Skip to main content
  • Short report
  • Open access
  • Published:

Deoxyhypusine synthase deficiency syndrome zebrafish model: aberrant morphology, epileptiform activity, and reduced arborization of inhibitory interneurons

Abstract

DHPS deficiency syndrome is an ultra-rare neurodevelopmental disorder (NDD) which results from biallelic mutations in the gene encoding the enzyme deoxyhypusine synthase (DHPS). DHPS is essential to synthesize hypusine, a rare amino acid formed by post-translational modification of a conserved lysine in eukaryotic initiation factor 5 A (eIF5A). DHPS deficiency syndrome causes epilepsy, cognitive and motor impairments, and mild facial dysmorphology. In mice, a brain-specific genetic deletion of Dhps at birth impairs eIF5AHYP-dependent mRNA translation. This alters expression of proteins required for neuronal development and function, and phenotypically models features of human DHPS deficiency. We studied the role of DHPS in early brain development using a zebrafish loss-of-function model generated by knockdown of dhps expression with an antisense morpholino oligomer (MO) targeting the exon 2/intron 2 (E2I2) splice site of the dhps pre-mRNA. dhps knockdown embryos exhibited dose-dependent developmental delay and dysmorphology, including microcephaly, axis truncation, and body curvature. In dhps knockdown larvae, electrophysiological analysis showed increased epileptiform activity, and confocal microscopy analysis revealed reduced arborisation of GABAergic neurons. Our findings confirm that hypusination of eIF5A by DHPS is needed for early brain development, and zebrafish with an antisense knockdown of dhps model features of DHPS deficiency syndrome.

Introduction

Post-translational modification is a core cellular strategy to rapidly alter protein activity in response to environmental stimuli. Over 200 post-translational modifications are mediated by enzymes which facilitate diverse reactions including phosphorylation, ubiquitination, glycosylation, palmitoylation, sulfation, methylation, small ubiquitin-like modifier (SUMO)-ylation, and nitrosylation, among others. A unique post-translational modification is hypusination, limited to eukaryotic translation factor 5 A (eIF5A) proteins [1]. Hypusination enables eIF5A activation via enzymatic conversion of a conserved lysine to the novel amino acid hypusine (Nϵ-4-amino-2-hydroxybutyl(lysine)). This reaction occurs in two steps: (1) lysine residue modification by deoxyhypusine synthase (DHPS), using the polyamine spermidine as a cofactor, to form the intermediate deoxyhypusine; (2) hydroxylation of this residue by deoxyhypusine hydroxylase (DOHH) in an oxygen-dependent reaction [2] to form hypusine.

Hypusinated eIF5A (eIF5AHYP) has key cellular functions: (1) facilitating initiation, elongation or termination during the translation of cell type-specific transcripts [1, 3,4,5,6]; (2) suppressing ribosomal stalling by stabilizing tRNA-ribosomal P-site interaction, facilitating peptide bond formation for consecutive polyprolines and other tripeptide motifs [7, 8]; and (3) nonsense-mediated decay (NMD) of mRNA transcripts with premature stop codons [9]. eIF5AHYP is critical to translate long polypeptides [10].

eIF5A, as well as the DHPS and DOHH enzymes required for activation by hypusination, are essential for eukaryotic cell viability and growth. In yeast, eIF5AHYP controls cell proliferation and is required for polarized cell growth during mating by regulating the translation of polyproline-rich formins [11]. In plants, eIF5A activation by hypusination is essential for growth during development and for environmental stress responses [12].

The DHPS-DOHH-eIF5A pathway relies on an oxygen-sensing mechanism. In yeast, DOHH deficiency impairs hydroxylation of the deoxyhypusine residue in eIF5A, decreasing N-terminal translation of proteins in mitochondrial respiration, oxidative stress response, and protein folding. eIF5A hypusination adapts cellular metabolism to oxygen levels [2].

In mammals, the DHPS-DOHH-eIF5A pathway is implicated in disease pathogenesis and in aging. In the Drosophila brain, hypusinated eIF5A levels decline with age, but can be increased by dietary spermidine, and genetic attenuation of eIF5AHYP levels induces premature aging (e.g., reduced mitochondrial respiration) [13]. Spermidine supplementation in mice boosts eIF5A hypusination and improves cognitive function [14].

Mice with T-cell-specific deletions of Dohh and Dhps develop severe intestinal inflammatory disease, supporting the role of hypusination in T cell activation and differentiation, long associated with polyamine synthesis [15]. Conversely, mice with a myeloid-specific deletion of Dhps revealed that eIF5AHYP promotes a pro-inflammatory macrophage M1-like phenotype [16].

Cancer-related signaling pathways regulated by eIF5AHYP include MYC, p53, and hypoxia-inducible factor 1-alpha (HIF1A). Overexpression of eIF5A is linked to colorectal, gastric, esophageal, lung, breast, ovarian, cervical, bladder, prostate, and hepatocellular cancers. Therefore, small-molecule inhibitors of hypusination that target DHPS or DOHH are potential anti-neoplastics [17].

In humans, mutations in the DHPS-DOHH-eIF5A pathway cause neurodevelopmental disorders. A rare, autosomal dominant disorder caused by heterozygous pathogenic EIF5A variants results in developmental delay, intellectual disability, microcephaly, and facial dysmorphism [18]. Rare, autosomal recessive biallelic pathogenic missense and truncating DOHH variants cause developmental delay, intellectual disability, microcephaly, facial dysmorphism, and epilepsy [19]. DHPS deficiency causes an ultra-rare, autosomal recessive disorder caused by biallelic pathogenic variants that reduce DHPS enzyme activity (~ 18–25% of normal), with features similar to the eIF5A and DOHH deficiency syndromes [20].

Homozygous knockout mouse models of DHPS, DOHH and eIF5A are early embryonic lethal, underscoring the essential role of this pathway in early development [21,22,23]. In mice, conditional knockout mouse models of these genes support their developmental roles. Mice with conditional genetic deletions of Dhps or Eif5a induced by the Emx1-Cre driver (primarily expressed in the cortex and hippocampus from E9.5 onwards) show gross defects in forebrain development, reduced growth, and premature death [24]. Mice with a brain-specific deletion of Dhps initiated at birth (via intraventricular injection of an adeno-associated virus with CMV-driven Cre expression) exhibited spontaneous seizures, impaired growth, and death before 6 weeks of age. Moreover, proteomic analysis of brain tissue using quantitative mass spectrometry revealed that these brain-specific Dhps knockout mice had changes in numerous proteins involved in neuronal growth, function, and secretion [3].

In patients and animal models, the DHPS-DOHH-eIF5A pathway disorders primarily impact the brain. While conditional mouse knockouts provide valuable data on the role of eIF5AHYP in postnatal brain development and function, zebrafish models can probe the role of hypusination in early brain development. A zebrafish model for eIF5A deficiency revealed mild microcephaly and micrognathia [18, 25]. A zebrafish model for DHPS deficiency focused on pancreas development [26]. Here, we characterize a zebrafish DHPS deficiency model, assessing the impact of reduced dhps expression on early brain development and activity.

Results

Generation of a zebrafish model for DHPS deficiency

Antisense morpholino oligomers (MOs) were designed targeting the AUG start codon of the zebrafish dhps mRNA (dhps AUG MO), and the E2I2 splice site of the zebrafish dhps pre-mRNA (dhps E2I2 MO) (Fig. 1A). Knockdown efficacy of the dhps E2I2 MO was analyzed by reverse transcriptase PCR (RT-PCR), which confirmed aberrant splicing (loss of exon 2) after microinjection of the dhps E2I2 MO, resulting in the appearance of a 172-bp amplicon and reduction of the 375-bp amplicon versus the control morpholino (Ctrl MO) and uninjected wild-type (Wt) larvae (Fig. 1B). Western blot analysis revealed a reduction in dhps protein levels in knockdown embryos generated using the dhps AUG MO and the dhps E212 MO. dhps knockdown embryos also had strongly reduced levels of both eif5aHYP and eif5aTOTAL, with a striking reduction of larger polypeptides (Fig. 1C and D).

Fig. 1
figure 1

Generation of the DHPS deficiency zebrafish model. (A) The first three exons and introns of the dhps gene and selected targeting sites of two antisense morpholino oligomers and PCR primers are shown, as well as the predicted amplicon lengths. (B) RT-PCR analysis confirmed the predicted lengths of the dhps mRNA amplicons from Wt, Ctrl MO and dhps E2I2 MO larvae with two biological repeats. While the length of the PCR amplicon covering the first three exons of dhps mRNA is 375 bp (b) in Wt, Ctrl MO, and dhps E2I2 MO larvae, in the latter two, additional bands appear (a and c). Appearance of the 172-bp amplicon (c), indicating deletion of exon 2 from the dhps mRNA, and reduction in amount of the 375-bp amplicon (b), together confirm partial knockdown of dhps. Appearance of an approximately 450-bp amplicon (a) in dhps E2I2 MO larvae may be due to partial activation of a cryptic splice site induced by antisense blockage of the E2I2 splice site. (C) Analysis of protein expression in dhps knockdown zebrafish. Zebrafish embryos (2 dpf) with knockdown by dhps E2I2 MO or dhps AUG MO were analyzed by Western blot for expression of DHPS, eIF5ATOTAL, eIF5AHYP, and total protein (as visualized by REVERT™). (D) Densitometric data for the relative expression of DHPS, eIF5ATOTAL, and eIF5AHYP, normalized on the basis of total protein as quantified by REVERT™ staining

Phenotypic analysis of dhps knockdown zebrafish

Phenotypic analysis of zebrafish larvae at 5 days post-fertilization (dpf), microinjected at one-cell stage with exon 2/intron 2 (E2I2) dhps E2I2 MO (7.92 ng) or with dhps AUG MO (11.55 ng), revealed that both antisense MOs caused similar dysmorphologies. dhps knockdown larvae were microcephalic with axis truncation, hyperpigmentation, cardiac edema, uninflated swim bladders, and body curvature, developmental delay and higher death rate at early stages compared to uninjected Wt and Ctrl MO (Fig. 2A). In control groups (Wt, n = 168 and Ctrl MO, n = 148), 96% of larvae exhibited normal development. In the dhps E2I2 MO group (n = 180), 13% of larvae exhibited significant dysmorphology, 38% moderate dysmorphology, and 16% mild dysmorphology. In the dhps AUG MO group (n = 126), 4% of larvae exhibited severe dysmorphology, 7% moderate dysmorphology, and 13% mild dysmorphology (Fig. 2B).

Fig. 2
figure 2

Phenotypic analysis of dhps knockdown zebrafish at five days post-fertilization (5 dpf). (A) Lateral view photographs of representative Wt, Ctrl MO, dhps E2I2 MO, and dhps AUG MO larvae, scale bar: 0.5 mm. (B) Chi-square analysis revealed significant differences between knockdown and control groups with regard to the percentages of larvae exhibiting normal development, mild and moderate dysmorphology (p<0.0001)

Partial rescue of dysmorphology in dhps knockdown larvae by expression of dhps mRNA

To control for antisense MO specificity, in vitro transcribed wild-type zebrafish dhps mRNA was co-injected with the dhps E2I2 MO to rescue the knockdown phenotype. Co-injection of in vitro transcribed dhps mRNA with dhps E2I2 MO reduced the number of larvae with dysmorphology and developmental delay, increasing in the percentage of normally developed larvae. In the dhps E2I2 MO group (n = 371), only 34% of embryos exhibited normal development, 41% mild dysmorphology, and 17% moderate dysmorphology, while in the group co-injected with dhps E2I2 MO and dhps mRNA (n = 179), 49% of embryos exhibited normal development, 31% mild dysmorphology, and 16% moderate dysmorphology. In control groups (Wt, n = 163 and dhps mRNA, n = 111) more than 94% of embryos exhibited normal development. These results demonstrate a partial rescue of the effects of dhps antisense MO (Fig. 3B, and C).

Fig. 3
figure 3

Partial rescue of dhps knockdown larvae by expression of dhps mRNA at 1 day post-fertilization (1 dpf). (A) Map of pIVT construct with zebrafish dhps cDNA. (B) mRNA overexpression partially shifted the more severe phenotypic categories of dhps knockdown to less severe categories. Chi-square analysis showed significant differences between control, knockdown and rescue groups with regard to the percentages of embryos exhibiting normal development, mild and moderate dysmorphology (P<0.0001). (C) Definition of observed phenotypic categories and sample size of each group

Electrophysiological analysis of dhps knockdown zebrafish larvae

Local field potential (LFP) recordings from the optic tectum revealed epileptiform events in 4-dpf dhps knockdown larvae. LFP recording of brain activity of 4-dpf dhps knockdown larvae revealed spontaneous electrographic discharges with high amplitude ( three-fold baseline) versus controls (Fig. 4A). Quantification of ictal-like events revealed that uninjected Wt larvae (n = 20) had a mean of 0.25 ± 0.14(SEM), Ctrl MO larvae (n = 22) had mean of 0.68 ± 0.24(SEM) and dhps E2I2 MO larvae (n = 23) had a mean of 3.74 ± 1.27(SEM). A significant increase was observed in the dhps knockdown group using the one-way ANOVA test (p ≤ 0.02) (Fig. 4B).

Fig. 4
figure 4

LFP recording of dhps knockdown larvae. (A) Snapshot of LFP signal recorded from 4-dpf Wt, Ctrl MO and dhps E2I2 MO larvae. (B) One-way ANOVA revealed that the frequency of ictal-like events in dhps knockdown larvae was significantly increased compared to control larvae (p ≤ 0.02). Each recording was performed over a period of 20 min. (C) Each pie chart shows percentage of larvae with more than 3 ictal-like events during 20 min of recording. While in Ctrl MO larvae less than 10% of larvae had more than 3 ictal-like events, in dhps E2I2 MO-injected larvae, more than 43% of larvae had more than 3 ictal-like events

Reduced GABAergic neuronal arborization in dhps knockdown larvae

Confocal microscopy analysis of dhps knockdown larvae derived from a transgenic reporter line with GABAergic-specific expression of mCherry assessed the effects of reduced dhps function on GABAergic neuron development. In zebrafish larvae, GABAergic neurons project arbors from the optic tectum to the tectal neuropil (Fig. 5A). Confocal microscopy revealed significantly reduced arborization of GABAergic neurons in dhps knockdown larvae (Fig. 5B). Quantification of the GABAergic neuronal arborization by Sholl analysis revealed a decreased dendritic arborization (p<0.0001, Table 1) in larvae with reduced dhps expression (Fig. 5C).

Fig. 5
figure 5

GABAergic neuronal dendritic arborization. (A) Larval head with location of optic tectum (blue) and neuropil (light yellow) where that GABAergic neurons project their arbors. (B) Confocal microscopy revealed that dhps knockdown larvae have less complex dendritic arborization in the GABAergic neurons. (C) GABAergic neuronal arbors were quantified in zebrafish larvae at three different ages (3, 5, and 7 dpf). At all three developmental ages, via one way ANOVA there was a significant reduction in the number of neuronal arbors in the dhps knockdown larvae (dhps E2I2 MO) versus both wild-type control larvae (Wt) and control MO-injected larvae (Crtl MO) (see also Table 1 below)

Table 1 Quantification of the number of neuronal arbors in wild-type control larvae (wt), control MO-injected larvae (Cntl MO), and dhps knockdown larvae (dhps E2I2 MO) at 3, 5 and 7 dpf

Discussion

Our zebrafish loss-of-function model of DHPS deficiency by antisense knockdown of dhps expression mimicked features of the human DHPS deficiency syndrome. The zebrafish showed developmental delayed with epileptiform discharges, as well as microcephaly, axis truncation, and body curvature. Electrophysiological analysis of dhps knockdown larvae showed increased epileptiform activity, while confocal microscopy analysis revealed significantly reduced arborisation and complexity in GABAergic neurons.

The epileptiform discharges in dhps knockdown larvae paralleled the epileptiform activity on electroencephalography (EEG) and seizures in DHPS deficiency syndrome patients [20], and in mice with a brain-specific knockout of Dhps induced by postnatal intraventricular injection of a Cre-expressing AAV vector [3]. Seizures were not reported in patients with loss-of-function EIF5A mutations [18], nor in an eIF5A-deficient zebrafish model [18, 25]. Patients with DOHH mutations have seizures [19]. Our findings suggest that for the seizure phenotype, the brain is more sensitive to reduced eIF5A hypusination caused by mutations in DHPS or DOHH, while patients with neurodevelopmental disorders caused by autosomal dominant EIF5A mutations do not exhibit seizures as remaining eIF5AHYP levels may be sufficient to prevent epileptiform activity and seizures.

Confocal microscopy analysis of dhps knockdown zebrafish using a transgenic reporter line revealed significantly reduced GABAergic neuron aborization. Loss of inhibitory GABAergic inputs may contribute to neuronal hyperexcitability, but mouse model studies are needed to confirm these findings. Moreover, cell type analyses in knockdown zebrafish brains could determine if this phenotype results from overall neuronal loss or a selective loss of GABAergic neurons. Brain magnetic resonance imaging scans of DHPS deficient patients are normal [OD et al., unreported data], suggesting that the brain morphology phenotypes in our zebrafish and other mouse models [3, 24] are more pronounced than the human phenotype.

Since hypusination of eIF5A is a key regulator of autophagy [14, 27], and autophagy is critical during axonal and presynaptic development [28], impaired autophagy during zebrafish and mouse brain development in the DHPS deficiency syndrome models should be explored as a pathogenic mechanism. Our analysis of protein expression in dhps knockdown zebrafish reveal a striking reduction in expression of larger proteins. Ongoing studies using proteomic and transcriptomic analysis of embryonic brains with reduced hypusination beginning during early development may identify key molecular and cellular changes at different developmental stages and may elucidate the role of eIF5AHYP in the translation of certain proteins (e.g., long polypeptides [10] and neurosecretory factors [5]). We need to better understand the reductions in total eIF5A and eIF5AHYP levels seen in dhps knockdown zebrafish, which are similar to those in conditional cell-specific mouse models of DHPS loss [4, 5].

Overexpression of dhps mRNA in wild-type embryos did not cause a deleterious phenotype, suggesting that gene therapy strategies that involve overexpression of DHPS may show benefit for DHPS deficiency syndrome patients. Conversely, only partial rescue of the dhps knockdown phenotype was achieved by overexpression of dhps mRNA, which may be due to the limited half-life of dhps mRNA after microinjection at the single-cell stage.

Our overall findings support that hypusination of eIF5A is important for early brain development, and zebrafish with reduced dhps expression are a useful model for DHPS deficiency syndrome. Future experiments with this model will evaluate anti-seizure medications using the seizure phenotype, as well as other therapeutic modalities and endpoints. Generation of zebrafish models of neurodevelopmental disorders caused by mutations in the DHPS-DOHH-eIF5A pathway that incorporate specific patient mutations into the genetically engineered lines could dissect the phenotypic differences observed between these related diseases.

Methods

Zebrafish husbandry

Wild-type (Wt) adult zebrafish (Danio rerio; AB strain; CVE-KIT) were maintained at 28.5 °C on a 14-h/10-h light/dark cycle under standard aquaculture conditions, and fertilized eggs were collected via natural spawning. Embryos were raised in embryo medium (E3; 1.5 mmol/L HEPES, pH 7.6, 17.4 mmol/L NaCl, 0.21 mmol/L KCl, 0.12 mmol/L MgSO4, and 0.18 mmol/L Ca[NO3]2), under the same conditions as adults. For all zebrafish experiments conducted at NCMM, larvae up to 7 days post-fertilization (dpf) were used.

Antisense morpholino oligomers (MOs) and microinjections

11.55 ng of a translation-blocking MO (dhps AUG MO: 5’ GGTTATGGATGTAAATCCGGCTTTT) targeting the AUG start codon of the zebrafish dhps mRNA and 7.92 ng of splice site-blocking MO targeting the exon 2/intron 2 splice site of the zebrafish dhps pre-RNA (dhps E2I2 MO: 5′ CACGATCAGTCTGTCACTCACCATC) were used to achieve partial knockdown of zebrafish dhps. Fluoresceinated standard control MO was used as a negative control (Ctrl MO) (11.55 and 7.92 ng respectively). MOs were designed and synthesized by Gene Tools LLC (Philomath, Oregon, USA) and injected into 1-2-cell stage embryos [26].

Reverse transcriptase PCR (RT-PCR)

Efficiency of knockdown was determined by RT-PCR, using primers that amplify across the predicted deletion: 5′ GCGCTGTGAAATGTGAGTGAAAC and 5′ GTTTGACGTGTAGCCCAGGAAT. The PCR amplicon was 385 bp in the control embryos and 172 bp in the dhps MO-injected embryos, and was visualized by standard agarose gel electrophoresis [26].

Western blot analysis

Zebrafish embryos were evaluated by Western blot analysis, adapting methods previously described [3]. Specifically, 30–40 zebrafish embryos (2 dpf) were lysed in 400 µL of buffer containing 50 mM Tris, pH 8.0, 150 mM NaCl, 0.05% deoxycholate, 0.1% IGEPAL CA-630, 0.1% SDS, 0.2% sarcosyl, 10% glycerol, 1 mM DTT, 1 mM EDTA, 10mM NaF, protease inhibitors (#11836170001, Roche), phosphatase inhibitors (#4906845001, Roche), 2 mM MgCl2, and 0.05% v/v Benzonase (Millipore) and were intermittently vortexed to facilitate protein extraction. Protein was quantified using the DC Protein Assay Kit II (#5000112, Bio-Rad) followed by SDS-PAGE (4–20% gel). Separated protein (20 µg) was transferred to PVDF membranes and blocked in Odyssey Blocking Buffer (#927-40100, LI-COR Biosciences) at room temperature for 1 h. Membranes were incubated with REVERT (#926-11016; LI-COR Biosciences) to permit visualization of total protein. Subsequent incubation with primary antibodies diluted in Intercept Blocking Buffer (#927-70001; LI-COR Biosciences) was performed overnight at 4 °C. Membranes were washed twice with TBST buffer prior to incubation with near infrared, fluorescent dye-conjugated secondary antibodies at room temperature for 1 h. Following additional washes with TBST buffer, the membranes were imaged using an Odyssey CLx Imaging System and images were analyzed using the CLx Image Studio Version 5.2 Software (LI-COR Biosciences).

The following primary antibodies were used at the dilutions indicated: mouse anti-deoxyhypusine synthase (1:2000; Santa Cruz, #sc-365077), mouse anti-eIF5ATOTAL (1:2000; BD Biosciences, #611977), and rabbit anti-eIF5AHYP (1:5000; Millipore, #ABS1064-I). Densitometric data are graphed as relative expression.

mRNA rescue

To generate dhps RNA, zebrafish dhps cDNA was cloned into the pIVT expression construct [Addgene plasmid 122139; 29], which was linearized through restriction at the 3’ end of the ORF and used as a template to generate dhps mRNA using T7 RNA polymerase in an in vitro transcription reaction.

Local field potential (LFP) recording

Recordings were obtained from tecta at 4 dpf [30]. Seizure detection was performed through visual inspection and automated using a custom-written R script to minimize bias and artifacts due to muscle contractions. Recorded frequencies were divided 1-100, 100–250, and 250–500 Hz bands. If amplitude exceeded 3× background, the event was considered a seizure based on high-frequency oscillations (> 100 Hz) [31]. Power spectrum was analyzed using Clampfit 10.7 software (Molecular Devices). 20-minute-long recordings were used to compute the power spectrum from larvae at 4 dpf, and each condition was averaged per group [32].

Confocal microscopy and quantification of arbors

To visualize GABAergic neurons, wild-type and dhps E2I2 MO-microinjected embryos were treated from 1 dpf with 0.003% phenylthiourea to prevent pigmentation. Larvae were anesthetized in 0.001% tricaine (Sigma), fixed for 3 h at room temperature with 4% paraformaldehyde, mounted on glass slides, and imaged using confocal microscopy. A dorsal z-stack of the optic tectum was collected using a 40x lens and a z-resolution of 0.44 μm. For Sholl analysis [33, 34], images were filtered using the 3D-Median filter in ImageJ. A z-projection of the tectum was generated, and the resulting image was converted to a thresholded binary image. Arborization extent was quantified using Sholl analysis (plug-in; http://imagej.net/Sholl_Analysis). The number of intersections was normalized against the number of neurons in the imaged area; this value was statistically analyzed [32].

Data availability

The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.

Abbreviations

DHPS:

Deoxyhypusine synthase

DOHH:

Deoxyhypusine hydroxylase

E2I2:

Exon 2/intron 2

EEG:

Electroencephalography

eIF5A:

Eukaryotic initiation factor 5 A

eIF5AHYP :

Hypusinated eIF5A

HIF1A:

Hypoxia-inducible factor 1-alpha

LFP:

Local field potential

MO:

Antisense morpholino oligomer

NDD:

Neurodevelopmental disorder

NMD:

Nonsense-mediated decay

SUMO:

Small ubiquitin-like modifier

RT-PCR:

Reverse transcriptase PCR

SEM:

Standard error of the mean

Wt:

Wild-type

References

  1. Park MH, Wolff EC. Hypusine, a polyamine-derived amino acid critical for eukaryotic translation. J Biol Chem. 2018. https://doi.org/10.1074/jbc.TM118.003341.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Zhang Y, Su D, Zhu J, et al. Oxygen level regulates N-terminal translation elongation of selected proteins through deoxyhypusine hydroxylation. Cell Rep. 2022. https://doi.org/10.1016/j.celrep.2022.110855.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Padgett LR, Shinkle MR, Rosario S, et al. Deoxyhypusine synthase mutations alter the post-translational modification of eukaryotic initiation factor 5A resulting in impaired human and mouse neural homeostasis. HGG Adv. 2023. https://doi.org/10.1016/j.xhgg.2023.100206.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Padgett LR, Robertson MA, Anderson-Baucum EK, et al. Deoxyhypusine synthase, an essential enzyme for hypusine biosynthesis, is required for proper exocrine pancreas development. FASEB J. 2021. https://doi.org/10.1096/fj.201903177R.

    Article  PubMed  Google Scholar 

  5. Connors CT, Villaca CBP, Anderson-Baucum EK, et al. A translational regulatory mechanism mediated by hypusinated eukaryotic initiation factor 5A facilitates β-cell identity and function. Diabetes. 2024. https://doi.org/10.2337/db23-0148.

    Article  PubMed  Google Scholar 

  6. Schuller AP, Wu CC, Dever TE, Buskirk AR, Green R. eIF5A functions globally in translation elongation and termination. Mol Cell. 2017. https://doi.org/10.1016/j.molcel.2017.03.003.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Pelechano V, Alepuz P. eIF5A facilitates translation termination globally and promotes the elongation of many non polyproline-specific tripeptide sequences. Nucleic Acids Res. 2017. https://doi.org/10.1093/nar/gkx479.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gutierrez E, Shin BS, Woolstenhulme CJ, et al. eIF5A promotes translation of polyproline motifs. Mol Cell. 2013. https://doi.org/10.1016/j.molcel.2013.04.021.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Hoque M, Park JY, Chang YJ, et al. Regulation of gene expression by translation factor eIF5A: Hypusine-modified eIF5A enhances nonsense-mediated mRNA decay in human cells. Translation (Austin). 2017. https://doi.org/10.1080/21690731.2017.1366294.

    Article  PubMed  Google Scholar 

  10. Abe T, Nagai R, Shimazaki S, et al. In vitro yeast reconstituted translation system reveals function of eIF5A for synthesis of long polypeptide. J Biochem. 2020. https://doi.org/10.1093/jb/mvaa022.

    Article  PubMed  Google Scholar 

  11. Li T, Belda-Palazón B, Ferrando A, Alepuz P. Fertility and polarized cell growth depends on eIF5A for translation of polyproline-rich formins in Saccharomyces cerevisiae. Genetics. 2014. https://doi.org/10.1534/genetics.114.166926.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Pálfi P P, Bakacsy L, Kovács H, Szepesi Á Á. Hypusination, a metabolic posttranslational modification of eIF5A in plants during development and environmental stress responses. Plants. 2021. https://doi.org/10.3390/plants1007126.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Liang Y, Piao C, Beuschel CB, et al. eIF5A hypusination, boosted by dietary spermidine, protects from premature brain aging and mitochondrial dysfunction. Cell Rep. 2021. https://doi.org/10.1016/j.celrep.2021.108941.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hofer SJ, Liang Y, Zimmermann A, et al. Spermidine-induced hypusination preserves mitochondrial and cognitive function during aging. Autophagy. 2021. https://doi.org/10.1080/15548627.2021.1933299.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Puleston DJ, Baixauli F, Sanin DE, et al. Polyamine metabolism is a central determinant of helper T cell lineage fidelity. Cell. 2021. https://doi.org/10.1016/j.cell.2021.06.007.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Anderson-Baucum E, Piñeros AR, Kulkarni A, et al. Deoxyhypusine synthase promotes a pro-inflammatory macrophage phenotype. Cell Metabol. 2021. https://doi.org/10.1016/j.cmet.2021.08.003.

    Article  Google Scholar 

  17. Sfakianos AP, Raven RM, Willis AE. The pleiotropic roles of eIF5A in cellular life and its therapeutic potential in cancer. Biochem Soc Trans. 2022. https://doi.org/10.1042/BST20221035.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Faundes V, Jennings MD, Crilly S, et al. Impaired eIF5A function causes a mendelian disorder that is partially rescued in model systems by spermidine. Nat Commun. 2021. https://doi.org/10.1038/s41467-021-21053-2.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ziegler A, Steindl K, Hanner AS, et al. Bi-allelic variants in DOHH, catalyzing the last step of hypusine biosynthesis, are associated with a neurodevelopmental disorder. Am J Hum Genet. 2022. https://doi.org/10.1016/j.ajhg.2022.06.010.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Ganapathi M, Padgett LR, Yamada K, et al. Recessive rare variants in Deoxyhypusine synthase, an enzyme involved in the synthesis of Hypusine, are Associated with a neurodevelopmental disorder. Am J Hum Genet. 2019. https://doi.org/10.1016/j.ajhg.2018.12.017.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Templin AT, Maier B, Nishiki Y, Tersey SA, Mirmira RG. Deoxyhypusine synthase haploinsufficiency attenuates acute cytokine signaling. Cell Cycle. 2011. https://doi.org/10.4161/cc.10.7.15206.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Nishimura K, Lee SB, Park JH, Park MH. Essential role of eIF5A-1 and deoxyhypusine synthase in mouse embryonic development. Amino Acids. 2012. https://doi.org/10.1007/s00726-011-0986-z.

    Article  PubMed  Google Scholar 

  23. Sievert H, Pällmann N, Miller KK, et al. A novel mouse model for inhibition of DOHH-mediated hypusine modification reveals a crucial function in embryonic development, proliferation and oncogenic transformation. Dis Model Mech. 2014. https://doi.org/10.1242/dmm.014449.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kar RK, Hanner AS, Starost MF, et al. Neuron-specific ablation of eIF5A or deoxyhypusine synthase leads to impairments in growth, viability, neurodevelopment, and cognitive functions in mice. J Biol Chem. 2021. https://doi.org/10.1016/j.jbc.2021.101333.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Carvalho CM, Vasanth S, Shinawi M, et al. Dosage changes of a segment at 17p13.1 lead to intellectual disability and microcephaly as a result of complex genetic interaction of multiple genes. Am J Hum Genet. 2014. https://doi.org/10.1016/j.ajhg.2014.10.006.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Mastracci TL, Robertson MA, Mirmira RG, Anderson RM. Polyamine biosynthesis is critical for growth and differentiation of the pancreas. Sci Rep. 2015. https://doi.org/10.1038/srep13269.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Gobert AP, Finley JL, Latour YL, et al. Hypusination orchestrates the Antimicrobial response of macrophages. Cell Rep. 2020. https://doi.org/10.1016/j.celrep.2020.108510.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Crawley O, Grill B. Autophagy in axonal and presynaptic development. Curr Opin Neurobiol. 2021. https://doi.org/10.1016/j.conb.2021.03.011.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Igarashi H, Knott JG, Schultz RM, Williams CJ. Alterations of PLCbeta1 in mouse eggs change calcium oscillatory behavior following fertilization. Dev Biol. 2007. https://doi.org/10.1016/j.ydbio.2007.09.028.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Afrikanova T, Serruys AS, Buenafe OE, et al. Validation of the zebrafish pentylenetetrazol seizure model: locomotor versus electrographic responses to antiepileptic drugs. PLoS ONE. 2013. https://doi.org/10.1371/journal.pone.0054166.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zijlmans M, Jiruska P, Zelmann R, Leijten FS, Jefferys JG, Gotman J. High-frequency oscillations as a new biomarker in epilepsy. Ann Neurol. 2012. https://doi.org/10.1002/ana.22548.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Tiraboschi E, Martina S, van der Ent W, et al. New insights into the early mechanisms of epileptogenesis in a zebrafish model of Dravet syndrome. Epilepsia. 2020. https://doi.org/10.1111/epi.16456.

    Article  PubMed  Google Scholar 

  33. Ferreira TA, Blackman AV, Oyrer J, et al. Neuronal morphometry directly from bitmap images. Nat Methods. 2014. https://doi.org/10.1038/nmeth.3125.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ristanović D, Milosević NT, Stulić V. Application of modified Sholl analysis to neuronal dendritic arborization of the cat spinal cord. J Neurosci Methods. 2006. https://doi.org/10.1016/j.jneumeth.2006.05.030.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M.H. Park, A. Mingorance, C. Olson, T. Olson, and W. van der Ent for helpful discussions.

Funding

This work was supported in part by funding to TLM from the National Institutes of Health (NIH) (R01DK121987).

Author information

Authors and Affiliations

Authors

Contributions

ADC, CVE, OD and TLM designed and/or supervised the study. ES, TLM and RS generated and collected data, designed and made diagrams, and performed analyses and interpretation. ES and ADC drafted the manuscript, and OD, TLM and CVE revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Alexander D. Crawford.

Ethics declarations

Ethics approval and consent to participate

The experiments were approved by Mattilsynet (Norwegian Food Safety Authority, FOTS ID 23935, Modeling severe neurological disorders in vivo for high-throughput drug discovery, valid from 01.07.2020 to 01.07.2024).

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

Shojaeinia, E., Mastracci, T.L., Soliman, R. et al. Deoxyhypusine synthase deficiency syndrome zebrafish model: aberrant morphology, epileptiform activity, and reduced arborization of inhibitory interneurons. Mol Brain 17, 68 (2024). https://doi.org/10.1186/s13041-024-01139-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13041-024-01139-w

Keywords