Capsule Summary
Impact of human FOXN1 variants on T cell development determined with transcriptional assays, protein localization studies, mouse models, and reaggregate thymus organ cultures. Variants with benign, partial-, complete- loss and gain- of function outcomes found.
Keywords: thymopoiesis, T cell immunodeficiency, TRECs, FOXN1, thymus epithelial cells, reaggregate thymus organ cultures
Background
The thymus supports T cell development via two well-defined epithelial cell subsets, cortical and medullary thymus epithelial cells (TECs). TEC development and function is controlled by the transcription factor, Forkhead Box N1 (FOXN1), which positively regulates the expression of nearly 500 genes. The genes include chemokines, cytokines, keratins, Notch ligands and components of the thymoproteosome. Autosomal recessive FOXN1 mutations in humans result in a combined nude and severe combined immunodeficiency phenotype. However, individuals with single allelic or compound heterozygous FOXN1 mutations often have less severe phenotypes. With >400 FOXN1 variants now reported, the impact of many on protein function and thymopoiesis remain unclear. Using functional studies, we classified FOXN1 variants into benign, loss- or gain- of function, and dominant negatives. Several FOXN1 variants reduced the nuclear distribution of the protein, revealing a nuclear localization signal. Mice developed to genocopy the compound heterozygous FOXN1 mutations, identified in a patient, exhibited a transient thymus hypoplasia during embryogenesis that normalized post-natally. To screen the consequences of diverse FOXN1 variants on thymopoiesis, reaggregate thymus organ cultures combined with TAT-Foxn1 fusion protein transduction procedures were developed. These diverse experimental strategies aid in the identification of FOXN1 variants with clinical consequences.
Introduction
The thymus produces the T cells of the immune system, a process that begins during embryogenesis and continues throughout life1. The magnitude of thymopoiesis is maximal around adolescence, followed by a progressive age-dependent decline in T cell output2, 3. The principal cells needed for maintaining thymus integrity and support T cell development are thymic epithelial cells (TEC)4. Such cells are categorized into 2 major subsets, cortical and medullary TECs. These TECs express chemokines, growth factors, and cell surface ligands, including self-peptides embedded in major histocompatibility complex (MHC) molecules to support T cell development1, 4. The presentation of self-peptide/self-MHC complexes by TECs provides selection cues for those T cells expressing T cell receptors recognizing self. This enables the T cells of the immune system to discriminate self from foreign antigens1. The differentiation, expansion, and function of cortical and medullary TECs is governed by the transcription factor, Forkhead Box N1 (FOXN1)5, 6. FOXN1 expression is induced following the initial specification of the thymus anlage within the 3rd pharyngeal pouch during embryogenesis7, 8. This occurs during the 7th week of gestation in humans, which corresponds to day 11 in mouse embryonic development9. Upon expression, FOXN1 supports cTEC and mTEC sub-lineage development in both the fetal and postnatal thymus. This is dependent on Foxn1 induced expression of a suite of genes needed for TEC functions. Among these are DLL4, CCL25, Cathepsin L, CD40, PAX1, and HLA/MHC Class II4, 10, 11 10, 12, 13. In addition to its role in the thymus, FOXN1 also maintains the structural rigidity of the hair shaft and keratinization of the nail plate14, 15. There are two defined domains in FOXN1, a centrally located DNA binding region and a carboxy terminal transactivation segment (Fig. 1A)10, 16–18.
Autosomal recessive (AR) FOXN1 mutations exist in humans, rats, mice, and cats, resulting in a nude/severe combined immunodeficiency (SCID) presentation19–24. The nude phenotype, or alopecia universalis, results from the hair shaft folding over at the level of the sebaceous gland and breaking prior to extrusion through the dermal layer14. A nude presentation is also observed in patients with HOXC13 mutations, with this transcription factor positively regulating FOXN1 expression in the skin14, 25, 26. AR FOXN1 mutations likewise impact the nails, with a layer of cornified/stratified squamous epithelium forming at the proximal end of the bed instead of the normal translucent layers14. FOXN1 autoregulates its own expression in the thymus10, 27. The SCID phenotype arises because AR FOXN1 mutations lead to significantly reduced TEC numbers along with their impaired functionality, resulting in a block in T cell development. AR loss of function mutations in FOXN1 and the consequent T cell lymphopenia result in life-threatening viral, fungal, and bacterial infections23, 24, 28. For such affected individuals, an allogenic thymus implant remains the only effective clinical treatment option29, 30. Since the original description of 3 distinct AR FOXN1 mutations affecting just a few individuals, increasing numbers of patients with single allelic and compound heterozygous mutations are being reported23, 28, 31. This increase is partly due to the widespread application of genomic sequencing to identify mutations in individuals identified with low T cell receptor excision circles (TRECs), which reveal reduced T cell output from the thymus32, 33. ClinVar database lists 400 mutations in human FOXN1, which are often designated as of uncertain clinical significance, likely benign, or conflicting interpretations of pathogenicity.
We analyzed the impact of 35 distinct human FOXN1 variants on protein function and thymopoiesis, selecting representative mutations throughout the protein, including those forming truncations. Our functional assays included transcriptional reporter assays and protein localization studies. The findings uncovered FOXN1 variants with functional changes categorized into benign, partial- to complete- loss of activity, dominant negative consequences, and gain of function outcomes. Several amino acid substitutions impacted the nuclear distribution of FOXN1, revealing a nuclear localization sequence within the DNA binding domain. To define the functional consequences of specific FOXN1 mutants on thymopoiesis, we used mouse models to genocopy compound heterozygous variants identified in a patient. The mouse model revealed a transient thymus hypoplasia evident during embryogenesis. Additionally, reaggregate thymus organ cultures coupled with transduction procedures using TAT-Foxn1 fusion proteins were developed to more rapidly screen variants for their impact on thymopoiesis. Taken together, these approaches are enabling a precise determination of the functional consequences of assorted FOXN1 variants on T cell development.
METHODS
Study Approval
The Institutional Review Board at UT Southwestern Medical Center approved this study (IRB# 072010–009 and IRB# 112010–013). M.T.d.l.M, C.S., and C.A.W. acquired clinical information for several patients with FOXN1 variants (all in the U.S.A). Sequence information on de-identified patients were obtained under a protocol declared exempt by Duke University Health System Institutional Review Board. Additional patients with FOXN1 variants were obtained from published reports and/or the ClinVar database23, 28, 34. Animal work described in this manuscript has been approved and conducted under the oversight of the UT Southwestern Institutional Animal Care and Use Committee (APN numbers 2015–101247 and 2015–101163). Mice were housed in a specific pathogen-free facility at UT Southwestern Medical Center. The Foxn1 targeted mice were developed entirely on a C57BL/6 background. While >40 founders screened for each construct, one independent line was expanded for each of the mutants generated by CRISPR/Cas; the Foxn11288 mutations (Foxn1-1288 #2; p.P430S) and Foxn11465 (Foxn1-1465 #43; p.Q489Rfs16). Foxn1933 #17 (p.D313Tfs) was previously described31. Mice used are listed in Supplemental Table 2. All the Foxn1 lines were crossed for 1–2 generations with C57BL/6 mice prior to intercrossing.
Antibodies, oligonucleotides, and plasmids
The antibodies used in the experiments, their sources and the plasmids used for transfections are listed in Supplemental Table 2. Oligonucleotide sequences used for genotyping, PCR reactions, qRT-PCR reactions, and sequencing are listed in Supplemental Table 3. Murine Foxn1 cDNA was cloned as previously described31. The EcoRI restriction site within the coding sequence of murine Foxn1 was eliminated by site-directed mutagenesis to facilitate subcloning into various vectors. Site-directed mutagenesis was performed to introduce human variants into the mouse Foxn1 cDNA. Human FOXN1 cDNA was obtained from Addgene (#153137) and subcloned into pCDNA3.1 using HindIII and XbaI restriction sites. For direct comparisons with the mouse Foxn1 constructs, the coding region of human FOXN1 was subcloned into the pCMV-FLAG vector, which was used originally used for murine Foxn1 (Cat. #635668, Clonetech Inc., now Thermo-Fisher). First, site-directed mutagenesis was used to mutate the internal BglII site in the human cDNA. Then primers were used to create new BglII and KpnI restriction sites at the 5’ and 3’ end of human FOXN1. The PCR product was digested and cloned into the BglII/KpnI sites in pCMV-FLAG. Site-directed mutagenesis was used to create the variants listed in the Table 1 and supplemental Table 1. Sanger sequencing was used to confirm the mutations in all the constructs. The pyruvate kinase gene includes the cDNA encoding a full-length pyruvate kinase construct with a Myc-tag at the 5’end. The pcDNAI-mycPKsec13 plasmid was kindly provided by B. Fontura (UTSWMC), with this plasmid sourced from Dr. M. Michael, University of California, San Diego, CA. The plasmid was digested with HindIII and ApaI, and the myc-PK-sec13 insert was subcloned into pcDNA3.1, digested with the same restriction enzymes. This enabled plasmid propagation in ampicillin selection media. The ApaI site was subsequently modified to incorporate a XhoI restriction site. With this, candidate Foxn1 cDNA sequences comprising a NLS were subcloned into the EcoRI/Xho I restriction sites within pcDNA3.1-mycPK plasmid to yield an in-frame extension of pyruvate kinase.
Table 1:
Patient Identifiers | DNA Sequence Variant | Protein Sequence Change | Luciferase Reporter Assaysa | Functional Impact | Protein Domain(s) Impactedb | ClinVar Designationc | Designation based current findingsd |
---|---|---|---|---|---|---|---|
Normal | NAe | None | 100% | NA | NA | ||
Du Study and current study | |||||||
Pt.1f | c.933_936dupACCC | p.D313fs169g | 0.9% | LOFh | DBD-TAD | Pathogenic | Pathogenic |
c.1089_1103del15 | p.W363Cdel5aa | 49% | LOF | DBD | Pathogenic | Pathogenic | |
Pt. 2f | c.1288C>T | p.P430S | 92% | No impact | - | Uncertain Significance | Benign |
c.1465delC | p.Q489Rfs60 | 5% | LOF | TAD | Pathogenic/Likely pathogenic | Pathogenic | |
Pts. 3–4 | c.1465delC | p.Q489Rfs60 | 5% | LOF | TAD | Pathogenic/Likely pathogenic | Pathogenic |
Pt. 5 | c.724C>T | p.P242S | 85% | N-term | Uncertain significance | Benign | |
Pt. 6 | c.958C>T | p.R320W | 2% | LOF | DBD | Uncertain significance | Pathogenic |
Pt. 7 | c.962A>G | p.H321R | 206% | GOFg | DBD | Uncertain significance | Pathogenic |
Pt. 8 | c.982T>C | p.C328R | 57% | Partial LOF | DBD | No entry | Likely benign |
Pt. 9 | c.1075G>A | p.E359K | 112% | GOF | DBD | Uncertain significance | Benign |
Pt. 10-Pt. 14 | c.1201_1206 | p.P401del2aa | 58% | Partial LOF | TAD | No entry | Likely Benign |
Pt. 15 | c.1293delC | p.P432fs118 | 0.4% | LOF | TAD | No entry | Pathogenic |
Pt. 16 | c.1418delC | p.P473Hfs77 | 1.5% | LOF | TAD | Pathogenic | Pathogenic |
New Pt. 17 | c.1567G>A | p.G523R | 84% | No impact | TAD | No entry | Benign |
New Pt. 18 | c.1628G>A | p.G543E | 107% | GOF | TAD | Uncertain significance | Benign |
New Pt. 19 | c.1364–1367del | p.Y455Cfs94 | 1.6% | LOF | TAD | Pathogenic/Likely pathogenic | Pathogenic |
Bosticardo Study | |||||||
P1 | c.505G>A | p.E169K | 78% | No impact | N-term | Uncertain significance | Benign |
P2 | c.907delG | p.E303Sfs247 | 1.2% | LOF | DBD-TAD | Uncertain significance | Pathogenic |
P4 | c.961C>A | p.H321N | 5% | LOF | DBD | Uncertain significance | Pathogenic |
P5 | c.1009delG | p.G337Efs213 | Not tested | Predict LOF | DBD-TAD | No entry | Not known |
P6 | c.1086_1087insA | p.W363Mfs118 | Not tested | Predict LOF | DBD-TAD | No entry | Not known |
P7 | c.1135+5G>C | not reported | Not tested | Predict LOF | No entry | Not known | |
Giardino Study | |||||||
P8 | c.1168_1168delG | pD390Kfs160 | Not tested | DBD-TAD | No entry | ||
P9 | c.1201_1216del16 | p.P401Afs144 | 1.4% | LOF | TAD | Pathogenic/Likely pathogenic | Pathogenic |
P14 | c.1205delC | p.P402Lfs148 | Not tested | Predict LOF | TAD | Pathogenic | Pathogenic |
P15 | c.1206delT | p.L404Cfs146 | Not tested | Predict LOF | TAD | Uncertain significance | Pathogenic |
P16 | c.1315delC | p.L439Cfs111 | Not tested | Predict LOF | TAD | Conflicting interpretations | Pathogenic |
P18 | c.1316delT | p.L439Rfs111 | Not tested | Predict LOF | TAD | Pathogenic | Pathogenic |
P19 | c.1418delC | p.P473Hfs77 | 1.4% | LOF | TAD | Pathogenic | Pathogenic |
P20 | c.1420C>T | p.Q474* | Not tested | Predict LOF | TAD | Pathogenic | Pathogenic |
P21 | c.1584delC | p.L529Wfs21 | 12.5% | LOF | TAD | No entry | Pathogenic |
P22 | c.974T>C | p.L325P | 1.5% | LOF | DBD | Uncertain significance | Pathogenic |
P23 | c.1392_1401del10 | p.P465Rfs82 | 1.4% | LOF | TAD | Pathogenic | Pathogenic |
P25 | c.1850_1854del5 | p.Y617Cfs157 | 32% | LOF | TAD | No entry | Pathogenic |
P31-P47 | c.763C>T | p.R255* | Not tested | LOF | DBD-TAD | Pathogenic | Pathogenic |
P8f | c.340C>T | p.R114X | Not tested | LOF | N-term | Pathogenic | Pathogenic |
Not Reported | p.E139fs | Not tested | Predict LOF | N-term | No entry | Pathogenic | |
P9f | Not Reported | p.C82* | Not tested | Predict LOF | N-term | No entry | Pathogenic |
c.1049C>T | p.P350L | 88% | No impact | DBD | Uncertain significance | Benign | |
P11 | Not Reported | p.T527fs | Not tested | Predict LOF | TAD | No entry | Pathogenic |
P12 | Not Reported | p.V294I | 18% | LOF | DBD | No entry | Pathogenic |
P13-P18 | c.1392–1401del10 | p.P465Rfs82 | 1.4% | LOF | TAD | Pathogenic | Pathogenic |
Rota Study | |||||||
3 Pts | c.1370delA | p.H457Pfs93 | 0.6% | LOF | TAD | Pathogenic | Pathogenic |
ClinVar | |||||||
c.205C>T | p.R69C | 91% | No impact | N-term | Benign | Benign | |
c.362C>T | p.A121V | 89% | No impact | N-term | Benign | Benign | |
c.689C>G | p.P230R | 103% | No impact | N-term | Uncertain significance | Benign | |
c.713G>A | p.G238D | 105% | No impact | DBD | Benign | Benign | |
c.1664C>T | p.A555V | 75% | No impact | TAD | Benign | Likely benign | |
Invitae | |||||||
c.1911C>A | p.Tyr637* | 72% | Partial LOF | C-term | Likely benign | ||
Variants developed for functional studies | |||||||
c.1584delC | p.D528fs20 | 12% | LOF | TAD | N/A | Pathogenic | |
c.1015–1016TC>GA | p.S339D | 83% | No impact | DBD | N/A | Benign | |
c.1465–1466delCA, c.1584–1585insC | p.Q489Gfs60 | 1.4% | LOF | TAD | N/A | Pathogenic | |
Site-directed mutations | p.K316A/R320W | 5.7% | LOF | DBD | N/A | Pathogenic | |
Site-directed mutations | K316A/R320W/K327A | 3.5% | LOF | DBD | N/A | Pathogenic | |
Site-directed mutations | p.K331A/N334A/K335A | 5.5% | LOF | DBD | N/A | Pathogenic | |
Site-directed mutations | p.K327A/K331AN334A/K335A | 2.1% | LOF | DBD | N/A | Pathogenic | |
Site-directed mutations | p.K331A/N334A/K335A/W363Adel5aa | 3.5% | LOF | DBD | N/A | Pathogenic |
Luciferase reporter assays performed in the current study with beta5t using human FOXN1 variants
DBD = DNA binding domain; TAD = transactivation domain; N-term = Amino terminal region of protein
Based on autosomal recessive presentation
Shaded box reveals updated designation from current findings using ClinVar terminology
NA = not applicable
Yellow shaded box defines patients with compound heterozygous FOXN1 mutations
In the murine cDNA, the ACCC insertion causes a D313Tfs12
Loss-of-function = LOF; Gain-of-function =GOF
Lymphocyte and epithelial cell preparations
Thymocytes and peripheral T cell populations were processed for flow cytometric analyses as published previously35, 36. Thymocyte subsets and peripheral T cells were analyzed for the cell surface expression of various proteins including CD3, CD4, CD8, CD11b, CD11c, CD19, CD25, CD44, CD45, CD69, B220, NK1.1, αβ TCR, γδ TCR, and Ter119. Antibody sources are documented in Supplemental Table 2 . Experimentally, the lymphoid tissues were placed in FACS buffer (PBS with 2% FBS). Single cell suspensions were made by mashing the tissues through a stainless-steel mesh (100 Mesh, 0.0045 inch woven #316). The cell suspension was collected, washed, counted and aliquots stained with antibodies for 20 minutes. For the characterization of the stromal cell populations in the thymus, the tissue was digested Liberase™ (Roche) containing DNase I (Roche) using procedures described37. For embryonic tissues obtained between e12-e17.5, the lobes were separated and independently processed for thymocyte or TEC isolations. In the case of thymocyte preparations, single cell suspensions were prepared by mashing one lobe under a nylon membrane. Such cells were stained with antibodies detecting CD4 and CD8 and the other markers as described above. To characterize mesenchymal, endothelial, and TECs, the whole tissue or one of the two lobes, if sufficiently large, was processed in 100 μl of Liberase™ (Roche) containing DNase I (Roche), with the digestion performed for 2–10 min. The single cells were stained with antibodies against CD45 (Tonbo Scientific), MHC II (I-A/I-E) (Tonbo Scientific), EpCAM (eBioscience), BP-1 (eBioscience), and UEA-1 (Vector Laboratories). Samples were analyzed on both FACSCaliber™ and FACSCantoII™ machines (BD Bioscience). FlowJo software (Tree Star Inc.) was used for data analysis. Thymic epithelial subsets were analyzed by electronically gating for CD45−EpCAM+ expression and selecting for UEA1+BP1− to define medullary TECs or BP1+UEA1− for cortical TECs. MHC class II high and low cells were used to discriminate between the subsets within cortical and two medullary TECs.
Immunohistochemistry and Western blotting
Thymic tissue was fixed in 4% paraformaldehyde (PFA) and used for imaging. H&E staining was performed under standard protocols and imaged on an Aziovert 200M inverted fluorescent microscope. For immunofluorescence staining, HeLa cells were grown on coverslips in 24-well plates followed by transfections. Forty-eight hours later, the cells were washed in PBS and then fixed with 2% PFA) for 10 minutes. These cells were permeabilized with 0.05% Triton X-100 and incubated with anti-mouse Foxn1 (1:100, Santa Cruz Biotech, CA) or anti-myc (9E11,1:100, Abcam Antibodies) along with Phyllandoin 488, all done at 4°C overnight. Cells were then washed with PBS and stained with donkey anti-mouse 594 and DAPI for 1 hour and mounted with ProLong™ Gold anti-fade mountant (Thermo Fisher Scientific). Images were taken on a Leica confocal microscope and/or Keyence and EVOS microscopes. For quantification of nuclear and cytoplasmic occupancy, 7 –10 random fields were taken for each construct, in each field number of cells with cytoplasmic occupancy were counted and divided with total number of transfected cells. Cytoplasmic occupancy means when Foxn1 protein can be seen outside the nucleus as well. In case of WT the probability of Foxn1 being also present in cytoplasm is less than 1%. For western blotting, 5 × 104 HeLa cells/well were plated in a 24 well plate. The cells were transfected with 500 ng to 1 μg of the various expression vectors containing either human or murine FOXN1 using Fugene HD reagent (Promega, Madison, WI). Twenty-four hours post-transfection, the cells were washed with PBS and lysed in RIPA buffer38. In our protocols, RIPA comprised 50 mM HEPES, pH 7.4, 150 mM NaCl, 0.5% w/v sodium deoxycholate, 0.1% SDS. This was supplemented with protease inhibitors (aprotinin, leupeptin, benzamidine, PMSF) and 1U/ml benzonase (digests DNA). Proteins were resolved on SDS-PAGE gels and transferred onto PVDF membranes for Western blotting. Immunoblotting was done with the indicated antibodies followed by secondary antibodies linked to horseradish peroxidase, as listed in Supplemental Table 239.
Luciferase reporter assays
A luciferase reporter containing the promoter of Psmb11 gene was generously provided by Drs. S. Zuklys and G. Holländer (University of Oxford, Oxford, UK)10. The Psmb11 luciferase reporter construct (1 μg) was co-transfected into HeLa cells (2 × 104 cells/well) along with the indicated expression containing murine or human FOXN1 wild type or mutants using 3 μl of Fugene HD reagent per transfectant (Promega, Madison, WI). A separate construct containing beta-galactosidase (0.1 μg), which is used as an internal control, was included in the transfections. Forty-eight hours post-transfection, the cells were washed in PBS, and 200 μl of Passive lysis buffer (1X) was added to the wells (Promega Incorp., Madison, WI). The plate was placed at −80°C for 30 minutes. The plate was thawed, and luciferase activity measured using luciferase assay kit (Promega). Luciferase activity was normalized to beta-galactosidase activity.
TAT-fusion protein production
The TAT-mFoxn1 wildtype expression vector was previously described40. With this plasmid, the T7 sequence of the pET45b vector was modified by adding 4 nucleotides (GAGA) to improve protein expression41. Site-directed mutagenesis was used to create the c.961C>A nucleotide substitution, introducing the p.H321N amino acid substitution. The plasmids were transduced into BL21-DE3 pLysS bacteria. Colonies were grown O/N in Luria broth containing ampicillin (50 μg/ml). The O/N cultures were used to inoculate 500 ml of LB using a 1/100 dilution of bacteria. After 4 hrs. at 37°C, IPTG was added to 0.5 mM final, and the bacteria were cultured for an additional 5 hrs. at 30°C. The cells were pelleted, washed once in PBS, and the pellet frozen at −80°C. The pellet was thawed and resuspended in 45 ml of 50 mM Tris-Cl, pH8.0, pH 8.0, 1 mM 2-ME supplemented with the protease inhibitors, aprotinin, leupeptin, phenyl methyl sulfonyl fluoride and benzamidine. DNase was added at 10 μg/ml along with lysozyme at 20 μg/ml. The cell lysate was incubated at RT for 30 min, and subsequently processed with an Emulsiflex C5 high pressure homogenizer for 6–8 min at 600 psi, with samples kept on ice. TX-100 was added to a final concentration of 1% (v/v). The lysate was cleared by centrifugation at 7500 g for 30 min. The residual supernatant was applied to 4 ml of Ni2+-NTA beads according to the manufacturers’ instructions (Qiagen Corp., Germantown, MD). After washing with 20 columns of 50 mM NaH2PO4, 0.3 M NaCl, 20 mM Imidazole, the proteins were batch eluted sequentially with 3 column volumes of 0.75, 0.1, 0.2 and 0.3 M imidazole in the wash buffer. Aliquots of the samples were resolved by SDS-PAGE and Coomassie staining was used to visualize TAT-Foxn1 (Supplemental Fig. 5). A 90–95% pure source of TAT-Foxn1 was detected in the 0.15 and 0.2 M imidazole eluates. These fractions were pooled and concentrated with a Pierce protein concentrator and buffer exchanged with 50 mM NaH2PO4, 0.3 M NaCl, pH 8.0. The purified protein (0.1–3 μg) was added to the cultures.
Reaggregate thymus organ cultures (RTOC)
Normal and hypoplastic fetal thymic lobes were isolated between e12.0–12.5 or e13–13.5 gestational ages, as indicated in the figure legends. The tissues were collected in thymus organ culture media (TOC). TOC comprised RPMI containing 10% fetal calf serum, and supplemented with HEPES, l-glutamine, sodium pyruvate, penicillin, streptomycin, 5 × 10−5 M 2-mercaptoethanol and non-essential amino acids. Pooled or individual lobes were washed with PBS. Embryonic day13–13.5 lobes were processed as described42. For e12.-12.5 thymus lobes, the tissues were trypsinized for 1–2 min in 0.25% trypsin, 0.02% EDTA at 37°C. Digestions were stopped by washing in TOC media. The cells were resuspended in volumes <250 μl/20 lobes and an aliquot counted. The cells were stained with antibodies specific for TECs (EpCam-FITC) and mesenchymal cells (Pdgfra-PE). After washing, the cells were sorted into three populations; TECs, mesenchymal and a pool of the remaining cell types; early thymus progenitors (ETPs), endothelial cells, macrophages (EpCAM−Pdgfra−). Sorting was performed on an Aria Zelda FACS machine. RTOC was performed by reaggregating the 3 cell populations, EpCAM+ cell (~20–40%), Pdgfra+ (~30–50%) and EpCAM− Pdgfra− (~30%) in a 1.5 ml microfuge tube. In certain experiments, specific cell subsets were substituted from the hypoplastic lobes with those from controls. For TAT transduction assays, between 0.1–3 μg of pure protein was added to the TECs and incubated for 30 min prior to reaggregations. For reaggregating the cells, the pool of different cell subsets was centrifuged consecutively for 5’ and 10’ at 1000 rpm and 2000 rpm, respectively. After the 2nd spin, the supernatant was aspirated until 2–4 μl of the aggregated cell pellet remained. This was placed on ice for 10 minutes. After gently dispersion, the mixture was drawn into a pulled glass pipette and placed as a single drop onto a Millipore nitrocellulose filter, sitting on top of a foam sponge (Gelfoam; 2 mm thick) in a 6-well tissue culture dish. The reaggregates were cultured for 10 days at 37°C in a 7.5% CO2 environment. The cells were processed for flow cytometry using previously described procedures3.
Statistics.
Statistical analyses were performed using GraphPad Prism version 9.4.1 (GraphPad Inc.). Differences between 2 groups were evaluated using two-tailed Student’s t tests. A P value of less than 0.05 was considered statistically significant. Ns denotes not significant. For comparisons between multiple samples, one-Way ANOVA was used. Since certain Foxn1 mutant thymuses had significantly lower cell numbers, Brown-Forsythe and Welch tests were applied.
RESULTS
Functional characterization of diverse human FOXN1 variants reveals benign and loss and gain-of-function consequences
The clinical impact of FOXN1 mutations in patients include the classic Nude/SCID phenotype, SCID without alopecia, or a mild T cell lymphopenia that normalizes over time23, 31, 43. An expanding number of FOXN1 variants are now reported, with 400 listed in ClinVar (Fig. 1A, Table 1). Presenting in patients as either autosomal recessive, single allelic or compound heterozygous variants, these FOXN1 mutations result in diverse amino acid substitutions, truncations, internal deletions and/or frameshifts in the protein (Fig. 1A). The first set of human FOXN1 variants studied corresponded to those in patients with low peripheral T cell counts, reported by the several groups listed (Table 1)23, 28, 31, 44. We undertook a systematic approach to define the impact of these diverse FOXN1 variants on protein function with luciferase reporter assays. The human FOXN1 variants were initially introduced into the murine cDNA (designated as Foxn1) as each amino acid impacted was conserved between these species. Cloning into the murine cDNA was first done as we had also developed several mouse lines genocopying the human variants31. Expression vectors containing Foxn1 variants were transfected into HeLa cells along with the beta5t-luciferase reporter construct, a well-defined transcriptional target of Foxn1 (Fig. 1B)10. Constructs containing beta-galactosidase were included for normalization. Statistically significant functional differences were noted with the variants relative to the wild-type Foxn1 control, whose activity is denoted with the dotted line (Fig. 1B). Among these were complete- and partial- loss-of function variants (Fig. 1B, Table 1). The complete loss of activity was defined as <10% normal activity (Table 1). The loss of activity for several variants concurred wherein the mutation led to a reduced protein size, seen by Western blotting with anti-Foxn1 specific antibodies (Fig. 1C, upper panel; Supplemental Fig. 1B). The same membranes were also probed with anti-Gapdh antibodies, confirming similar protein extractions from the cells (Fig. 1C, lower panel; Supplemental Fig. 1B). Foxn1 p.D313Tfs12 had the lowest molecular mass of 34 kDa due to a frameshift causing a premature stop codon prior to the DNA binding domain (Fig. 1C). Foxn1 p.W363Cdel5aa, lacking a 5 amino acid stretch at the end of the DNA binding domain, had a molecular mass slightly less than the wild-type control (75 kDa) (Fig. 1C). Many FOXN1 variants had normal transcriptional activities (Fig. 1B, Table 1). In addition, two gain of function variants were found, FOXN1 p.H321R and p.E359K, defined as activities >110% wildtype values. Foxn1 p.H321R exhibited a 2-fold higher activity compared to the wildtype control (Fig. 1B, Table 2). For comparative purposes, we also tested several previously designated benign variants (p.R69C, p.A121V, p.230R, p.G238D, p.A555V) along with some listed as of “uncertain significance” (p.H321N, p.L325P, p.350L) in ClinVar (Table 1, Supplemental Fig. 1A). The results from such transcriptional reporter assays enabled the classification of numerous variants into either pathogenic or benign (Table 1 and Supplemental Table 1).
Human FOXN1 frameshift variants have a species-specific dominant negative function
Most FOXN1 mutations listed in ClinVar are monoallelic, with only 4 individuals to date reported with compound heterozygous mutations23, 28, 31. A recent study identified 3 individuals with the same FOXN1 c.1370delA frameshift variant with a dominant negative functional effect44. Interestingly, there are a significant number of frameshifts lised in ClinVar, some of which are listed in Table 1. The FOXN1 c.1370delA frameshift mutation creates a p.H457Pfs93 change, resulting in a de novo amino acid sequence of 92 amino acids ending with a premature stop codon at amino acid 550 (Supplemental Fig. 1A–B). To determine how different single allelic human FOXN1 frameshift variants impacted the function of the wildtype protein, the transcriptional assays were done with wildtype human FOXN1 tested in the presence of increasing amounts of the different variants. Wildtype human FOXN1 activity was suppressed to less than 5% normal levels when equivalent amounts of p.H457Pfs93 were expressed, consistent with a dominant negative consequence (Fig. 2A). In contrast, the same mutation created in murine Foxn1 only resulted in a 40% inhibition of wildtype Foxn1 activity (Fig. 2A). Of note, the murine c.1370delA was considered a partial dominant negative per a previous report44. The species comparisons were repeated with the FOXN1 c.1465delC frameshift deletion (p.Q489Rfs60). Human FOXN1 p.Q489Rfs60 acted as a dominant negative against the wildtype, when transfected at a 1:1 ratio (Fig. 2A). Murine Foxn1 p.Q489Rfs16 only acted as a competitive inhibitor, suggesting a species selective difference (Fig. 2A).
To define the basis of the species-specific dominant negative activity, an additional series of human FOXN1 frameshift variants were cloned and tested in the reporter assays. Among the variants tested were FOXN1 c.1201–1216del (p.P401Afs144), c.1293del (p.P432fs118), c.1364–1367del (p.Y455Cfs94), c.1392–1401del (p.P465Rfs82), c.1418del (p.P473Hfs77), c.1584delC (p.D528fs20) and c.907delG (p.E303Sfs247) (Fig. 2B, Supplemental Fig. 2A–D). Additionally, a premature stop codon (FOXN1 c.1138C>T, p.E379stop) was introduced immediately after DNA binding domain to determine the minimal length of protein needed for a dominant negative effect. All the human FOXN1 frameshift variants tested were dominant negative except for c.1584delC (p.D528fs20) and p.E379stop (Supplemental Fig. 2E). None of the equivalent murine Foxn1 variants were dominant negative. The human FOXN1 frameshift variants with dominant negative activity formed de novo amino acid sequences of varying lengths (Supplemental Fig. 2A). To determine if this de novo sequence was responsible for the dominant negative activity, a distinct frameshift was engineered in human FOXN1 to change the de novo coding sequence. This was done by introducing a 2-nucleotide deletion at c.1465–1466 and adding a new nucleotide insertion downstream to maintain the same stop codon position at amino acid 550 while changing the codon usage (Human FOXN1 p.Q489Gfs60) (Fig. 2C). Human FOXN1 p.Q489Gfs60 showed dominant negative activity, revealing that the de novo sequence itself was not causal to the phenotype. Consequently, various FOXN1 frameshifted constructs were aligned to characterize commonalities among the dominant negatives (Fig. 2D). This alignment revealed that the dominant negative effect required the expression of a 549 amino acid protein along with missense mutations at several Asp amino acid acids, previously shown essential for transactivation activity (Fig. 2E)18. The murine Foxn1 frameshifted variants were distinct as an earlier stop codon was formed at amino acid position 505. To assess whether the species-selective dominant negative function was related to the length of the frameshifted protein, a human FOXN1 c.1370delC (p.H457Pfs93) dominant negative construct was titrated against wildtype murine Foxn1. Human FOXN1 p.H457Pfs93 construct did not act as a dominant negative against murine Foxn1 (Supplemental Fig. 2F). Taken together, these results establish that multiple human FOXN1 frameshift variants form a species-specific dominant negative. This requires an intact DNA binding domain followed by a truncated protein that forms missense mutations at the key Asp residues responsible for transactivation activity. Such results suggest that any single allelic FOXN1 frameshift variant formed after the DNA binding domain and prior to the transactivation region can be considered a dominant negative.
Nuclear localization sequence exists within the DNA binding domain of FOXN1
Wildtype FOXN1 is entirely nuclear in location, and due to its molecular mass (75 kDa) can only enter by a nuclear localization signal (NLS). However, no reported NLS has been identified in FOXN1. Given the many FOXN1 missense variants with amino acid substitutions throughout the protein, we screened 19 different variants for those with potential effects on the nuclear distribution of the protein (Supplemental Fig. 3A–B). For this, HeLa cells were transfected with selected murine Foxn1 variants that matched the human nucleotide mutations. The nuclear versus cytosolic distribution of the Foxn1 variants were compared by immunohistochemistry (IHC). An antibody specific for the NH2-terminal domain enabled an analysis of every variant. DAPI staining was used to delineate the nucleus while FITC-labeled Phalloidin visualized the actin cytoskeleton (cytosol). Four Foxn1 variants severely or slightly impacted nuclear localization; p.D313Tfs, p.R320W, p.L325P and p.C328R (Fig. 3A–B, Supplemental Figure 3A–B). All these variants were localized before or within the DNA binding domain (Fig. 1A). Foxn1 p.D313Tfs, which lacks the DNA binding and transactivation domains, was entirely cytosolic in all transfected cells. Foxn1 p.R320W, p.L325P and p.C328R had a cytosolic localization in smaller percentage of transfected cells (Fig. 3A–B). Foxn1 p.P430S, which lies outside the DNA binding domain, retained a nuclear location. The results suggest that a nuclear localization sequence (NLS) exists within the highly conserved DNA binding domain (Fig. 3C). To localize the sequence(s) mediating nuclear localization, different subregions of Foxn1 (aa 271–455, aa 271–378, aa 271–344, aa 271–378, aa 314–344, aa 331–395, and aa 415–497) were cloned, in-frame, to the 3’ end of the gene encoding pyruvate kinase (Fig. 3D, Supplemental Fig. 4A). Pyruvate kinase is a cytosolic protein that can redistribute to the nucleus if a NLS is added to its COOH-terminal end, such as that present in sec1345. Pyruvate kinase-Sec13 was localized in the nucleus (Fig. 3E, Supplemental Fig. 4C). A pyruvate kinase fusion protein containing the Foxn1 region encompassing amino acids 271–455 also entered the nucleus (Supplemental Fig. 4). Consequently, shorter regions were tested, revealing that an amino acid stretch between 314–344 was sufficient to enable nuclear entry (Fig. 3E, Supplemental Fig. 4C). A pyruvate kinase fusion protein containing amino acids 331–395 had only partial entry while 414–497 of Foxn1 did not enter the nucleus, and instead exhibiting a cytosolic distribution (Fig. 3E, Supplemental Fig. 4). Western blotting with anti-myc antibodies confirmed expression of the various pyruvate kinase fusion proteins, with pyruvate kinase-Sec13 (aa 1–322) had a molecular mass of 75 kDa while those containing various sequences of Foxn1 had sizes ranging from ~48–55 kDa (Supplemental Fig. 4). Anti-Gapdh Western blotting confirmed similar protein loading (Supplemental Fig. 4). These findings confirm the existence of a NLS within the DNA binding domain of Foxn1. A NLS typically contains a cluster of basic amino acids (Arg, Lys) that enable nuclear entry via importins46. Sequence alignments with human versus mouse FOXN1, which are 98% sequence identical in the DNA binding domain, and the amino acid stretch between 314–344 in human FOXN1 and its paralog, FOXN4, revealed several candidate basic amino acids (Fig. 3F–G). We substituted numerous charged amino acids with Ala residues at varying locations in human FOXN1 (Fig. 3H). Notably, these variants eliminated the functional activity of the protein in luciferase assays (Supplemental Fig. 4D). The impact of these substitutions on nuclear localization was determined by IHC. Combinatorial substitutions of what we had considered the strongest candidates for mediating nuclear entry had a minimal impact on the FOXN1’s location (p.K331A/N334A/K335A) (Fig. 3H). In contrast, human FOXN1 p.K316A/R320W/K327A protein had a higher percentage of cells with just a cytosolic distribution, confirmed by quantitation (Fig. 3H–I). Taken together, these findings reveal that the human FOXN1 NLS resides within the DNA binding domain, with at least 3 basic amino acids involved in nuclear localization.
Selected compound heterozygous Foxn1 mutations reduce the efficiency of thymopoiesis during embryogenesis
We reported previously on two patients with compound heterozygous FOXN1 mutations who each presented with a T−/loB+NK+ phenotype with normal hair extrusion (Table 1)31. Pt.1 had two allelic variants, forming distinct proteins, FOXN1 p.D313fs169 and p.W363Cdel5aa. Pt.2 had c.1288C>T and c.1465delC on different alleles, resulting in the formation of two proteins, p.P430S and p.Q489Rfs60, respectively (Fig. 4A). We compared the impact of these various compound heterozygous mutations on the transcriptional activities of the human and murine wildtype proteins. In the case of Pt.1, the 35% normal activity of human FOXN1 p.W363Cdel5aa was reduced ~50% when co-expressed at a 1:1 ratio with p.D313fs169 (Fig. 4B)31. For Pt.2, human FOXN1 p.Q489Rfs functioned as a dominant negative against p.P430S (Fig. 4B). Introducing the same mutations into the mouse cDNA revealed that the murine Foxn1 p.D313Tfs12 had a minimal impact on the p.W363Cdel5aa protein (Fig. 4C). Matching the variants in Pt.2, the murine Foxn1 p.Q489Rfs only acted as a competitive inhibitor against p.P430S (Fig. 4C).
We next assessed the impact of the Pt.2 FOXN1 mutations on thymopoiesis. For this, two new mouse lines were developed using CRISPR/Cas9 technologies (Fig. 4D). The mice (Foxn1Wt/P430S and c.Foxn1Wt/Q489Rfs) were intercrossed to obtain various single allelic and compound heterozygous Foxn1 lines along with littermate controls. The Foxn1P430S/Q489Rfs mice that genocopied Pt.2 phenotypically resembled wildtype and heterozygous littermate controls, as evidenced by their normal body size along with the presence of fur and whiskers (Fig. 4E). Homozygous mutant mice with a complete loss of function variant (Foxn1Q489Rfs/Q489Rfs) had a nude and small size phenotype (Fig. 44D–E)18, 31. Foxn1Wt/P430S and Foxn1Wt/Q489Rfs mice were interbred to determine the impact of the compound heterozygous Foxn1 variants on thymopoiesis. In adult mice, thymuses from Foxn1P430S/Q489Rfs compound heterozygous mice closely resembled littermate controls and those with single allelic mutations (Foxn1Wt/P430S, Foxn1Wt/Q489Rfs ), evidenced by similar thymus weights, cellularity, and comparable percentages of DN, DP, SP and DN1-DN4 thymocyte subsets (Supplemental Fig. 5A–E). Only the Foxn1Q489Rfs/Q489Rfs nude mice had significantly reduced numbers and percentages of thymocyte subsets at the adult stages.
Pt.2 had a T−/loB+NK+ phenotype within the first year of life. Moreover, many patients with single allelic FOXN1 mutations have low T cell numbers at birth that normalize over time23. Given these observations, we considered the possibility that a thymus hypoplasia might be more evident during embryonic stages. This was previously revealed in the Foxn1Wt/nu heterozygous mice relative to controls23. To assess this, Foxn1Wt/P430S and Foxn1Wt/Q489Rfs mice were intercrossed, and thymuses isolated from embryonic day 16–16.5 (e16–16.5). Thymus cellularity along with DP thymocyte and TEC percentages were analyzed. Five different genotypes were compared with the diverse intercrosses, Foxn1Wt/Wt, Foxn1Wt/P430S, Foxn1Wt/Q489Rfs, Foxn1Q489Rfs/Q489Rfs and Foxn1P430S/Q489Rfs. Thymuses from the compound heterozygous mice matching Pt.2 (Foxn1P430S/Q489Rfs) had a slightly lower cell number compared to the single allelic variants, including a reduced DP percentage, and a slightly higher percentage of TECs (EpCam+) (Fig. 5A–C). The mild phenotypes in the mice genocopying Pt.2 were in sharp contrast to the severe block in T cell development noted with the autosomal recessive Foxn1Q489Rfs/Q489Rfs line (Fig. 5A–C). For example, the Foxn1Q489Rfs/Q489Rfs autosomal recessive mice had a 100-fold reduction in cellularity, with no DP cells evident (Fig. 5A–C). In the Foxn1P430S/Q489Rfs embryonic thymuses, the higher percentage of TECs was consistent with a delayed or reduced expansion of the DP thymocytes. Some of the statistically significant different suggested that the mice genocopying Pt.2 could be more severe at earlier developmental stages. Consequently, we next examined thymopoiesis at e13–13.5. The thymuses from the Foxn1P430S/Q489Rfs embryos were significantly smaller at this early developmental stage (Fig. 5D). Two representative examples are shown (Fig. 5D, e.g., 1 and 2). To identify the differences among the embryonic thymuses, the percentage of early thymus progenitors (ETPs) and mesenchymal and thymic epithelial cells was determined following flow cytometry using antibodies specific for the cell surface proteins CD117, CD45 (ETPs), Pdgfra (Mesenchymal cells) and EpCam (TECs) (Fig. 5E–F). Comparing the numbers of cells and the percentages of the cell subsets revealed that the Foxn1P430S/Q489Rfs had statistically significant reductions in the total cell number and percentage of ETPs (Fig. 5G). The percentage of TECs and mesenchymal cells was not significantly different, implying a functional defect in the TECs (Fig. 5G). Taken together, these findings suggest the compound heterozygous Foxn1P430S/Q489Rfs mutations cause a transient hypoplasia that corrects post-natally, consistent with the TloB+NK+ presentation revealed in many patients with single allelic FOXN1 variants, including those with dominant negative activities23.
Thymopoiesis in autosomal recessive FOXN1 mutant embryonic thymuses assays is restored with a TAT-Foxn1 wildtype fusion protein
Assessing the impact of FOXN1 mutations on thymopoiesis currently necessitates the development of mouse knock-in lines, which can take 1–2 years to characterize (Fig. 4). To develop a more rapid screening system to define the impact of the FOXN1 variants on thymopoiesis, we capitalized on our success with fetal thymus organ cultures (FTOC) and reaggregated thymus organ cultures (RTOC)42. At e11.5 of embryogenesis (week 7 in humans), the thymus anlage is formed and Foxn1 expression is initiated7. Foxn1 expression is not needed to reach this stage of development8. However, as the thymus remains too under-developed to isolate, we used e12–12.5 fetal thymuses. To eliminate endogenous Foxn1 functions, Foxn1Wt/T313fs mice were intercrossed to obtain embryos with varying genotypes, including those with autosomal recessive Foxn1 mutations (Foxn1D313Tfs/D313Tfs). At e12–12.5, thymuses from the Foxn1 mutant embryos were much smaller that littermate controls (Fig. 6A, dotted area). To develop RTOC procedures that could be used with the hypoplastic tissues, we selected normal thymuses as a source of different cell subsets. Control thymuses from e12–12.5 were processed into single cell suspensions and stained with mAbs detecting TECs (EpCam+) and mesenchymal cells (Pdgfra+). This enabled a flow sorting of cells into 3 distinct cell subgroups, mesenchymal (Group I), TECs (Group 2) and a remaining pool of early thymus progenitors (ETPs), endothelial cells, and macrophages (Group III) (Fig. 6B). Reaggregate thymus organ cultures were prepared using these cell subsets along with cells prepared from pooled hypoplastic thymuses (Fig. 6C)47. Cells from normal and hypoplastic thymuses were reaggregated, overlayed onto membranes, and cultured for 10-days to monitor thymocyte growth and T cell differentiation (Fig. 6C). Reaggregates of control embryonic thymuses expanded in size and supported CD4+CD8+ T cell development (Fig. 6D). Equivalent numbers of reaggregated cells using Foxn1D313Tfs/D313Tfs embryonic thymuses failed to expand and CD4+CD8+ thymocytes development did not occur (Fig. 6D). To correct for the functional deficiency of Foxn1 in Foxn1D313Tfs/D313Tfs reaggregates, a TAT-FOXN1 fusion protein was purified from bacteria and added to the cells (Supplemental Fig. 6A–B). The TAT domain enables Foxn1 entry into TECs, first revealed by intrathymic injections of the protein40. We confirmed that pure TAT-Foxn1 wildtype protein entered primary embryonic e12–12.5 TECs by Western immunoblotting with anti-Foxn1 mAbs using lysates from ~20,000 transduced cells (Supplemental Fig. 6C–D). Since our prior experiments suggested that the Foxn1-deficient thymuses had limited numbers of ETPs, reaggregates of the Foxn1T313fs/T313fs embryonic thymuses were supplemented with Group III cells (with ETPs) from normal tissues. In such reaggregates, the TAT-Foxn1 wildtype protein supported thymus growth and CD4+CD8+ T cell development (Fig. 6D). In one RTOC culture, the TAT-Foxn1 wildtype protein enabled equivalent cell expansion as the normal controls (Fig. 6E). This was not always seen, an issue we suggest is due to the technical challenges of aggregating very small numbers of cells. Also noteworthy, the addition of Group III cells alone supported some growth for the Foxn1D313Tfs/D313Tfs reaggregates (Fig. 6D). We hypothesize this is facilitated by the presence of endothelial cells that retain TEC progenitor potential. In future directions, we will use primary TECs from Foxn1D313Tfs/D313Tfs embryonic thymuses that are expanded in culture in the presence/absence of TAT-Foxn1 followed by reaggregate cultures with normal ETPs.
Our goal was to define the impact of selected TAT-Foxn1 variants on thymopoiesis. To address this, we next assessed whether a Foxn1 variant could antagonize normal RTOC growth. For this purpose, the p.H321N mutation was engineered into the TAT-Foxn1 wildtype protein (Supplemental Fig. 6A–B). RTOCs of normal e12–12.5 thymuses cell subsets were cultured either in media alone, with the TAT-Foxn1 wildtype protein, or with TAT-Foxn1 H321N (Supplemental Fig. 7). In such RTOCs, TAT-Foxn1 H321N variants had variable suppressive effects on thymus growth (Supplemental Fig. 7C). This variability likely depends on the variant selected, as Foxn1 p.H321N does not bind to DNA as effectively as the wildtype protein16. This suggests that TAT-Foxn1 variants may be more effectively screened for partial or complete restoration of organoid growth with the Foxn1T313fs/T313fs hypoplastic thymuses. Overall, our proof of concept experiments reveal a novel methodology to evaluate the impact of human FOXN1 variants on thymopoiesis. Such RTOC assays involve a 10-day culture period, which will enable a rapid screen for thymopoietic impacts of Foxn1. The development of primary TEC cultures and bio banked ETPs will likely make this technology more widely available for investigators.
DISCUSSION
Several human diseases result in a thymus hypoplasia due to genetic mutations that impact the stromal cell populations3, 42, 48. Humans with autosomal recessive, single allelic and compound heterozygous FOXN1 mutations can have impaired TEC functions, distinct from the mesenchymal cell problems occurring in 22q11.2 deletion syndrome patients (DiGeorge)20, 24, 42. There are ~400 human FOXN1 variants listed in the ClinVar database, with the clinical impacts of these variants designated as benign, likely benign, uncertain significance, pathogenic, or conflicting interpretations of pathogenicity. To improve on our understanding of how these varied mutations affect protein functions, we characterized 35 of the FOXN1 variants using transcriptional reporter assays, revealing those with benign, partial- and complete- loss and gain- of function activities (Tables 1, Supplemental Table 1).
Interpreting the consequences of the diverse FOXN1 variants on human thymopoiesis necessitates several considerations. First, knowledge of the allelic presentation is required. For autosomal recessive FOXN1 variants, the functional impact directly correlates with the level of activity revealed in transcriptional reporter assays (Fig. 1). For single allelic variants, the functional consequence depends on how a particular variant influences the wildtype allele. For example, human FOXN1 frameshift variants resulting from nucleotide deletions after the DNA binding domain and prior to the transactivation domain form dominant negatives (Fig. 2, Supplemental Fig. 2). Variants fitting this category include FOXN1 c.1201–1216del, c.1205delC, c.1206delT, c.1293del, c.1315delC, c.1364–1367del, c.1370delA, c.1392–1401del, c.1418delC, c.1465delC, along with the others listed in the ClinVar database containing single nucleotide deletions between c.1135-c.1538 (Table 1). Noteworthy, the dominant negative effect is human specific as similar frameshifts created in the murine cDNA only act as competitive inhibitors. A key feature of the human dominant negative activity is the elimination of the acidic amino acids needed for transactivation functions. The dominant negative activity of human FOXN1 is not completely understood but may relate to protein dimerization and/or protein aggregation changes that alter the nuclear distribution of the wildtype protein44. Additionally, nucleotide deletions prior to the region encoding DNA binding domain are likely benign when affecting only one allele. This is because the protein that is formed would lack the DNA binding domain and lack the capacity to enter the nucleus.
Among the diverse hFOXN1 variants characterized were several that reduced the nuclear localization of the protein (Fig. 3; p.R320W, p.L325P and p.C328R). This enabled us to identify a NLS within the highly conserved DNA binding domain (aa 314–344). Using homology searches with diverse NLS sequences, a strong sequence homology was evident between amino acids 331–344 of FOXN1 with the corresponding region in FOXN4, a paralog. In addition, some sequence similarity with the NLS of Pho4 was found46, 49. Our studies also revealed gain of function consequences for two unrelated FOXN1 variants (FOXN1 p.H321R, p.E359K) revealed. FOXN1 p.H321R had a 2-fold increase in activity. We suggest introduction of a positively charged Arg residue may have increased the DNA binding affinity.
For patients with single allelic variant or compound heterozygous mutations, co-titration experiments with luciferase reporter assays provides an excellent experimental strategy to define how one allelic variant impacts either the wildtype FOXN1 or second allelic variant. For example, any FOXN1 variants that have frameshifts or stop codons eliminating the DNA binding domain (e.g., FOXN1 p.D313fs) or missense mutations impacting amino acids required for DNA binding (e.g., FOXN1 p.R320W) have weak impacts on the transcriptional activity of the normal allele16. In vivo, the impact of such single allelic FOXN1 variants may be even less severe if the mRNA undergoes non-sense mediated decay. This is difficult to assess in transfected cells as the expression vectors enforce mRNA over-expression, which may override normal decay processes. It is also important to note that most TEC cell lines lose expression of the endogenous FOXN1, suggesting that co-transfection experiments still remain more informative. The best approach for testing the RNA stability is to generate knock-in mice and/or create cell lines wherein the mutation is introduced into the genome. We have used this technology to characterize two distinct Foxn1 compound heterozygous mice genocopying Pt.1 and Pt.2, respectively. In our previous report, the Foxn1 mutant mice genocopying Pt. 1 matched the T−B+NK+ phenotype with normal hair31. In the case of Pt.2, the resulting compound heterozygous Foxn1 mutations introduced in mice (Foxn1P320S/Q489Rfs) resulted in a relatively mild and transient hypoplasia of the thymus that was very pronounced during embryonic stages. Per clinical information, Pt.2 had low TRECs at birth, and consequently, a peripheral T cell lymphopenia (~100 CD3+ T cells/μl)31. The species selective differences we described between human and mouse FOXN1 may partly explain the less severe impact of the Pt.2 mutations genocopied in mice. First, murine Foxn1 is more active transcriptionally than human FOXN1, even when both cDNAs have been engineered with the same position of the FLAG epitope and their expression driven by the same expression vector (Supplemental Fig. 2G). Second, the human FOXN1 Q489Rfs in Pt.2 is a dominant negative, attenuating the function of the wildtype allele. It does not function as a dominant negative in mice nor in transcriptional reporter assays. Third, human FOXN1 can form dimers to mediate transcriptional functions44. It remains unclear if murine Foxn1 forms such dimers. Finally, we speculate that epigenetic modifications in some TEC cells in the expanding post-natal thymus in human could undergo clonal selection since higher expression of the wildtype FOXN1 allele may provide a growth advantage. This could explain the improved T cell numbers that occurs over time for some patients with single allelic FOXN1 variants23. Such increased T cell output should preclude an early decision for a thymus transplant in a patient, since a delay permits more careful monitoring of T cell recovery23, 48, 50.
In the current manuscript, a new experimental strategy was developed to more rapidly define the impact of selected FOXN1 variants on thymopoiesis. Reaggregate thymus organ cultures were modified by using embryonic thymuses lacking a functional Foxn1 gene (Foxn1D313Tfs/D313Tfs) as a source of Foxn1-deficient TECs. Supplementing these RTOCs with purified TAT-Foxn1 fusion proteins revealed restoration of thymopoiesis within a 10-day culture. This strategy also required the addition of normal ETPs, as these cell were limiting in the hypoplastic thymuses, even when the latter were pooled to achieve cell numbers equivalent to controls. This need is consistent with the Foxn1D313Tfs/D313Tfs TECs expressing reduced chemokines, particularly Ccl25, which recruits ETPs31. Some additional modifications we are testing includes biobanking the various cell subsets needed for RTOC assays. Expanding the TECs in defined culture media prior to the biobanking would override the extreme cell number limitation currently faced with the Foxn1 mutant TECs. Such changes would enable high-thru put screening strategies to define the impact of the many FOXN1 variants not yet studied.
In summary, a combination of in vitro functional assays, imaging analyses, mouse lines genocopying patients, and reaggregate thymus organ cultures establish the impact of human FOXN1 variants on thymopoiesis. These approaches have yielded key insights into the impact of single allelic, compound heterozygous and autosomal recessive FOXN1 mutations on T cell development in humans. Such information will help select appropriate clinical interventions for patients with diverse thymic stromal cell deficiencies leading to in-born errors of immunity.
Supplementary Material
Key Messages.
Human FOXN1 variants impact T cell development by modulating transcriptional activity, nuclear localization, DNA binding, or form dominant negatives
FOXN1 mutations have distinct consequences on thymopoiesis when present as autosomal recessive, compound heterozygous or single allelic variants
ACKNOWLEDGEMENTS
We appreciate the technical support from Alyssa Guzman, Kaitlyn Hornfischer, and Angela Mobley who were extremely helpful in perfecting flow sorting for the recovery of small numbers of cells from embryos (UT Southwestern Medical Center flow cytometry core). John McAnally from the Eric Olson lab at UT Southwestern Medical Center gave excellent technical insights into applying small volumes of cell aggregates onto membranes for RTOCs. We also thank Dr. Ivan Chinn (Baylor College of Medicine, Houston, TX), Dr. Britt Johnson (Invitae Corp., San Francisco, CA) and Justyne Ross (U. North Carolina, NC) for their helpful discussions and assistance in defining the impact of human FOXN1 variants.
This work was supported by an NIH grant (NvO, AI42953), UT Southwestern Medical Center Internal Funds (NvO, Beecherl) and the Jeffery Modell Foundation (CAW)
Abbreviations:
- AR
Autosomal recessive
- DN
Dominant negative
- FTOC
Fetal thymic organ cultures
- FOXN1
Forkhead Box N1
- OMIM
Online Mendelian inheritance in man
- NLS
nuclear localization signal
- RTOC
Reaggregate thymus organ cultures
- SCID
Severe combined immunodeficiency
- TECs
thymic epithelial cells
- TCR
T cell receptor
- TRECs
T cell receptor excision circles
REFERENCES
- 1.Klein L, Kyewski B, Allen PM, Hogquist KA. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat Rev Immunol 2014; 14:377–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dixit VD. Thymic fatness and approaches to enhance thymopoietic fitness in aging. Curr Opin Immunol 2010; 22:521–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bhalla P, Su DM, van Oers NSC. Thymus Functionality Needs More Than a Few TECs. Front Immunol 2022; 13:864777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Alawam AS, Anderson G, Lucas B. Generation and Regeneration of Thymic Epithelial Cells. Front Immunol 2020; 11:858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Vaidya HJ, Briones Leon A, Blackburn CC. FOXN1 in thymus organogenesis and development. Eur J Immunol 2016; 46:1826–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Morimoto R, Swann J, Nusser A, Trancoso I, Schorpp M, Boehm T. Evolution of thymopoietic microenvironments. Open Biol 2021; 11:200383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gordon J, Manley NR. Mechanisms of thymus organogenesis and morphogenesis. Development 2011; 138:3865–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gordon J, Bennett AR, Blackburn CC, Manley NR. Gcm2 and Foxn1 mark early parathyroid- and thymus-specific domains in the developing third pharyngeal pouch. Mech Dev 2001; 103:141–3. [DOI] [PubMed] [Google Scholar]
- 9.Farley AM, Morris LX, Vroegindeweij E, Depreter ML, Vaidya H, Stenhouse FH, et al. Dynamics of thymus organogenesis and colonization in early human development. Development 2013; 140:2015–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zuklys S, Handel A, Zhanybekova S, Govani F, Keller M, Maio S, et al. Foxn1 regulates key target genes essential for T cell development in postnatal thymic epithelial cells. Nat Immunol 2016; 17:1206–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Frantzeskakis M, Takahama Y, Ohigashi I. The Role of Proteasomes in the Thymus. Front Immunol 2021; 12:646209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Newman JA, Aitkenhead H, Gravard A, Rota IA, Handel AE, Hollander GA, et al. The structural basis for forkhead box family specificity revealed by the crystal structure of human FOXN1 in complex with DNA. bioRxiv 2018:428011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Romano R, Palamaro L, Fusco A, Giardino G, Gallo V, Del Vecchio L, et al. FOXN1: A Master Regulator Gene of Thymic Epithelial Development Program. Front Immunol 2013; 4:187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Potter CS, Pruett ND, Kern MJ, Baybo MA, Godwin AR, Potter KA, et al. The nude mutant gene Foxn1 is a HOXC13 regulatory target during hair follicle and nail differentiation. J Invest Dermatol 2011; 131:828–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Schorpp M, Schlake T, Kreamalmeyer D, Allen PM, Boehm T. Genetically separable determinants of hair keratin gene expression. Dev Dyn 2000; 218:537–43. [DOI] [PubMed] [Google Scholar]
- 16.Newman JA, Aitkenhead H, Gavard AE, Rota IA, Handel AE, Hollander GA, et al. The crystal structure of human forkhead box N1 in complex with DNA reveals the structural basis for forkhead box family specificity. J Biol Chem 2020; 295:2948–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Schlake T, Schorpp M, Nehls M, Boehm T. The nude gene encodes a sequence-specific DNA binding protein with homologs in organisms that lack an anticipatory immune system. Proc Natl Acad Sci U S A 1997; 94:3842–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Schuddekopf K, Schorpp M, Boehm T. The whn transcription factor encoded by the nude locus contains an evolutionarily conserved and functionally indispensable activation domain. Proc Natl Acad Sci U S A 1996; 93:9661–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nehls M, Pfeifer D, Schorpp M, Hedrich H, Boehm T. New member of the winged-helix protein family disrupted in mouse and rat nude mutations. Nature 1994; 372:103–7. [DOI] [PubMed] [Google Scholar]
- 20.Vigliano I, Gorrese M, Fusco A, Vitiello L, Amorosi S, Panico L, et al. FOXN1 mutation abrogates prenatal T-cell development in humans. J Med Genet 2011; 48:413–6. [DOI] [PubMed] [Google Scholar]
- 21.Abitbol M, Bosse P, Thomas A, Tiret L. A deletion in FOXN1 is associated with a syndrome characterized by congenital hypotrichosis and short life expectancy in Birman cats. PLoS One 2015; 10:e0120668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chou J, Massaad MJ, Wakim RH, Bainter W, Dbaibo G, Geha RS. A novel mutation in FOXN1 resulting in SCID: a case report and literature review. Clin Immunol 2014; 155:30–2. [DOI] [PubMed] [Google Scholar]
- 23.Bosticardo M, Yamazaki Y, Cowan J, Giardino G, Corsino C, Scalia G, et al. Heterozygous FOXN1 Variants Cause Low TRECs and Severe T Cell Lymphopenia, Revealing a Crucial Role of FOXN1 in Supporting Early Thymopoiesis. Am J Hum Genet 2019; 105:549–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pignata C, Fiore M, Guzzetta V, Castaldo A, Sebastio G, Porta F, et al. Congenital Alopecia and nail dystrophy associated with severe functional T-cell immunodeficiency in two sibs. Am J Med Genet 1996; 65:167–70. [DOI] [PubMed] [Google Scholar]
- 25.Ali RH, Habib R, Ud-Din N, Khan MN, Ansar M, Ahmad W. Novel mutations in the gene HOXC13 underlying pure hair and nail ectodermal dysplasia in consanguineous families. Br J Dermatol 2013; 169:478–80. [DOI] [PubMed] [Google Scholar]
- 26.Godwin AR, Capecchi MR. Hoxc13 mutant mice lack external hair. Genes Dev 1998; 12:11–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Handel AE, Hollander GA. Comment on “Identification of an Intronic Regulatory Element Necessary for Tissue-Specific Expression of Foxn1 in Thymic Epithelial Cells”. J Immunol 2019; 203:2355. [DOI] [PubMed] [Google Scholar]
- 28.Giardino G, Sharapova SO, Ciznar P, Dhalla F, Maragliano L, Radha Rama Devi A, et al. Expanding the Nude SCID/CID Phenotype Associated with FOXN1 Homozygous, Compound Heterozygous, or Heterozygous Mutations. J Clin Immunol 2021; 41:756–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Markert ML, Gupton SE, McCarthy EA. Experience with cultured thymus tissue in 105 children. J Allergy Clin Immunol 2022; 149:747–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kreins AY, Bonfanti P, Davies EG. Current and Future Therapeutic Approaches for Thymic Stromal Cell Defects. Front Immunol 2021; 12:655354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Du Q, Huynh LK, Coskun F, Molina E, King MA, Raj P, et al. FOXN1 compound heterozygous mutations cause selective thymic hypoplasia in humans. J Clin Invest 2019; 129:4724–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Puck JM. Newborn screening for severe combined immunodeficiency and T-cell lymphopenia. Immunol Rev 2019; 287:241–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.van der Spek J, Groenwold RH, van der Burg M, van Montfrans JM. TREC Based Newborn Screening for Severe Combined Immunodeficiency Disease: A Systematic Review. J Clin Immunol 2015; 35:416–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Du Q, Hoover AR, Dozmorov I, Raj P, Khan S, Molina E, et al. MIR205HG Is a Long Noncoding RNA that Regulates Growth Hormone and Prolactin Production in the Anterior Pituitary. Dev Cell 2019; 49:618–31 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Belkaya S, Murray SE, Eitson JL, de la Morena MT, Forman JA, van Oers NSC. Transgenic expression of microRNA-185 causes a developmental arrest of T cells by targeting multiple genes including Mzb1. J Biol Chem 2013; 288:30752–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hoover AR, Dozmorov I, MacLeod J, Du Q, de la Morena MT, Forbess J, et al. MicroRNA-205 Maintains T Cell Development following Stress by Regulating Forkhead Box N1 and Selected Chemokines. J Biol Chem 2016; 291:23237–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Williams KM, Mella H, Lucas PJ, Williams JA, Telford W, Gress RE. Single cell analysis of complex thymus stromal cell populations: rapid thymic epithelia preparation characterizes radiation injury. Clin Transl Sci 2009; 2:279–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Holden P, Horton WA. Crude subcellular fractionation of cultured mammalian cell lines. BMC Res Notes 2009; 2:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.DeFord-Watts LM, Dougall DS, Belkaya S, Johnson BA, Eitson JL, Roybal KT, et al. The CD3 zeta subunit contains a phosphoinositide-binding motif that is required for the stable accumulation of TCR-CD3 complex at the immunological synapse. J Immunol 2011; 186:6839–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Song Y, Su M, Zhu J, Di W, Liu Y, Hu R, et al. FOXN1 recombinant protein enhances T-cell regeneration after hematopoietic stem cell transplantation in mice. Eur J Immunol 2016; 46:1518–28. [DOI] [PubMed] [Google Scholar]
- 41.Shilling PJ, Mirzadeh K, Cumming AJ, Widesheim M, Kock Z, Daley DO. Improved designs for pET expression plasmids increase protein production yield in Escherichia coli. Commun Biol 2020; 3:214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bhalla P, Du Q, Kumar A, Xing C, Moses A, Dozmorov I, et al. Mesenchymal cell replacement corrects thymic hypoplasia in murine models of 22q11.2 deletion syndrome. J Clin Invest 2022; 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Giardino G, Borzacchiello C, De Luca M, Romano R, Prencipe R, Cirillo E, et al. T-Cell Immunodeficiencies With Congenital Alterations of Thymic Development: Genes Implicated and Differential Immunological and Clinical Features. Front Immunol 2020; 11:1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rota IA, Handel AE, Maio S, Klein F, Dhalla F, Deadman ME, et al. FOXN1 forms higher-order nuclear condensates displaced by mutations causing immunodeficiency. Sci Adv 2021; 7:eabj9247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Enninga J, Levay A, Fontoura BM. Sec13 shuttles between the nucleus and the cytoplasm and stably interacts with Nup96 at the nuclear pore complex. Mol Cell Biol 2003; 23:7271–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lu J, Wu T, Zhang B, Liu S, Song W, Qiao J, et al. Types of nuclear localization signals and mechanisms of protein import into the nucleus. Cell Commun Signal 2021; 19:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Blackburn CC, Augustine CL, Li R, Harvey RP, Malin MA, Boyd RL, et al. The nu gene acts cell-autonomously and is required for differentiation of thymic epithelial progenitors. Proc Natl Acad Sci U S A 1996; 93:5742–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bhalla P, Wysocki CA, van Oers NSC. Molecular Insights Into the Causes of Human Thymic Hypoplasia With Animal Models. Front Immunol 2020; 11:830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kaffman A, Rank NM, O’Shea EK. Phosphorylation regulates association of the transcription factor Pho4 with its import receptor Pse1/Kap121. Genes Dev 1998; 12:2673–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yamazaki Y, Urrutia R, Franco LM, Giliani S, Zhang K, Alazami AM, et al. PAX1 is essential for development and function of the human thymus. Sci Immunol 2020; 5:eaax1036. [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.