Phenotypic Mutation 'Longs_peak' (pdf version)
AlleleLongs_peak
Mutation Type missense
Chromosome11
Coordinate53,666,762 bp (GRCm39)
Base Change T ⇒ A (forward strand)
Gene Irf1
Gene Name interferon regulatory factor 1
Synonym(s) Irf-1
Chromosomal Location 53,660,841-53,669,200 bp (+) (GRCm39)
MGI Phenotype FUNCTION: [Summary is not available for the mouse gene. This summary is for the human ortholog.] IRF1 encodes interferon regulatory factor 1, a member of the interferon regulatory transcription factor (IRF) family. IRF1 serves as an activator of interferons alpha and beta transcription, and in mouse it has been shown to be required for double-stranded RNA induction of these genes. IRF1 also functions as a transcription activator of genes induced by interferons alpha, beta, and gamma. Further, IRF1 has been shown to play roles in regulating apoptosis and tumor-suppressoion. [provided by RefSeq, Jul 2008]
PHENOTYPE: Homozygous disruption of this gene leads to reduced CD8+ T cell number and altered response to viral infection and may cause alterations in cytokine levels, CD4+ cell subset homeostasis, blood vessel healing, DNA repair, and susceptibility to induced lymphomas, arthritis and autoimmune encephalitis. [provided by MGI curators]
Accession Number

NCBI RefSeq: NM_008390, NM_001159396, NM_001159393; MGI: 96590

MappedYes 
Amino Acid Change Tryptophan changed to Arginine
Institutional SourceBeutler Lab
Gene Model predicted gene model for protein(s): [ENSMUSP00000019043] [ENSMUSP00000104548] [ENSMUSP00000104550] [ENSMUSP00000122101] [ENSMUSP00000116656] [ENSMUSP00000118314] [ENSMUSP00000114315] [ENSMUSP00000118795]
AlphaFold P15314
PDB Structure INTERFERON REGULATORY FACTOR 1 (IRF-1) COMPLEX WITH DNA [X-RAY DIFFRACTION]
SMART Domains Protein: ENSMUSP00000019043
Gene: ENSMUSG00000018899
AA Change: W247R

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
low complexity region 120 138 N/A INTRINSIC
Predicted Effect probably benign

PolyPhen 2 Score 0.267 (Sensitivity: 0.91; Specificity: 0.88)
(Using ENSMUST00000019043)
SMART Domains Protein: ENSMUSP00000104548
Gene: ENSMUSG00000018899
AA Change: W247R

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
low complexity region 120 138 N/A INTRINSIC
Predicted Effect probably benign

PolyPhen 2 Score 0.267 (Sensitivity: 0.91; Specificity: 0.88)
(Using ENSMUST00000108920)
SMART Domains Protein: ENSMUSP00000104550
Gene: ENSMUSG00000018899
AA Change: W247R

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
low complexity region 120 138 N/A INTRINSIC
Predicted Effect probably benign

PolyPhen 2 Score 0.159 (Sensitivity: 0.92; Specificity: 0.87)
(Using ENSMUST00000108922)
SMART Domains Protein: ENSMUSP00000122101
Gene: ENSMUSG00000018899

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
low complexity region 120 138 N/A INTRINSIC
Predicted Effect probably benign
SMART Domains Protein: ENSMUSP00000116656
Gene: ENSMUSG00000018899

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
Predicted Effect probably benign
SMART Domains Protein: ENSMUSP00000118314
Gene: ENSMUSG00000018899

DomainStartEndE-ValueType
IRF 1 62 2.41e-8 SMART
Predicted Effect probably benign
SMART Domains Protein: ENSMUSP00000114315
Gene: ENSMUSG00000018899

DomainStartEndE-ValueType
IRF 1 114 6.75e-62 SMART
low complexity region 120 138 N/A INTRINSIC
Predicted Effect probably benign
SMART Domains Protein: ENSMUSP00000118795
Gene: ENSMUSG00000018899

DomainStartEndE-ValueType
Pfam:IRF 5 48 4.8e-11 PFAM
Predicted Effect probably benign
Meta Mutation Damage Score 0.2982 question?
Is this an essential gene? Non Essential (E-score: 0.000) question?
Phenotypic Category Unknown
Candidate Explorer Status loading ...
Single pedigree
Linkage Analysis Data
Penetrance  
Alleles Listed at MGI

All Mutations and Alleles(12) : Chemically induced (ENU)(1) Chemically induced (other)(1) Gene trapped(4) Targeted(5) Transgenic(1)

Lab Alleles
AlleleSourceChrCoordTypePredicted EffectPPH Score
IGL01355:Irf1 APN 11 53665187 missense probably benign
IGL01743:Irf1 APN 11 53665277 missense probably benign 0.39
endeka UTSW 11 53663717 missense probably damaging 1.00
Endeka2 UTSW 11 53662148 missense probably damaging 1.00
R0981:Irf1 UTSW 11 53664548 makesense probably null
R1861:Irf1 UTSW 11 53665183 missense possibly damaging 0.65
R2511:Irf1 UTSW 11 53664617 missense probably damaging 1.00
R5828:Irf1 UTSW 11 53666762 missense probably benign 0.27
R6514:Irf1 UTSW 11 53662148 missense probably damaging 1.00
R6986:Irf1 UTSW 11 53664966 missense probably damaging 1.00
R7108:Irf1 UTSW 11 53665238 missense probably damaging 1.00
R7696:Irf1 UTSW 11 53667162 missense probably benign 0.07
R9055:Irf1 UTSW 11 53667196 missense probably benign
Mode of Inheritance Unknown
Local Stock
Repository
Last Updated 2019-09-04 9:37 PM by Anne Murray
Record Created 2018-04-29 12:58 PM by Bruce Beutler
Record Posted 2018-12-18
Phenotypic Description
Figure 1. Longs_peak mice exhibit an increase in the CD4 to CD8 T cell ratio. Flow cytometric analysis of peripheral blood was utilized to determine T cell frequencies. Normalized data are shown. Abbreviations: WT, wild-type; REF, homozygous reference mice; HET, heterozygous variant mice; VAR, homozygous variant mice. Mean (μ) and standard deviation (σ) are indicated.

Figure 2. Longs_peak mice exhibit increased frequencies of peripheral CD4+ T cells in CD3+ T cells. Flow cytometric analysis of peripheral blood was utilized to determine T cell frequency. Normalized data are shown. Abbreviations: WT, wild-type; REF, homozygous reference mice; HET, heterozygous variant mice; VAR, homozygous variant mice. Mean (μ) and standard deviation (σ) are indicated.

Figure 3. Longs_peak mice exhibit reduced frequencies of peripheral CD8+ T cells in CD3+ T cells. Flow cytometric analysis of peripheral blood was utilized to determine T cell frequency. Normalized data are shown. Abbreviations: WT, wild-type; REF, homozygous reference mice; HET, heterozygous variant mice; VAR, homozygous variant mice. Mean (μ) and standard deviation (σ) are indicated.

The Longs_peak phenotype was identified among G3 mice of the pedigree R5828, some of which showed an increase in the CD4 to CD8 T cell ratio (Figure 1) due to increased frequencies of CD4+ T cells in CD3+ T cells (Figure 2) and concomitant reduced frequencies of CD8+ T cells in CD3+ T cells (Figure 3) in the peripheral blood.

Nature of Mutation

Figure 4. Linkage mapping of reduced CD8+ T cell frequency using an additive model of inheritance. Manhattan plot shows -log10 P values (Y-axis) plotted against the chromosome positions of 61 mutations (X-axis) identified in the G1 male of pedigree R5828. Normalized phenotype data are shown for single locus linkage analysis without consideration of G2 dam identity. Horizontal pink and red lines represent thresholds of P = 0.05, and the threshold for P = 0.05 after applying Bonferroni correction, respectively.

Whole exome HiSeq sequencing of the G1 grandsire identified 61 mutations. All of the above anomalies were linked by continuous variable mapping to a mutation in Irf1:  a T to A transversion at base pair 53,775,936 (v38) on chromosome 11, or base pair 5,923 in the GenBank genomic region NC_000077. The strongest association was found with an additive model of inheritance to the CD8+ T cell phenotype, wherein two variant homozygotes and 15 heterozygous mice departed phenotypically from 10 homozygous reference mice with a P value of 3.449 x 10-6 (Figure 3).  

The mutation corresponds to residue 1,008 in the mRNA sequence NM_008390 within exon 9 of 10 total exons.

993 TTTGAACAGTCTGAGTGGCAGCCGACACACATC
242 -F--E--Q--S--E--W--Q--P--T--H--I-

The mutated nucleotide is indicated in red. The mutation results in a tryptophan to arginine substitution at position 247 (W247R) in the IRF1 protein, and is strongly predicted by Polyphen-2 to be benign (score = 0.267).

Illustration of Mutations in
Gene & Protein
Protein Prediction
FIgure 5. Domain organization of IRF1.The two regions to which the IAD2 (pink) have been mapped are shown together as one block. The transactivation domain overlaps the region containing IAD2. L1, L2, and L3 are indicated. The Longs_peak mutation results in a tryptophan to arginine substitution at position 247 (W247R). This image is interactive. Other mutations found in IRF1 are noted. Click on each mutation for more information.

Interferon regulatory factor (IRF)-1 is one of nine members of the IRF family of transcription factors, which regulate the transcription of type I interferons (IFN) and IFN-inducible genes during immune system development, homeostasis and activation by microbes. 

Mouse IRF1 contains 329 amino acids and functions as a transcriptional activator. As in the other IRFs, the N-terminal half of IRF1 (residues 1-113) serves as the DNA binding region, and is characterized by the presence of five tryptophans spaced ten to eighteen amino acids apart in a “tryptophan cluster” (residues 11, 26, 38, 58, and 77) (1). The DNA binding region bears similarity to that of the c-Myb oncoprotein that also contains a tryptophan cluster (2), but not to any other transcription factor classes. IRF family proteins share sequence and structural homology in their DNA binding regions, and all bind to a similar DNA motif (A/G NGAAANNGAAACT) called the IFN-stimulated response element (ISRE) (3) or IFN regulatory element (4), found within positive regulatory domain I and III (PRD I and PRD III) of the IFN-β promoter. 

The C-terminal half of IRF1 contains a nuclear localization signal (amino acids 115-139) (5). A transactivation domain is also reported to exist (amino acids 185-256) (5).  The C-terminal halves of all IRF family members contain either an IRF association domain 1 (IAD1) or an IAD2, with which they bind to IRF other family members, other transcription factors, or self-associate. The IAD1 is approximately 177 amino acids in length, and is conserved in all IRFs except IRF1 and IRF2 (6-8). IAD2 domains are found only in IRF1 and IRF2 (7). In IRF1, the IAD2 has been mapped in vitro using electrophoretic mobility shift (EMSA) and GST pull-down assays to two overlapping regions (amino acids 164-219 or 201-263) (5;7)

The Longs_peak mutation at position 247 of IRF1 is within the transactivation domain and IAD2. 

For more information about Irf1, see the record for Endeka.

Putative Mechanism

IRF1 regulates transcription in response to diverse signals received during development and homeostasis of the myeloid and lymphoid compartments, and upon activation of innate and adaptive immune receptors by microbial infection. In particular, IRF1 is required for the development and function of dendritic cells (DC), granulocytes, NK and NKT cells, CD4+ T cells, and CD8+ T cells. IRF1 functions in innate immune signaling from Toll-like receptor (TLR) 9 to activate a select group of genes. IRF1 also acts as a tumor suppressor, and its inactivation contributes to oncogenesis. For more details on these functions, see the record for Endeka.

The targets of IRF1 continue to be investigated. IRF1 is reported to bind to ISREs in many IFN-inducible gene promoters, such as those for inducible nitric oxide synthase (iNOS) (9), cyclooxygenase-2 (Cox-2) (10), class II transactivator (CIITA) (11), and guanylate-binding protein (GBP) in IFN-stimulated macrophages (12).  Transcription of IRF1 itself is also induced by IFN-β (13). Downstream of TLR9, IRF1 induces IL-12p40, IL-12p40, iNOS, IL-18, and IFN-β (11;14). The target genes of IRF1 responsible for apoptotic responses may include genes encoding Caspase 1 (15), Caspase 7 (16), and TNF-related apoptosis-inducing ligand (TRAIL) (17).

Human IRF1 is located on chromosome 5q31.1 (18), within a region frequently deleted in human leukemias and the preleukemic myelodysplastic syndromes (MDS, OMIM #153550) (19). Of the genes in the 5q31.1 region, only IRF1 was consistently deleted at one or both alleles in thirteen cases of leukemia or myelodysplasia associated with 5q31 abnormalities, providing evidence that IRF1 is the tumor suppressor mutated in these diseases (20). In another study, twelve of fourteen patients with 5q deletions and acute myeloid leukemia or MDS had loss of one allele of IRF1 (21). Loss of an IRF1 allele has also been reported to occur in gastric and esophageal cancers (22;23).

Irf1-/- mice have an increased population of pDC and a selective reduction of the CD8α+ subset of cDC (24). Splenic DC from Irf1-/- mice are impaired in their ability to produce proinflammatory cytokines such as IL-12, but express high levels of IL-10, TGF-β and the tolerogenic enzyme indoleamine 2,3 dioxygenase. Irf1-/- DC have a reduced ability to stimulate the proliferation of allogeneic T cells, and induce an IL-10-mediated suppressive activity in allogeneic CD4+CD25+ regulatory T cells. Bone marrow from Irf1-/- mice contains an increased number of immature granulocytic precursors, and a decreased number of mature granulocytes compared to wild type bone marrow cells, as determined by cell staining for Gr-1 or CD11b (25). The colony-forming ability of Irf1-/- progenitors from the bone marrow in response to treatment with G-CSF and M-CSF is also reduced. Irf1-/- mice exhibit a severe deficiency of NK cells, which lack in vitro cytotoxic activity in response to LCMV infection or poly I:C treatment in vivo and fail to produce IFN-γ upon IL-12 stimulation in vitro (26-28). NKT and intestinal intraepithelial lymphocytes (IELs) are also greatly reduced in Irf1-/- mice (27). Interestingly, Irf+/- mice display an intermediate reduction in NK, NKT and IEL numbers. Irf1-/- mice display a 10-fold reduction of mature CD8+ T cells in the thymus, spleen, lymph nodes and blood, while CD4+ T cells are present in normal numbers (29;30). Although Irf1-/- CD4+ T cells mature normally, they exclusively undergo Th2 differentiation (28;31). This is manifested by a skewed profile of cytokine production (dominated by IL-4 and lacking IFN-γ) when Irf1-/- mice are infected with Leishmania major in vivo and when Irf1-/- splenic cells are stimulated with TCR-specific peptide in vitro (28;31). The failure of Th1 differentiation is due in part to the inability of Irf1-/- mice to both produce and respond to IL-12.

Primers PCR Primer
Longs_peak_pcr_F: ACTCATGCAGGTTCTGTGC
Longs_peak_pcr_R: TAATCTGGCTCTGGCTCTGC

Sequencing Primer
Longs_peak_seq_F: CATGCAGGTTCTGTGCCAGAG
Longs_peak_seq_R: GGCTCTGCCTCCAACAG
Genotyping

PCR program

1) 94°C 2:00
2) 94°C 0:30
3) 55°C 0:30
4) 72°C 1:00
5) repeat steps (2-4) 40x
6) 72°C 10:00
7) 4°C hold


The following sequence of 434 nucleotides is amplified (chromosome 11, + strand):


1   actcatgcag gttctgtgcc agagggggac agcgacacac aggaggggcg ggggccagca
61  gggcccaggt cgctttgtgc tcccctccct agacagtttt gcatattgtt acccataaac
121 agtcaaggtt atgatcctgc ttgggaaggg ttaggtcacc caagggaagg gccacagggt
181 gagtgcactg gcaggcagct gatggcttca tgctctctcc ccagctcttt gaacagtctg
241 agtggcagcc gacacacatc gatggcaagg gatacttgct caatgagcca gggacccagc
301 tctcttctgt ctatggagac ttcagctgca aagaggaacc agagattgac agccctcgag
361 gtaagaggct tctaggtcct tctgggttct gggagaaggg ggaagctgtt ggaggcagag
421 ccagagccag atta


Primer binding sites are underlined and the sequencing primers are highlighted; the mutated nucleotide is shown in red.

References
Science Writers Anne Murray
Illustrators Diantha La Vine
AuthorsJin Huk Choi, Xue Zhong, and Bruce Beutler