• TCR-triggered T-cell and Fc receptor–triggered NK-cell assays are most accurate for diagnosing defective cytotoxic lymphocyte exocytosis.

  • The standard K562 cell assay has high interindividual variability and is affected by expanded NK-cell subsets and transportation stress.

Abstract

Primary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening disorder associated with autosomal recessive variants in genes required for perforin-mediated lymphocyte cytotoxicity. A rapid diagnosis is crucial for successful treatment. Although defective cytotoxic T lymphocyte (CTL) function causes pathogenesis, quantification of natural killer (NK)–cell exocytosis triggered by K562 target cells currently represents a standard diagnostic procedure for primary HLH. We have prospectively evaluated different lymphocyte exocytosis assays in 213 patients referred for evaluation for suspected HLH and related hyperinflammatory syndromes. A total of 138 patients received a molecular diagnosis consistent with primary HLH. Assessment of Fc receptor–triggered NK-cell and T-cell receptor (TCR)–triggered CTL exocytosis displayed higher sensitivity and improved specificity for the diagnosis of primary HLH than routine K562 cell–based assays, with these assays combined providing a sensitivity of 100% and specificity of 98.3%. By comparison, NK-cell exocytosis after K562 target cell stimulation displayed a higher interindividual variability, in part explained by differences in NK-cell differentiation or large functional reductions after shipment. We thus recommend combined analysis of TCR-triggered CTL and Fc receptor–triggered NK-cell exocytosis for the diagnosis of patients with suspected familial HLH or atypical manifestations of congenital defects in lymphocyte exocytosis.

Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory disorder characterized by unremitting fever, splenomegaly, cytopenia, hypertriglyceridemia, hypofibrinogenemia, hemophagocytosis, as well as high serum ferritin and soluble CD25.1-4 Early-onset, familial forms of HLH (FHL) are associated with autosomal recessive variants in PRF1, UNC13D, STXBP2, and STX11 encoding perforin, Munc13-4, Munc18-2, and syntaxin-11, respectively. Perforin expression is mainly restricted to nature killer (NK) cells as well as subsets of CD8+ T cells, where it is contained in cytotoxic granules.5,6 In contrast, Munc13-4, Munc18-2, and syntaxin-11 are cytosolic proteins that mediate exocytosis by hematopoietic cells. The similarities in clinical presentation of patients with perforin-, Munc13-4–, Munc18-2–, and syntaxin-11–deficiency underscore their pivotal role in facilitating target cell killing by cytotoxic T lymphocytes (CTLs) and NK cells.7-10 Individuals with autosomal recessive variants in RAB27A, LYST, and AP3B1 also display defective cytotoxic granule exocytosis and frequently develop HLH, with a majority manifesting characteristic hypopigmentation.11-13 Impaired cytotoxic granule exocytosis has also been reported in patients with autosomal recessive AP3D1, MADD, ORAI1, RHOG, and STIM1.14-18 Individuals with variants in SH2D1A, XIAP, CD27, or ITK may also manifest with HLH but lack a gross defect in lymphocyte cytotoxicity.19-23 Individuals diagnosed with other inborn errors of immunity (IEI) may also develop HLH without displaying defective lymphocyte cytotoxicity.24,25 HLH may also present in the context of infections or malignancy in individuals seemingly lacking a genetic predisposition.26 

Primary defects in lymphocyte cytotoxicity can be rapidly fatal unless promptly treated.27 Although first-line treatment involves immunosuppression, the only currently available cure is allogeneic hematopoietic stem cell transplantation. Gene therapeutic approaches are being investigated.1,28,29 Advances in DNA sequencing technology can facilitate rapid molecular diagnoses but may miss pathogenic noncoding genetic aberrations.30-33 Moreover, variants of unknown significance require functional validation. Thus, functional analysis of lymphocyte cytotoxicity can provide timely and necessary answers that support vital treatment decisions.4,8 

According to the HLH-2004 guidelines, defective NK-cell activity, as measured by the 51Cr-labeled K562 target cell lysis assay, represents 1 criterion for the diagnosis of HLH.1 Using radioactive isotope, which is prohibited in many laboratories, this assay requires relatively large numbers of peripheral blood mononuclear cells (PBMCs). Results are also dependent on the frequency of NK cells among PBMCs, which may be influenced by inflammation. Hence, the 51Cr-labeled K562 target cell lysis assay does not unequivocally reflect the killing capacity of NK cells.34,35 Laboratories have therefore adopted assays quantifying intracellular perforin expression as well as induced surface expression of CD107a (also known as lysosomal-associated membrane protein 1) in response to K562 cells for the diagnosis of patients with biallelic loss-of-function (LoF) variants in PRF1 or UNC13D, STX11, STXBP2, RAB27A, and LYST.21,36-38 

Prf1 knockout mice have demonstrated that defective CTL activity is key to the pathogenesis of HLH, whereas defective NK-cell cytotoxicity may hamper control of adaptive immune responses and exacerbate disease.39,40 Thus, measurements of CTL lytic activity may be of particular value for the diagnosis of primary patients with HLH. Additionally, stimulation can lead to functional recovery of NK- and T-cell exocytosis in patients with LoF variants in RAB27A, STX11, STXBP2, and LYST.8-10,21,41-43 Based on phenotypic characterization of peripheral blood CTL as CD8+CD57+ T cells, we previously published a protocol for sensitive evaluation of CTL exocytosis.6 This assay aided the diagnosis of patients with UNC13D variants, in which NK-cell assays yielded contradictory results.44 Importantly, the efficacy of different NK-cell and CTL exocytosis assays for the overall diagnosis of patients with defects in lymphocyte cytotoxicity has not been systematically evaluated.

Here, we detail interindividual differences in cytotoxic lymphocyte exocytosis in a large cohort of healthy individuals and prospectively evaluate the efficacy of CTL and NK-cell exocytosis assays for patients with suspected primary HLH. Quantification of CTL exocytosis triggered by T-cell receptor (TCR) engagement or NK-cell exocytosis triggered by Fc receptor CD16 engagement provided the highest levels of accuracy for the diagnosis of primary defects in cytotoxic lymphocyte exocytosis, with K562 cell–triggered exocytosis assays being less sensitive. Combined evaluation of distinct cytotoxic lymphocyte subsets provided the most reliable and accurate diagnosis of patients with defects in cytotoxic lymphocyte exocytosis.

Cells

This study was approved by the Stockholm Regional Ethical Review Board. Patient samples were collected between November 2011 and May 2022. Healthy individual samples were obtained from the Karolinska University Hospital Blood Bank. For methods on sequencing, please refer to the supplemental Data, available on the Blood website.

Heparinized blood was received at room temperature, and PBMCs were extracted via density gradient centrifugation (Lymphoprep; Axis-Shield). From adults, 10 mL of blood was requested, whereas between 1 and 10 mL was obtained from infants and children. Live nucleated cells were counted (Muse; Cytek) and plated overnight unstimulated in complete medium (RPMI 1640 medium supplemented with 10% fetal bovine serum and 2 mM L-glutamate; all Hyclone). K562 cells (CCL243; American Type Culture Collection [ATCC]) and P815 cells (TIB64; ATCC) were maintained in complete medium. In total, 8 × 105 PBMCs were generally used for individual analyses. A detailed protocol is specified in the supplemental Material.

Flow cytometric assays

Assessment of exocytosis was performed as previously described.6 Briefly, PBMCs were stimulated for 3 hours at with either K562 cells (CCL-243; ATCC), P815 cells (TIB-64; ATCC) coated with anti-CD16 antibody (3G8; BD Biosciences), or P815 cells coated with anti-CD3 (OKT3, BD Biosciences or S4.1; Thermo Fisher) antibody at 1:1 effector-to-target cell ratio. Cells were then surfaced stained with antibodies to CD3 (OKT3), CD8 (SK1), CD56 (HCD56), CD57 (HNK1), as well as an amine reactive viability dye (Thermo Fisher) and CD107a (H4A3). This test was also performed in combination with intracellular staining of signaling proteins in which surface staining was followed by intracellular staining with anti-FcεRγ (polyclonal rabbit; Upstate), anti-EAT-2 (polyclonal rabbit; Proteintech), and anti-SYK (clone 4D10.2; eBioscience) antibodies, as previously detailed.45 Cells were acquired on a 4 laser BD Fortessa instrument. Flow cytometry data were analyzed with Flowjo (v9.9, Treestar).21 Gates for NK cells (Live CD3CD56+) and CTL (Live CD3+CD8+CD57+) were made based on the mock stimulated cells. These were then juxtaposed onto the various stimulated tubes, in which the delta value of CD107a expression was calculated by deducting the levels of background.

Statistical analysis

Statistics were generated with Prism (v7, GraphPad) as well as pROC R (v3.0.2) package for the generation of receiver operating characteristic curves.46 Youden index and discriminant power (DP) were calculated as described.47,48 Assay accuracy was defined as follows: (true positive + true negative)/(true positive + false positive + false negative + true negative). Unpaired t tests are reported as ∗P < .05; ∗∗P < .01; ∗∗∗P < .001; and ∗∗∗∗P < .0001. Boxes and whisker plots indicate quartiles (25th, 50th, and 75th) and 2.5th and 97.5th percentiles for whiskers, and points were plotted individually for outliers. Bar graphs show lines representing standard deviation. Specific statistics used are reported in the figure legends.

This study was approved by the Stockholm Regional Ethical Review Board.

Performance of different cytotoxic lymphocyte exocytosis assays in healthy adults

In our initial description of a sensitive T-cell exocytosis assay, analysis of PBMCs from 14 healthy adult volunteers revealed superior responses by CD8+CD57dim and CD8+CD57bright T cells after TCR engagement compared with other T-cell as well as NK-cell subsets after antibody-mediated engagement of CD16 or K562 cell stimulation.6 To validate these findings from a large number of healthy adults, we assessed cytotoxic lymphocyte exocytosis in 198 healthy adult volunteers (Figure 1A). Substantiating previous findings, assessment of CD107a surface expression in response to engagement of the TCR on CD8+CD57dim and CD8+CD57bright T cells or CD16 on NK cells using anti-CD3 or anti-CD16 monoclonal antibodies, respectively, demonstrated the strongest responses with a coefficient of variation of 0.22, 0.14, and 0.20, respectively. By comparison, CD3CD56dim NK-cell responses to K562 cells and CD8+CD57 T-cell responses to TCR engagement were weaker and displayed greater donor variability (coefficient of variation of 0.33 and 0.33, respectively). In summary, the exocytic response of cytotoxic CD8+CD57+ T cells upon TCR engagement or CD3CD56dim NK cells in response to CD16 engagement was the most robust.

Figure 1.

Analysis of experimental interassay variability in evaluation of cytotoxic lymphocyte exocytosis. (A) PBMCs from 198 healthy adult volunteers were incubated for 3 hours in medium or with target cells and monoclonal antibodies (mAbs) as indicated. Induced CD107a surface expression (ΔCD107a) values for distinct cytotoxic lymphocyte subsets and stimulations, as indicated, in 198 healthy adult volunteers. Graph depicts mean values with bars indicating standard deviation (SD). (B-F) Compiled induced CD107a surface expression values for distinct cytotoxic lymphocyte subsets and stimulations, as indicated, from 2 healthy adult volunteers run 17 (donor 1) and 23 times (donor 2) using multiple frozen PBMC vials thawed over a period of 3 months. Samples were run using the same exocytosis protocol. Shown are the ΔCD107a for CD3CD56dim NK cells after K562 or anti-CD16 stimulation and CD8+CD57+ T cells (CTL) after anti-CD3 stimulation. Results plotted against time for donor 1 (B) and donor 2 (C), displaying results of the 3 different exocytosis stimulations. The same data were normalized by dividing each value by respective mean and plotted against time for donor 1 (D) and donor 2 (E), demonstrating the range of deviation for each assay and donor. (F) Accumulated normalized data for each assay. Bars indicate SD.

Figure 1.

Analysis of experimental interassay variability in evaluation of cytotoxic lymphocyte exocytosis. (A) PBMCs from 198 healthy adult volunteers were incubated for 3 hours in medium or with target cells and monoclonal antibodies (mAbs) as indicated. Induced CD107a surface expression (ΔCD107a) values for distinct cytotoxic lymphocyte subsets and stimulations, as indicated, in 198 healthy adult volunteers. Graph depicts mean values with bars indicating standard deviation (SD). (B-F) Compiled induced CD107a surface expression values for distinct cytotoxic lymphocyte subsets and stimulations, as indicated, from 2 healthy adult volunteers run 17 (donor 1) and 23 times (donor 2) using multiple frozen PBMC vials thawed over a period of 3 months. Samples were run using the same exocytosis protocol. Shown are the ΔCD107a for CD3CD56dim NK cells after K562 or anti-CD16 stimulation and CD8+CD57+ T cells (CTL) after anti-CD3 stimulation. Results plotted against time for donor 1 (B) and donor 2 (C), displaying results of the 3 different exocytosis stimulations. The same data were normalized by dividing each value by respective mean and plotted against time for donor 1 (D) and donor 2 (E), demonstrating the range of deviation for each assay and donor. (F) Accumulated normalized data for each assay. Bars indicate SD.

Close modal

Interassay reproducibility of cytotoxic lymphocyte exocytosis assays

Interassay variability may represent a confounding factor in the diagnosis of HLH. Therefore, we repeatedly tested the same 2 donors over 3 months, examining exocytosis by CD8+CD57+ T cells in response to TCR engagement and CD3CD56dim NK cells in response to CD16 engagement or K562 cell stimulation. PBMCs cryopreserved in multiple vials were thawed and run at different time points. Results were comparable throughout the period (Figure 1B-C). Although numerical values for exocytosis varied more for TCR engagement or CD16 engagement (Figure 1B-C; supplemental Table 1), the greatest relative variability was observed for CD3CD56dim NK-cell responses to K562 cells (Figure 1D-F). Nonetheless, the overall high interassay consistency provided confidence when comparing data accumulated from independent experiments collected over several years.

Variables associated with low cytotoxic lymphocyte exocytosis in healthy adults

With available data on sex, age, cytomegalovirus (CMV) serostatus, cell numbers, and differentiation of the 198 healthy adult volunteers for whom cytotoxic lymphocyte function was examined, additional factors influencing the magnitude of response in distinct cell subsets could be interrogated. When stratified by gender or age, no statistical difference was found among the 3 exocytosis assays (supplemental Figure 1).

Forming latent infection, CMV imprints human immune cell constitution and function, foremost promoting effector CD8+ T-cell differentiation, elevating serum interleukin-6 (IL-6) and IL-10, and influencing responses to these cytokines.49,50 Notably, CD8+CD57+ T-cell exocytosis upon TCR engagement was somewhat higher in CMV seropositive individuals, whereas NK-cell responses to CD16 engagement or K562 cells were not correlated to CMV serostatus (Figure 2A).

Figure 2.

Analysis of the variability in cytotoxic lymphocyte exocytosis in healthy adults. (A-H) PBMCs from 198 healthy adult volunteers were incubated for 3 hours in medium or with target cells and mAbs as indicated. (A) Graph displays levels of exocytosis in CD8+CD57+ T cells or CD3CD56dim NK cells stratified according to CMV serostatus. Boxes and whiskers display quartiles, limits, as well as outliers, with plus signs representing means. Pairs statistically different by Student t test are indicated. (B) Flow cytometry plots show CD3CD56dim NK-cell CD107a surface expression in relation to intracellular expression of FcεRγ or EAT-2, after indicated stimulations. The percentages of cells in each quadrant are indicated. Only 5.4% of FcεRγ NK cells exocytosed after K562 stimulation, compared with 21.7% of FcεRγ+ NK cells. Similarly, 6.7% of EAT-2 NK cells exocytosed when stimulated with K562, compared with 25% of EAT-2+ NK cells. (C-D) Graphs displaying the relationship between CD3CD56dim NK-cell–induced CD107a surface expression and frequencies of cells lacking expression of FcεRγ, EAT-2, or SYK after stimulation with P815 cells added anti-CD16 mAb (C) or K562 cells (D). Diagonal lines represent linear regression and with 95% confidence intervals in bold, whereas the dashed horizontal line indicates the cutoff percentage deemed abnormal in regard to K562 cell–induced NK-cell exocytosis. (E) The graph depicts frequencies of CD107a-expressing NK cells according to stimulation and expression of FcεRγ, SYK, and EAT-2 in CD3CD56dim NK-cell subsets, with bars representing SD. (F-H) Graphs correlating exocytic responses in cytotoxic lymphocyte subsets in healthy individuals, as specified. Donors with low CD107a response after K562 stimulation and expanded FcεRγ NK-cell populations are highlighted with red and orange dots, whereas donors with low CD107a responses to anti-CD3 and anti-CD16 are highlighted with blue dots.

Figure 2.

Analysis of the variability in cytotoxic lymphocyte exocytosis in healthy adults. (A-H) PBMCs from 198 healthy adult volunteers were incubated for 3 hours in medium or with target cells and mAbs as indicated. (A) Graph displays levels of exocytosis in CD8+CD57+ T cells or CD3CD56dim NK cells stratified according to CMV serostatus. Boxes and whiskers display quartiles, limits, as well as outliers, with plus signs representing means. Pairs statistically different by Student t test are indicated. (B) Flow cytometry plots show CD3CD56dim NK-cell CD107a surface expression in relation to intracellular expression of FcεRγ or EAT-2, after indicated stimulations. The percentages of cells in each quadrant are indicated. Only 5.4% of FcεRγ NK cells exocytosed after K562 stimulation, compared with 21.7% of FcεRγ+ NK cells. Similarly, 6.7% of EAT-2 NK cells exocytosed when stimulated with K562, compared with 25% of EAT-2+ NK cells. (C-D) Graphs displaying the relationship between CD3CD56dim NK-cell–induced CD107a surface expression and frequencies of cells lacking expression of FcεRγ, EAT-2, or SYK after stimulation with P815 cells added anti-CD16 mAb (C) or K562 cells (D). Diagonal lines represent linear regression and with 95% confidence intervals in bold, whereas the dashed horizontal line indicates the cutoff percentage deemed abnormal in regard to K562 cell–induced NK-cell exocytosis. (E) The graph depicts frequencies of CD107a-expressing NK cells according to stimulation and expression of FcεRγ, SYK, and EAT-2 in CD3CD56dim NK-cell subsets, with bars representing SD. (F-H) Graphs correlating exocytic responses in cytotoxic lymphocyte subsets in healthy individuals, as specified. Donors with low CD107a response after K562 stimulation and expanded FcεRγ NK-cell populations are highlighted with red and orange dots, whereas donors with low CD107a responses to anti-CD3 and anti-CD16 are highlighted with blue dots.

Close modal

CMV infection is associated with clonal expansions of long-lived, adaptive NK cells that lack expression of certain cytosolic signaling proteins in a variegated manner.45,51-54 Such adaptive NK cells typically constitute a small subset of CD56dim NK cells in most CMV seropositive individuals but may in some individuals dominate the NK-cell repertoire. Because adaptive NK cells display altered signaling and target cell recognition, we hypothesized that variability in NK-cell responses to K562 cells may be associated with adaptive NK-cell expansions. Indeed, NK cells lacking FcεRγ or SYK expression displayed less exocytosis in response to K562 cell stimulation, whereas responses to engagement of CD16 were maintained (Figure 2B). Among the 198 healthy individuals examined, expansions of NK cells lacking FcεRγ, EAT-2, or SYK expression did not affect CD3CD56dim NK-cell responses to CD16 engagement (Figure 2C). However, highly reduced FcεRγ or SYK expression correlated with low CD3CD56dim NK-cell exocytosis upon K562 cell stimulation (Figure 2D). A few healthy individuals with large adaptive CD3CD56dimFcεRγ NK-cell subsets displayed K562 cell–induced exocytosis below the 10% threshold that has been deemed abnormal (Figure 2D).21 Lack of FcεRγ, EAT-2, and SYK expression is stochastically interlinked.45 Boolean gating of adaptive NK-cell subsets demonstrated that lack of FcεRγ, but not EAT-2 or SYK, specifically correlated with reduced NK-cell exocytosis in response to K562 cell stimulation (Figure 2E). Thus, NK-cell responses to K562 cells were diminished in individuals with a high proportion of adaptive NK cells lacking expression of FcεRγ.

Finally, we compared TCR-triggered exocytosis in CD8+CD57+ T cells with CD16-triggered exocytosis in CD3CD56dim NK cells. Notably, a few donors displayed low responses to both these stimuli and additionally displayed low CD3CD56dim NK-cell exocytosis in response to K562 cells (Figure 2F-G) but were distinct from low responders with respect to K562 cell recognition, explained by epigenetic regulation of signaling protein expression (Figure 2H). Thus, combined assays may discriminate healthy adult individuals with overall mild impairments in cytotoxic lymphocyte exocytosis.

Prospective evaluation of cytotoxic lymphocyte exocytosis assays in hyperinflammatory patients

To gain insights to the efficacy of different cytotoxic lymphocyte exocytosis assays for the diagnosis of FHL, we prospectively evaluated cohorts of patients fulfilling HLH criteria, carrying HLH-associated LoF variants, or manifesting related hyperinflammatory disorders. Patients who exhibited reduced exocytosis according to established guidelines, or with at least 5 of 8 HLH criteria fulfilled, or with a family history of HLH were sequenced for HLH-associated genes.21 Furthermore, in patients with hypopigmentation, LYST, AP3B1, and RAB27A were sequenced.

Notably, a total of 92 patients analyzed harbored biallelic UNC13D, STX11, STXBP2, LYST, RAB27A, AP3B1, or RHOG LoF variants (termed EXO). Moreover, a total of 58 patients with biallelic or hemizygous PRF1, SH2D1A, XIAP, CD27, GATA2, MAGT1, ZNFX1, CYBA, or ITK LoF variants were analyzed (termed IEI). Thirty-three patients fulfilling HLH criteria but lacking pathogenic variants in genes associated with familial HLH or IEI and 30 patients diagnosed with macrophage-activation syndrome or systemic-onset juvenile idiopathic arthritis were also analyzed (termed HYPINF). These results were compared with those from 84 unrelated adult transport control samples or 198 healthy adult controls derived from the local blood bank. Three exocytosis stimulations were performed, namely, K562 cell–triggered NK-cell exocytosis, Fc receptor–triggered anti-CD16 antibody CD3CD56dim NK-cell exocytosis, and TCR-triggered anti-CD3 antibody CTL exocytosis. Overall, we compared 5 sample groups and 3 stimulations (Table 1; Figure 3).

Table 1.

Categorization and genetic characterization of samples

CategorySubgroupNumber of individualsTotal
EXO UNC13D (FHL3) 35  
STX11 (FHL4) 12  
STXBP2 (FHL5) 13  
LYST (CHS) 19  
RAB27A (GS2) 10  
AP3B1 (HPS2)  
RHOG 92 
IEI (non-EXO) PRF1 (FHL2) 30  
SH2D1A (XLP1)  
XIAP (XLP2)  
CD27  
GATA2  
MAGT1 (XMEN)  
ZNFX1  
CYBA  
ITK 58 
HYPINF Secondary HLH 33  
soJIA/MAS 30 63 
HD-CTRL Healthy donors 198 198 
TRP-CTRL Transport controls 84 84 
Grand total   495 
CategorySubgroupNumber of individualsTotal
EXO UNC13D (FHL3) 35  
STX11 (FHL4) 12  
STXBP2 (FHL5) 13  
LYST (CHS) 19  
RAB27A (GS2) 10  
AP3B1 (HPS2)  
RHOG 92 
IEI (non-EXO) PRF1 (FHL2) 30  
SH2D1A (XLP1)  
XIAP (XLP2)  
CD27  
GATA2  
MAGT1 (XMEN)  
ZNFX1  
CYBA  
ITK 58 
HYPINF Secondary HLH 33  
soJIA/MAS 30 63 
HD-CTRL Healthy donors 198 198 
TRP-CTRL Transport controls 84 84 
Grand total   495 

Number and categories of patients included in this study by their respective groups retrospectively compiled from 1 center over 10.5 years. EXO, individuals with NK/CTL exocytosis defect; HD-CTRL, healthy adult volunteers; HYPINF, individuals with a hyperinflammatory syndrome fulfilling HLH criteria; IEI, individuals with other inborn errors of immunity excluding EXO; TRP-CTRL, healthy adult transport controls; XLP, X-linked lymphoproliferative disease; XMEN, X-linked MAGT1 deficiency with increased susceptibility to Epstein-Barr virus infection and N-linked glycosylation defect.

Figure 3.

Exocytosis is defective in cytotoxic lymphocytes of FHL3-5, CHS, and GS2 patients. (A-C) Percentages of CD8+CD57+ T cells (A) or CD3CD56dim NK cells (B-C) expressing surface CD107a after incubation with P815 cells with anti-CD3 antibody (A), P815 cells with anti-CD16 antibody (B), or K562 cells (C), shown for 5 different groups, as indicated. Values represent the difference in CD107a levels of stimulated against control unstimulated cells. Boxes display quartiles and whiskers display 2.5th and 97.5th percentiles, as well as outliers. Student t test comparing EXO against the other groups; ∗∗∗∗P < .0001. (D-E) Exocytosis data of specific patient categories from the “impaired exocytosis” and “other IEI patients” groups are shown individually. Each individual patient value is represented by a symbol, with violin plots indicating quartiles. Colored markings depict normal ranges from 198 healthy adults, with boxes displaying quartiles and whiskers displaying 2.5th and 97.5th percentiles. (E) Student t test comparing patients with 198 heathy adult controls; n.s., not significant; ∗P < .05; ∗∗P < .01; ∗∗∗∗P < .0001. CHS, Chediak-Higashi syndrome; FHL3, familial hemophagocytic lymphohistioccytosis type 3; XLP1, X-linked lymphoproliferative disease type 2.

Figure 3.

Exocytosis is defective in cytotoxic lymphocytes of FHL3-5, CHS, and GS2 patients. (A-C) Percentages of CD8+CD57+ T cells (A) or CD3CD56dim NK cells (B-C) expressing surface CD107a after incubation with P815 cells with anti-CD3 antibody (A), P815 cells with anti-CD16 antibody (B), or K562 cells (C), shown for 5 different groups, as indicated. Values represent the difference in CD107a levels of stimulated against control unstimulated cells. Boxes display quartiles and whiskers display 2.5th and 97.5th percentiles, as well as outliers. Student t test comparing EXO against the other groups; ∗∗∗∗P < .0001. (D-E) Exocytosis data of specific patient categories from the “impaired exocytosis” and “other IEI patients” groups are shown individually. Each individual patient value is represented by a symbol, with violin plots indicating quartiles. Colored markings depict normal ranges from 198 healthy adults, with boxes displaying quartiles and whiskers displaying 2.5th and 97.5th percentiles. (E) Student t test comparing patients with 198 heathy adult controls; n.s., not significant; ∗P < .05; ∗∗P < .01; ∗∗∗∗P < .0001. CHS, Chediak-Higashi syndrome; FHL3, familial hemophagocytic lymphohistioccytosis type 3; XLP1, X-linked lymphoproliferative disease type 2.

Close modal

Patients in the EXO group displayed reduced exocytosis upon stimulation of CTL and NK cells compared with IEI or HYPINF patients, healthy adults, or transport controls (Figure 3A-B). Patients diagnosed with FHL3 (UNC13D), FHL4 (STX11), FHL5 (STXBP2), CHS (LYST), and GS2 (RAB27A) displayed defective exocytosis in all assays (Figure 3D), whereas patients diagnosed with FHL2 (PRF1), XLP1 (SH2D1A), XLP2 (XIAP), or other IEI generally displayed exocytosis almost at the level of healthy adults controls (Figure 3E). Notably, trend lines for exocytosis in IEI or HYPINF patients plotted vs age at analysis showed similar levels of exocytosis in pediatric patients to that of healthy adult control samples (supplemental Figure 2A).

Sensitivity and specificity of individual cytotoxic lymphocyte exocytosis assays

Our exocytosis data were used to plot receiver operating characteristic curves (Figure 4A-D; supplemental Figure 2B-E). The area under the curve was larger for anti-CD3 CTL and anti-CD16 NK-cell exocytosis assays. The K562 cell exocytosis assay returned 93.1% sensitivity and 87.0% specificity at a 5.3% exocytosis cutoff. By comparison, the anti-CD16 stimulation of NK cells returned 96.6% sensitivity and 92.4% specificity at a 12.5% exocytosis, and the anti-CD3 stimulation of CTL was 94.8% sensitivity and 92.4% specificity at 23.5% exocytosis. The accuracy of the anti-CD3 and anti-CD16 assays was 93.3% and 98.7%, respectively, compared with 89.3% for the K562 cell assay (Table 2). The DP and Youden index showed a similar trend with K562 cell–induced NK-cell exocytosis having poorer results than the anti-CD3 CTL or anti-CD16 NK-cell exocytosis assay (Table 2).

Figure 4.

Combination receiver operating characteristic (ROC) curves from this study. (A-D) Combined ROC curves for different exocytosis stimulations (K562 cell stimulation for NK cells, anti-CD16 antibody for NK cells, and anti-CD3 antibody for CTL) when comparing 92 impaired exocytosis (EXO) patients against 4 other control groups consisting of 58 other IEI patients (A), 63 hyperinflammatory (HYPINF) patients (B), 198 healthy adults controls (C), or 84 healthy adults transport controls (D). Cutoff values with sensitivity and specificity are indicated in brackets for each comparison. (E-F) Plots show exocytosis values for different cytotoxic lymphocyte subsets and stimulations, as indicated. Each circle represents an individual patient color coded according to indicated key.

Figure 4.

Combination receiver operating characteristic (ROC) curves from this study. (A-D) Combined ROC curves for different exocytosis stimulations (K562 cell stimulation for NK cells, anti-CD16 antibody for NK cells, and anti-CD3 antibody for CTL) when comparing 92 impaired exocytosis (EXO) patients against 4 other control groups consisting of 58 other IEI patients (A), 63 hyperinflammatory (HYPINF) patients (B), 198 healthy adults controls (C), or 84 healthy adults transport controls (D). Cutoff values with sensitivity and specificity are indicated in brackets for each comparison. (E-F) Plots show exocytosis values for different cytotoxic lymphocyte subsets and stimulations, as indicated. Each circle represents an individual patient color coded according to indicated key.

Close modal
Table 2.

Statistics on specific sample group comparisons

StimulationCutoff CD107a+ (%)SensitivitySpecificityYoudenDPAccuracy
EXO vs IEI       
K562 5.3 93.1 87.0 0.81 1.08 89.3 
Anti-CD16 12.5 96.6 92.4 0.89 1.40 98.7 
Anti-CD3 23.5 94.8 92.4 0.88 1.29 93.3 
EXO vs HYPINF       
K562 8.0 81.0 95.7 0.78 1.09 89.7 
Anti-CD16 14.3 98.4 94.6 0.93 1.67 96.1 
Anti-CD3 21.2 98.4 91.3 0.90 1.55 94.2 
EXO vs HD-CTRL       
K562 9.9 97.8 98.0 0.96 1.84 97.9 
Anti-CD16 26.3 98.9 99.0 0.98 2.18 99.0 
Anti-CD3 40.9 98.9 99.5 0.98 2.34 99.0 
EXO vs TRP-CTRL       
K562 7.8 91.7 95.7 0.88 1.32 93.2 
Anti-CD16 15.2 98.8 94.6 0.93 1.74 96.6 
Anti-CD3 21.3 95.2 91.3 0.87 1.28 93.2 
StimulationCutoff CD107a+ (%)SensitivitySpecificityYoudenDPAccuracy
EXO vs IEI       
K562 5.3 93.1 87.0 0.81 1.08 89.3 
Anti-CD16 12.5 96.6 92.4 0.89 1.40 98.7 
Anti-CD3 23.5 94.8 92.4 0.88 1.29 93.3 
EXO vs HYPINF       
K562 8.0 81.0 95.7 0.78 1.09 89.7 
Anti-CD16 14.3 98.4 94.6 0.93 1.67 96.1 
Anti-CD3 21.2 98.4 91.3 0.90 1.55 94.2 
EXO vs HD-CTRL       
K562 9.9 97.8 98.0 0.96 1.84 97.9 
Anti-CD16 26.3 98.9 99.0 0.98 2.18 99.0 
Anti-CD3 40.9 98.9 99.5 0.98 2.34 99.0 
EXO vs TRP-CTRL       
K562 7.8 91.7 95.7 0.88 1.32 93.2 
Anti-CD16 15.2 98.8 94.6 0.93 1.74 96.6 
Anti-CD3 21.3 95.2 91.3 0.87 1.28 93.2 

Optimal cutoff for exocytosis assay (CD107a+ percentage), sensitivity, and specificity obtained through receiver operating characteristic plots comparing 3 different exocytosis assays for patients with variants associated with defective exocytosis (EXO) vs 4 other control groups (IEI, other HYPINFs, 198 HD-CTRLs, and TRP-CTRLs). Youden index, DP, and accuracy were calculated based on the obtained values.HD-CTRL, healthy adult volunteers; TRP-CTRL, healthy adult transport controls.

Exocytosis values for the EXO, IEI, and HYPINF patient groups were plotted for the 3 different pair-wise comparisons (Figure 4E-G). Overall, the correlation between the exocytosis values obtained in the different assays was high, with a few exceptions of patients with low NK-cell exocytosis induced by K562 cells but with higher exocytosis induced by anti-CD16 NK-cell or anti-CD3 CTL assays.

In summary, the anti-CD3 CTL and anti-CD16 NK assays were more accurate than the current standard K562 cell–triggered NK-cell assay in predicting primary genetic defects affecting cytotoxic lymphocyte exocytosis.

Predictive power of combined analyses for the diagnosis of defective cytotoxic lymphocyte exocytosis

Combining analysis of distinct cell types could potentially increase the accuracy of detecting primary defects in lymphocyte cytotoxicity. Therefore, we performed combined analyses of the sensitivity and specificity values of 2 assays, that is K562 cells with anti-CD16 NK-cell assays, anti-CD16 NK cell with anti-CD3 CTL assays, or K562 cell NK cell with anti-CD3 CTL assays. To have both true negative and true positive samples, the EXO and IEI groups were pooled. Taking cutoff values of 5.3% for K562 cell–triggered NK-cell exocytosis, 12.5% for anti-CD16 NK-cell assays, and 23.5% for anti-CD3 CTL assays, the results were evaluated with “AND” or “OR” Boolean functions. Using “AND” did not improve the accuracy, but anti-CD16 NK-cell OR anti-CD3 CTL assays returned 100% sensitivity and 98.3% specificity for a combined 99.3% accuracy (Table 3). Thus, combined analyses can enhance the diagnostic accuracy of functional diagnostic approaches.

Table 3.

Increased accuracy provided by combined assessment of exocytosis in cytotoxic T-cell and NK-cell subsets

SensitivitySpecificityYoudenAccuracy
K562 with anti-CD16     
OR 100.0 98.3 >10 99.3 
AND 87.0 91.4 1.02 88.7 
K562 with anti-CD3     
OR 97.8 98.3 1.88 98.0 
AND 81.5 91.4 0.92 85.3 
Anti-CD16 with anti-CD3     
OR 100.0 98.3 >10 99.3 
AND 92.4 93.1 1.22 92.7 
SensitivitySpecificityYoudenAccuracy
K562 with anti-CD16     
OR 100.0 98.3 >10 99.3 
AND 87.0 91.4 1.02 88.7 
K562 with anti-CD3     
OR 97.8 98.3 1.88 98.0 
AND 81.5 91.4 0.92 85.3 
Anti-CD16 with anti-CD3     
OR 100.0 98.3 >10 99.3 
AND 92.4 93.1 1.22 92.7 

Combining 2 different exocytosis assays leads to improved sensitivity or specificity. Displayed are calculated sensitivity, specificity, Youden index, and accuracy after combining results from 2 exocytosis assays with “AND” or “OR” Boolean gating.

Robustness of exocytosis assays in relation to transport duration

Exocytosis testing is typically analyzed in larger experienced centers, necessitating sample transport. Some clinical laboratories have strict cutoff time points of <24 hours transport, but this is not always feasible from remote locations. Retrospectively, exocytosis assays from IEI patients were found impartial to shipment duration up to 48 hours (supplemental Figure 3A). Samples from healthy unrelated controls arriving <48 hours generally did not show any bias toward lower exocytosis results (supplemental Figure 3B). Familial controls (supplemental Figure 3D) show slightly poorer exocytosis at the >48-hour time point. When grouped by time of arrival (supplemental Figure 3C,E), a difference in function was observed when comparing samples arriving <24 hours with those arriving >48 hours. Notably, anti-CD16–triggered NK-cell exocytosis appeared the most time stable test. We further investigated the stability on blood samples from 10 healthy adult volunteers left for 1, 24, 48, and 72 hours before PBMC isolation. A significant reduction of exocytosis potential with all 3 stimulations was observed within 24 hours after venipuncture (supplemental Figure 3F). For K562 cell–triggered NK-cell exocytosis, the responses continuously dropped up to 48 hours, whereas anti-CD16–triggered assays remained stable during that time followed by a drop in function at >48 hours. The anti-CD3 CTL assay was the most robust with no statistically significant reduction in function even in samples stored >48 hours. The contrast in results between shipped and laboratory controls highlighted the difficulty in controlling for transportation stress because it is unique for each shipment and may include not just time but also variables such as temperature and agitation. In summary, because the anti-CD3 CTL exocytosis assay has inherently high signal-to-noise ratio and assay sturdiness, although not ideal, evaluation of cytotoxic lymphocyte function is possible with samples arriving after >48 hours.

We previously optimized an assay to evaluate exocytosis by freshly isolated human CTL, without prior stimulation or prolonged incubation.6 Here, we assessed the performance of different NK-cell and T-cell exocytosis assays in a large cohort of healthy individuals and prospectively evaluated patients with suspected primary HLH collected over 10 years. Our results demonstrate that assessment of CD8+CD57+ T-cell exocytosis in response to TCR engagement and CD3CD56dim NK-cell exocytosis in response to CD16 engagement provided the highest diagnostic sensitivity. Importantly, combined analysis provided the highest diagnostic accuracy and safeguarded results in challenging patients when ambiguous results, long or suboptimal shipment, or low blood CTL or NK-cell frequencies might lead to assay failure.

Evaluation of 51Cr-labeled K562 target cell lysis has historically represented a gold standard for the evaluation of cytotoxic lymphocyte activity and diagnosis of primary HLH.55,56 However, this test carries a number of drawbacks that may yield false positive results.1 Hence, quantification of surface CD107a upregulation on NK cells after K562 cell stimulation was proposed as a diagnostic assay for patients with defective cytotoxic lymphocyte exocytosis.21,57 Initially identified as an activation-induced surface protein on PBMC subsets, CD107a surface expression could play a role in self-protection against released cytolytic proteins and has been used to quantify cytotoxic T-cell, NK-cell, basophil, and mast cell exocytosis.58-64 The K562 cell–triggered NK-cell exocytosis represents a safe, rapid, easily performed, and robust assay with good sensitivity and specificity,21,37,65 but our efforts demonstrate that even higher diagnostic accuracy can be achieved with additional stimulations.

Primary HLH is commonly viewed as a T-cell–driven pathological response to infection.39 The main aim of our study was to assess the efficacy of T-cell exocytosis assay for the diagnosis of congenital defects in lymphocyte exocytosis. The assessment of T-cell blast exocytosis was previously tested but may display a low signal-to-noise ratio especially with milder defects.21,66 Confirming a pilot study, our data from 198 healthy adult donors revealed uniformly strong exocytic responses of CTL with relatively low interindividual variability.6 In comparison with assays evaluating NK- and T-cell exocytosis in response to engagement of CD16 or the TCR complex, respectively, the K562–NK-cell test displayed lower signal and higher interindividual variability. Notably, with our modification of the exocytosis assay examining 3 readouts of cytotoxic lymphocyte exocytosis, the cellular input is still lower than that typically used for 51Cr-release assays. Hence, flow cytometric exocytosis assays are more sensitive and require lower numbers of PBMCs than 51Cr-release assays, potentially facilitating measurements in prenatal infants or severely lymphopenic patients. CTL assays can facilitate assessment of patients with few NK cells, which we find represents an issue in ∼3% of pediatric and 6% of adolescent and adult patient samples.

Low NK-cell reactivity toward K562 cells among healthy individuals correlated with outliers harboring high proportions of CMV-associated adaptive NK cells lacking FcεRγ expression. Our results implicate FcεRγ-associated activating NKp30 and NKp46 receptors in recognition of K562 cells. Congruently, the NKp30-ligand B7-H6 is highly expressed on K562 cells and antibody-mediated NKp30 blockade inhibits NK-cell recognition of K562 cells.67 Our results demonstrate that a high proportion of FcεRγ adaptive NK cells can negatively skew the overall NK-cell response to K562 cell stimulation. Although adaptive NK-cell expansions are associated with CMV seropositivity, in a European population, only 15% of healthy blood volunteers manifested sizable adaptive NK-cell expansions constituting >30% of the total CD56dim NK-cell population.45 CMV seropositivity is thus a poor indicator of adaptive NK-cell expansions. However, in a sub-Saharan population, adaptive NK-cell expansions were much more prevalent and constituted >50% of the total CD56dim NK-cell population in a majority of participants.68 Inflammation can promote adaptive NK-cell expansions.69,70 Notably, patients with GATA2 variants occasionally present with HLH and can have extremely high frequencies of adaptive NK cells.53,71,72 As such, diminished NK-cell exocytosis in response to K562 cell stimulation could reflect epigenetic cell differentiation processes rather than primary HLH and be especially prevalent in individuals with severe inflammation or underlying primary genetic defects. Quantification of NK or CTL exocytosis triggered by anti-CD16 or anti-CD3, respectively, can thus safeguard K562 cell–based analyses.

Prospectively, we evaluated 213 patients with suspected primary HLH. Patients with biallelic UNC13D, STX11, STXBP2, LYST, RAB27A, AP3B1, and RHOG variants consistently displayed strongly impaired CTL exocytosis compared with patients with PRF1, SH2D1A, XIAP, CD27, GATA2, MAGT1, ZNFX1, CYBA, or ITK variants, which have all been associated with HLH or low NK-cell numbers. Of note, healthy adult controls do not take into account several factors that might influence cellular analyses, including differences in age, time from sampling to analysis, medication/therapy, and blood inflammatory status. Thus, samples from patients diagnosed with other pathogenic HLH variants that do not impair exocytosis, secondary HLH, or other inflammatory syndromes represent better groups for comparison with patients with primary defects in cytotoxic lymphocyte exocytosis. Moreover, the interpretation of patient results is normally performed by juxtaposing a reference range obtained from a group of healthy adult donors. This study instead focuses on estimating NK and CTL exocytosis ranges of various genetically confirmed groups. Our patient data demonstrated superior sensitivity of the anti-CD3 CTL and anti-CD16 NK-cell exocytosis assays for the identification of patients with biallelic LoF variants in genes required for cytotoxic lymphocyte exocytosis compared with the established K562 cell stimulation assay, providing a high DP and Youden index, signifying the assays’ ability to discriminate between the 2 populations and avoid false positives and negatives, respectively.

Most cases of primary HLH are acute and require urgent diagnosis. Offering more confidence in findings, running the anti-CD3 CTL and anti-CD16 NK-cell exocytosis assays side by side increased accuracy and provided up to 100% sensitivity and 98% specificity. Infants aged <3 months normally have low numbers of CD57+ T cells, and it is not uncommon for patients to have too few NK cells for evaluation, suggesting a place for testing expanded T-cell exocytosis.18,21,73,74 Independent assays also lessen the risk of human error leading to assay failure. Thus, running multiple assays increases the probability that at least 1 result is informative, avoiding delays and lowering the risk of misdiagnosis. Importantly, combining ≥2 assays also strengthens the overall diagnostic prediction. Generally, implementing changes in laboratory assays in a clinical setting can be costly in terms of efforts to fulfill national regulatory requirements, validate assays, and establish reference ranges. In terms of operating time and costs for implementing Fc receptor stimulation of NK cells and TCR stimulation of CTLs, use of multichannel pipettes and flow cytometers with plate readers should imply no drastic increase in operator time to run assays, whereas reagent costs are deemed to double due to an increase from 2 to 4 wells for each tested individual.

Although we report specific cutoff percentages for the 3 exocytosis assays, results will vary among laboratories. Moreover, our values were estimated by comparing controls and confirmed patient data, with the latter being quite rare and thus challenging for smaller laboratories to clinically validate. Determination of specific normal values are necessary for each laboratory and protocol. Results showing defective/absent exocytosis below the cutoff should be repeated to confirm, whereas those with reduced but not defective exocytosis should also be repeated and sequenced because data here suggest some patients with hypomorphic variants in genes required for exocytosis have lowered but not absent exocytosis. Here, the K562 cell exocytosis cutoff of 5.3% is in line with the previously stipulated 5%,21 even with protocol changes and extension from 2 to 3 hours of incubation. Importantly, the CTL exocytosis assay is also more robust than the K562 cell assay when dealing with suboptimal samples, due to long or harsh transportation.31 Large numbers of familial and healthy unrelated controls sent together with patient samples were minimally affected by transport times up to 48 hours, indicating that successful diagnosis of samples from farther locales or long shipping duration is feasible if well controlled.

We have demonstrated the accuracy of various exocytosis assays in diagnosing different primary genetic defects causative of defective exocytosis. We recommend laboratories run a combination of 2 exocytosis assays along with other panels to investigate the levels of perforin, SLAM-associating protein, X-linked inhibitor of apoptosis protein, CD27, NKT cells, and TCRα/β–double negative T cells to encompass a wider range of primary genetic defects linked to HLH, as illustrated in our suggested laboratory diagnosis algorithm (Figure 5).22,23,75-83 We have previously reported how IL-2 stimulation can discriminate certain forms of FHL.6,8,21,41-43 In this study, we focused analyses on freshly isolated samples in which functional assays can promptly identify patients with defective exocytosis and potentially direct treatment. With respect to molecularly categorizing patients, we advocate that genetic tests are a more reliable way to stratify patients with FHL and gain important genotype-functional correlations.

Figure 5.

Proposed laboratory diagnostic algorithm for patients presenting with HLH. Because the spectrum and our understanding of primary diseases linked to HLH grow, laboratories would need to constantly update themselves with the latest diagnostic assays. It is recommended that individual laboratories conduct tests to determine local cutoff values for the respective assays. Functional XIAP assay75 should be considered in both males and females in light of recent findings,76,77 reduced iNKT numbers a hallmark of ITK23,78 but cautionary for SAP deficiency with highly variable levels in young children.79,80 Elevated α/β-DNT cells raise suspicion of ALPS.81 The total absence CD27 in all lymphocyte subgroups is indicative of its deficiency,22,82 whereas absence of CD27 specifically in the B-cell compartment is characteristic of SAP deficiency.83 Although functional XIAP and NK/CTL exocytosis assays should be performed on heparinized blood, if EDTA blood is available, it is preferable for phenotyping tests (perforin, SAP, XIAP, CD27, and α/β-DNT) because it is more stable with transport, can be used for DNA extraction, and plasma collected for analyzing cytokine levels. ALPS, autoimmune lymphoproliferative syndrome; DNT, double negative T cells; iNKT, invariant NK T cells; ITK, interleukin-2–inducible T-cell kinase; SAP, SLAM-associating protein; XIAP, X-linked inhibitor of apoptosis protein.

Figure 5.

Proposed laboratory diagnostic algorithm for patients presenting with HLH. Because the spectrum and our understanding of primary diseases linked to HLH grow, laboratories would need to constantly update themselves with the latest diagnostic assays. It is recommended that individual laboratories conduct tests to determine local cutoff values for the respective assays. Functional XIAP assay75 should be considered in both males and females in light of recent findings,76,77 reduced iNKT numbers a hallmark of ITK23,78 but cautionary for SAP deficiency with highly variable levels in young children.79,80 Elevated α/β-DNT cells raise suspicion of ALPS.81 The total absence CD27 in all lymphocyte subgroups is indicative of its deficiency,22,82 whereas absence of CD27 specifically in the B-cell compartment is characteristic of SAP deficiency.83 Although functional XIAP and NK/CTL exocytosis assays should be performed on heparinized blood, if EDTA blood is available, it is preferable for phenotyping tests (perforin, SAP, XIAP, CD27, and α/β-DNT) because it is more stable with transport, can be used for DNA extraction, and plasma collected for analyzing cytokine levels. ALPS, autoimmune lymphoproliferative syndrome; DNT, double negative T cells; iNKT, invariant NK T cells; ITK, interleukin-2–inducible T-cell kinase; SAP, SLAM-associating protein; XIAP, X-linked inhibitor of apoptosis protein.

Close modal

In conclusion, screening for anti-CD3 antibody–induced exocytosis of CTL and anti-CD16 antibody–induced exocytosis of NK cells is more accurate than the current standard K562 cell stimulation for the diagnosis of congenital defects in lymphocyte exocytosis. The data presented here thus suggest a change in the HLH-2004 criteria from low or absent NK-cell activity to low NK-cell or CTL exocytosis, except for FHL2/perforin deficiency. Performing 2 different exocytosis stimulations simultaneously gives high accuracy and acts as confirmatory assays. Such assays with increased sensitivity can aid the understanding of immunodeficiencies that may partially impair exocytosis.

The authors thank Felicia Er ZhiXin for technical assistance, MedH Flow Cytometry Facility, Uppsala Multidisciplinary Center for Advanced Computational Science, all patients and their families for contributing samples, as well as collaborative clinicians from around the world.

This work was supported by Deutsche Forschungsgemeinschaft SFB1160 (256073931; S.E.) TPA1; and by grants from the Swedish Research Council (2022-01178), Cancer Foundation (23 3090 Pj), Childhood Cancer Foundation, the Knut and Alice Wallenberg Foundation, and the Center for Innovative Medicine (all to Y.T.B.).

Contribution: S.C.C.C. designed the study, performed exocytosis assays, evaluated the data, and wrote the manuscript; B.T. and L.E.C. performed sequencing, evaluated the data, and performed receiver operating characteristic analyses; H.S. performed exocytosis assays with EAT-2 and and FcεRγ staining; J.T. partially performed statistical analysis; T.M.C., J.N.-Z., T.S., and S.W. performed exocytosis assays; M.M. performed sequencing and evaluated the data; K.M., W.A.-H., H.H.A., F.B.B., M.Y.C., O.D., T.A., M.I., I.M., M.S., E.U., S.U., W.J.I., K.K., K.C.G., and S.E. cared for patients and/or provided material; all pHLH collaborators cared for patients and provided clinical information; H.-G.L., M.N., A.H., and J.-I.H. revised the manuscript; Y.T.B. designed the study, evaluated the data, and wrote the manuscript; and all authors have critically read the manuscript before submission.

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

A complete list of all pHLH collaborators appears in “Appendix.”

Correspondence: Yenan T. Bryceson, Karolinska Institute, Department of Medicine, Center for Hematology and Regenerative Medicine, Hälsovägen 9, 14186 Stockholm, Sweden; email: yenan.bryceson@ki.se; and Samuel C. C. Chiang, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229; email: sam.chiang@cchmc.org.

The members of the pHLH collaboration are Miguel R Abboud, American University of Beirut, Beirut, Lebanon; Sevkiye Selin Aytac, Hacettepe Univerity Faculty of Medicine, Hacettepe, Turkey; Franziskus Johannes Bosse, Barne og Ungdomsklinikken, Haukeland Universitetssjukehus, Bergen, Norway; Sharon Choo, Royal Children's Hospital, Melbourne, Australia; Katarzyna Drabko, Klinika Onkologii, Hematologii i Transplantologii Pediatrycznej, Uniwersytet Medyczny w Poznaniu, Szpital Kliniczny im. Karola Jonschera, Instytut Pediatrii, Poznań, Poland; Reem Elfeky, Great Ormond Street Hospital, London, United Kingdom, and Children's hospital Ain Shams University, Cairo, Egypt; Dalia Helmy El-Ghoneimy, Children's Hospital, Ain Shams University, Cairo, Egypt; Zehra Fadoo, Aga Khan University, Karachi, Pakistan; Tatiana Greenwood, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden, and Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden; Britt Gustafsson, Karolinska Institutet, Stockholm, Sweden; Stefan Hagelberg, Astrid Lindgren's Children's Hospital, Karolinska University Hospital, Stockholm, Sweden; Henrik Hasle, Aarhus University Hospital, Aarhus, Denmark; Johanna Hästbacka, New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Oleg Jadrešin, Children's Hospital Zagreb, Zagreb, Croatia; Martin Jädersten, Karolinska University Hospital, and, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden; Zuhre Kaya, Gazi University Faculty of Medicine, Ankara, Turkey; Ranon Lecumberri, Clínica Universidad de Navarra, Pamplona, Spain; Laura Marques, Centro Materno Infantil do Norte, CHUdSA, Porto, Portugal; Naureen Mushtaq, Aga Khan University Hospital, Karachi, Pakistan; Ahmed Naqvi, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; João Farela Neves, Hospital Dona Estefânia, CHULC-EPE, Comprehensive Health Research Centre and Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal; Susana Nunes, Centro Hospitalar Universitário São João, Porto, Portugal; Martin Paucar, Karolinska University Hospital, Stockholm Sweden; Jeanette H. Payne, Sheffield Childrens Hospital, Sheffield, United Kingdom; Jelena Rascon, Vilnius University Hospital Santaros Klinikos, and Clinics of Children’s Diseases, Faculty of Medicine, Vilnius University, Vilnius, Lithuania; Terhi Susanna Ruuska, University of Oulu and Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland; Ebru Tugrul Saribeyoglu, FÄ für Kinder- und Jugendmedizin, SP Pädiatrische Onkologie/Hämatologie, ZB Palliativemedizin, Charité-Universitätsmedizin Berlin, Berlin, Germany; Mikael C. Sundin, Karolinska University Hospital Huddinge and Clintec, Karolinska Institutet, Stockholm, Sweden; Jenny Svedenkrans, Karolinska University Hospital, Stockholm, Sweden; Natalia Świderska, Medical University of Gdańsk, Gdańsk, Poland; Ulf Tedgård, Skåne University Hospital, Lund, Sweden; Tor Henrik Tvedt, Oslo University Hospital, Oslo, Norway; Ayşegül Ünüvar, İstanbul School of Medicine, İstanbul University, Istanbul, Turkey; Jan A. M. Van Laar, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands; Sheila Weitzman, The Hospital for Sick Children, Toronto, Canada; Jacek Winiarski, Karolinska University Hospital Huddinge and Clintec, Karolinska Institutet, Stockholm, Sweden; Muhamma Zohaib Yaseen, Children's Hospital Karachi, Karachi, Pakistan; Mehmet Yildiz, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey; Daniela Zantomio, Austin Health, Melbourne, Australia; Ingrid Øra, Lund University, Lund, Sweden; and Torstein Øverland, Oslo University Hospital, Oslo, Norway.

1.
Henter
JI
,
Horne
A
,
Arico
M
, et al
.
HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis
.
Pediatr Blood Cancer
.
2007
;
48
(
2
):
124
-
131
.
2.
Jordan
MB
,
Allen
CE
,
Greenberg
J
, et al
.
Challenges in the diagnosis of hemophagocytic lymphohistiocytosis: recommendations from the North American Consortium for Histiocytosis (NACHO)
.
Pediatr Blood Cancer
.
2019
;
66
(
11
):
e27929
.
3.
Canna
SW
,
Marsh
RA
.
Pediatric hemophagocytic lymphohistiocytosis
.
Blood
.
2020
;
135
(
16
):
1332
-
1343
.
4.
Chinnici
A
,
Beneforti
L
,
Pegoraro
F
, et al
.
Approaching hemophagocytic lymphohistiocytosis
.
Front Immunol
.
2023
;
14
:
1210041
.
5.
Chattopadhyay
PK
,
Betts
MR
,
Price
DA
, et al
.
The cytolytic enzymes granyzme A, granzyme B, and perforin: expression patterns, cell distribution, and their relationship to cell maturity and bright CD57 expression
.
J Leukoc Biol
.
2009
;
85
(
1
):
88
-
97
.
6.
Chiang
SC
,
Theorell
J
,
Entesarian
M
, et al
.
Comparison of primary human cytotoxic T-cell and natural killer cell responses reveal similar molecular requirements for lytic granule exocytosis but differences in cytokine production
.
Blood
.
2013
;
121
(
8
):
1345
-
1356
.
7.
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
;
115
(
4
):
461
-
473
.
8.
Bryceson
YT
,
Rudd
E
,
Zheng
C
, et al
.
Defective cytotoxic lymphocyte degranulation in syntaxin-11 deficient familial hemophagocytic lymphohistiocytosis 4 (FHL4) patients
.
Blood
.
2007
;
110
(
6
):
1906
-
1915
.
9.
Cote
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
;
119
(
12
):
3765
-
3773
.
10.
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
;
85
(
4
):
482
-
492
.
11.
Barbosa
MD
,
Nguyen
QA
,
Tchernev
VT
, et al
.
Identification of the homologous beige and Chediak-Higashi syndrome genes
.
Nature
.
1996
;
382
(
6588
):
262
-
265
.
12.
Menasche
G
,
Pastural
E
,
Feldmann
J
, et al
.
Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome
.
Nat Genet
.
2000
;
25
(
2
):
173
-
176
.
13.
Gil-Krzewska
A
,
Murakami
Y
,
Peruzzi
G
, et al
.
Natural killer cell activity and dysfunction in Hermansky-Pudlak syndrome
.
Br J Haematol
.
2017
;
176
(
1
):
118
-
123
.
14.
Maul-Pavicic
A
,
Chiang
SC
,
Rensing-Ehl
A
, et al
.
ORAI1-mediated calcium influx is required for human cytotoxic lymphocyte degranulation and target cell lysis
.
Proc Natl Acad Sci U S A
.
2011
;
108
(
8
):
3324
-
3329
.
15.
Ammann
S
,
Schulz
A
,
Krageloh-Mann
I
, et al
.
Mutations in AP3D1 associated with immunodeficiency and seizures define a new type of Hermansky-Pudlak syndrome
.
Blood
.
2016
;
127
(
8
):
997
-
1006
.
16.
Parry
DA
,
Holmes
TD
,
Gamper
N
, et al
.
A homozygous STIM1 mutation impairs store-operated calcium entry and natural killer cell effector function without clinical immunodeficiency
.
J Allergy Clin Immunol
.
2016
;
137
(
3
):
955
-
957.e8
.
17.
Kalinichenko
A
,
Perinetti Casoni
G
,
Dupre
L
, et al
.
RhoG deficiency abrogates cytotoxicity of human lymphocytes and causes hemophagocytic lymphohistiocytosis
.
Blood
.
2021
;
137
(
15
):
2033
-
2045
.
18.
Schutze
K
,
Gross
M
,
Cornils
K
, et al
.
MAP kinase activating death domain deficiency is a novel cause of impaired lymphocyte cytotoxicity
.
Blood Adv
.
2023
;
7
(
8
):
1531
-
1535
.
19.
Rigaud
S
,
Fondaneche
MC
,
Lambert
N
, et al
.
XIAP deficiency in humans causes an X-linked lymphoproliferative syndrome
.
Nature
.
2006
;
444
(
7115
):
110
-
114
.
20.
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
;
117
(
1
):
53
-
62
.
21.
Bryceson
YT
,
Pende
D
,
Maul-Pavicic
A
, et al
.
A prospective evaluation of degranulation assays in the rapid diagnosis of familial hemophagocytic syndromes
.
Blood
.
2012
;
119
(
12
):
2754
-
2763
.
22.
Salzer
E
,
Daschkey
S
,
Choo
S
, et al
.
Combined immunodeficiency with life-threatening EBV-associated lymphoproliferative disorder in patients lacking functional CD27
.
Haematologica
.
2013
;
98
(
3
):
473
-
478
.
23.
Ghosh
S
,
Bienemann
K
,
Boztug
K
,
Borkhardt
A
.
Interleukin-2-inducible T-cell kinase (ITK) deficiency - clinical and molecular aspects
.
J Clin Immunol
.
2014
;
34
(
8
):
892
-
899
.
24.
Bode
SF
,
Ammann
S
,
Al-Herz
W
, et al
.
The syndrome of hemophagocytic lymphohistiocytosis in primary immunodeficiencies: implications for differential diagnosis and pathogenesis
.
Haematologica
.
2015
;
100
(
7
):
978
-
988
.
25.
Tesi
B
,
Sieni
E
,
Neves
C
, et al
.
Hemophagocytic lymphohistiocytosis in 2 patients with underlying IFN-gamma receptor deficiency
.
J Allergy Clin Immunol
.
2015
;
135
(
6
):
1638
-
1641
.
26.
Lofstedt
AH
,
Jadersten
M
,
Meeths
M
,
Henter
JI
.
Malignancy-associated hemophagocytic lymphohistiocytosis in Sweden: incidence, clinical characteristics and survival
.
Blood
.
2024
;
143
(
3
):
233
-
242
.
27.
Alsohime
F
,
Temsah
MH
,
Alotaibi
RM
, et al
.
Presentations and outcomes of familial hemophagocytic lymphohistiocytosis in the pediatric intensive care units (PICUs)
.
Front Pediatr
.
2023
;
11
:
1152409
.
28.
Fischer
A
,
Hacein-Bey-Abina
S
.
Gene therapy for severe combined immunodeficiencies and beyond
.
J Exp Med
.
2020
;
217
(
2
):
e20190607
.
29.
Dettmer-Monaco
V
,
Weissert
K
,
Ammann
S
, et al
.
Gene editing of hematopoietic stem cells restores T-cell response in familial hemophagocytic lymphohistiocytosis
.
J Allergy Clin Immunol
.
2024
;
153
(
1
):
243
-
255.e14
.
30.
Meeths
M
,
Chiang
SC
,
Wood
SM
, et al
.
Familial hemophagocytic lymphohistiocytosis type 3 (FHL3) caused by deep intronic mutation and inversion in UNC13D
.
Blood
.
2011
;
118
(
22
):
5783
-
5793
.
31.
Entesarian
M
,
Chiang
SC
,
Schlums
H
, et al
.
Novel deep intronic and missense UNC13D mutations in familial haemophagocytic lymphohistiocytosis type 3
.
Br J Haematol
.
2013
;
162
(
3
):
415
-
418
.
32.
Seo
JY
,
Song
JS
,
Lee
KO
, et al
.
Founder effects in two predominant intronic mutations of UNC13D, c.118-308C>T and c.754-1G>C underlie the unusual predominance of type 3 familial hemophagocytic lymphohistiocytosis (FHL3) in Korea
.
Ann Hematol
.
2013
;
92
(
3
):
357
-
364
.
33.
Qian
Y
,
Johnson
JA
,
Connor
JA
, et al
.
The 253-kb inversion and deep intronic mutations in UNC13D are present in North American patients with familial hemophagocytic lymphohistiocytosis 3
.
Pediatr Blood Cancer
.
2014
;
61
(
6
):
1034
-
1040
.
34.
Vossen
MT
,
Biezeveld
MH
,
de Jong
MD
, et al
.
Absence of circulating natural killer and primed CD8+ cells in life-threatening varicella
.
J Infect Dis
.
2005
;
191
(
2
):
198
-
206
.
35.
Mao
H
,
Tu
W
,
Qin
G
, et al
.
Influenza virus directly infects human natural killer cells and induces cell apoptosis
.
J Virol
.
2009
;
83
(
18
):
9215
-
9222
.
36.
Abdalgani
M
,
Filipovich
AH
,
Choo
S
, et al
.
Accuracy of flow cytometric perforin screening for detecting patients with FHL due to PRF1 mutations
.
Blood
.
2015
;
126
(
15
):
1858
-
1860
.
37.
Rubin
TS
,
Zhang
K
,
Gifford
C
, et al
.
Perforin and CD107a testing is superior to NK cell function testing for screening patients for genetic HLH
.
Blood
.
2017
;
129
(
22
):
2993
-
2999
.
38.
Chiang
SCC
,
Bleesing
JJ
,
Marsh
RA
.
Current flow cytometric assays for the screening and diagnosis of primary HLH
.
Front Immunol
.
2019
;
10
:
1740
.
39.
Jordan
MB
,
Hildeman
D
,
Kappler
J
,
Marrack
P
.
An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder
.
Blood
.
2004
;
104
(
3
):
735
-
743
.
40.
Sepulveda
FE
,
Maschalidi
S
,
Vosshenrich
CA
, et al
.
A novel immunoregulatory role for NK-cell cytotoxicity in protection from HLH-like immunopathology in mice
.
Blood
.
2015
;
125
(
9
):
1427
-
1434
.
41.
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
;
116
(
15
):
2635
-
2643
.
42.
Chiang
SCC
,
Wood
SM
,
Tesi
B
, et al
.
Differences in granule morphology yet equally impaired exocytosis among cytotoxic T cells and NK cells from Chediak-Higashi syndrome patients
.
Front Immunol
.
2017
;
8
:
426
.
43.
Tesi
B
,
Rascon
J
,
Chiang
SCC
, et al
.
A RAB27A 5' untranslated region structural variant associated with late-onset hemophagocytic lymphohistiocytosis and normal pigmentation
.
J Allergy Clin Immunol
.
2018
;
142
(
1
):
317
-
321.e8
.
44.
Hori
M
,
Yasumi
T
,
Shimodera
S
, et al
.
A CD57(+) CTL degranulation assay effectively identifies familial hemophagocytic lymphohistiocytosis type 3 patients
.
J Clin Immunol
.
2017
;
37
(
1
):
92
-
99
.
45.
Schlums
H
,
Cichocki
F
,
Tesi
B
, et al
.
Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function
.
Immunity
.
2015
;
42
(
3
):
443
-
456
.
46.
Robin
X
,
Turck
N
,
Hainard
A
, et al
.
pROC: an open-source package for R and S+ to analyze and compare ROC curves
.
BMC Bioinformatics
.
2011
;
12
:
77
.
47.
Youden
WJ
.
Index for rating diagnostic tests
.
Cancer
.
1950
;
3
(
1
):
32
-
35
.
48.
Blakeley
DD
,
Oddone
EZ
,
Hasselblad
V
,
Simel
DL
,
Matchar
DB
.
Noninvasive carotid artery testing. A meta-analytic review
.
Ann Intern Med
.
1995
;
122
(
5
):
360
-
367
.
49.
Klenerman
P
,
Oxenius
A
.
T cell responses to cytomegalovirus
.
Nat Rev Immunol
.
2016
;
16
(
6
):
367
-
377
.
50.
Wang
EC
,
Taylor-Wiedeman
J
,
Perera
P
,
Fisher
J
,
Borysiewicz
LK
.
Subsets of CD8+, CD57+ cells in normal, healthy individuals: correlations with human cytomegalovirus (HCMV) carrier status, phenotypic and functional analyses
.
Clin Exp Immunol
.
1993
;
94
(
2
):
297
-
305
.
51.
Lee
J
,
Zhang
T
,
Hwang
I
, et al
.
Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals
.
Immunity
.
2015
;
42
(
3
):
431
-
442
.
52.
Corat
MA
,
Schlums
H
,
Wu
C
, et al
.
Acquired somatic mutations in PNH reveal long-term maintenance of adaptive NK cells independent of HSPCs
.
Blood
.
2017
;
129
(
14
):
1940
-
1946
.
53.
Schlums
H
,
Jung
M
,
Han
H
, et al
.
Adaptive NK cells can persist in patients with GATA2 mutation depleted of stem and progenitor cells
.
Blood
.
2017
;
129
(
14
):
1927
-
1939
.
54.
Santosa
EK
,
Kim
H
,
Ruckert
T
, et al
.
Control of nutrient uptake by IRF4 orchestrates innate immune memory
.
Nat Immunol
.
2023
;
24
(
10
):
1685
-
1697
.
55.
Roder
JC
,
Haliotis
T
,
Klein
M
, et al
.
A new immunodeficiency disorder in humans involving NK cells
.
Nature
.
1980
;
284
(
5756
):
553
-
555
.
56.
Arico
M
,
Nespoli
L
,
Maccario
R
, et al
.
Natural cytotoxicity impairment in familial haemophagocytic lymphohistiocytosis
.
Arch Dis Child
.
1988
;
63
(
3
):
292
-
296
.
57.
Marcenaro
S
,
Gallo
F
,
Martini
S
, et al
.
Analysis of natural killer-cell function in familial hemophagocytic lymphohistiocytosis (FHL): defective CD107a surface expression heralds Munc13-4 defect and discriminates between genetic subtypes of the disease
.
Blood
.
2006
;
108
(
7
):
2316
-
2323
.
58.
Kannan
K
,
Stewart
RM
,
Bounds
W
, et al
.
Lysosome-associated membrane proteins h-LAMP1 (CD107a) and h-LAMP2 (CD107b) are activation-dependent cell surface glycoproteins in human peripheral blood mononuclear cells which mediate cell adhesion to vascular endothelium
.
Cell Immunol
.
1996
;
171
(
1
):
10
-
19
.
59.
Betts
MR
,
Brenchley
JM
,
Price
DA
, et al
.
Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation
.
J Immunol Methods
.
2003
;
281
(
1-2
):
65
-
78
.
60.
Alter
G
,
Malenfant
JM
,
Altfeld
M
.
CD107a as a functional marker for the identification of natural killer cell activity
.
J Immunol Methods
.
2004
;
294
(
1-2
):
15
-
22
.
61.
Grutzkau
A
,
Smorodchenko
A
,
Lippert
U
,
Kirchhof
L
,
Artuc
M
,
Henz
BM
.
LAMP-1 and LAMP-2, but not LAMP-3, are reliable markers for activation-induced secretion of human mast cells
.
Cytometry A
.
2004
;
61
(
1
):
62
-
68
.
62.
Bryceson
YT
,
March
ME
,
Barber
DF
,
Ljunggren
HG
,
Long
EO
.
Cytolytic granule polarization and degranulation controlled by different receptors in resting NK cells
.
J Exp Med
.
2005
;
202
(
7
):
1001
-
1012
.
63.
Hennersdorf
F
,
Florian
S
,
Jakob
A
, et al
.
Identification of CD13, CD107a, and CD164 as novel basophil-activation markers and dissection of two response patterns in time kinetics of IgE-dependent upregulation
.
Cell Res
.
2005
;
15
(
5
):
325
-
335
.
64.
Cohnen
A
,
Chiang
SC
,
Stojanovic
A
, et al
.
Surface CD107a/LAMP-1 protects natural killer cells from degranulation-associated damage
.
Blood
.
2013
;
122
(
8
):
1411
-
1418
.
65.
Ammann
S
,
Lehmberg
K
,
Zur Stadt
U
, et al
.
Effective immunological guidance of genetic analyses including exome sequencing in patients evaluated for hemophagocytic lymphohistiocytosis
.
J Clin Immunol
.
2017
;
37
(
8
):
770
-
780
.
66.
Enders
A
,
Zieger
B
,
Schwarz
K
, et al
.
Lethal hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type II
.
Blood
.
2006
;
108
(
1
):
81
-
87
.
67.
Brandt
CS
,
Baratin
M
,
Yi
EC
, et al
.
The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans
.
J Exp Med
.
2009
;
206
(
7
):
1495
-
1503
.
68.
Hart
GT
,
Tran
TM
,
Theorell
J
, et al
.
Adaptive NK cells in people exposed to Plasmodium falciparum correlate with protection from malaria
.
J Exp Med
.
2019
;
216
(
6
):
1280
-
1290
.
69.
Ruckert
T
,
Romagnani
C
.
Extrinsic and intrinsic drivers of natural killer cell clonality
.
Immunol Rev
.
2024
;
323
(
1
):
80
-
106
.
70.
Tesi
B
,
Schlums
H
,
Cichocki
F
,
Bryceson
YT
.
Epigenetic regulation of adaptive NK cell diversification
.
Trends Immunol
.
2016
;
37
(
7
):
451
-
461
.
71.
Grossman
J
,
Cuellar-Rodriguez
J
,
Gea-Banacloche
J
, et al
.
Nonmyeloablative allogeneic hematopoietic stem cell transplantation for GATA2 deficiency
.
Biol Blood Marrow Transplant
.
2014
;
20
(
12
):
1940
-
1948
.
72.
Spinner
MA
,
Ker
JP
,
Stoudenmire
CJ
, et al
.
GATA2 deficiency underlying severe blastomycosis and fatal herpes simplex virus-associated hemophagocytic lymphohistiocytosis
.
J Allergy Clin Immunol
.
2016
;
137
(
2
):
638
-
640
.
73.
Brauer
N
,
Maruta
Y
,
Lisci
M
, et al
.
Immunodeficiency with susceptibility to lymphoma with complex genotype affecting energy metabolism (FBP1, ACAD9) and vesicle trafficking (RAB27A)
.
Front Immunol
.
2023
;
14
:
1151166
.
74.
Ohishi
Y
,
Ammann
S
,
Ziaee
V
, et al
.
Griscelli syndrome type 2 sine albinism: unraveling differential RAB27A effector engagement
.
Front Immunol
.
2020
;
11
:
612977
.
75.
Ammann
S
,
Elling
R
,
Gyrd-Hansen
M
, et al
.
A new functional assay for the diagnosis of X-linked inhibitor of apoptosis (XIAP) deficiency
.
Clin Exp Immunol
.
2014
;
176
(
3
):
394
-
400
.
76.
Holle
JR
,
Marsh
RA
,
Holdcroft
AM
, et al
.
Hemophagocytic lymphohistiocytosis in a female patient due to a heterozygous XIAP mutation and skewed X chromosome inactivation
.
Pediatr Blood Cancer
.
2015
;
62
(
7
):
1288
-
1290
.
77.
Yang
X
,
Hoshino
A
,
Taga
T
, et al
.
A female patient with incomplete hemophagocytic lymphohistiocytosis caused by a heterozygous XIAP mutation associated with non-random X-chromosome inactivation skewed towards the wild-type XIAP allele
.
J Clin Immunol
.
2015
;
35
(
3
):
244
-
248
.
78.
Qi
Q
,
Kannan
AK
,
August
A
.
Tec family kinases: Itk signaling and the development of NKT alphabeta and gammadelta T cells
.
FEBS J
.
2011
;
278
(
12
):
1970
-
1979
.
79.
Nichols
KE
,
Hom
J
,
Gong
SY
, et al
.
Regulation of NKT cell development by SAP, the protein defective in XLP
.
Nat Med
.
2005
;
11
(
3
):
340
-
345
.
80.
Ralph
E
,
Evans
J
,
Booth
C
,
Gilmour
K
.
Patients with XLP type 1 have variable numbers of NKT cells
.
Br J Haematol
.
2022
;
198
(
1
):
151
-
154
.
81.
Oliveira
JB
,
Bleesing
JJ
,
Dianzani
U
, et al
.
Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop
.
Blood
.
2010
;
116
(
14
):
e35
-
e40
.
82.
van Montfrans
JM
,
Hoepelman
AI
,
Otto
S
, et al
.
CD27 deficiency is associated with combined immunodeficiency and persistent symptomatic EBV viremia
.
J Allergy Clin Immunol
.
2012
;
129
(
3
):
787
-
793.e6
.
83.
Ma
CS
,
Hare
NJ
,
Nichols
KE
, et al
.
Impaired humoral immunity in X-linked lymphoproliferative disease is associated with defective IL-10 production by CD4+ T cells
.
J Clin Invest
.
2005
;
115
(
4
):
1049
-
1059
.

Author notes

L.E.C. and B.T. contributed equally to this study.

Data are available on request from the corresponding authors, Yenan T. Bryceson (yenan.bryceson@ki.se) and Samuel C. C. Chiang (sam.chiang@cchmc.org).

The online version of this article contains a data supplement.

There is a Blood Commentary on this article in this issue.

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.

Sign in via your Institution