Skip to main content

Genome-wide transcriptomic analysis of the forebrain of postnatal Slc13a4+/− mice

Abstract

Objective

Sulfation is an essential physiological process that regulates the function of a wide array of molecules involved in brain development. We have previously shown expression levels for the sulfate transporter Slc13a4 to be elevated during postnatal development, and that sulfate accumulation in the brains of Slc13a4+/− mice is reduced, suggesting a role for this transporter during this critical window of brain development. In order to understand the pathways regulated by cellular sulfation within the brain, we performed a bulk RNA-sequencing analysis of the forebrain of postnatal day 20 (P20) Slc13a4 heterozygous mice and wild-type litter mate controls.

Data description

We performed an RNA transcriptomic based sequencing screen on the whole forebrain from Slc13a4+/− and Slc13a4+/+mice at P20. Differential expression analysis revealed 90 differentially regulated genes in the forebrain of Slc13a4+/− mice (a p-value of 0.1 was considered as significant). Of these, 55 were upregulated, and 35 were downregulated in the forebrain of heterozygous mice. Moreover, when we stratified further with a ± 1.2 fold-change, we observed 38 upregulated, and 16 downregulated genes in the forebrain of heterozygous mice. This resource provides a useful tool to interrogate which pathways may require elevated sulfate levels to drive normal postnatal development of the brain.

Objective

Sulfate is an abundant anion in circulation, and its enzymatic conjugation (sulfation) to a variety of molecules is a biotransformation widely utilised to regulate biological activity [1]. Within the developing brain, sulfation reactions alter the functions of extracellular matrix components, in turn regulating local growth factor interactions critical for neurogenesis or perineuronal net formation [2, 3]. Sulfate is also a substrate for brain cerebrosides and neurotransmitters [1]. To provide sufficient sulfate for these critical sulfation reactions, cells either metabolize sulfur-containing amino acids to release intracellular sulfate, or uptake inorganic sulfate across the plasma membrane via transporters. The maintenance of a low ratio of cerebrospinal fluid (CSF)/serum sulfate level indicates the selective transport of sulfate to maintain brain levels within a tight range [4]. Yet despite its importance, sulfate levels are not typically measured clinically, and therefore the contribution of sulfate deficiencies to neurodevelopmental disorders and disease is poorly appreciated and understood.

Recently, we reported that haploinsufficiency for the sulfate transporter Slc13a4 resulted in abnormal social behaviours, memory deficits and altered neurogenesis in mice [5, 6]. SLC13A4 is expressed predominantly within the choroid plexus and pia mater of the brain, in an orientation that suggests a role in transporting sulfate from the blood into the CSF [5]. Expression of this transporter therefore appears counter to the notion that sulfate is actively pumped out of the CSF [1]. Nevertheless, Slc13a4+/− mice, when injected systemically through the tail vein, accumulate ~ 50% less radiolabelled sulfate within their brains than do Slc13a4+/+ mice [5]. Fitting with the model of active transport of sulfate out of the CSF, SLC13A4 activity is not essential for adult brain function, as conditional deletion of Slc13a4 in adult mice does not result in the onset of cellular or behavioural phenotypes. However, deletion of one Slc13a4 allele in early postnatal development does result in the onset of abnormal social behaviours, memory deficits and increased neurogenesis, suggesting elevated sulfate transport into the brain is critical during early developmental stages of brain formation [5]. Indeed, gene expression analysis indicates that expression of multiple sulfate transporters peak in postnatal mouse development [5], implying a critical role for sulfate availability and metabolism during this developmental window. In line with this, newborn humans have higher serum sulfate levels than children at 3-years of age or adults [7].

The question remains: what critical pathways require elevated sulfate availability during the postnatal period for normal brain development? To address this question, we have undertaken an unbiased RNA-Seq screen to compare the transcriptome of the forebrain of Slc13a4+/− mice to control Slc13a4−/− littermates at postnatal day 20.

Data description

The rationale behind this profiling experiment was to understand the differential gene expression that arise during postnatal development in the mouse forebrain when one allele of Slc13a4 gene is absent. To do this, Slc13a4+/− mice and control wild-type littermates were used. These mice were maintained on a C57BL6 background. To generate Slc13a4+/− mice, Slc13a4+/− male mice were crossed to wild-type female mice. Polymerase chain reaction (PCR) was used to identify the genotype of the offspring (PCR primers are available on request). P20 Slc13a4+/− and control mice were cervically dislocated and whole brains were removed and placed on ice. The forebrain was isolated and snap frozen using dry ice. An RNeasy Micro Kit (QIAGEN) was used to extract total RNA from these samples, and 5–10 μg RNA in a total volume of 20 μl was sent to the Institute for Molecular Biosciences Sequencing Facility (The University of Queensland). The sequencing facility assessed sample quality using a Bioanalyzer. All samples passed the quality control with an RNA integrity number > 8. A second analysis was performed to measure the purity of the RNA using a spectrophotometer to determine the OD 260/280 ratio; all samples had values ~ 2.

RNA-Seq libraries were prepared using the Illumina TruSeq Stranded Total RNA LT (Ribo-Zero Gold) Sample Prep Kit (Illumina, RS-122-2301/RS-122-2302), according to the standard manufacturer’s protocol (Illumina, 15031048 Rev. E October 2013) described briefly as follows. To enrich for mRNA, 1 µg of total RNA was depleted of rRNA using Ribo-Zero Gold. The enriched mRNA was then subjected to a heat fragmentation step aimed at producing fragments between 130 and 290 base pairs (average 185 base pairs). cDNA was synthesised from the fragmented RNA using SuperScript II Reverse Transcriptase (Invitrogen, 18064014) and random primers. The resulting cDNA was converted into dsDNA in the presence of dUTP to prevent subsequent amplification of the second strand and thus maintaining the ‘strandedness’ of the library. Following 3’ adenylation and adaptor ligation, libraries were subjected to 15 cycles of PCR to produce libraries ready for sequencing. The libraries were quantified on the Perkin Elmer LabChip GX with the DNA High Sensitivity Reagent kit (Perkin Elmer, CLS760672). Libraries were pooled in equimolar ratios, and the pool was quantified by qPCR using the KAPA Library Quantification Kit—Illumina/Universal (KAPA Biosystems, KK4824) in combination with the Life Technologies Viia 7 real time PCR instrument.

Bulk RNA-sequencing was performed using the Illumina NextSeq500 (NextSeq control software v2.1.0/Real Time Analysis v2.4.11). The library pool was diluted and denatured according to the standard NextSeq protocol (Document # 15048776 v02) and sequenced to generate paired-end 76 base pair reads using a 150 cycle NextSeq500/550 High Output reagent Kit v2 (Illumina, FC-404-2002). After sequencing, fastq files were generated using the bcl2fastq2 (v2.18.0, demultiplexed option used, available from Illumina) and received from the Institute for Molecular Bioscience Sequencing Facility (University of Queensland). Salmon (v1.2.0; validate mappings and gcBias options used) [8] was used for quantifying transcript abundance. The count data was loaded into R (v4.0.3) using tximeta (v1.8.3; default import option used) [9]. Differential gene expression analysis between Slc13a4+/− and wild-type samples was carried out in R using the DeSeq2 pipeline (v1.30.0; estimate size factors and walt test options used) [10]. Gene expression levels between Slc13a4+/− and wild-type samples were compared using Wald test as implemented in the DESeq2 pipeline. p-values were corrected using Benjamini–Hochberg adjustment. A statistically significant difference in gene expression between cohorts was represented by an adjusted p-value < 0.1. Furthermore, to stratify differentially expressed transcripts, a fold change cut-off of >  ± 1.2 was employed.

The data repositories where the work presented in this manuscript can be found are listed in Table 1. The raw sequencing files have also been lodged, and are available at GEO (Data set 1) [11]. Differential gene analysis revealed 90 differentially regulated genes in the forebrain of Slc13a4+/− mice (Data file 2) [12]. Of these, 55 were upregulated, and 35 were down downregulated in comparison to controls (Data file 3) [13].

Table 1 Overview of data files

Limitations

This work complements our previously published work [5, 6] adding to our understanding of the critical pathways required during the postnatal period for normal brain development. Moreover we have attempted to identify those pathways functioning in the presence of elevated sulfate levels during this critical window demonstrating an important role for this sulfate transporter SLC13A4 in regulating brain development. There are a number of limitations to this work, however. Firstly our analysis of the transcriptomic changes in the forebrain of Slc13a4+/− was only conducted at one postnatal age P20. Investigating consecutive ages during the early developmental window would provide further context around the genetic landscape that requires elevated sulfate levels for brain genesis. In turn, this could highlight common genes/pathways that may be essential drivers of normal brain development throughout this period. Secondly, although the expression of SLC13A4 is predominantly within the choroid plexus and pia mater of the forebrain at this age [5], our design was not based on a cell specific approach. Performing single cell RNA-sequencing in future could circumvent this limitation to pinpoint at a cell specific level the gene requirements for elevated sulfate levels to maintain normal brain development over the postnatal period.

Availability of data and materials

The raw sequencing data described in this Data note can be freely and openly accessed through GEO (Data file 1) [11]. Data file 2 and Data file 3 are available on Figshare (https://figshare.com) [12, 13]. Please see Table 1 for links to the data.

Abbreviations

SLC13A4:

Solute Carrier Family 13 (Sodium/Sulphate Symporters) Member 4

RNA-Seq:

RNA-Sequencing

P:

Postnatal day

PCR:

Polymerase chain reaction

References

  1. Cole DE, Evrovski J. The clinical chemistry of inorganic sulfate. Crit Rev Clin Lab Sci. 2000;37(4):299–344.

    Article  CAS  Google Scholar 

  2. Miyata S, Komatsu Y, Yoshimura Y, Taya C, Kitagawa H. Persistent cortical plasticity by upregulation of chondroitin 6-sulfation. Nat Neurosci. 2012;15(3):414–22.

    Article  CAS  Google Scholar 

  3. Yamaguchi Y. Heparan sulfate proteoglycans in the nervous system: their diverse roles in neurogenesis, axon guidance, and synaptogenesis. Semin Cell Dev Biol. 2001;12(2):99–106.

    Article  CAS  Google Scholar 

  4. Cole DE, Shafai J, Scriver CR. Inorganic sulfate in cerebrospinal fluid from infants and children. Clin Chim Acta. 1982;120(1):153–9.

    Article  CAS  Google Scholar 

  5. Zhang Z, Dawson PA, Piper M, Simmons DG. Postnatal N-acetylcysteine administration rescues impaired social behaviors and neurogenesis in Slc13a4 haploinsufficient mice. EBioMedicine. 2019;43:435–46.

    Article  Google Scholar 

  6. Zhang Z, Jhaveri D, Sharmin S, Harvey TJ, Dawson PA, Piper M, Simmons DG. Cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Biol Open. 2020;9(7):bio053132.

    Article  CAS  Google Scholar 

  7. Cole DE, Scriver CR. Age-dependent serum sulfate levels in children and adolescents. Clin Chim Acta. 1980;107(1–2):135–9.

    Article  CAS  Google Scholar 

  8. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017;14(4):417–9.

    Article  CAS  Google Scholar 

  9. Love MI, Soneson C, Hickey PF, Johnson LK, Pierce NT, Shepherd L, Morgan M, Patro R. Tximeta: Reference sequence checksums for provenance identification in RNA-seq. PLoS Comput Biol. 2020;16(2):e1007664.

    Article  CAS  Google Scholar 

  10. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.

    Article  Google Scholar 

  11. Harvey TJ, Ayala Davila R, Vidovic D, Sharmin S, Sharmin S, Piper M, Simmons DG. Expression data from P20 wild type and Slc13a4 heterozygous mouse forebrains. Gene Exp Omnibus 2021;GSE171765.

  12. Harvey TJ, Ayala Davila R, Vidovic D, Sharmin S, Sharmin S, Piper M, Simmons DG. Differentially regulated genes, P20 cerebral cortex, Slc13a4 heterozygous versus wild type mice, figshare. 2021. https://doi.org/10.6084/m9.figshare.13088978.

  13. Harvey TJ, Ayala Davila R, Vidovic D, Sharmin S, Sharmin S, Piper M, Simmons DG. Volcano plot of differentially regulated genes in Slc13a4 heterozygous mice, figshare. 2021. https://doi.org/10.6084/m9.figshare.14099321.

Download references

Acknowledgements

We would like to thank the staff at the UQ Biological Resources for their assistance with colony maintenance.

Funding

This work was funded by an Australian Research Council (ARC) Discovery Project to MP (DP180100017).

Author information

Authors and Affiliations

Authors

Contributions

MP, TJH, and DGS designed the experiments; TJH and SS collected the tissue and conducted the sequencing experiment; TJH, RAD, and DV interrogated the sequencing data. All authors contributed to the writing of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to David G. Simmons.

Ethics declarations

Ethics approval and consent to participate

All animal procedures were performed with approval from the University of Queensland Animal Ethics Committee (SBMS/147/17 and SBMS/034/17) and experiments carried out in accordance with the Australian Code of Practice for the Care and Use of Animals for Scientific Purposes.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Harvey, T.J., Davila, R.A., Vidovic, D. et al. Genome-wide transcriptomic analysis of the forebrain of postnatal Slc13a4+/− mice. BMC Res Notes 14, 269 (2021). https://doi.org/10.1186/s13104-021-05687-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13104-021-05687-5

Keywords