pineal gland, we found heterogeneous onset of nr1d1 expression in which each cell undergoes circadian oscillation superimposed over cell-type specific developmental trajectory. Furthermore, we found that single-cell expression of nr1d1 showed synchronous circadian oscillation under light-dark cycle. Remarkably, single-cell oscillations were lost in animals raised under constant darkness while developmental trend still persists. It suggests that light exposure in early clock development initializes cellular clocks rather than synchronizes existing individual oscillators as previously believed.



Fig. 1 Fus is a circadian regulated gene. a-c. Western blot showing the protein expression of FUS in the liver (a), hypothalamus (b) and cortex (c) of free-running wild-type mice (CT: circadian time), the quantification was shown on the left (mean ± s.e.m.; N = 5 mice were sacrificed at each time point). d mRNA expression level of Fus in the whole brain of sleep-deprived mice using microarray datasets [31] (mean ± s.e.m.; N = 3 experiments, t-test, *:P≤0.05). (e, f) mRNA expression level of Fus and Tdp-43 in the liver [37] (e) and brain stem [38] (f) of mouse. The lowest value for the dataset in each graph was set as 1 to determine relative fold change
Fig. 2 REV-ERBα activates the circadian expression of FUS. a ChIP-seq analysis showing REV-ERBα binding signals on the Fus promoter. The black bar below indicates the Fus promoter region (WT-P in Fig. 2c) used in the Fus promoter-luciferase construct, while the grey area in the middle of black bar indicates the region harboring the REV-ERBα-binding site based on ChIP-seq data [32]. b ChIP-qPCR showing the binding of FLAG-REV-ERBα to the Fus promoter in Neuro-2a cells (Fus-1 and Fus-2 are two pairs of primers specific for regions located in the predicted REV-ERBα binding sites in Fig. 2a; FLAG-GFP was used as the control; mean ± s.e.m.; N = 4 experiments; t-test; *:P≤0.05). c Luciferase activity of the intact (WT-P) and REV-ERBα-binding site deleted (Del-P) Fus promoter-luciferase constructs in Neuro-2a cells after siRNA silencing (mean ± s.e.m.; N = 4 experiments; t-test; ***:P≤0.001). The right panel showed the knock-down efficiency of Nr1d1-targeting siRNA (mean ± s.e.m.; N = 4 experiments; t-test; ***:P≤0.001; Ctrl represents scrambled control siRNA; NS: non-significant). d FUS expression level in synchronized wild-type or Nr1d1 knock-out (KO) MEFs. Quantification result was shown in the bar graph (mean ± s.e.m.; three lines of wild-type MEFs and four lines of Nr1d1 KO MEFs were generated from two pregnant Nr1d1 heterozygous mice, t-test; **: P≤0.01). e FUS expression level in the liver of free-running wild-type and Nr1d1 knock-out mouse at indicated time point, quantification result was shown in the right bar graph (N = 3 pairs of littermates for CT-0 hr and N = 2 pairs for CT-12 hr; mean ± s.e.m.; two-way ANOVA with Sidak's multiple comparison test, *:P≤0.05, **:P≤0.01). f Activating/repressive functional prediction analysis based on the published REV-ERBα ChIP-seq data [32] and transcriptional profile in Nr1d1 knock-out mice [53]. Genes are cumulated by the rank on the basis of the regulatory potential score from high to low according REV-ERBα ChIP-seq data (x-axis). The red and purple lines represent the percentage of up-regulated (UP) or down-regulated (DOWN) genes that harbor REV-ERBα binding sites from Nr1d1 knock-out microarray data, respectively. The black dashed line indicates the non-differentially (NON) expressed genes among REV-ERBα-binding genes. P values that represent the significance of the UP or DOWN group distributions are compared with the NON group by the Kolmogorov-Smirnov test. The right panel is an example showing the fraction of up-regulated (red) or down-regulated genes (purple) that contain REV-ERBα binding sites when the top 2,000 peaks from the ChIP-seq data were included (gray dash line in the left panel). The cumulative fractions of genes that are down-regulated in Nr1d1 knock-out mice indicate that REV-ERBα could also act as an activator
Fig. 3 FUS regulates the expression of core circadian genes. a, b mRNA expression levels of Per2 (a) and Cry1 (b) in synchronized Neuro-2a cells after siRNA silencing (mean ± s.e.m.; N = 4-6 experiments; two-way ANOVA with Sidak's multiple comparison test, * represents the significant P-value of overall two-way ANOVA analysis, ***:P≤0.001, # represents the significant P-value of Sidak's multiple comparison test, #:P≤0.05, ##:P≤0.01, ###:P≤0.001). c Western blot showing the knock-down efficiency of Fus-targeting siRNA (mean ± s.e.m.; N = 3-4 experiments; t-test; ****: P≤0.0001). d, e RT-qPCR showing the mRNA expression of Per2 and Cry1 in cells with indicated transfection conditions in serum shock-synchronized Neuro-2a cells (mean ± s.e.m.; N = 8-9 experiments; One-way ANOVA with Newman-Keuls multiple comparisons test; *:P≤0.05; **:P≤0.01)
Neurodegeneration-associated FUS is a novel regulator of circadian gene expression

December 2018

·

269 Reads

·

13 Citations

Translational Neurodegeneration

Background Circadian rhythms are oscillating physiological and behavioral changes governed by an internal molecular clock, and dysfunctions in circadian rhythms have been associated with ageing and various neurodegenerative diseases. However, the evidence directly connecting the neurodegeneration-associated proteins to circadian control at the molecular level remains sparse. Methods Using meta-analysis, synchronized animals and cell lines, cells and tissues from FUS R521C knock-in rats, we examined the role of FUS in circadian gene expression regulation. Results We found that FUS, an oscillating expressed nuclear protein implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), exerted a novel feedback route to regulate circadian gene expression. Nr1d1-encoded core circadian protein REV-ERBα bound the Fus promoter and regulated the expression of Fus. Meanwhile, FUS was in the same complex as PER/CRY, and repressed the expression of E box-containing core circadian genes, such as Per2, by mediating the promoter occupancy of PSF-HDAC1. Remarkably, a common pathogenic mutant FUS (R521C) showed increased binding to PSF, and caused decreased expression of Per2. Conclusions Therefore, we have demonstrated FUS as a modulator of circadian gene expression, and provided novel mechanistic insights into the mutual influence between circadian control and neurodegeneration-associated proteins. Electronic supplementary material The online version of this article (10.1186/s40035-018-0131-y) contains supplementary material, which is available to authorized users.





Citations (6)


... Thymus (10) (4) (7), (12) bens, in the olfactory system with areas such as the olfactory bulb, olfactory tubercle, nucleus of the lateral olfactory tract and cortical regions related to olfaction (entorhinal and piriform cortex) (3,(5)(6)(7)15,22). A recent article by our team showed that APLNR is expressed in the subventricular region located in the caudate nucleus, an area related to neurogenesis (22). ...

Reference:

Distribution and Expression of Apelin/APLNR System in the Mammalian Body
Transcriptome Response and Spatial Pattern of Gene Expression in the Primate Subventricular Zone Neurogenic Niche After Cerebral Ischemia
Frontiers in Cell and Developmental Biology

Frontiers in Cell and Developmental Biology

... 28,29 In support of this idea, studies in zebrafish showed that cyclic expression of a reporter gene driven by the nr1d1 promoter starts in the pineal photoreceptors and then spreads to other brain regions. 30 In addition, per2 expression has been shown to be higher in the pineal gland than in other regions, 31 as are many accessory clock genes. 32 Pineal-less mutants also show reduced amplitude in behavioural rhythms, 33 a similar effect to that found after blocking its molecular oscillator (Ben-Moshe Livne et al. 19 see below). ...

Single-cell in vivo imaging of cellular circadian oscillators in zebrafish
PLOS Biology

PLOS Biology

... At present, Tip60 participates in many cellular processes such as transcriptional regulation, cell cycle control, apoptosis, autophagy, and DNA repair [23][24][25][26][27]. Based on previous reports, Tip60 expression is high in the hippocampal CA1 region in adult mice [28]. Tamoxifen-induced conditional Tip60 deficiency in postmitotic excitatory neurons of the adult forebrain reduced H4K12 acetylation and the dysregulation of gene expression in the hippocampal CA1 region [29]. Inflammation and neurodegeneration in the CA1 region gradually progressed through Tip60 deficiency, and Tip60-deficient mice showed modest memory impairment at a later time point [29]. ...

TIP60/KAT5 is required for neuronal viability in hippocampal CA1

Scientific Reports

... Physiological FUS is a transcription factor [523] which has been identified to regulate circadian gene expression via a novel feedback effect [601]. FUS mutations interfere with RNA metabolic pathways and suppress protein translation [602]. ...

Neurodegeneration-associated FUS is a novel regulator of circadian gene expression

Translational Neurodegeneration

... With the discovery of new FUS mutations, researchers continue to establish several novel transgenic animal models to gain a more comprehensive understanding of how FUS mutations contribute to ALS. These models not only involve the overexpression of mutant forms but may also incorporate precise gene editing technologies such as CRISPR/Cas9 (Zhang et al., 2018). Researchers are dedicated to delving into the interactions between FUS and other proteins such as TDP-43 in the pathogenesis of ALS (Loughlin and Wilce, 2019). ...

Sleep and circadian abnormalities precede cognitive deficits in R521C FUS knockin rats
  • Citing Article
  • September 2018

Neurobiology of Aging

... PSMD5 gene has also been found to be associated with residual feed intake in pigs (Liu et al., 2016). PHF19 gene has been found to be associated with nervous system development (Liu et al., 2014). TRAF1 gene has been found to be associated with immune response in swine (Galindo and Alonso, 2017). ...

Reconstruction of the Gene Regulatory Network Involved in the Sonic Hedgehog Pathway with a Potential Role in Early Development of the Mouse Brain