Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Feb 15.
Published in final edited form as: Dev Biol. 2019 Nov 14;458(2):189–199. doi: 10.1016/j.ydbio.2019.10.039

Nubp2 is required for cranial neural crest survival in the mouse.

Andrew DiStasio 1, David Paulding 1, Praneet Chaturvedi 2, Rolf W Stottmann 1,2,3,4,#
PMCID: PMC6995770  NIHMSID: NIHMS1545373  PMID: 31733190

Abstract

The N-ethyl-N-nitrosourea (ENU) forward genetic screen is a useful tool for the unbiased discovery of novel mechanisms regulating developmental processes. We recovered the dorothy mutation in such a screen designed to recover recessive mutations affecting craniofacial development in the mouse. Dorothy embryos die prenatally and exhibit many striking phenotypes commonly associated with ciliopathies, including a severe midfacial clefting phenotype. We used exome sequencing to discover a missense mutation in nucleotide binding protein 2 (Nubp2) to be causative. This finding was confirmed by a complementation assay with the dorothy allele and an independent Nubp2 null allele (Nubp2null). We demonstrated that Nubp2 is indispensable for embryogenesis. NUBP2 is implicated in both the cytosolic iron/sulfur cluster assembly pathway and negative regulation of ciliogenesis. Conditional ablation of Nubp2 in the neural crest lineage with Wnt1-cre recapitulates the dorothy craniofacial phenotype. Using this model, we found that the proportion of ciliated cells in the craniofacial mesenchyme was unchanged, and that markers of the SHH, FGF, and BMP signaling pathways are unaltered. Finally, we show evidence that the phenotype results from a marked increase in apoptosis within the craniofacial mesenchyme.

Keywords: Nubp2, neural crest, ENU mutagenesis, cilia

Summary Statement

An ENU screen identifies a novel allele of Nubp2 and a requirement in cranial neural crest survival and proper midfacial development.

Introduction

Vertebrate craniofacial development is a dynamic process highly dependent on a fascinating cell population known as the neural crest. Craniofacial neural crest cells (CNCC) are a multipotent and migratory cell type which delaminate from the dorsal aspect of the closing neural tube and travel ventrolaterally into the frontonasal prominence and pharyngeal arches where they become the mesenchyme of the emerging facial primordia [1, 2]. On each side of the mouse face, proliferation of the post-migratory neural crest causes medial and lateral nasal prominences to swell from the frontonasal prominence around embryonic day (E) 10, resulting in paired horseshoe-shaped outgrowths with visible depressed nasal pits in the center. The outgrowth of these prominences is coupled with that of the maxillary prominences to bring the three primordia into contact [3, 4]. This contact and the subsequent fusion of the medial nasal prominence, lateral nasal prominence, and maxillary prominence create the nostrils and upper lip [5]. As development continues past E10.5, further growth brings the paired medial nasal prominences into contact at the midface, where they fuse and give rise to the nasal septum, philtrum, and premaxilla.

During these early stages of craniofacial development, the CNCC rely on signaling cues from surrounding tissues to regulate their survival and the proliferation. These include Sonic Hedgehog (SHH) and Fibroblast Growth Factor 8 (FGF8) secreted by the surface ectoderm [6-9]. Loss of these signals, or an inability of CNCC to properly transduce ligand binding within the cell, impairs the outgrowth and subsequent fusion of the facial primordia, often ultimately leading to midline defects of the face [10]. The CNCC are also a particularly vulnerable cell population as evidenced by their marked susceptibility to ribosome biogenesis insults, sensitivity to the teratogenic effects of alcohol and many examples of genetic mutations leading to elevated CNCC apoptosis [11-17].

Despite significant advances in understanding the signaling pathways and transcriptional networks involved in craniofacial developmental disorders, their etiology often remains unclear. Unbiased forward genetics approaches in model organisms allow new insights and investigations into previously unexamined roles for genes and pathways in embryogenesis [18]. Here we report a new allele from an N-ethyl-N-nitrosourea (ENU) screen in the mouse that is integral to craniofacial development. We found the causal mutation to be in the gene nucleotide binding protein 2 (Nubp2). Nubp2 is a highly conserved P-loop NTPase first described in early sequencing and mapping of cDNA isolated from E7.5 mouse embryos [19]. In Saccharomyces cerevisiae it was identified as cytosolic Fe-S cluster deficient (Cfd1) in a screen for genes able to convert Iron regulatory protein 1 into c-aconitase [20] [21]. These and subsequent studies in yeast and animal cell lines established that NUBP2 is an integral component of the cytosolic iron-sulfur cluster assembly pathway where it acts along with its homologous binding partner NUBP1 as a scaffold for transfer of the iron-sulfur cofactor to non-mitochondrial apoproteins [22-24]. Later studies revealed that knockdown of Nubp1, or both Nubp1 and Nubp2, led to excessive centrosome duplication in vitro [25]. Further work indicated that silencing of Nubp1 and Nubp2 in cell culture led to supernumerary cilia formation [26]. An ENU screen isolated a mouse allele of Nubp1 with interrupted lung budding and centriole over-duplication [27].

This implication of Nubp2 in regulation of centriole duplication and ciliary dynamics, and the importance of the primary cilia to intercellular signaling, led us to hypothesize aberrant ciliogenesis would disrupt crucial CNCC signaling pathways, resulting in loss of mitogenic and/or survival signals and subsequent midfacial fusion defects. On the contrary, here we demonstrate that loss of Nubp2 from the CNCC does not lead to ciliogenesis or signaling defects, but causes a rapid onset of apoptosis throughout the craniofacial mesenchyme following CNCC migration which underlies the distinctive craniofacial presentation of the dorothy mutant. This represents the first example of a role for iron-sulfur cluster assembly pathway components in midfacial development.

Materials and Methods

Animal husbandry

All animals were housed and cared for in accordance with the Guide for the Care and Use of Laboratory Animals (National Institutes of Health), and all animal-associated activity was approved by the Institutional Animal Care and Use Committee of the Cincinnati Children’s Hospital Medical Center (Protocol # IACUC2016-0098). All animals were monitored daily by registered veterinary technicians and additional care provided where necessary. Euthanasia was performed via Isoflurane sedation followed by cervical dislocation and secondary euthanasia. Mice were housed in ventilated cages provided with water and chow ad libitum. During timed mating, noon of the day a copulation plug was detected was considered embryonic day (E) 0.5 (E0.5).

ENU mutagenesis

ENU mutagenesis was performed as previously published [18]. Wildtype C57BL/6J (B6; Jackson Laboratory, Bar Harbor ME) 6- to 8-week-old males were given a fractionated dose of ENU (three weekly dosages of 90-100 mg/kg ENU, Sigma). Mutagenized males were bred with Tg(Etv1-EGFP)B2192 GSat females (FVB/N TaC background) as part of a larger effort to identify potential mutants in cortical neuron patterning [28]. Following a three-generation backcross breeding scheme [18], G3 litters were dissected at late embryonic stages to detect gross developmental anomalies. The allele was propagated with test crosses onto the FVB background (to facilitate mapping if needed) until the causal allele was identified.

Variant detection

Exome sequencing was carried out in the CCHMC Genetic Variation and Gene Discovery Core Facility (Cincinnati, OH, USA). An exome library was prepared from genomic DNA using the NimbleGen EZ Exome V2 kit (Madison, WI, USA). Sequencing of the resulting exome library was done with an Illumina Hi Seq 2000 (San Diego, CA, USA). The Broad Institute’s GATK algorithm was used for alignment and variant detection. Variants were then filtered as described in Table 3. Mutations were validated by Sanger sequencing.

Table 3:

Explanation of exome sequencing result filtering

Total variants 176,315
Filter out non-homozygous variants 36,590
Filter out non-SNP variants 30,733
Filter out variants listed in dbSNP 5,800
Filter out "Low impact" variants 267
Filter out genes with multiple variants 116
Filter out known polymorphisms 4
Filter out variants seen in CCHMC control exomes 3
Filter out genes with no null phenotype 2
Filter out variant that doesn't segregate with phenotype 1

Alleles and genotyping

Nubp2Dor (Nubp2 c.626T>A; Nubp2V209D; dorothy) was discovered in an ENU mutagenesis screen as described above and maintained on a mixed background. Dorothy genotyping was performed using the TaqMan™ Sample-to-SNP™ Kit using proprietary primers designed by the vendor targeting Nubp2:c:626T>A or Zbtb12:c:1061C>A (Thermo Fisher Scientific, Waltham, MA, USA). The Nubp2tm1a (Nubp2null) allele was imported from the European Conditional Mouse Mutagenesis Program (EUCOMM, EM:08954) [29] using standard in vitro fertilization protocols. We excised the reporter gene trap cassette to create a traditional floxed allele Nubp2tm1c (Nubp2fx) by crossing the Nubp2tm1a with carriers of the Flippase enzyme [30]. PCR primers amplified a 470bp amplicon unique to the Nubp2tm1a allele (Nubp2tm1a F: TCATGCTGGAGTTCTTCGCC, Nubp2tm1a R: ATTCTTCCGCCTACTGCGAC). A separate primer pair amplify a 263bp sequence in the wildtype Nubp2 allele and 457bp band in the Nubp2fx allele (Nubp2WtFxF: GGATCCCAGTGCTGAGCTTT, Nubp2WtFxR: ACCACCTGCTAGCACTCAAC). For conditional ablations, we used the Wnt1-cre allele (B6.Cg-H2afvTg(Wnt1-cre)11RthTg(Wnt1-GAL4)11Rth/J) [31] which was genotyped using a standard Cre detection PCR reaction (CreF: GCGGTCTGGCAGTAAAAACTATC, CreR: GTGAAACAGCATTGCTGTCACTT). Visualization of neural crest cells targeted by Wnt1-cre was accomplished using the mT/mG double-fluorescent Cre reporter allele developed by Muzumdar et al. [32].

Cell culture

Human Embryonic 293T (HEK293T) cells were cultured in Dulbecco’s modified eagle medium (DMEM, Sigma-Aldrich) supplemented with 10% Fetal Bovine Serum and 1% Penicillin-Streptomycin (Sigma-Aldrich). C- and N- terminal GFP-tagged Nubp1 and Nubp2 expression plasmids [33] were transfected using Lipofectamine® 3000 (Thermo Fisher Scientific, Waltham, MA, USA) and imaged using a Nikon C2 Confocal microscope.

Histology and Immunohistochemistry

Embryos designated for hematoxylin and eosin (H&E) staining were dissected and fixed overnight in 10% neutral buffered formalin solution (Sigma-Aldrich) at room temperature (RT) and then dehydrated in 70% ethanol for 48h. Following fixation and dehydration, samples were submitted to the CCHMC Pathology Research Core (Cincinnati, OH, USA) for paraffin embedding. Embedded samples were sectioned at a thickness of 10-14μM, stained with hematoxylin and eosin, and sealed with Cytoseal Mounting Medium (Thermo-Scientific). Samples designated for Immunohistochemical (IHC) analysis were dissected and fixed overnight in 4% paraformaldehyde at 4°C followed by immersion in 30% sucrose solution at 4°C for 24-48 hours until sufficiently dehydrated as observed by changes in sample buoyancy. Embryos were then embedded sagittally (E9.5) or coronally (E10.5) in OCT compound (Tissue-Tek) and cryosectioned at a thickness of 10-14μM. Sections were dried on a slide warmer at 42°C for 30-45 minutes for general IHC or dried at room temperature for 15-30 minutes when cilia were to be imaged. Samples were then soaked in boiling citrate buffer for 45-60 minutes for antigen retrieval followed by three PBS washes and blocked for 1 hour at RT in 4% normal goat serum in PBST. After blocking, sections were incubated overnight at 4°C with the following primary antibodies as indicated: Cleaved Caspase 3 (1:400, Cell Signaling 9661), PHH3 (1:500, Sigma H0412), AP2 (1:20, Developmental Studies Hybridoma Bank), Laminin (1:250, Sigma L9393), γ-Tubulin (1:1000, Sigma T6557), Arl13b (1:250, Proteintech 17711-1-AP). The following morning, sections were washed three times in PBS and incubated at RT in AlexaFluor 488 goat anti-mouse and AlexaFluor 594 goat anti-rabbit secondary (1:500, Life Technologies) for 1 hour at RT, washed 3 times in PBS and incubated in DAPI (1:1000, Sigma) for 20 minutes at RT. After 3 more PBS washes slides were sealed with ProLong Gold Antifade Reagent (Life Technologies) and imaged using a Nikon C2 confocal microscope. IHC images depicting cell death and proliferation were quantified using ImageJ (Rasband, W.S., ImageJ, U. S. National Institutes of Health, Bethesda, Maryland, USA, https://imagej.nih.gov/ij/, 1997-2018). Quantification of primary cilia was performed using NIS elements software (Nikon Instruments, Melville, NY, USA).

In Situ Hybridization

Whole mount in situ hybridization was performed as described previously [34] with a hybridization temperature of 65°C. Probes were transcribed from linearized plasmids using T7 RNA polymerase: Etv5 (Xin Sun/ Gail Martin), Ptc1 [35], Msx1 [36], FGF8 [37], and Sox10 [38]

RNA Sequencing

3 WT and 3 Wnt1-cre; Nubp2cKO mutant E9.5 embryos were dissected from the yolk sac, and heads were snap frozen on dry ice. RNA was isolated and the individual samples were used for paired-end bulk-RNA sequencing (BGI-Americas, Cambridge, MA). Results for one of the 3 mutants were of low quality and were discarded at the start of analysis. RNA-Seq analysis pipeline steps were performed using CSBB [Computational Suite for Bioinformaticians and Biologists: https://github.com/csbbcompbio/CSBB-v3.0]. CSBB has multiple modules, RNA-Seq module is focused on carrying out analysis steps on sequencing data, which comprises of quality check, alignment, quantification and generating mapped read visualization files. Quality check of the sequencing reads was performed using FASTQC (http://www.bioinformatics.bbsrc.ac.uk/projects/fastqc). Reads were mapped (to mm10 version of Mouse genome) and quantified using RSEM-v1.3.0 [39].

RESULTS

Dorothy mutants were recovered in an ENU mutagenesis experiment.

We performed a forward Ethyl-N-Nitrosourea (ENU) genetic screen in the mouse to discover new alleles important for craniofacial development [18]. Dorothy (dor) mutants were isolated from this screen and readily identified by a characteristic craniofacial phenotype (Figure 1, Tables 1 and 2). At E16.5, dorothy mutants displayed a striking midfacial cleft phenotype in which the rostral-most nasal cartilage appears to be absent while the remaining nasal and maxillary structures were shifted laterally (Figure 1: A-F). We also noted micromelia and oligodactyly in each mutant embryo and commonly observed blood pooling in the distal hindlimbs reminiscent of the eponymous ruby slippers [40] (Table 2). While all surviving embryos (6/6) collected at E16.5 had midline clefting and truncated limb phenotypes, mandibular development was variably affected and ranged from apparently normal development in 3/6 to complete agenesis in 1/6 non-necrotic E16.5 embryos observed. Curiously, despite the dysmorphic nature of the dorothy face, histological analysis revealed the presence of tooth buds in relatively appropriate positions, albeit with slightly delayed development (Figure 1: C, F, white dotted outlines). We also noted a significant portion of dorothy mutants did not survive beyond organogenesis stages. These commonly featured extensive hematomas and appeared morphologically to have arrested around E9.0-E9.5 (Figure 1: G-N, Table 1,2). During maintenance of the dorothy colony, living embryos past E10.5 appropriate for phenotypic analysis became increasingly rare. We hypothesize this was due to the increasingly inbred genetic background as the propagation of line was initially onto an FVB background in the event genetic mapping would be needed to clone the causal gene. The initial B6/FVB hybrid environment in the first matings may have led to increased dorothy mutant survival which declined with subsequent homogeneity on the B6 background.

Figure 1: The Dorothy mutants have variable craniofacial phenotypes.

Figure 1:

Whole-mount and histological analysis of E16.5 control (A-C) and dorothy (D-F) mutant embryos. Mutants consistently present with midfacial clefting, micromelia and oligodactyly. The nasal cartilage (black arrows) is underdeveloped and ectopically located. Accumulation of blood or other fluid under the epithelium are also often noted (red asterisk). (F) Coronal sections also reveal cleft palate (black asterisks). Tooth buds are present but tooth development appears delayed (yellow dotted outlines). (G-N) Dorothy mutants with more severe phenotypes are developmentally arrested at the start of organogenesis (G-M), and often show signs of pervasive hematoma (red arrows). (Scale bars in B,C,E,F = 1mm, G-N = 200μm).

Table 1:

Recovery of dorothy mutants at different stages of embryogenesis

E9.5 E10.5 E11.5 E16.5
Genotype Observed Expected Observed Expected Observed Expected Observed Expected
Nubp2+/+ 23 20.25 34 30.5 6 5 51 45.25
Nubp2dor/+ 40 40.5 69 61 10 10 120 90.5
Nubp2dor/dor 18 20.25 19 30.5 4 5 10 45.25
p= 0.730 p= 0.055 p= 0.819 p= 6.172E-09

Table 2:

dorothy phenotyping at different stages of embryogenesis

E9.5
Term MGI ID (n) (%)
hematoma MP: 0008817 13 72.22%
abnormal first pharyngeal arch morphology MP: 0006337 11 61.11%
small frontonasal prominence MP: 0030249 7 38.89%
embryonic growth retardation MP: 0003984 1 5.56%
E10.5
abnormal lateral nasal prominence morphology MP: 0009902 19 100.00%
abnormal medial nasal prominence morphology MP: 0009903 19 100.00%
hematoma MP: 0008817 15 78.95%
embryonic growth retardation MP: 0003984 15 78.95%
small mandibular prominence MP: 0030346 15 78.95%
small maxillary prominence MP: 0030342 8 42.11%
absent maxillary prominence MP: 0030344 7 36.84%
E11.5
small mandibular prominence MP: 0030346 4 100%
hematoma MP: 0008817 3 75%
abnormal lateral nasal prominence morphology MP: 0009902 3 75%
abnormal medial nasal prominence morphology MP: 0009903 3 75%
abnormal eye development MP: 0001286 3 75%
absent maxillary prominence MP: 0030344 2 50%
E16.5
embryonic lethality during organogenesis MP: 0006207 4 40%
Remaining phenotypes scored from 6/10 embryos not clearly necrotic
abnormal digit development MP: 0006280 6 100%
micromelia MP: 0008736 6 100%
oligodactyly MP: 0000565 6 100%
midline facial cleft MP: 0000108 4 66%
embryonic lethality during organogenesis MP: 0006207 4 66%
hematoma MP: 0008817 3 50%
protruding tongue MP: 0009908 3 50%
absent snout MP: 0000450 2 33%
absent mandible MP: 0000087 2 33%
embryonic growth arrest MP: 0001730 1 16%
encephalomeningocele MP: 0012260 1 16%

The dorothy allele is a missense mutation in nucleotide binding protein 2 (Nupb2).

We performed exome sequencing to identify the causal allele in dorothy mutants. We sequenced three mutants and initially identified approximately 176,000 variants after quality control and read depth filters were applied. We first filtered out variants that were heterozygous in one or more mutants or that didn’t result from single nucleotide changes working under the assumption the dorothy allele is a recessive SNP, bringing the list of potential variants from ~176,000 to ~30,000 (Table 3). After filtering out SNPs known to be strain polymorphisms and variants predicted to have “low” impact, along with genes containing multiple variants (assumed to be highly polymorphic genes), 3 variants remained. One of these was in Pbx2 which has been deleted in mouse with no resulting phenotype [41]. The remaining variants were in nucleotide binding protein 2 (Nubp2) and zinc finger and BTB domain containing 12 (Zbtb12). We further genotyped ten mutants and only homozygosity for the Nubp2 was completely concordant with the phenotype as the Zbtb12 mutation was also homozygous in 9 unaffected embryos (Table 3). Thus, Nubp2:c.626T>A; p.Val209Asp was identified as a candidate causal variant.

The Val209Asp missense mutation changes the coding for a highly conserved nonpolar valine into a charged aspartic acid residue five amino acids downstream from a pair of iron-interacting cysteine residues [23] (Figure 2A). Sanger sequencing confirmed the nucleotide change c626T>A in dorothy embryos (Figure 2B). Transfection of HEK293T cells with GFP-tagged hNUBP2wt and hNUBP2Dor expression constructs [33] did not identify a change in translation or cellular localization of hNUBP2V209D (Figure 2C,D).

Figure 2: The dorothy mutation is an allele of Nubp2.

Figure 2:

(A) Amino acid sequence of NUPB2 in multiple model organisms. Nonpolar side chains (valine and isoleucine) at the position of the affected residue (red box, highlighted in yellow) are heavily conserved and located in close proximity to known functional Iron-interaction sites (highlighted in purple). In the dorothy mutant (p.Val209Asp; NUBP2V209D), this is replaced with an aspartate residue (red) harboring an electrically charged side chain. (B) Sanger sequencing confirms that Nubp2:c.626T>A is homozygous in embryos with the dorothy phenotype. (C,D) transfection of HEK293T cells with pAcGFP-C1-hNUBP2 (C) and pAcGFP-C1-hNUBP2DOR (D) expression constructs revealed that the corresponding fusion proteins were both expressed and localized in roughly the same pattern, suggesting that the Nubp2dor mutation does not preclude translation of NUBP2 protein.

While the dorothy phenotype was completely concordant with homozygosity for the NUBP2V209D allele, we sought to further confirm that the causal allele was in Nubp2 via a complementation test with an independently derived null allele of Nubp2. We imported the Nubp2tm1a (Nubp2null) allele from the international mouse phenotyping consortium [29, 42]. We first demonstrated that the Nubp2null/null embryos were not viable and were never recovered at early organogenesis stages (Table 4). Consistent with this, we noted frequent resorptions during these experiments, consistent with an early developmental lethality phenotype. We then intercrossed Nubp2dor/wt with Nubp2null/wt and never recovered Nubp2null/dor embryos at E16.5 (Table 5). This failure of the null allele to complement the ENU allele is the strongest possible genetic evidence that dorothy is an allele of Nupb2.

Table 4:

Embryonic lethality of Nubp2 ablation

E7.5-E10.5 embryo recovery
Genotype Expected Observed
Nubp2+/+ 8.75 20
Nubp2Null/+ 17.5 15
Nubo2Null/Nul1 8.75 0
p= 7.613E-06

Table 5:

Complementation test of Nubp2dor as the causative allele

Embryo recovery
E16.5 E10.5
Genotype Expected Observed Expected Observed
Nubp2+/+ 4.25 6 7.25 14
Nubp2Null/+ 4.25 3 7.25 8
Nubp2dor/+ 4.25 8 7.25 7
Nubp2Null/dor 4.25 0 7.25 0
p= 0.0344 p= 0.00347

Nubp2 deletion from the neural crest lineage recapitulates the dorothy craniofacial phenotypes.

Given the dorothy craniofacial phenotype, we hypothesized a unique requirement for Nubp2 in the CNCC lineage. In order to test the hypothesis of cell-autonomous requirement for Nubp2 in the CNCC and circumvent the early lethality and phenotypic variability of the dorothy allele, we used Wnt1-cre to ablate Nubp2 in just the neural crest [31]. We first generated the Nubp2tm1c (Nubp2fx) allele by crossing the Nubp2tm1a with a FLP transgenic [30] . We then mated Nubp2null/wt with Wnt1-cre to generate Wnt1-cre Nubp2null/wt. These were again mated to Nubp2fx/wt or Nubp2fx/fx to create the Wnt1-cre; Nubp2cKO embryos. We also performed similar crosses to generate Wnt1-cre; Nubpfx/fx. The two genotypes did not result in significantly different phenotypes and are collectively referred to here as Wnt1-cre; Nupb2cKO (Supplemental Table 1). The resulting Wnt1-cre; Nupb2cKO embryos had phenotypes remarkably similar to the dorothy homozygous mutants (Figure 3A-H). At E15.5, embryos were recovered with midfacial clefts and undersized nasal, mandibular, and maxillary structures. Histological analysis revealed severe hypoplasia of the cartilage primordia of the nasal septum along with agenesis of the lateral cartilage primordia of the nasal capsule (Figure 3C,D,G,H, black and yellow dotted outlines, respectively). Combined with the cleft palate, this seems to lead to a situation where the olfactory epithelium is continuous with the oral cavity and, as in the dorothy mutant, the brain is shifted rostrally. We next set out to determine the developmental stage at which mutants are first phenotypically distinct from littermates. Virtually all Wnt1-cre; Nubp2cKO embryos (34/35) were anatomically indistinguishable from littermate controls at E9.5 (Figure 3: I-L; Table 6). In contrast, a majority (14/17) of conditional mutants at E10.5 are easily identified by their conspicuously undersized facial primordia (Figure 3: M-P; Table 6). Thus, the Wnt1-cre; Nubp2cKO phenotype appears to emerge after CNCC migration, during early expansion stages in the pharyngeal arch, and results in a loss of craniofacial tissue.

Figure 3: Conditional ablation of Nubp2 in the neural crest lineage causes midfacial defects and recapitulates the craniofacial phenotype of the dorothy mutant.

Figure 3:

(A-H) Wnt1-cre; Nubp2cKO embryos recovered at E15.5 have severe craniofacial abnormalities including midline clefting and mandibular hypoplasia. (C, G) H&E stained coronal sections demonstrate palatal shelves fail to elevate. (D, H) higher magnification reveals that the cartilage primordium of the nasal septum is severely truncated (black dotted outline), while cartilage primordium of the nasal capsule lateral to the septum (yellow dotted outline) appears to be completely absent in the mutant. (I-L) At E9.5 Wnt1-cre; Nubp2cKO mutants are indistinguishable from wild-type (M-P) but at E10.5 the phenotype becomes apparent with hypoplastic nasal prominences, maxillary and mandibular prominences). (Scale bars in C, G = 500μm; D,H=50μm; I-P 100μm.)

Table 6:

Phenotypic description of Wnt1-cre; Nubp2cKO embryos

E9.5
Term MGI ID (n) (%)
no abnormal phenotype detected MP: 0002169 34/35 97%
abnormal first pharyngeal arch morphology MP: 0006337 1/35 3%
 
E10.5
Term MGI ID (n) (%)
abnormal lateral nasal prominence morphology MP: 0009902 14/17 82.35%
abnormal medial nasal prominence morphology MP: 0009903 14/17 82.35%
small maxillary prominence MP: 0030342 12/17 70.59%
small mandibular prominence MP: 0030346 4/17 28.57%
no abnormal phenotype detected MP: 0002169 3/17 17.65%
embryonic growth retardation MP: 0003984 1/17 5.88%
 
E11.5
Term MGI ID (n) (%)
abnormal lateral nasal prominence morphology MP: 0009902 10/12 83.33%
abnormal medial nasal prominence morphology MP: 0009903 10/12 83.33%
small maxillary prominence MP: 0030342 10/12 83.33%
absent mandible MP: 0000087 9/12 75.00%
hematoma MP: 0008817 6/12 33.33%
no abnormal phenotype detected MP: 0002169 2/12 16.67%

Deletion of Nubp2 in the CNCC does not affect centriole duplication or ciliogenesis.

Nubp2 has previously been implicated in the regulation of centriole duplication and ciliogenesis [25, 26]. We therefore hypothesized that similar disruptions in primary cilia biology may compromise the CNCC-derived mesenchyme. We used immunohistochemistry with confocal fluorescence microscopy to image and quantify centrosomes and primary cilia within the E9.5 craniofacial mesenchyme (Figure 4). We used antibodies for γ-tubulin to mark centrioles and ARL13B to highlight the ciliary axoneme. The cilia in Wnt1-cre; Nubp2cKO mutants were qualitatively indistinct from those in littermates (Figure 4, A-H). We then quantified the number of cilia and centrioles per nucleus for conditional mutants and controls. We observed slight increase in average cilia/cell (Figure 4I, p=0.009) but no difference in centriole number (Figure 4J; p=0.422).

Figure 4: Craniofacial mesenchymal cells have normal primary cilia and centrioles.

Figure 4:

(A-H) representitive fields showing immunostaining with fluorescent antibodies against γ-Tubulin and Arl13b in E9.5 craniofacial mesenchyme. No gross abnormalities are observed in mutant cilia (white arrows) or centrioles (white arrowheads). Mitotic cells can be observed with two centrioles (white M). (I) a minor, but statistically significant increase in the proportion of cilia per cell was measured in mutants. (J) There was no observed difference in the proportion of centrioles per cell in mutants. Scale bars: 5μm.

Loss of Nupb2 does not alter SHH, FGF or BMP signaling in craniofacial neural crest cells.

The midfacial defects found in the Wnt1-cre; Nubp2cKO mutants suggested a number of developmental signaling pathways may be adversely affected. Appropriate levels of SHH, FGF and BMP signaling are all known to be crucial for proper CNCC development. We addressed each of these individually with whole mount in situ hybridization on selected markers as canonical transcriptional targets of each signaling pathway.

Hedgehog signaling is well known to be a survival signal for the CNCC [43]. Genetic ablation of smoothened in the neural crest using the same Wnt1-cre allele used here led to a phenotype highly reminiscent of the dorothy phenotype [6]. However, Ptch1 expression was qualitatively unaffected in Wnt1-cre; Nubp2cKO mutants at E9.5 (Figure 5, A-D). FGF signaling is required from the surface ectoderm to the neural crest-derived facial mesenchyme during early craniofacial development [8, 9]. Etv5 as a measure of FGF signaling was expressed at normal levels (Figure 5, E-H). Finally, disturbances in BMP signaling during craniofacial development have been associated with defective fusion of the facial primordia [44], and mutation of the downstream transcription factor Msx1 causes cleft lip/palate. However, Msx1 levels also appeared unchanged in conditional mutants (Figure 5, I-L).

Figure 5: Major developmental signaling pathways are unaffected in Wnt1-cre; Nubp2cKO embryos.

Figure 5:

(A-L) Whole mount in situ hybridizations of E9.5 embryos and (M) normalized TPM values derived from RNA-sequencing of dissected E9.5 heads show that the SHH (A-D), FGF (E-H) and BMP (I-L) signaling pathways are unaltered at E9.5 in Wnt1-cre; Nubp2cKO embryos (Scale bars =100μm).

In order to further assess these canonical pathways and perform an unbiased transcriptional analysis of mutants, we dissected E9.5 heads from three controls and two mutants and performed bulk RNA sequencing. We first measured levels of SHH signaling (Ptch1, Gli1, Gli2, Gli3), FGF signaling (Fgf8, Etv5, Spry1, Spry2), and BMP signaling (Msx1, Lef1) and saw no significant changes between wild-type and mutant embryos, consistent with the whole mount in situ results (Figure 5, M, Table S2). Nubp2 transcripts were reduced by approximately 60% in Wnt1-cre; Nubp2cKO heads. This residual Nubp2 expression can likely be attributed to non-CNCC tissues which don’t express the Wnt1-cre recombinase. Additionally, mutants used for this experiment had a copy of the Nubp2null (Nubp2tm1a) allele, a conditional gene trap allele which could easily produce some sequence reads mapped to Nubp2 in the RNA-Seq analysis. After selecting for genes with a log fold change greater than ± 0.5 and false discovery rate ≤ 0.1, 61 genes were found to be differentially expressed (Table S2). Multiple analyses failed to identify a disrupted core molecular pathway or process from this list of differentially expressed genes.

Neural crest-specific ablation of Nubp2 does not significantly alter migration or proliferation, but increases apoptosis.

In the absence of intercellular signaling defects, we hypothesized that the failure of facial primordia to sufficiently expand may be due to differences in the ability of mutant neural crest cells to migrate, proliferate, and/or survive. To highlight migrating neural crest cells, we performed whole mount in situ hybridization on E9.5 (n=4) and E10.5 (n=2) Wnt1-cre; Nubp2cKO embryos using a Sox10 riboprobe (supplemental Figure 1 A-D). Sox10 is expressed by migratory NCC streams at E9.5 and in the developing cranial ganglia and dorsal root ganglia at E10.5. There were no significant changes in Sox10 expression patterns between mutants and littermate controls. We also made use of the mT/mG cre reporter allele [32] to visualize the NCC distribution in Nubp2cKO mutants at E10.5 (n=1, supplemental Figure 1 E-F). Again, the distribution of GFP signal in peripheral CNCC-derived structures Wnt1-cre; Nubp2cKO; mT/mG was similar to littermate controls. However, loss of GFP signal is obvious in the nasal prominences and pharyngeal arches which are the affected tissues in mutants populated by post-migratory CNCC.

Following migration of the CNCC, the craniofacial mesenchyme must be highly proliferative in order to produce the maxillary, mandibular, and nasal prominences and to ensure that the paired prominences meet at the midline to form the intermaxillary segment. At E9.5 the ratio of proliferating cells in the craniofacial mesenchyme as measured by a mitotic index of Phosphorylated Histone H3 to DAPI-stained nuclei is unchanged in Wnt1-cre; Nubp2cKO mutants (Figure 6, A-C; p=0.453). This ratio is mildly elevated at E10.5, when the lateral nasal prominences and medial nasal prominences are noticeably truncated in comparison to littermate controls (Figure 6, D-F; p=0.060).

Figure 6: Loss of Nubp2 leads to decreased survival of craniofacial neural crest.

Figure 6:

(A-E’) immunofluorescent staining for Phosphorylated Histone 3 (PHH3) at E9.5 (A-C; p=0.453) and E10.5 (D-F; p=0.060) shows no significant change in the proportion of proliferating Wnt1-cre; Nubp2cKO craniofacial mesenchymal cells at either stage. (G-L) in contrast, staining for CC3 at E9.5 (G-I; p=0.040) shows a slight increase in apoptotic mesenchyme and by E10.5 (J-L; p<0.0001) there is a striking and statistically significant increase. Scale bars: 100μm.

We then assayed levels of apoptosis by calculating the ratio of Cleaved Caspase 3 to DAPI-stained nuclei. We observed increased apoptosis at E9.5 (Figure 6, G-I; p=0.040) and a drastic increase at E10.5 (Figure 6, J-L; p<0.0001). This increase was more substantial in the MNPs than the LNPs (Fig. 6K). We do note that there is a wide range of rates of apoptosis in the mutants. This is consistent with the phenotypic variability where some embryos appear normal for a day or so longer in development than most littermates. In fact, we further quantified the rates of apoptosis and proliferation in “mild” and “severe” mutants and do note a striking difference in the apoptotic index (Supplemental Figure 2). Taken together, these data show CNCC lacking Nubp2 are capable of migrating into the frontonasal prominence and pharyngeal arches, correctly regulating ciliogenesis, responding properly to intercellular signaling, and proliferating at a normal rate, but undergo abrupt apoptosis between E9.5 and E10.5.

DISCUSSION

In this report, we describe our identification of the dorothy mutation and determine it to be an allele of Nubp2. The dorothy mutant has a variable phenotype with highly penetrant micromelia, syndactyly, dysmorphic nasal cartilage, and midfacial clefting (Figure 1, Tables 1, 2). Exome sequencing identified a variant in Nubp2 as a candidate causal mutation (Figure 3, Table 3). We used a null allele to show Nubp2 is required for early development (Table 4) and a complementation test to further demonstrate dorothy is a hypomorphic allele of Nubp2 (Table 5). We then determined that the dorothy craniofacial phenotype is due to a cell-autonomous requirement for Nubp2 in the neural crest lineage (Wnt1-cre; Nubp2cKO), with a phenotype emerging between E9.5 and E10.5 (Figure 3). We used these Wnt1-cre; Nupb2cKO embryos to show that ciliogenesis is only mildly affected in the craniofacial mesenchyme (Figure 4), despite previously demonstrated roles for Nubp2 as a regulator of ciliogenesis [26]. Several canonical signaling pathways required for craniofacial development and the proportion of proliferating cells are unaltered in Wnt1-cre; Nupb2cKO embryos (Figure 5, 6). Ultimately, we observed a conspicuous increase in apoptosis among the post-migratory CNCC mesenchyme of the craniofacial primordia starting around E9.5 (Figure 6, G-L). We hypothesize that it is this ectopic cell death which prevents the craniofacial primordia from growing sufficiently to bring the opposing medial nasal prominences into contact, resulting in severe midfacial clefting and hypoplasia of the mandible, nasal septum and nasal capsule. This study is the first to demonstrate a requirement for Nubp2 in development with a more specific role in craniofacial development.

The discovery of this role for Nubp2 illustrates the enduring power of unbiased forward genetic screens, as it is unlikely the gene would have otherwise been selected for study in craniofacial development. To our knowledge, no clinical disorders have been associated with NUBP2 variants. Moreover, this report is the first to show a CNCC defect resulting from mutations in a member of the iron-sulfur cluster assembly pathway. During the course of this study, the International Mouse Phenotyping Consortium characterized the Nubp2tm1b/tm1b mouse as “preweaning lethal with complete penetrance” (Nubp2tm1b is derived from the Nubp2null allele we described above) [42]. ENU mutagenesis creates missense mutations that are often hypo-morphic, providing more insight into how essential genes function in specific developmental processes. For instance, ENU-induced mutations of Hsd17b7 and Grhl2 lead to informative craniofacial and neural phenotypes despite the fact that total deletion of either gene is lethal before completion of organogenesis [45-48]. In many such cases the effects of point mutations cannot be inferred from knockout studies alone, and forward screens can provide a genetic model more relevant to the natural population. For example, patients with mutations in both alleles of POLR1C present with Treacher Collins Syndrome, while Plr1cnull/null mice are embryonic lethal [42, 49, 50].

NUBP1 and NUBP2 participate in an early step of the iron-sulfur cluster assembly pathway as a scaffold for the nascent cluster, which is transferred to target proteins via a process involving ATP hydrolysis [22, 23]. Depletion of Nubp2 in human cell lines compromises iron-sulfur cluster assembly pathway function [51]. The NUBP1NISW mutant is compromised in its ability to bind NUBP2, thus preventing formation of the canonical heteromeric scaffold complex [27]. This may explain some of the phenotypic similarities between the Nubp1Nisw/Nisw and dorothy alleles, such as micromelia, syndactyly, and defects in ocular development. Cataracts are also associated with mutations in Xpd/Ercc2 and Spry2, both of which are extramitochondrial iron-sulfur proteins [52-55]. Loss of Iop1 in the mouse, which operates downstream of NUBP1/NUBP2 in the pathway, causes lethality prior to E10.5 as does loss of Nubp2 [56, 57]. These data taken together suggest that aspects of the dorothy and Nubp1Nisw/Nisw phenotypes may be attributed to decreased iron-sulfur cluster assembly pathway activity, but also that complete loss of the pathway may be lethal prior to organogenesis. This is consistent with our inability to recover Nubp2null/nul1 embryos. Unfortunately, we were unable to directly assay iron sulfur cluster assembly pathway function during this study as MEFs derived from dorothy mutants did not expand in vitro. It is possible that NUBP2 homocomplexes are more important for iron-sulfur cluster maturation in the craniofacial mesenchyme, but recent in vitro studies using yeast proteins indicate that the homodimer is unable to catalyze ATP hydrolysis and therefore is presumably unable to transfer mature clusters to target proteins [58, 59]. It’s possible that in order to become catalytically active the NUBP2 homodimer requires interaction with a component not included in the cited studies or that the complex functions differently in mammals.

Another possibility to consider is if NUBP2 has acquired a secondary role during mammalian evolution. Such cases of “protein moonlighting” can be established when proteins acquire mutations or new post-translational modifications during evolution and new interactions arise [60]. One relevant example is the case of Iron regulatory protein 1. When the cytosolic iron-sulfur cluster pathway is able to act on the protein, it becomes enzymatically active as an aconitase. However, in the absence of cofactor, Iron regulatory protein 1 becomes an active RNA-binding protein modifying the turnover of target mRNAs [61]. Presumably these functions evolved as a way for the cell to couple the activity of an important iron-sulfur protein to regulation of transcripts relevant to iron homeostasis. Could Nubp2 have another function outside of the cytosolic iron-sulfur cluster assembly pathway that explains the presence of midfacial clefting in Nubp2, but not Nubp1 mutants? Our RNAseq experiment showed that pleckstrin homology like domain family A member 3 (Phlda3) transcripts were significantly upregulated in E9.5 Wnt1-cre; Nubp2cKO heads, while transcripts related to the iron-sulfur cluster assembly pathway, ciliary function, and all major signaling pathways remained unchanged (Supplemental Table 2). PHLDA3 is a proapoptotic repressor of the oncogene Akt [62], which is consistent with the increased apoptotic index of Wnt1-cre; Nubp2cKO craniofacial mesenchyme. The activation of a pro-apoptotic transcriptional landscape in CNCC at an embryonic stage before any other significant changes are detectable could indicate a more direct role for NUBP2 than previously considered. NUBP2 may interact with transcriptional regulators to promote CNCC survival. A similar function for Nubp1 in different contexts could explain why an increased expression of NUBP1 protein is associated with poor prognosis in certain types of cancer [63]. Future studies focused on Nubp1 and Nubp2 could attempt to identify novel binding partners for the proteins in mammalian tissue, which would not only address this alternate hypothesis but might help shed more mechanistic light on their roles in centrosome duplication and ciliogenesis and show whether these latter functions involve other iron-sulfur cluster assembly pathway components.

Supplementary Material

1

Supplemental Figure 1: Loss of Nubp2 does not interfere with neural crest migration. (A-D) Sox10 whole mount in situ hybridization at E9.5 (A-B) and E10.5 (C-D) shows normal organization of NCC migratory streams and peripheral nervous system derivatives in Wnt1-cre; Nubp2cKO mutants. (E-F) Fluorescent imaging of Wnt1-cre; Nubp2cKO; mT/mG and Wnt1-cre; mT/mG littermate control E10.5 embryos shows intact and normally organized peripheral NCC and dorsal root ganglia, while the frontonasal prominence and pharyngeal arches (populated by post-migratory CNCC mesenchyme) appear truncated and have diminished GFP signal in the mutant.

2

Supplemental Figure 2: Phenotypic variation among Wnt1-cre; Nubp2cKO E10.5 embryos corresponds to variation in the apoptotic, but not proliferative index. Sorting proliferation (A) and apoptosis (B) data based on severity of mutant phenotypes at E10.5 reveals that mutants with no abnormal phenotype detectable by sight (mild mutant) are indisinct from controls. On the other hand, mutants which can be easily distinguished by hypoplastic craniofacial primordia (severe mutants) have significantly higher apoptotic indices.

3
4
  • The ENU mutant dorothy exhibits midfacial clefting, micromelia, and syndactyly

  • Dorothy is caused by Nubp2:c:626T>A which codes NUBP2:p:Val209Asp (Nubp2dor)

  • Nubp2 is required for organogenesis, Nubp2null/null and Nubp2null/dor are lethal

  • Midfacial cleft in Wnt1-cre; Nubp2cKO embryos is caused by neural crest apoptosis

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Bronner ME, Simoes-Costa M, The neural crest migrating into the 21st century. Curr Top Dev Biol, 2016. 116: p. 115–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cordero DR, et al. , Cranial Neural Crest Cells on the Move: Their Roles in Craniofacial Development. American Journal of Medical Genetics Part A, 2011. 155a(2): p. 270–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Yoon H, et al. , Development of the lip and palate in staged human embryos and early fetuses. Yonsei Med J, 2000. 41(4): p. 477–84. [DOI] [PubMed] [Google Scholar]
  • 4.Kim CH, et al. , Early development of the nose in human embryos: a stereomicroscopic and histologic analysis. Laryngoscope, 2004. 114(10): p. 1791–800. [DOI] [PubMed] [Google Scholar]
  • 5.Jiang R, Bush JO, and Lidral AC, Development of the upper lip: morphogenetic and molecular mechanisms. Dev Dyn, 2006. 235(5): p. 1152–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jeong J, et al. , Hedgehog signaling in the neural crest cells regulates the patterning and growth of facial primordia. Genes Dev, 2004. 18(8): p. 937–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Billmyre KK and Klingensmith J, Sonic hedgehog from pharyngeal arch 1 epithelium is necessary for early mandibular arch cell survival and later cartilage condensation differentiation. Dev Dyn, 2015. 244(4): p. 564–76. [DOI] [PubMed] [Google Scholar]
  • 8.Shao M, et al. , FGF8 signaling sustains progenitor status and multipotency of cranial neural crest-derived mesenchymal cells in vivo and in vitro. J Mol Cell Biol, 2015. 7(5): p. 441–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Trumpp A, et al. , Cre-mediated gene inactivation demonstrates that FGF8 is required for cell survival and patterning of the first branchial arch. Genes & Development, 1999. 13(23): p. 3136–3148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Suzuki A, et al. , Molecular mechanisms of midfacial developmental defects. Dev Dyn, 2016. 245(3): p. 276–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Weaver KN, et al. , Acrofacial Dysostosis, Cincinnati Type, a Mandibulofacial Dysostosis Syndrome with Limb Anomalies, Is Caused by POLR1A Dysfunction. Am J Hum Genet, 2015. 96(5): p. 765–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang P, et al. , Alcohol exposure induces chick craniofacial bone defects by negatively affecting cranial neural crest development. Toxicol Lett, 2017. 281: p. 53–64. [DOI] [PubMed] [Google Scholar]
  • 13.Smith SM, et al. , Neural crest development in fetal alcohol syndrome. Birth Defects Res C Embryo Today, 2014. 102(3): p. 210–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Berres ME, et al. , Transcriptome Profiling Identifies Ribosome Biogenesis as a Target of Alcohol Teratogenicity and Vulnerability during Early Embryogenesis. Plos One, 2017. 12(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hasegawa S, et al. , Apoptosis in neural crest cells by functional loss of APC tumor suppressor gene. Proc Natl Acad Sci U S A, 2002. 99(1): p. 297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wang J, et al. , Defective ALK5 signaling in the neural crest leads to increased postmigratory neural crest cell apoptosis and severe outflow tract defects. BMC Dev Biol, 2006. 6: p. 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vieceli FM and Bronner ME, Leukocyte receptor tyrosine kinase interacts with secreted midkine to promote survival of migrating neural crest cells. Development, 2018. 145(20). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Stottmann RW and Beier DR, Using Enu Mutagenesis for Phenotype-Driven Analysis of the Mouse Methods in Enzymology, Vol 477: Guide to Techniques in Mouse Development, Part B: Mouse Molecular Genetics, Second Edition, 2010. 477: p. 329–348. [DOI] [PubMed] [Google Scholar]
  • 19.Nakashima H, et al. , Two novel mouse genes--Nubp2, mapped to the t-complex on chromosome 17, and Nubp1, mapped to chromosome 16--establish a new gene family of nucleotide-binding proteins in eukaryotes. Genomics, 1999. 60(2): p. 152–60. [DOI] [PubMed] [Google Scholar]
  • 20.Roy A, et al. , A novel eukaryotic factor for cytosolic Fe-S cluster assembly. Embo j, 2003. 22(18): p. 4826–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Voss H, et al. , Nucleotide sequence and analysis of the centromeric region of yeast chromosome IX. Yeast, 1995. 11(1): p. 61–78. [DOI] [PubMed] [Google Scholar]
  • 22.Netz DJ, et al. , The Cfd1-Nbp35 complex acts as a scaffold for iron-sulfur protein assembly in the yeast cytosol. Nat Chem Biol, 2007. 3(5): p. 278–86. [DOI] [PubMed] [Google Scholar]
  • 23.Netz DJA, et al. , A Bridging [4Fe-4S] Cluster and Nucleotide Binding Are Essential for Function of the Cfd1-Nbp35 Complex as a Scaffold in Iron-Sulfur Protein Maturation. Journal of Biological Chemistry, 2012. 287(15): p. 12365–12378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Camire EJ, et al. , The Yeast Nbp35-Cfd1 Cytosolic Iron-Sulfur Cluster Scaffold Is an ATPase. J Biol Chem, 2015. 290(39): p. 23793–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Christodoulou A, et al. , Motor protein KIFC5A interacts with Nubp1 and Nubp2, and is implicated in the regulation of centrosome duplication. J Cell Sci, 2006. 119(Pt 10): p. 2035–47. [DOI] [PubMed] [Google Scholar]
  • 26.Kypri E, et al. , The nucleotide-binding proteins Nubp1 and Nubp2 are negative regulators of ciliogenesis. Cell Mol Life Sci, 2014. 71(3): p. 517–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Schnatwinkel C and Niswander L, Nubp1 Is Required for Lung Branching Morphogenesis and Distal Progenitor Cell Survival in Mice. Plos One, 2012. 7(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cionni M, Menke C, and Stottmann RW, Novel genetic tools facilitate the study of cortical neuron migration. Mamm Genome, 2016. 27(1-2): p. 8–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Friedel RH, et al. , EUCOMM--the European conditional mouse mutagenesis program. Brief Funct Genomic Proteomic, 2007. 6(3): p. 180–5. [DOI] [PubMed] [Google Scholar]
  • 30.Dixon JE, Shaikh AC, and Sadowski PD, The Flp recombinase cleaves Holliday junctions in trans. Mol Microbiol, 1995. 18(3): p. 449–58. [DOI] [PubMed] [Google Scholar]
  • 31.Danielian PS, et al. , Modification of gene activity in mouse embryos in utero by a tamoxifen-inducible form of Cre recombinase. Curr Biol, 1998. 8(24): p. 1323–6. [DOI] [PubMed] [Google Scholar]
  • 32.Muzumdar MD, et al. , A global double-fluorescent Cre reporter mouse. Genesis, 2007. 45(9): p. 593–605. [DOI] [PubMed] [Google Scholar]
  • 33.Okuno T, Yamabayashi H, and Kogure K, Comparison of intracellular localization of Nubp1 and Nubp2 using GFP fusion proteins. Molecular Biology Reports, 2010. 37(3): p. 1165–1168. [DOI] [PubMed] [Google Scholar]
  • 34.Belo JA, et al. , Cerberus-like is a secreted factor with neutralizing activity expressed in the anterior primitive endoderm of the mouse gastrula. Mech Dev, 1997. 68(1-2): p. 45–57. [DOI] [PubMed] [Google Scholar]
  • 35.Johnson RL, et al. , Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science, 1996. 272(5268): p. 1668–1671. [DOI] [PubMed] [Google Scholar]
  • 36.Hill RE, et al. , A new family of mouse homeo box-containing genes: molecular structure, chromosomal location, and developmental expression of Hox-7.1. Genes Dev, 1989. 3(1): p. 26–37. [DOI] [PubMed] [Google Scholar]
  • 37.Crossley PH and Martin GR, The mouse Fgf8 gene encodes a family of polypeptides and is expressed in regions that direct outgrowth and patterning in the developing embryo. Development, 1995. 121(2): p. 439–51. [DOI] [PubMed] [Google Scholar]
  • 38.Southard-Smith EM, Kos L, and Pavan WJ, Sox10 mutation disrupts neural crest development in Dom Hirschsprung mouse model. Nat Genet, 1998. 18(1): p. 60–4. [DOI] [PubMed] [Google Scholar]
  • 39.Li B and Dewey CN, RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics, 2011. 12: p. 323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Baum LF, The Wonderful Wizard of Oz The Oz books. 1900: George M. Hill Company. [Google Scholar]
  • 41.Selleri L, et al. , The TALE homeodomain protein Pbx2 is not essential for development and long-term survival. Mol Cell Biol, 2004. 24(12): p. 5324–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dickinson ME, et al. , High-throughput discovery of novel developmental phenotypes. Nature, 2016. 537(7621): p. 508–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ahlgren SC and Bronner-Fraser M, Inhibition of sonic hedgehog signaling in vivo results in craniofacial neural crest cell death. Curr Biol, 1999. 9(22): p. 1304–14. [DOI] [PubMed] [Google Scholar]
  • 44.Liu W, et al. , Distinct functions for Bmp signaling in lip and palate fusion in mice. Development, 2005. 132(6): p. 1453–61. [DOI] [PubMed] [Google Scholar]
  • 45.Stottmann RW, et al. , Cholesterol metabolism is required for intracellular hedgehog signal transduction in vivo. PLoS Genet, 2011. 7(9): p. e1002224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shehu A, et al. , Prolactin receptor-associated protein/17beta-hydroxysteroid dehydrogenase type 7 gene (Hsd17b7) plays a crucial role in embryonic development and fetal survival. Mol Endocrinol, 2008. 22(10): p. 2268–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Menke C, et al. , Grhl2 is Required in Nonneural Tissues for Neural Progenitor Survival and Forebrain Development. Genesis, 2015. 53(9): p. 573–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rifat Y, et al. , Regional neural tube closure defined by the Grainy head-like transcription factors. Dev Biol, 2010. 345(2): p. 237–45. [DOI] [PubMed] [Google Scholar]
  • 49.Dauwerse JG, et al. , Mutations in genes encoding subunits of RNA polymerases I and III cause Treacher Collins syndrome. Nat Genet, 2011. 43(1): p. 20–2. [DOI] [PubMed] [Google Scholar]
  • 50.Ghesh L, et al. , Autosomal recessive Treacher Collins syndrome due to POLR1C mutations: Report of a new family and review of the literature. Am J Med Genet A, 2019. [DOI] [PubMed] [Google Scholar]
  • 51.Stehling O, et al. , Function and crystal structure of the dimeric P-loop ATPase CFD1 coordinating an exposed [4Fe-4S] cluster for transfer to apoproteins. Proc Natl Acad Sci U S A, 2018. 115(39): p. E9085–e9094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kunze S, et al. , New mutation in the mouse Xpd/Ercc2 gene leads to recessive cataracts. PLoS One, 2015. 10(5): p. e0125304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Tan X, et al. , Sprouty2 Suppresses Epithelial-Mesenchymal Transition of Human Lens Epithelial Cells through Blockade of Smad2 and ERK1/2 Pathways. PLoS One, 2016. 11(7): p. e0159275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rudolf J, et al. , The DNA repair helicases XPD and FancJ have essential iron-sulfur domains. Mol Cell, 2006. 23(6): p. 801–8. [DOI] [PubMed] [Google Scholar]
  • 55.Wu X, et al. , Mammalian sprouty proteins assemble into large monodisperse particles having the properties of intracellular nanobatteries. Proc Natl Acad Sci U S A, 2005. 102(39): p. 14058–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Song D and Lee FS, Mouse knock-out of IOP1 protein reveals its essential role in mammalian cytosolic iron-sulfur protein biogenesis. J Biol Chem, 2011. 286(18): p. 15797–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Seki M, et al. , IOP1 protein is an external component of the human cytosolic iron-sulfur cluster assembly (CIA) machinery and functions in the MMS19 protein-dependent CIA pathway. J Biol Chem, 2013. 288(23): p. 16680–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Grossman JD, et al. , The Cfd1 Subunit of the Nbp35-Cfd1 Iron Sulfur Cluster Scaffolding Complex Controls Nucleotide Binding. Biochemistry, 2019. 58(12): p. 1587–1595. [DOI] [PubMed] [Google Scholar]
  • 59.Grossman JD, et al. , Coupling Nucleotide Binding and Hydrolysis to Iron-Sulfur Cluster Acquisition and Transfer Revealed through Genetic Dissection of the Nbp35 ATPase Site. Biochemistry, 2019. 58(15): p. 2017–2027. [DOI] [PubMed] [Google Scholar]
  • 60.Copley SD, An evolutionary perspective on protein moonlighting. Biochem Soc Trans, 2014. 42(6): p. 1684–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Volz K, The functional duality of iron regulatory protein 1. Curr Opin Struct Biol, 2008. 18(1): p. 106–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kawase T, et al. , PH domain-only protein PHLDA3 is a p53-regulated repressor of Akt. Cell, 2009. 136(3): p. 535–50. [DOI] [PubMed] [Google Scholar]
  • 63.Liu W, et al. , Expression of family with sequence similarity 172 member A and nucleotide-binding protein 1 is associated with the poor prognosis of colorectal carcinoma. Oncol Lett, 2017. 14(3): p. 3587–3593. [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

1

Supplemental Figure 1: Loss of Nubp2 does not interfere with neural crest migration. (A-D) Sox10 whole mount in situ hybridization at E9.5 (A-B) and E10.5 (C-D) shows normal organization of NCC migratory streams and peripheral nervous system derivatives in Wnt1-cre; Nubp2cKO mutants. (E-F) Fluorescent imaging of Wnt1-cre; Nubp2cKO; mT/mG and Wnt1-cre; mT/mG littermate control E10.5 embryos shows intact and normally organized peripheral NCC and dorsal root ganglia, while the frontonasal prominence and pharyngeal arches (populated by post-migratory CNCC mesenchyme) appear truncated and have diminished GFP signal in the mutant.

2

Supplemental Figure 2: Phenotypic variation among Wnt1-cre; Nubp2cKO E10.5 embryos corresponds to variation in the apoptotic, but not proliferative index. Sorting proliferation (A) and apoptosis (B) data based on severity of mutant phenotypes at E10.5 reveals that mutants with no abnormal phenotype detectable by sight (mild mutant) are indisinct from controls. On the other hand, mutants which can be easily distinguished by hypoplastic craniofacial primordia (severe mutants) have significantly higher apoptotic indices.

3
4

RESOURCES