Familial hemophagocytic lymphohistiocytosis (FHL) is an autosomal recessive, often-fatal hyperinflammatory disorder. Mutations in PRF1, UNC13D, STX11, and STXBP2 are causative of FHL2, 3, 4, and 5, respectively. In a majority of suspected FHL patients from Northern Europe, sequencing of exons and splice sites of such genes required for lymphocyte cytotoxicity revealed no or only monoallelic UNC13D mutations. Here, in 21 patients, we describe 2 pathogenic, noncoding aberrations of UNC13D. The first is a point mutation localized in an evolutionarily conserved region of intron 1. This mutation selectively impairs UNC13D transcription in lymphocytes, abolishing Munc13-4 expression. The second is a 253-kb inversion straddling UNC13D, affecting the 3′-end of the transcript and likewise abolishing Munc13-4 expression. Carriership of the intron 1 mutation was found in patients across Europe, whereas carriership of the inversion was limited to Northern Europe. Notably, the latter aberration represents the first description of an autosomal recessive human disease caused by an inversion. These findings implicate an intronic sequence in cell-type specific expression of Munc13-4 and signify variations outside exons and splice sites as a common cause of FHL3. Based on these data, we propose a strategy for targeted sequencing of evolutionary conserved noncoding regions for the diagnosis of primary immunodeficiencies.

Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory disorder characterized by unremitting fever, hepatosplenomegaly, hyperferritinemia, cytopenia, and sometimes hemophagocytosis.1-4  HLH can be divided into primary and secondary forms.4-6  Without a known familial history or a molecular diagnosis, it is difficult to distinguish between such primary and secondary HLH. For the diagnosis of HLH, at least 5 of the 8 defined criteria need to be fulfilled.7  First-line treatment of HLH is directed at controlling the hyperinflammation caused by aberrant immune activation.7  However, the only cure for primary HLH is hematopoietic stem cell transplantation.8  Thus, for treatment, it is important to obtain a rapid molecular diagnosis.

Familial hemophagocytic lymphohistiocytosis (FHL), which has an autosomal recessive inheritance, has so far been linked to 5 different loci. No gene has yet been associated with FHL1, whereas mutations in PRF1, UNC13D, STX11, and STXBP2 are causative of FHL2, 3, 4, and 5, respectively.9-13  These genes all encode proteins required for lymphocyte cytotoxicity,14-16  and defective NK cell cytotoxicity is one of the 8 diagnostic criteria for HLH.7  In addition, mutations in UNC13D and STXBP2, encoding Munc13-4 and syntaxin binding protein 2, respectively, also impair platelet granule release.17,18  Furthermore, patients with X-linked lymphoproliferative syndrome (XLP) may present with severe Epstein-Barr virus infections, hypogammaglobulinemia, lymphoma, or HLH.19  Mutations in SH2D1A and XIAP are associated with XLP1 and XLP2, respectively.20,21  Although the syndromes share clinical phenotypes, recent reports emphasize differences between the syndromes, such as the frequent development of lymphoma in XLP1 patients in contrast to XLP2 patients.22,23  In addition, Griscelli syndrome type 2 and Chediaki-Higashi syndrome, both with autosomal recessive inheritance and caused by mutations in RAB27A and LYST, respectively, are associated with development of HLH in addition to manifesting partial albinism.24,25 

The relative frequency of mutations in various genes causative of FHL differs among ethnicities.26,27  With the advance of knowledge concerning genetic associations with FHL, a molecular diagnosis can be attained for an increasing proportion of patients. In this regard, Côte et al recently reported that only 10% of the FHL cases they have studied are without a molecular diagnosis.13  However, in our and other cohorts of infants from Northern Europe with HLH and defective NK cell cytotoxicity,26  sequencing of exons and exon-intron boundaries in the genes associated with HLH have not provided a molecular diagnosis for many patients.

In this study, we describe 2 different aberrations in UNC13D that together can explain the majority of the FHL cases in Scandinavia as well as several other cases across Europe. These findings improve diagnostics and carrier testing and may also offer the possibility of prenatal testing for a number of families. Moreover, they also define an intronic sequence involved in transcriptional control of Munc13-4 expression in lymphocytes.

Patients and controls

In this study, Swedish infants presenting with a known familial history of HLH and/or defective cytotoxicity and/or degranulation between December 2005 and January 2011 were included. In addition, the study also encompassed all infants at our unit identified with UNC13D mutations from Sweden, Denmark, Norway, Finland, Slovenia, and Croatia. The studies were approved by the ethics committee at the Karolinska Institutet. Written consent was obtained from all families in accordance with the Declaration of Helsinki.

DNA extraction, amplification, and sequence analysis

Genomic DNA was isolated from peripheral blood or cultured fibroblasts according to standard procedures. Specific primers were designed and used for amplification of selected regions by PCR. The amplified products were subsequently used for direct sequencing according to standard procedures (BigDye Version 3.1; Applied Biosystems). Sequencing reactions were analyzed by capillary electrophoresis (ABI 3730 Genetic Analyzer; Applied Biosystems). Data were analyzed using SeqScape software (Version 2.5; Applied Biosystems) and a UNC13D reference sequence template (NCBI Accession NM_199242.2). Primers, PCR conditions, and sequencing reaction conditions are available on request.

RNA extraction, cDNA synthesis, amplification, and analyses

Total RNA was extracted from WBCs (RNeasy, QIAGEN), and cDNA was synthesized with oligo(dT)20 primed reverse transcription (SuperScript III, Invitrogen) according to the manufacturer's protocol. For evaluation of UNC13D splicing, 9 primer pairs were used for amplification of overlapping cDNA fragments. For 3′-rapid amplification of cDNA ends PCR, cDNA was synthesized using oligo(dT)20 primers marked with an M13R-tag. A gene specific forward primer and a M13R reverse primer were used for amplification of the 3′-end of the UNC13D transcript. Amplified products were separated by agarose gel electrophoresis, extracted (QIAquick gel extraction kit; QIAGEN), and cloned (TOPO-TA cloning kit, Invitrogen). Plasmid DNA was isolated (GeneJET plasmid miniprep kit, Fermentas) and sequenced.

Allele-specific quantitative RT-PCR of isolated cell populations

PBMCs were isolated by density gradient centrifugation. Specific cell populations were consecutively isolated by magnetic positive selection using anti–CD14, anti–CD4, anti–CD8, and anti–CD56 mAb-coated beads, respectively (Miltenyi Biotec). Purity of the isolated cell populations was assessed by flow cytometry using fluorochrome-conjugated mAbs against CD3 (clone UHCT1; eBioscience), CD4 (clone 53.5; Invitrogen), CD8 (clone 3B5; Invitrogen), CD14 (clone MϕP9; BD Bioscience), CD45 (clone 2D1; BD Bioscience), and CD56 (clone NCAM16.2; BD Bioscience). RNA was isolated (TRIzol; Invitrogen) and cDNA synthesized as described. For quantitative real-time PCR quantification of allele-specific transcription, 2 different reverse primers that could discriminate a single nucleotide polymorphism, c.888G > C located in exon 11, were designed. The specificity of these primers was evaluated by agarose gel electrophoresis and by examining real-time PCR dissociations curves. Relative quantification was assessed using the standard curve method. All samples were run in triplicates. For each reaction, 100 ng cDNA was mixed with 10 μL of Power SYBR Green PCR master mix (Applied Biosystems) and 10 pmol of forward and reverse primers in a total volume of 20 μL. The reactions were analyzed by real-time PCR (ABI 7900 HT; Applied Biosystems) and analyzed with SDS software (Version 2.2.1; Applied Biosystems).

Western blot analysis of Munc13-4 expression

PBMCs from patients and healthy controls were lysed in lysis buffer (20mM Tris, pH 7.4, 1mM EDTA, 1% Triton X-100, 150mM NaCl) with protease inhibitors (Roche Diagnostics). Cells were disrupted and centrifuged for 15 minutes at 14 000g. Protein content was determined by the Bradford assay (Bio-Rad), and 50 μg protein was loaded per lane and analyzed using SDS-PAGE separation and Western blotting (NuPAGE; Invitrogen). The rabbit polyclonal antibodies to Munc13-4 (Protein Technologies Group, raised against amino acids 1-236) and ERK1 (Santa Cruz Biotechnology) were used for Western blotting.

Analysis of cytotoxic lymphocyte function

For assessment of NK cell-mediated cytotoxicity, a standard 4-hour 51Cr assay was used.28  PBMCs were isolated by density gradient centrifugation and used as effector cells either after incubation overnight in complete medium (RPMI 1640 supplemented with 10% FBS, l-glutamine, penicillin, and streptomycin, all from Invitrogen) or after activation with IL-2 (Proleukin; Novartis) for 36 to 60 hours. 51Cr-labeled K562 target cells (ATCC) were used as target cells. Lymphocyte cytotoxicity was calculated as lytic units (LU) at 25% lysis. A value < 10 LU was considered pathologic.7  NK cell degranulation was assessed by flow cytometric quantification of induction of CD107a surface expression on CD3CD56+ gated cells in response to incubation with K562 target cells, as previously described.29 

Genotyping of microsatellite markers

PCR was performed according to standard procedures with fluorescently labeled primers. PCR products were separated by electrophoresis on an ABI 3730 DNA Analyzer (Applied Biosystems) with GeneScan 400HD Rox (Applied Biosystems) as size standard and analyzed with Peak Scanner Version 1.0 software (Applied Biosystems).

HLH patients

Between December 2005 and January 2011, 13 infants from Sweden (age, 0-3 years) came to our attention who fulfilled the current criteria for HLH, specifically including defective NK cell cytotoxicity.7  Within this group, one infant (of Afghani origin) carried a novel, homozygous PRF1 mutation, c.658G > C (p.Gly220Arg), whereas another infant (of Somali origin) carried a novel homozygous UNC13D mutation, c.2381delT (p.Leu794ArgfsX2). A pair of twins carried heterozygous compound mutations in RAB27A, and another infant carried a heterozygous compound mutation in PRF1, all with mutations that have been described.28,30  Of the remaining 8 patients, 2 infants had monoallelic mutations in UNC13D that have been described.26,31  Hence, the majority (62%) of these infants from Sweden were without a molecular diagnosis after sequencing of the coding region and splice sites of genes associated with FHL. Through sequencing other infants referred to us from Europe with HLH and defective NK cell cytotoxicity, we identified an additional 5 infants from 4 families with monoallelic UNC13D mutations (Table 1). Two brothers carried a novel missense mutation, c.2642T > C, p.Leu881Pro predicted to be pathogenic using 2 different protein prediction programs (PolyPhen, SIFT32,33 ), whereas the other mutations are previously described.10,26,31  Notably, a German study also reported 6 infants who developed HLH in which monoallelic UNC13D mutations were identified.26  This prompted us to search for mutations in UNC13D outside of exons and splice sites.

Table 1

Clinical and laboratory findings in patients with the UNC13D intron 1 mutation, c.118-308C > T, and/or the 253-kb inversion

Patient APatient B:1Patient B:2Patient CPatient DPatient EPatient FPatient GPatient HPatient IPatient J:1Patient J:2Patient KPatient LPatient MPatient NPatient O:1Patient O:2Patient PPatient QPatient R
Ethnical origin Croatia Slovenia Slovenia Sweden Denmark/Ukraine Croatia Sweden Denmark Sweden Finland Ethnical origin Sweden Sweden Sweden Sweden Sweden Sweden Sweden Sweden Sweden/USA Sweden Norway 
Familial disease No Yes Yes No No No No No  No Familial disease Yes Yes No Yes No No Yes Yes No No No 
Parental consanguinity No No No No No No No No No No Parental consanguinity No No No  No No No No No  No 
Sex Male Male Male Male Male Female Male Female Female Male Sex Female Female Female Female Male Female Male Female Female Female Male 
Allele 1 753 + 1G, splice error 2642T > C, Leu881Pro 2642T > C, Leu881Pro 532delC, Gln178ArgfsX71 2346_2349delGGAG, Arg782SerfsX12 118-308C > T Inversion Inversion Inversion Inversion Allele 1 Inversion  Inversion Inversion Inversion Inversion  Inversion Inversion Inversion Inversion 
Allele 2 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 2346_2349delGGAG, Arg782SerfsX12 2176C > T, Gln726X Allele 2 Inversion  Inversion Inversion Inversion Inversion  Inversion Inversion Inversion Inversion 
Age at diagnosis of HLH, days 55 195 229 57 150 133 87 198 104 148 Age at diagnosis of HLH, days 365 79 459 104 127 56 134 73 388 
Fever Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Fever Yes No Yes Yes Yes Yes Yes Yes Yes No Yes 
Splenomegaly Yes Yes Yes Yes Yes Yes Yes Yes No Yes Splenomegaly Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes 
Hepatomegaly Yes Yes No Yes Yes Yes Yes Yes Yes Yes Hepatomegaly Yes Yes Yes Yes Yes Yes Yes No Yes Yes Yes 
Hb, g/L 100 115 105 70 61 81 60 95 46 97 Hb, g/L 83 138 62 100 87 100 58 65 82 61 62 
Neutrophils, × 109/L 0.3 0.8 1.0 1.0 0.4 0.0 0.2 0.6 0.4 0.9 Neutrophils, × 109/L 2.9 2.9 1.4 0.8 0.7 0.4 0.1 0.7 0.2 0.3 0.8 
Platelets, × 109/L 28 143 34 13 22 31 72 17 59 Platelets, × 109/L 38 15 32 15 12 37 76 18 10 33 
Triglycerides, mM 2.8 12 4.5 2.3 7.3 3.9 2.4 6.1 4.2 5.4 Triglycerides, mM 3.0 2.7 11 2.6 3.4 2.7 3.4 17.3 5.5 1.9 4.4 
Fibrinogen, g/L 0.9 1.5 4.2 2.1 0.9 0.6 1.2 2.3 0.6  Fibrinogen, g/L 0.5 0.6 1.2 2.4 0.6 0.7 < 0.5 1.7 0.8 0.7 2.9 
Hemophagocytosis Yes Yes No No No Yes No Yes Yes Yes Hemophagocytosis Yes No No No No Yes ND No Yes (CSF) No Yes 
Ferritin, μg/L 5000 6482 315 2800 2399 1020 1540 437 4148 1858 Ferritin, μg/L 2920 > 15 000 5433 6597 7828 2387 11 372 208 8100 1147 1162 
sCD25, U/mL ND ND ND > 7500 ND ND 20 380 ND ND 30 486 sCD25, U/mL 21 976 ND 5666 14 110 ND 11 861 ND 8802 > 7500 > 7500 Elevated 
NK cell activity* Borderline (11 LU) ND Defective Defective Defective Defective ND Defective Borderline (13 LU) Defective NK cell activity* Defective Defective Defective Defective Defective Defective ND Defective Defective Defective Defective 
NK cell degranulation Defective ND Defective Low Defective Defective ND Defective Defective ND NK cell degranulation Defective Defective Defective Defective Defective ND ND ND Defective Defective ND 
Neurologic manifestations Yes No No No Yes No No No No Yes Neurologic manifestations Yes Yes Yes Yes Yes Yes Yes No Yes No No 
Pathologic CSF Yes No No No Yes Yes Yes Yes Yes Yes Pathologic CSF Yes ND Yes Yes Yes ND Yes Yes Yes ND Yes 
Treatment active disease HLH-2004 HLH-2004 HSCT HLH-2004 HLH-2004 HLH-2004 HLH-94 HLH-2004 HLH-2004 HLH-94 Treatment active disease HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-94 HLH-2004 HLH-2004 HLH-2004 HLH-94 
Remission at 2 months Remission, but possible CNS progression No NA No Remission, but possible CNS progression Yes No No Yes No Remission at 2 months No No No No No No No Yes No No Yes 
Age at HSCT, days 245 191 273 128 297 653 177 373 419 471 Age at HSCT 705 No 195 No 224 371/853 No 197 150 86 843 
Outcome Alive Deceased 226 days Deceased 289 days Alive Deceased 312 days Alive Alive Deceased 460 days Alive Alive Outcome Alive Deceased 71 days Alive Deceased Alive Alive Deceased 143 days Alive Alive Deceased Alive 
Patient APatient B:1Patient B:2Patient CPatient DPatient EPatient FPatient GPatient HPatient IPatient J:1Patient J:2Patient KPatient LPatient MPatient NPatient O:1Patient O:2Patient PPatient QPatient R
Ethnical origin Croatia Slovenia Slovenia Sweden Denmark/Ukraine Croatia Sweden Denmark Sweden Finland Ethnical origin Sweden Sweden Sweden Sweden Sweden Sweden Sweden Sweden Sweden/USA Sweden Norway 
Familial disease No Yes Yes No No No No No  No Familial disease Yes Yes No Yes No No Yes Yes No No No 
Parental consanguinity No No No No No No No No No No Parental consanguinity No No No  No No No No No  No 
Sex Male Male Male Male Male Female Male Female Female Male Sex Female Female Female Female Male Female Male Female Female Female Male 
Allele 1 753 + 1G, splice error 2642T > C, Leu881Pro 2642T > C, Leu881Pro 532delC, Gln178ArgfsX71 2346_2349delGGAG, Arg782SerfsX12 118-308C > T Inversion Inversion Inversion Inversion Allele 1 Inversion  Inversion Inversion Inversion Inversion  Inversion Inversion Inversion Inversion 
Allele 2 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 118-308C > T 2346_2349delGGAG, Arg782SerfsX12 2176C > T, Gln726X Allele 2 Inversion  Inversion Inversion Inversion Inversion  Inversion Inversion Inversion Inversion 
Age at diagnosis of HLH, days 55 195 229 57 150 133 87 198 104 148 Age at diagnosis of HLH, days 365 79 459 104 127 56 134 73 388 
Fever Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Fever Yes No Yes Yes Yes Yes Yes Yes Yes No Yes 
Splenomegaly Yes Yes Yes Yes Yes Yes Yes Yes No Yes Splenomegaly Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes Yes 
Hepatomegaly Yes Yes No Yes Yes Yes Yes Yes Yes Yes Hepatomegaly Yes Yes Yes Yes Yes Yes Yes No Yes Yes Yes 
Hb, g/L 100 115 105 70 61 81 60 95 46 97 Hb, g/L 83 138 62 100 87 100 58 65 82 61 62 
Neutrophils, × 109/L 0.3 0.8 1.0 1.0 0.4 0.0 0.2 0.6 0.4 0.9 Neutrophils, × 109/L 2.9 2.9 1.4 0.8 0.7 0.4 0.1 0.7 0.2 0.3 0.8 
Platelets, × 109/L 28 143 34 13 22 31 72 17 59 Platelets, × 109/L 38 15 32 15 12 37 76 18 10 33 
Triglycerides, mM 2.8 12 4.5 2.3 7.3 3.9 2.4 6.1 4.2 5.4 Triglycerides, mM 3.0 2.7 11 2.6 3.4 2.7 3.4 17.3 5.5 1.9 4.4 
Fibrinogen, g/L 0.9 1.5 4.2 2.1 0.9 0.6 1.2 2.3 0.6  Fibrinogen, g/L 0.5 0.6 1.2 2.4 0.6 0.7 < 0.5 1.7 0.8 0.7 2.9 
Hemophagocytosis Yes Yes No No No Yes No Yes Yes Yes Hemophagocytosis Yes No No No No Yes ND No Yes (CSF) No Yes 
Ferritin, μg/L 5000 6482 315 2800 2399 1020 1540 437 4148 1858 Ferritin, μg/L 2920 > 15 000 5433 6597 7828 2387 11 372 208 8100 1147 1162 
sCD25, U/mL ND ND ND > 7500 ND ND 20 380 ND ND 30 486 sCD25, U/mL 21 976 ND 5666 14 110 ND 11 861 ND 8802 > 7500 > 7500 Elevated 
NK cell activity* Borderline (11 LU) ND Defective Defective Defective Defective ND Defective Borderline (13 LU) Defective NK cell activity* Defective Defective Defective Defective Defective Defective ND Defective Defective Defective Defective 
NK cell degranulation Defective ND Defective Low Defective Defective ND Defective Defective ND NK cell degranulation Defective Defective Defective Defective Defective ND ND ND Defective Defective ND 
Neurologic manifestations Yes No No No Yes No No No No Yes Neurologic manifestations Yes Yes Yes Yes Yes Yes Yes No Yes No No 
Pathologic CSF Yes No No No Yes Yes Yes Yes Yes Yes Pathologic CSF Yes ND Yes Yes Yes ND Yes Yes Yes ND Yes 
Treatment active disease HLH-2004 HLH-2004 HSCT HLH-2004 HLH-2004 HLH-2004 HLH-94 HLH-2004 HLH-2004 HLH-94 Treatment active disease HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-2004 HLH-94 HLH-2004 HLH-2004 HLH-2004 HLH-94 
Remission at 2 months Remission, but possible CNS progression No NA No Remission, but possible CNS progression Yes No No Yes No Remission at 2 months No No No No No No No Yes No No Yes 
Age at HSCT, days 245 191 273 128 297 653 177 373 419 471 Age at HSCT 705 No 195 No 224 371/853 No 197 150 86 843 
Outcome Alive Deceased 226 days Deceased 289 days Alive Deceased 312 days Alive Alive Deceased 460 days Alive Alive Outcome Alive Deceased 71 days Alive Deceased Alive Alive Deceased 143 days Alive Alive Deceased Alive 

Patient B:1 and B:2 carried a novel missense mutation, c.2642T > C, p.Leu881Pro predicted to be pathogenic using 2 different protein prediction programs (PolyPhen, SIFT30,31 ), whereas the other mutations are previously described.9,25,32 

ND indicates no data; NA, not applicable; CSF, cerebrospinal fluid; CNS, central nervous system; and HSCT, hematopoietic stem cell transplantation.

*

Defective: ≤ 10 lytic units.

Reported at some point during the course of the disease.

UNC13D intron 1 mutation

To identify mutations in noncoding regions of UNC13D, evolutionary conservation of nucleotides was evaluated using a phylogenetic hidden Markov model based on sequence from 17 vertebrates, including mammalian, amphibian, bird, and fish species (AlaMut, Interactive Biosoftware34 ; Figure 1A). Aside from the highly conserved coding regions, 4 intronic regions in UNC13D displayed a high degree of evolutionary conservation. In an attempt to identify mutations in the patients carrying a monoallelic UNC13D mutation, these conserved noncoding regions were sequenced. A variant, c.118-308C > T, was identified in a conserved region of intron 1 in 5 of the 7 patients carrying monoallelic UNC13D mutations (4 families, Figure 1B). This variant has previously been reported in a heterozygous state in a single patient diagnosed with FHL3.35  Besides c.118-308C > T, sequencing of the entire intron 1 identified 2 other variants, c.117 + 59C > T and c.118–176G > C. The most upstream variant, c.117 + 59C > T, is a common polymorphism (rs3744010) and thus unlikely to cause disease. The variant c.118–176G > C was found in a homozygous state in 3 and in a heterozygous state in 12 of 96 healthy blood donors. In contrast, the variant c.118-308C > T is located in a conserved region and was not identified in any of the healthy controls, suggesting that this nucleotide variant might represent a disease-causing mutation.

Figure 1

Characterization of a UNC13D intron 1 mutation. (A) Evolutionary nucleotide conservation (gray bars) as predicted by the AlaMut algorithm relative to intronic (black line) and exonic (black boxes) regions of UNC13D. (B) The nucleotide exchange c.118-308C > T is located in a highly conserved region of intron 1. The c.118-308C > T mutation was identified in 7 patients (6 families) in a heterozygous state and in one patient in a homozygous state. (C) PBMCs from patient C and a healthy control were lysed, and protein content was analyzed by SDS-PAGE and Western blotting. Rabbit polyclonal antibodies for detection of Munc13-4 and ERK1 and 2 were used. (D) Nine different primer pairs were used for amplification of overlapping, consecutive UNC13D cDNA fragments. The amplified fragments from patient E, carrying the intron 1 mutation c.118-308C > T in a homozygous state, and a healthy control were separated by gel electrophoresis and are shown relative to their position in the UNC13D transcript.

Figure 1

Characterization of a UNC13D intron 1 mutation. (A) Evolutionary nucleotide conservation (gray bars) as predicted by the AlaMut algorithm relative to intronic (black line) and exonic (black boxes) regions of UNC13D. (B) The nucleotide exchange c.118-308C > T is located in a highly conserved region of intron 1. The c.118-308C > T mutation was identified in 7 patients (6 families) in a heterozygous state and in one patient in a homozygous state. (C) PBMCs from patient C and a healthy control were lysed, and protein content was analyzed by SDS-PAGE and Western blotting. Rabbit polyclonal antibodies for detection of Munc13-4 and ERK1 and 2 were used. (D) Nine different primer pairs were used for amplification of overlapping, consecutive UNC13D cDNA fragments. The amplified fragments from patient E, carrying the intron 1 mutation c.118-308C > T in a homozygous state, and a healthy control were separated by gel electrophoresis and are shown relative to their position in the UNC13D transcript.

Close modal

In addition to the initial 7 patients with monoallelic UNC13D mutations, 2 infants of Swedish and Danish origin, respectively, with HLH and defective NK cell cytotoxicity were identified carrying the c.118-308C > T mutation in a heterozygous state. Importantly, one infant from Croatia with HLH and defective NK cell cytotoxicity was homozygous for the c.118-308C > T mutation (patient E; Table 1). Western blot analyses of whole cell lysates prepared from PBMCs did not show Munc13-4 expression in patients carrying the c.118-308C > T mutation (Figure 1C), indicating that the conserved intronic sequence may interfere with UNC13D splicing, as hypothesized by Santoro et al,35  or alternatively, regulate UNC13D transcription.

To distinguish between defects in splicing or transcription, UNC13D transcripts were analyzed in patients and heterozygous carriers of the c.118-308C > T mutation in intron 1. First, to determine whether this mutation interfered with UNC13D splicing, overlapping fragments of the UNC13D transcript were amplified in patient E, homozygous for the c.118-308C > T mutation. In this patient, all fragments were readily amplified and displayed the predicted length (Figure 1D). Notably, sequence analysis of the fragments revealed correctly spliced products. Thus, data indicated that the intron 1 mutation did not interfere with UNC13D splicing. To evaluate whether the mutation affected UNC13D transcript levels, allele-specific quantitative real-time PCR was performed on persons heterozygous for the c.118-308C > T mutation that were also heterozygous for a linked, common polymorphism, c.888G > C, in exon 11. For comparison, allele-specific transcription was quantified in healthy controls that did not carry the c.118-308C > T mutation but were heterozygous for the c.888G > C polymorphism. On analysis of allele transcription in WBCs, UNC13D transcripts with the c.888C allele were as frequent as those with the c.888G allele in persons that did not carry the c.118-308C > T mutation (Figure 2A). In contrast, in parents that where heterozygous carriers of the c.118-308C > T mutation, transcription of the allele carrying the linked c.888G > C polymorphism was significantly diminished, representing 27% of total UNC13D transcripts (Figure 2A). Accordingly, a homozygous carrier of the mutation would theoretically have 37% of the normal amount of transcript. In transplanted FHL patients, relapse of HLH is rarely, if ever, observed unless the percentage of donor lymphocytes decreased to < 10%.36,37  Accordingly, such a decrease in UNC13D transcription might not be expected to cause disease.

Figure 2

Allele-specific quantitative real-time PCR in heterozygous carriers of the UNC13D intron 1 mutation. (A) Allele-specific expression analysis of UNC13D was assessed in WBCs from healthy controls and heterozygous carriers of the intron 1 mutation, c.118-308C > T, using real-time PCR (standard curve method). Primers were designed for specific amplification of either the c.888G or the c.888C allele, a single nucleotide polymorphism located in exon 11. Both healthy controls (n = 3) and carriers of the intron 1 mutation (n = 4) carried the SNP, c.888G > C, in a heterozygous state. In all heterozygous carriers of the intron 1 mutation, c.118-308C > T, was located on the c.888C allele. (B) For evaluation of allele-specific transcription in different cell subsets, CD14+, CD4+, CD8+, and CD56+ cells were isolated consecutively by positive magnetic selection. Allele specific real-time PCR was performed in healthy controls (n = 4) and heterozygous carriers of the intron 1 mutation, c.118-308C > T (n = 4). Statistical significance was analyzed using the 2-tailed Mann-Whitney U test. *P < .05.

Figure 2

Allele-specific quantitative real-time PCR in heterozygous carriers of the UNC13D intron 1 mutation. (A) Allele-specific expression analysis of UNC13D was assessed in WBCs from healthy controls and heterozygous carriers of the intron 1 mutation, c.118-308C > T, using real-time PCR (standard curve method). Primers were designed for specific amplification of either the c.888G or the c.888C allele, a single nucleotide polymorphism located in exon 11. Both healthy controls (n = 3) and carriers of the intron 1 mutation (n = 4) carried the SNP, c.888G > C, in a heterozygous state. In all heterozygous carriers of the intron 1 mutation, c.118-308C > T, was located on the c.888C allele. (B) For evaluation of allele-specific transcription in different cell subsets, CD14+, CD4+, CD8+, and CD56+ cells were isolated consecutively by positive magnetic selection. Allele specific real-time PCR was performed in healthy controls (n = 4) and heterozygous carriers of the intron 1 mutation, c.118-308C > T (n = 4). Statistical significance was analyzed using the 2-tailed Mann-Whitney U test. *P < .05.

Close modal

Thus, we hypothesized that the c.118-308C > T mutation might more severely affect UNC13D transcription in cell types that mediate lymphocyte cytotoxicity, such as CD3+CD8+ T cells and CD3CD56+ NK cells. To test this, allele-specific transcription was examined in CD14+, CD4+, CD8+, and CD56+ cells isolated sequentially from controls or carriers of the c.118-308C >T mutation that all were heterozygous carriers of the c.888G >C polymorphism. Strikingly, the allele carrying the c.888G >C polymorphism represented < 10% of UNC13D transcripts in CD4+, CD8+, and CD56+ cells from persons carrying the c.118–308C > T mutation, indicating a more than a 9-fold reduction in transcription of the mutated UNC13D allele relative to the wild-type in lymphocytes (Figure 2B). The relative frequency of the transcript from the mutated allele in CD4+, CD8+, and CD56+ cells was significantly less than that observed in CD14+ cells. In persons that did not carry the c.118-308C > T mutation, the frequency of the alleles with or without the c.888G > C polymorphism was similar (Figure 2B). Altogether, the data indicate that the c.118-308C > T mutation specifically impairs UNC13D transcription in lymphocytes.

UNC13D 253-kb inversion

The identification of 2 patients with the monoallelic UNC13D intron 1 mutation, in addition to the 2 previous patients with monoallelic UNC13D mutations that did not harbor the intron 1 mutation, indicated other noncoding UNC13D aberrations. Examination of exonic and intronic single nucleotide polymorphisms throughout UNC13D in these patients revealed a shared haplotype on the second allele in 2 patients of Swedish origin, one of Danish origin, and one of Finnish origin. Remarkably, other nonrelated Scandinavian HLH patients with defective NK cell cytotoxicity were homozygous for this haplotype.

To determine whether these patients carried a common disease-causing mutation affecting UNC13D splicing, overlapping fragments of the UNC13D transcript were amplified in 3 patients homozygous for the second haplotype where RNA was available. Whereas the first 7 fragments were readily amplified in these 3 patients, the last 2 fragments were not detected (Figure 3A). By comparison, all fragments were readily amplified in healthy control samples (Figure 3A). Thus, the data indicated an aberration affecting splicing of the 3′-end of UNC13D. To obtain the 3′-end sequence of the transcript in the patients homozygous for the second haplotype, 3′-rapid amplification of cDNA ends PCR was performed. Three different products were amplified. Cloning and sequencing of the 3 PCR products revealed UNC13D sequence up to the end of exon 30. In the first product, also observed in healthy controls, the sequence of exon 30 was followed by sequence from intron 30. In the 2 other PCR products, specific to the patients, sequence following exon 30 lacked homology to UNC13D. Thus, these 3′-end sequences were blasted against the human genome using the BLAT algorithm.38  The highest homology for both products was located on chromosome 17, 253 kb upstream of UNC13D. Thus, the sequence identity suggested an inversion of approximately 253 kb straddling the UNC13D locus. To confirm the existence of such an inversion in the genomic DNA from the patients, primers were designed in the vicinity of the exon 30 splice site and from the sequence located 253 kb upstream of UNC13D. One primer pair specifically amplified product in the patients, but not in healthy, unrelated controls. PCR products from the patients were isolated, cloned, and sequenced, thereby providing sequence over one of the break points. Similarly, the second break point was amplified with a different set of primers and subsequently cloned and sequenced. In this way, the breakpoints were mapped down to 2 25-bp Alu elements that were identical in sequence (chromosome 17: 73825638-73825663 and 73573065-73573090, human, February 2009, GRCh37/hg19 assembly; Figure 3B). No gain or loss of DNA seemed to have occurred. For rapid detection of the 253-kb inversion, a multiplex PCR assay using 2 different forward primers and 2 different reverse primers was developed (Figure 4). The inversion was detected in a heterozygous state in one of 190 healthy blood donors, in line with this aberration being a cause of FHL in the Scandinavian population.

Figure 3

Characterization of an UNC13D inversion. (A) Nine different primer pairs were used for amplification of overlapping, consecutive UNC13D cDNA fragments. The amplified fragments from a patient (representative of 3 analyzed patients) and a healthy control were separated by gel electrophoresis and are shown relative to their position in the UNC13D transcript. (B) Schematic illustration of a 253-kb inversion identified in patients with FHL. The breakpoints are located in Alu elements containing an identical sequence of 25 bp. Two elements, AluSc8 and AluSx1 (yellow, orange), are located on the reverse strand in intron 30 UNC13D, and an AluY (red) element is located 253 kb upstream of UNC13D on the forward strand. (C) PBMCs from patient O, patient L, and healthy controls were lysed, and protein content was analyzed by SDS-PAGE and Western blotting. Rabbit polyclonal antibodies for detection of Munc13-4 and ERK1 and 2 were used.

Figure 3

Characterization of an UNC13D inversion. (A) Nine different primer pairs were used for amplification of overlapping, consecutive UNC13D cDNA fragments. The amplified fragments from a patient (representative of 3 analyzed patients) and a healthy control were separated by gel electrophoresis and are shown relative to their position in the UNC13D transcript. (B) Schematic illustration of a 253-kb inversion identified in patients with FHL. The breakpoints are located in Alu elements containing an identical sequence of 25 bp. Two elements, AluSc8 and AluSx1 (yellow, orange), are located on the reverse strand in intron 30 UNC13D, and an AluY (red) element is located 253 kb upstream of UNC13D on the forward strand. (C) PBMCs from patient O, patient L, and healthy controls were lysed, and protein content was analyzed by SDS-PAGE and Western blotting. Rabbit polyclonal antibodies for detection of Munc13-4 and ERK1 and 2 were used.

Close modal
Figure 4

Multiplex PCR assay for detection of the UNC13D inversion. A multiplex PCR assay with 2 different forward and reverse primers was designed for rapid detection of the UNC13D inversion. (A) Primer sequences and schematic representation of primer pair positioning on wild-type (Wt) and inverted (Inv) UNC13D genomic sequence. (B) Gel electrophoresis of multiplex PCR products on genomic DNA from individuals wild-type, heterozygous, or homozygous for the inversion straddling UNC13D. In homozygous carriers of the inversion, 2 products of 725 and 922 bp are amplified. In persons that do not carry the inversion, 2 products of 466 and 1220 bp are amplified. Consequently, in heterozygous carriers of the inversion, all 4 products are amplified.

Figure 4

Multiplex PCR assay for detection of the UNC13D inversion. A multiplex PCR assay with 2 different forward and reverse primers was designed for rapid detection of the UNC13D inversion. (A) Primer sequences and schematic representation of primer pair positioning on wild-type (Wt) and inverted (Inv) UNC13D genomic sequence. (B) Gel electrophoresis of multiplex PCR products on genomic DNA from individuals wild-type, heterozygous, or homozygous for the inversion straddling UNC13D. In homozygous carriers of the inversion, 2 products of 725 and 922 bp are amplified. In persons that do not carry the inversion, 2 products of 466 and 1220 bp are amplified. Consequently, in heterozygous carriers of the inversion, all 4 products are amplified.

Close modal

Western blot analysis of whole cell lysates prepared from PBMCs did not detect Munc13-4 expression in patients carrying the inversion (Figure 3C), suggesting that the truncated UNC13D transcript generates an unstable protein product.

Effect of mutations on lymphocyte cytotoxic function

Decreased NK cell activity, typically assessed by quantifying lysis of K562 target cells using freshly isolated PBMCs as effector cells, is a diagnostic criterion for HLH.7  A value of ≤ 10 LU is indicative of HLH. With freshly isolated PBMCs, NK cell-mediated lysis of K562 target cells was defective compared with controls and below the limit set as pathologic in most patients tested (Figure 5A). For patients with biallelic mutations in UNC13D and with at least 1 allele of the intron 1 mutation, all but 1 (patient A, 11 lytic units) displayed pathologic NK cell activity (Figure 5A). Similarly, of the patients with biallelic aberrations in UNC13D and with at least 1 allele of the inversion, all but 1 (patient H, 13 LU) displayed pathologic NK cell activity (Figure 5A). This pathologic NK cell activity was evident despite normal or close to normal NK cell frequencies, apart from for 2 patients, who displayed much reduced frequencies of circulating NK cells (supplemental Table 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). Furthermore, elevated K562 cell lysis coincided with high NK cell frequencies (supplemental Table 1). Degranulation by freshly isolated CD3CD56+ NK cells in response to K562 cells in FHL2 patients carrying biallelic mutations in PRF1 was comparable with that of healthy adult and infant controls, whereas degranulation was < 3.0% in all patients with the intron 1 mutation and most patients carrying the inversion (Figure 5B). Three patients tested during flares displayed degranulation > 3.0% (patients C, Q, and P; supplemental Table 1). On repeated analysis during HLH-2004 therapy or remission, degranulation in these patients was < 3.0%.

Figure 5

Freshly isolated NK cell cytotoxicity and degranulation in FHL3 patients carrying the intron 1 mutation or the 253-kb inversion. Resting PBMCs (A-B) or PBMCs stimulated with IL-2 for 48 hours (C-D) from healthy adult and infant donors or FHL3 patients were evaluated for cytotoxicity and degranulation toward K562 cells. (A,C) NK cell cytotoxicity was evaluated in a 4-hour 51Cr-release assay. Plots represent lytic units calculated at 25% lysis. (B,D) NK cell degranulation was evaluated in a 2-hour flow cytometric assay. The cells were stained with fluorochrome-conjugated anti–CD3, anti–CD56, and anti–CD107a mAbs. Lymphocytes were gated on forward/side scatter plots, followed by gating on CD3 versus CD56 plots. Plots represent induced CD107a surface expression (ΔCD107a+) on CD3CD56+ NK cells. Boxes represent 25th, 50th, and 75th percentiles; pluses, mean values; and error bars, 95% CI. For FHL3 patients, each point represents one person. Intron 1 mutant alleles (blue), inversion alleles (red), and alleles with other mutations (gray) are indicated. Bars represent mean and 95% CI.

Figure 5

Freshly isolated NK cell cytotoxicity and degranulation in FHL3 patients carrying the intron 1 mutation or the 253-kb inversion. Resting PBMCs (A-B) or PBMCs stimulated with IL-2 for 48 hours (C-D) from healthy adult and infant donors or FHL3 patients were evaluated for cytotoxicity and degranulation toward K562 cells. (A,C) NK cell cytotoxicity was evaluated in a 4-hour 51Cr-release assay. Plots represent lytic units calculated at 25% lysis. (B,D) NK cell degranulation was evaluated in a 2-hour flow cytometric assay. The cells were stained with fluorochrome-conjugated anti–CD3, anti–CD56, and anti–CD107a mAbs. Lymphocytes were gated on forward/side scatter plots, followed by gating on CD3 versus CD56 plots. Plots represent induced CD107a surface expression (ΔCD107a+) on CD3CD56+ NK cells. Boxes represent 25th, 50th, and 75th percentiles; pluses, mean values; and error bars, 95% CI. For FHL3 patients, each point represents one person. Intron 1 mutant alleles (blue), inversion alleles (red), and alleles with other mutations (gray) are indicated. Bars represent mean and 95% CI.

Close modal

On stimulation with IL-2 for 30 to 60 hours, K562 cell lysis and NK cell degranulation increased markedly in healthy controls (Figure 5C-D). By comparison, lysis and degranulation were weak in FHL3 patients. Notably, in patients carrying at least 1 allele of the inversion, degranulation by IL-2–stimulated NK cells was significantly augmented compared with FHL3 patients carrying the intron 1 mutation or with other UNC13D mutations (Figure 5D). Accordingly, in FHL3 patients, mean cytotoxicity of IL-2–stimulated NK cells was higher in patients carrying allele(s) of the inversion relative to patients carrying the intron 1 mutation or with other UNC13D mutations (Figure 5C).

Clinical characteristics, neurologic manifestations, and outcome

Altogether, 21 infants with at least one allele with the intron 1 mutation, c.118-308C > T, or the 253-kb inversion were included in this study (Table 1). All patients fulfilled the diagnostic criteria (n = 19) according to the HLH-2004 guidelines7  or had a familial disease (n = 2). The median age at diagnosis of patients with at least one allele with the intron 1 mutation was 4.7 months (142 days; mean, 138 days; range, 55-229 days, n = 8), and the age at diagnosis of the one patient with the mutation in homozygous was 133 days. The median age at diagnosis in the group of patients with the 253-kb inversion in a homozygous state was 3.4 months (104 days; mean 158 days; range, 1-459 days, n = 12). By comparison, the age at diagnosis is similar to patients with 2 disruptive mutations in UNC13D (3 months).39  Thus, although a slight improvement in degranulation and cytotoxicity was seen in the patients carrying the inversion, no difference was seen in terms of age at onset in this group of patients compared with other FHL3 patients.

Consistent with a severe presentation of HLH, neurologic alterations were reported in 11 of 20 patients (55%) of the patients, whereas 15 of 18 (83%) had abnormal cerebrospinal fluid (cells and/or protein levels) during the course of the disease.

Sixteen of 21 patients were treated according to the HLH-2004 protocol, 4 were treated according to HLH-94,7,40  and one patient went directly to hematopoietic stem cell transplantation. Only 4 of 19 patients (21%) were reported in remission at 2 months after treatment. Thirteen of 21 patients (62%) are alive after hematopoietic stem cell transplantation.

Geographic distribution of new UNC13D aberrations

Further examination of the geographic distribution and frequency of different UNC13D aberrations in FHL3 patients from Scandinavian countries Croatia, and Slovenia revealed a high allele frequency of the intron 1 mutation in Croatia and Slovenia, but the mutation was also found in patients from Denmark and southern Sweden (Figure 6A). Microsatellite markers indicated a common haplotype for the intron 1 mutation (Figure 6B), suggesting a founder mutation that has become widely geographically distributed. In contrast, the 253-kb UNC13D inversion was found only in patients of Scandinavian ancestry. All these affected persons shared a common haplotype (Figure 6C). Moreover, a gradient was observed with particularly high frequency of the inversion in patients from northern Scandinavia, suggesting a founder effect. Thus, the geographic distribution and microsatellite analysis revealed distinct prevalence and origins of the 2 relatively frequent UNC13D disease-causing aberrations.

Figure 6

Geographic distribution and microsatellite marker analyses of noncoding UNC13D mutations. (A) Frequency of distinct UNC13D mutations in families from different European countries. All patients with UNC13D studied from Sweden, Denmark, Norway, Finland, Croatia, and Slovenia are included. Pie charts indicate the relative frequency of the intron 1 mutation (blue), the inversion (red), or other mutations (gray). Numbers represent alleles. (B) Haplotype analyses using microsatellite markers of patients with the c.118-308C > T mutation. (C) Haplotype analyses using microsatellite markers of patients with the inversion. (D) Relative frequency of mutated alleles in infants that fulfilled the diagnostic criteria for HLH, including defective NK cell cytotoxicity in Sweden between December 2005 and January 2011. The pie chart includes 13 patients and 26 mutated alleles.

Figure 6

Geographic distribution and microsatellite marker analyses of noncoding UNC13D mutations. (A) Frequency of distinct UNC13D mutations in families from different European countries. All patients with UNC13D studied from Sweden, Denmark, Norway, Finland, Croatia, and Slovenia are included. Pie charts indicate the relative frequency of the intron 1 mutation (blue), the inversion (red), or other mutations (gray). Numbers represent alleles. (B) Haplotype analyses using microsatellite markers of patients with the c.118-308C > T mutation. (C) Haplotype analyses using microsatellite markers of patients with the inversion. (D) Relative frequency of mutated alleles in infants that fulfilled the diagnostic criteria for HLH, including defective NK cell cytotoxicity in Sweden between December 2005 and January 2011. The pie chart includes 13 patients and 26 mutated alleles.

Close modal

We here describe 2 aberrations outside the coding region of UNC13D that together explain HLH in 21 infants with primary HLH included in this study. These aberrations are not detectable by normal sequencing of the exons and exon/intron boundaries. The findings increase the sensitivity of a molecular diagnosis in patients with suspected FHL, as well as extended prenatal diagnostics and carrier testing for families affected by FHL. Furthermore, the results provide insight into lymphocyte-specific transcriptional regulation of UNC13D.

The first aberration, a point mutation, c.118-308C > T, was identified in 7 patients in a heterozygous state and in one patient in a homozygous state, all with early onset HLH. The mutation was widely distributed in patients originating from Croatia, Slovenia, Denmark, and Sweden. Microsatellite analysis revealed a common haplotype. Together, these data suggest a distant mutation event that has become widely geographically distributed and that may explain a number of HLH cases in whites. The mutation was located in an evolutionary conserved region of intron 1. Defective NK cell degranulation and cytotoxicity, as well as absent Munc13-4 expression, was consistent with a loss-of-function mutation in UNC13D. Interestingly, further experiments revealed that this mutation did not impair splicing, as previously suggested,35  but rather reduced UNC13D transcription specifically in lymphocytes. The conserved intronic sequence in which the c.118-308C > T mutation is located displays homology to STAT4 and Ets-1 consensus binding sites, which are disrupted by the mutation. Thus, this sequence may represent an intronic enhancer. STAT4 and Ets-1 are highly expressed in lymphocytes,41,42  may promote perforin expression,43  but have not previously been implicated in regulation of Munc13-4 expression. Binding of transcription factors to this putative regulatory site remains to be explored experimentally, but it is well documented that such intronic enhancers can control cell-specific transcription, as exemplified by the B cell-specific expression of the immunoglobulin locus.44,45 

The second aberration, a 253-kb inversion with one of the breakpoints located within intron 30 of UNC13D, causes a disruption of the gene. Defective NK cell degranulation and cytotoxicity, as well as absent Munc13-4 expression, was consistent with a loss-of-function mutation in UNC13D. Notable, this aberration appears to be the most common cause of FHL in Swedish infants, constituting ∼ 50% of the mutated alleles. The inversion was also identified in HLH patients from neighboring countries. The incidence and geographic distribution of patients carrying this allele suggest a founder effect in Northern Scandinavia. Most likely, the genomic rearrangement occurred by pairing of the inverted Alu repeats, as described in the X-linked recessive hemophilia A and Alport syndrome.46,47  To our knowledge, this is the first paracentric inversion alone that causes an autosomal recessive disease in humans. Functionally, IL-2 stimulation slightly increased degranulation by NK cells carrying the UNC13D inversion relative to NK cells from patients with other UNC13D mutations. UNC13D missense mutations have been associated with milder impairments in NK cell degranulation and cytotoxicity, as well as later disease onset.48,49  Moreover, mutations in STX11 and STXBP2 are associated with partial gain of degranulation after IL-2 stimulation as well as later disease onset relative to patients with nonsense mutations in PRF1 or UNC13D.12,13,28,50  In contrast, we did not find evidence for any later onset of disease in our group of patients with the UNC13D inversion. Thus, the disease relevance of these functional observations is not clear.

Remarkably, with the discovery of the 2 noncoding UNC13D aberrations, a molecular diagnosis could be provided for all 13 infants from Sweden that presented with HLH and defective NK cell cytotoxicity between December 2005 and January 2011 (Figure 6D). The UNC13D c.118-308C > T mutation in intron 1 corresponded to 3.8% of the mutated alleles in Sweden, whereas the UNC13D 253-kb inversion represented 50% of the mutated alleles in this patient cohort. When only considering patients of European ancestry, the c.118-308C > T mutation corresponded to 4.5% of the mutated alleles in Sweden, whereas the 253-kb inversion represented 59.1% of the mutated alleles. Although our analysis suggests a relatively confined geographic distribution of the UNC13D inversion to Northern Sweden, this aberration may also exist in the Northern American population with Scandinavian ancestry. The newly developed multiplex PCR assay for detection of the UNC13D inversion can facilitate a rapid molecular diagnosis of patients carrying this aberration and can be implemented for future Guthrie-based neonatal screening.

Taken together, identification of the 2 genetic aberrations described here provides a molecular diagnosis for the majority of HLH cases among infants in northern Europe. To our knowledge, the UNC13D inversion represents the first paracentric inversion causing an autosomal recessive disease in humans. Furthermore, with its widespread distribution and abundance across Europe, the UNC13D intron 1 mutation may explain many nonconsanguineous white HLH cases. This mutation identified an intronic sequence required for UNC13D transcription in lymphocytes, advancing our knowledge of the genetic regulation of lymphocyte cytotoxicity. These findings highlight aberrations outside the coding regions as a cause of disease and make a case for sequencing of evolutionary conserved noncoding regions in diagnostics of primary immunodeficiency syndromes.

An Inside Blood analysis of this article appears at the front of this issue.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the patients and their families for participation and Sigrid Sahlén and Mårten Winge for technical assistance.

This work was supported by the Swedish Children's Cancer Foundation, Swedish Research Council, Cancer and Allergy Foundation of Sweden, Swedish Cancer Foundation, Mary Béve Foundation, Märta and Gunnar V Philipson Foundation, David and Astrid Hageléns Foundation, Histiocytosis Association of America, Tobias Foundation, Åke Olsson Foundation for Hematologic Research, Swedish Society for Medical Research, Clas Groschinsky's Memorial Fund, Shizu Matsumara's Donation, the Karolinska Institute Research Foundation, and the Stockholm County Council (ALF project). M.M. was supported by Karolinska Institutet (MD/PhD scholarship and research internship).

Contribution: M.M. designed research and performed genetic analysis, analyzed and interpreted data, and drafted the manuscript; S.C.C.C. performed evaluations of NK cell activity and interpreted data; S.M.W. performed evaluations of NK cell activity and Western blot and interpreted data; M.E. performed and interpreted genetic analyses, including microsatellite analyses; H.S. performed FACS analysis; B.B. and E.N. performed genetic analysis; C.B., G.J., J.J., H.H., B.-M.H., L.R., S.P., S.R., M.S., T.T.S., T.S., and J.W. cared for patients and provided clinical data; H.-G.L., B.F., and M.N. designed research and contributed to drafting the manuscript; J.-I.H. initiated the study, designed research, analyzed and interpreted data, and drafted the manuscript; and Y.T.B. designed research, analyzed and interpreted data, and drafted the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Marie Meeths, Childhood Cancer Research Unit, CMM L8:02, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden; e-mail: marie.meeths@ki.se; and Yenan T. Bryceson, Center for Infectious Medicine, F59, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, S-141 86 Stockholm, Sweden; e-mail: yenan.bryceson@ki.se.

1
Henter
 
JI
Tondini
 
C
Pritchard
 
J
Histiocyte disorders.
Crit Rev Oncol Hematol
2004
, vol. 
50
 
2
(pg. 
157
-
174
)
2
Janka
 
GE
Hemophagocytic syndromes.
Blood Rev
2007
, vol. 
21
 
5
(pg. 
245
-
253
)
3
Filipovich
 
AH
Hemophagocytic lymphohistiocytosis and related disorders.
Curr Opin Allergy Clin Immunol
2006
, vol. 
6
 
6
(pg. 
410
-
415
)
4
Gupta
 
S
Weitzman
 
S
Primary and secondary hemophagocytic lymphohistiocytosis: clinical features, pathogenesis and therapy.
Expert Rev Clin Immunol
2010
, vol. 
6
 
1
(pg. 
137
-
154
)
5
Henter
 
JI
Arico
 
M
Elinder
 
G
Imashuku
 
S
Janka
 
G
Familial hemophagocytic lymphohistiocytosis: primary hemophagocytic lymphohistiocytosis.
Hematol Oncol Clin North Am
1998
, vol. 
12
 
2
(pg. 
417
-
433
)
6
Janka
 
G
Imashuku
 
S
Elinder
 
G
Schneider
 
M
Henter
 
JI
Infection- and malignancy-associated hemophagocytic syndromes: secondary hemophagocytic lymphohistiocytosis.
Hematol Oncol Clin North Am
1998
, vol. 
12
 
2
(pg. 
435
-
444
)
7
Henter
 
JI
Horne
 
A
Arico
 
M
, et al. 
HLH-2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis.
Pediatr Blood Cancer
2007
, vol. 
48
 
2
(pg. 
124
-
131
)
8
Jordan
 
MB
Filipovich
 
AH
Hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis: a journey of a thousand miles begins with a single (big) step.
Bone Marrow Transplant
2008
, vol. 
42
 
7
(pg. 
433
-
437
)
9
Stepp
 
SE
Dufourcq-Lagelouse
 
R
Le Deist
 
F
, et al. 
Perforin gene defects in familial hemophagocytic lymphohistiocytosis.
Science
1999
, vol. 
286
 
5446
(pg. 
1957
-
1959
)
10
Feldmann
 
J
Callebaut
 
I
Raposo
 
G
, et al. 
Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3).
Cell
2003
, vol. 
115
 
4
(pg. 
461
-
473
)
11
zur Stadt
 
U
Schmidt
 
S
Kasper
 
B
, et al. 
Linkage of familial hemophagocytic lymphohistiocytosis (FHL) type-4 to chromosome 6q24 and identification of mutations in syntaxin 11.
Hum Mol Genet
2005
, vol. 
14
 
6
(pg. 
827
-
834
)
12
zur Stadt
 
U
Rohr
 
J
Seifert
 
W
, et al. 
Familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) is caused by mutations in Munc18–2 and impaired binding to syntaxin 11.
Am J Hum Genet
2009
, vol. 
85
 
4
(pg. 
482
-
492
)
13
Côte
 
M
Menager
 
MM
Burgess
 
A
, et al. 
Munc18–2 deficiency causes familial hemophagocytic lymphohistiocytosis type 5 and impairs cytotoxic granule exocytosis in patient NK cells.
J Clin Invest
2009
, vol. 
119
 
12
(pg. 
3765
-
3773
)
14
Nichols
 
KE
In FHL4, NK cells pack less of a punch.
Blood
2007
, vol. 
110
 
6
(pg. 
1705
-
1706
)
15
de Saint Basile
 
G
Menasche
 
G
Fischer
 
A
Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules.
Nat Rev Immunol
2010
, vol. 
10
 
8
(pg. 
568
-
579
)
16
Cetica
 
V
Pende
 
D
Griffiths
 
GM
Arico
 
M
Molecular basis of familial hemophagocytic lymphohistiocytosis.
Haematologica
2010
, vol. 
95
 
4
(pg. 
538
-
541
)
17
Sandrock
 
K
Nakamura
 
L
Vraetz
 
T
Beutel
 
K
Ehl
 
S
Zieger
 
B
Platelet secretion defect in patients with familial hemophagocytic lymphohistiocytosis type 5 (FHL-5).
Blood
2010
, vol. 
116
 
26
(pg. 
6148
-
6150
)
18
Ren
 
Q
Wimmer
 
C
Chicka
 
MC
, et al. 
Munc13-4 is a limiting factor in the pathway required for platelet granule release and hemostasis.
Blood
2010
, vol. 
116
 
6
(pg. 
869
-
877
)
19
Booth
 
C
Gilmour
 
KC
Veys
 
P
, et al. 
X-linked lymphoproliferative disease due to SAP/SH2D1A deficiency: a multicenter study on the manifestations, management and outcome of the disease.
Blood
2011
, vol. 
117
 
1
(pg. 
53
-
62
)
20
Coffey
 
AJ
Brooksbank
 
RA
Brandau
 
O
, et al. 
Host response to EBV infection in X-linked lymphoproliferative disease results from mutations in an SH2-domain encoding gene.
Nat Genet
1998
, vol. 
20
 
2
(pg. 
129
-
135
)
21
Rigaud
 
S
Fondaneche
 
MC
Lambert
 
N
, et al. 
XIAP deficiency in humans causes an X-linked lymphoproliferative syndrome.
Nature
2006
, vol. 
444
 
7115
(pg. 
110
-
114
)
22
Marsh
 
RA
Madden
 
L
Kitchen
 
BJ
, et al. 
XIAP deficiency: a unique primary immunodeficiency best classified as X-linked familial hemophagocytic lymphohistiocytosis and not as X-linked lymphoproliferative disease.
Blood
2010
, vol. 
116
 
7
(pg. 
1079
-
1082
)
23
Pachlopnik Schmid
 
J
Canioni
 
D
Moshous
 
D
, et al. 
Clinical similarities and differences of patients with X-linked lymphoproliferative syndrome type 1 (XLP-1/SAP deficiency) versus type 2 (XLP-2/XIAP deficiency).
Blood
2011
, vol. 
117
 
5
(pg. 
1522
-
1529
)
24
Menasche
 
G
Pastural
 
E
Feldmann
 
J
, et al. 
Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome.
Nat Genet
2000
, vol. 
25
 
2
(pg. 
173
-
176
)
25
Barbosa
 
MD
Barrat
 
FJ
Tchernev
 
VT
, et al. 
Identification of mutations in two major mRNA isoforms of the Chediak-Higashi syndrome gene in human and mouse.
Hum Mol Genet
1997
, vol. 
6
 
7
(pg. 
1091
-
1098
)
26
zur Stadt
 
U
Beutel
 
K
Kolberg
 
S
, et al. 
Mutation spectrum in children with primary hemophagocytic lymphohistiocytosis: molecular and functional analyses of PRF1, UNC13D, STX11, and RAB27A.
Hum Mutat
2006
, vol. 
27
 
1
(pg. 
62
-
68
)
27
Horne
 
A
Ramme
 
KG
Rudd
 
E
, et al. 
Characterization of PRF1, STX11 and UNC13D genotype-phenotype correlations in familial hemophagocytic lymphohistiocytosis.
Br J Haematol
2008
, vol. 
143
 
1
(pg. 
75
-
83
)
28
Bryceson
 
YT
Rudd
 
E
Zheng
 
C
, et al. 
Defective cytotoxic lymphocyte degranulation in syntaxin-11 deficient familial hemophagocytic lymphohistiocytosis 4 (FHL4) patients.
Blood
2007
, vol. 
110
 
6
(pg. 
1906
-
1915
)
29
Bryceson
 
YT
Fauriat
 
C
Nunes
 
JM
, et al. 
Functional analysis of human NK cells by flow cytometry.
Methods Mol Biol
2010
, vol. 
612
 (pg. 
335
-
352
)
30
Meeths
 
M
Bryceson
 
YT
Rudd
 
E
, et al. 
Clinical presentation of Griscelli syndrome type 2 and spectrum of RAB27A mutations.
Pediatr Blood Cancer
2010
, vol. 
54
 
4
(pg. 
563
-
572
)
31
Santoro
 
A
Cannella
 
S
Bossi
 
G
, et al. 
Novel Munc13-4 mutations in children and young adult patients with haemophagocytic lymphohistiocytosis.
J Med Genet
2006
, vol. 
43
 
12
(pg. 
953
-
960
)
32
Ramensky
 
V
Bork
 
P
Sunyaev
 
S
Human non-synonymous SNPs: server and survey.
Nucleic Acids Res
2002
, vol. 
30
 
17
(pg. 
3894
-
3900
)
33
Ng
 
PC
Henikoff
 
S
Predicting deleterious amino acid substitutions.
Genome Res
2001
, vol. 
11
 
5
(pg. 
863
-
874
)
34
Siepel
 
A
Bejerano
 
G
Pedersen
 
JS
, et al. 
Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes.
Genome Res
2005
, vol. 
15
 
8
(pg. 
1034
-
1050
)
35
Santoro
 
A
Cannella
 
S
Trizzino
 
A
, et al. 
Mutations affecting mRNA splicing are the most common molecular defect in patients with familial hemophagocytic lymphohistiocytosis type 3.
Haematologica
2008
, vol. 
93
 
7
(pg. 
1086
-
1090
)
36
Marsh
 
RA
Vaughn
 
G
Kim
 
MO
, et al. 
Reduced-intensity conditioning significantly improves survival of patients with hemophagocytic lymphohistiocytosis undergoing allogeneic hematopoietic cell transplantation.
Blood
2010
, vol. 
116
 
26
(pg. 
5824
-
5831
)
37
Ouachee-Chardin
 
M
Elie
 
C
de Saint Basile
 
G
, et al. 
Hematopoietic stem cell transplantation in hemophagocytic lymphohistiocytosis: a single-center report of 48 patients.
Pediatrics
2006
, vol. 
117
 
4
(pg. 
e743
-
e750
)
38
Kent
 
WJ
BLAT–the BLAST-like alignment tool.
Genome Res
2002
, vol. 
12
 
4
(pg. 
656
-
664
)
39
Sieni
 
E
Cetica
 
V
Santoro
 
A
, et al. 
Genotype-phenotype study of familial haemophagocytic lymphohistiocytosis type 3.
J Med Genet
2011
, vol. 
48
 
5
(pg. 
343
-
352
)
40
Henter
 
JI
Arico
 
M
Egeler
 
RM
, et al. 
HLH-94: a treatment protocol for hemophagocytic lymphohistiocytosis. HLH study Group of the Histiocyte Society.
Med Pediatr Oncol
1997
, vol. 
28
 
5
(pg. 
342
-
347
)
41
Dittmer
 
J
The biology of the Ets1 proto-oncogene.
Mol Cancer
2003
, vol. 
2
 pg. 
29
 
42
Bacon
 
CM
Petricoin
 
EF
Ortaldo
 
JR
, et al. 
Interleukin 12 induces tyrosine phosphorylation and activation of STAT4 in human lymphocytes.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 
16
(pg. 
7307
-
7311
)
43
Glimcher
 
LH
Townsend
 
MJ
Sullivan
 
BM
Lord
 
GM
Recent developments in the transcriptional regulation of cytolytic effector cells.
Nat Rev Immunol
2004
, vol. 
4
 
11
(pg. 
900
-
911
)
44
Gillies
 
SD
Morrison
 
SL
Oi
 
VT
Tonegawa
 
S
A tissue-specific transcription enhancer element is located in the major intron of a rearranged immunoglobulin heavy chain gene.
Cell
1983
, vol. 
33
 
3
(pg. 
717
-
728
)
45
Maniatis
 
T
Goodbourn
 
S
Fischer
 
JA
Regulation of inducible and tissue-specific gene expression.
Science
1987
, vol. 
236
 
4806
(pg. 
1237
-
1245
)
46
Lakich
 
D
Kazazian
 
HH
Antonarakis
 
SE
Gitschier
 
J
Inversions disrupting the factor VIII gene are a common cause of severe haemophilia A.
Nat Genet
1993
, vol. 
5
 
3
(pg. 
236
-
241
)
47
Hertz
 
JM
Persson
 
U
Juncker
 
I
Segelmark
 
M
Alport syndrome caused by inversion of a 21 Mb fragment of the long arm of the X-chromosome comprising exon 9 through 51 of the COL4A5 gene.
Hum Genet
2005
, vol. 
118
 
1
(pg. 
23
-
28
)
48
Rudd
 
E
Bryceson
 
YT
Zheng
 
C
, et al. 
Spectrum, and clinical and functional implications of UNC13D mutations in familial haemophagocytic lymphohistiocytosis.
J Med Genet
2008
, vol. 
45
 
3
(pg. 
134
-
141
)
49
Meeths
 
M
Entesarian
 
M
Al-Herz
 
W
, et al. 
Spectrum of clinical presentations in familial hemophagocytic lymphohistiocytosis type 5 patients with mutations in STXBP2.
Blood
2010
, vol. 
116
 
15
(pg. 
2635
-
2643
)
50
Rohr
 
J
Beutel
 
K
Maul-Pavicic
 
A
, et al. 
Atypical familial hemophagocytic lymphohistiocytosis due to mutations in UNC13D and STXBP2 overlaps with primary immunodeficiency diseases.
Haematologica
2010
, vol. 
95
 
12
(pg. 
2080
-
2087
)

Author notes

*

S.C.C.C. and S.M.W. contributed equally to this study.

J.-I.H. and Y.T.B. share senior authorship.

Sign in via your Institution