Skip to main content
Erschienen in: Journal of Clinical Immunology 7/2021

Open Access 02.06.2021 | Original Article

Lineage-Specific Chimerism and Outcome After Hematopoietic Stem Cell Transplantation for DOCK8 Deficiency

verfasst von: Johannes Raedler, Thomas Magg, Meino Rohlfs, Christoph Klein, Tanja Vallée, Fabian Hauck, Michael H. Albert

Erschienen in: Journal of Clinical Immunology | Ausgabe 7/2021

Abstract

Bi-allelic variants in the dedicator of cytokinesis 8 (DOCK8) gene cause a combined immunodeficiency, characterized by recurrent sinopulmonary and skin infections, food allergies, eczema, eosinophilia, and elevated IgE. Long-term outcome is poor given susceptibility to infections, malignancy, and vascular complications. Allogeneic hematopoietic stem cell transplantation is currently the only curative treatment option and has shown promising outcome. The impact of mixed chimerism on long-term outcome is unclear. We reasoned that reversal of disease phenotype would depend on cell lineage-specific chimerism. DOCK8 variants were confirmed by Sanger and/or exome sequencing and immunoblot and/or intracellular flow cytometry. Donor chimerism was analyzed by XY-fluorescence in situ hybridization or quantitative short tandem repeat PCR. Outcome was assessed by laboratory tests, lymphocyte subsets, intracellular DOCK8 protein flow cytometry, T-cell proliferation analysis, and multiparameter immunoblot allergy screening. We report on nine patients, four of whom with mixed chimerism, with a median follow-up of 78 months after transplantation. Overall, we report successful transplantation with improvement of susceptibility to infections and allergies, and resolution of eczema in all patients. Immunological outcome in patients with mixed chimerism suggests a selective advantage for wild-type donor T-cells but lower donor B-cell chimerism possibly results in a tendency to hypogammaglobulinemia. No increased infectious and allergic complications were associated with mixed chimerism. Aware of the relatively small cohort size, we could not demonstrate a consistent detrimental effect of mixed chimerism on clinical outcomes. We nevertheless advocate aiming for complete donor chimerism in treating DOCK8 deficiency, but recommend reduced toxicity conditioning.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10875-021-01069-5.
Johannes Raedler, Thomas Magg, as well as Fabian Hauck and Michael H. Albert contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The inborn error of immunity (IEI) caused by bi-allelic dedicator of cytokinesis 8 (DOCK8) deficiency was first described in 2009 by Zhang et al. [1]. The resulting combined T- and B-cell immunodeficiency in DOCK8 deficiency presents with food allergies, eczema, eosinophilia, and elevated IgE, explaining why it was historically categorized as an autosomal-recessive hyper-IgE syndrome (HIES) [24]. Recurrent sinopulmonary infections with bronchiectasis, skin infections, lymphopenia, and hypogammaglobulinemia are other frequent findings [2, 3, 5]. Common infectious agents are bacteria and fungi, DNA viruses, or molluscum contagiosum. A TH1-deficient phenotype and cytokine abnormalities with TH2 activation may explain eosinophilia and IgE overproduction in DOCK8 deficiency [6, 7]. Diagnosis may be delayed or missed in patients with milder or varying phenotypes or near-normal flow cytometric DOCK8 expression caused by missense mutations or somatic reversions [3, 5, 8, 9]. The long-term natural disease outcome is poor, given infectious or vascular complications and an increased susceptibility to malignancy, predominantly of hematological or epithelial origin. Median survival is less than 20 years and almost all patients experience a life-threatening complication by the age of 25, despite prophylactic treatment [10].
DOCK8 is an atypical guanine nucleotide exchange factor of the DOCK 180 superfamily, which interact with Rho GTPases regulating cytoskeletal rearrangements [1, 5, 10, 11], partially explaining phenotypical overlap with other IEIs caused by cytoskeletal dysregulation, especially Wiskott-Aldrich syndrome (WAS) and CARMIL2 deficiency [12, 13]. Interaction between DOCK8 and WASP in NK-cells has been reported [2, 14, 15]. In comparison, the IEI caused by STAT3 deficiency, autosomal-dominant HIES, presents with different phenotypical features, such as connective tissue, dental, and skeletal abnormalities, and typically exhibits a less severe immunodeficiency [2, 3, 5, 1618].
Previous reports suggest that allogeneic hematopoietic stem cell transplantation (HSCT) offers a curative therapeutic option for DOCK8 deficiency [2, 12, 1922]. A recent retrospective analysis of a large cohort confirmed favorable outcomes after HSCT and suggested good safety and efficacy of reduced toxicity conditioning regimens. A detailed analysis of donor chimerism and immunological reconstitution was however not possible in this multi-center retrospective analysis [23]. Treosulfan or reduced-dose busulfan-based reduced toxicity regimens offer less short-term and possibly long-term toxicity [24]. However, they do not always result in complete donor chimerism of all cell lineages, which—depending on the underlying condition—may or may not result in reversal of the disease phenotype [25]. For example, in familial hemophagocytic lymphohistiocytosis, a donor T-cell chimerism of about 20–30% is deemed sufficient [26], regardless of the chimerism in other cell lineages, while in WAS complete donor chimerism is necessary for full disease correction [27]. Reports on immunological outcome of DOCK8 patients after HSCT are relatively scarce, especially for patients with mixed chimerism [19, 28, 29].
Given the fact that DOCK8 deficiency affects many hematopoietic lineages, we reasoned that reversal of disease phenotype after HSCT would depend on the level of cell lineage-specific donor chimerism. We here report detailed analysis of long-term clinical outcome, lineage-specific chimerism, laboratory parameters, and pulmonary function tests of nine patients who underwent HSCT for DOCK8 deficiency between 2004 and 2017, four of whom with ensuing variable degrees of mixed chimerism.

Methods

Data Acquisition

Data were retrieved from archived patients’ files. All laboratory analyses were part of the regular long-term follow-up schedule. All patients or their caregivers consented to scientific analysis of their data. P4 was lost to follow-up after 2.5 years. All patients have been part of previous publications [2, 20, 23].

Flow Cytometry

Flow cytometry for T-, B-, and NK-cell subsets was performed as described [13]. DOCK8 intracellular staining flow cytometry was performed using Cytofix/Cytoperm buffer set (Becton Dickinson, San Jose, USA (BD)), anti-DOCK8 (G-2, 1:20, Santa Cruz, Dallas, USA), and mouse FITC-conjugated secondary antibody (RMG1-1, 1:200, BioLegend, San Diego, USA) followed by blocking with normal mouse IgG (Thermo Fisher Scientific, Waltham, USA) and surface staining with PE-anti-CD3 (SK7, 1:10, BD), PC7-anti-CD19 (J3-119, 1:50, Beckman Coulter, Brea, USA), and APC-anti-CD56 (NCAM16.2, 1:50, BD).

Immunoblot Analysis

For immunoblot analysis, whole cell lysates were prepared from T-lymphoblasts as described [13]. Total protein was resolved on SDS-PAGE, transferred to nitrocellulose membrane, and probed with anti-DOCK8 (G-2, 1:1000), anti-GAPDH (6C5, 1:3000), and goat anti-mouse IgG-HRP (sc-2005, 1:10,000, all Santa Cruz).

Proliferation

PBMC were labeled with 2.5 µM carboxyfluorescein succinimidyl ester (CFSE, Thermo Fisher Scientific, Waltham, MA) and stimulated with anti-CD3-coupled beads (anti-Biotin MACSiBeads, Miltenyi Biotec, Bergisch Gladbach, Germany, coupled with Biotin-anti-CD3, OKT3) at a ratio of 5:1 with and without 1 µg/ml CD28 (CD28.2, both Thermo Fisher Scientific) or with 0.5 ng/ml PMA and 1 µM ionomycin (Sigma-Aldrich, St. Louis, MO). T-cell proliferation was measured by labeling PBMC with APC-H7-anti-CD3 (SK7, 1:50), APC-anti-CD4 (SK3, 1:50), PacB-anti-CD8 (RPA-T8, 1:50), and PE-anti-CD25 (M-A251, 1:25, all BD) 5 days after stimulation as described [13].

Exome and Sanger Sequencing

Exome sequencing was performed at the Dr. von Hauner Children’s Hospital NGS facility as previously described [13]. Briefly, genomic DNA from whole blood was used for preparation of whole-exome libraries using the SureSelect XT Human All Exon V6 + UTR kit (Agilent Technologies, Ratingen, Germany) and subsequently sequenced with a NextSeq 500 platform (Illumina, San Diego, CA) to an average coverage depth of 90 × . Bioinformatic analysis used Burrows-Wheeler Aligner (BWA 0.7.15), Genome Analysis ToolKit (GATK 3.6), and Variant Effect Predictor (VEP 89). Frequency filtering was done against public (e.g., GnomAD, ExAC, and GME) and in-house databases. Potentially causative variants were confirmed by Sanger sequencing.

Laboratory and Statistical Analyses

Donor chimerism for opposite-sex transplantations was analyzed by XY-fluorescence in situ hybridization (FISH), and by quantitative short tandem repeat PCR for same-sex transplantations, after MACS-based cell sorting, respectively. Allergy screening was performed by multiparameter immunoblots (Euroimmune, Lübeck, Germany).
Statistical analysis was performed with Prism version 7.0 (GraphPad, La Jolla, CA) using unpaired two-tailed T-tests with Welch’s correction not assuming equal variances, accepting P < 0.05 as significant. Central tendencies are reported as median values. Mean values are provided for selected results.

Results

Patients and HSCT

This report comprises nine patients with DOCK8 deficiency, confirmed by bi-allelic DOCK8 variants by Sanger and/or exome sequencing, who received allogeneic HSCT after reduced toxicity conditioning at our institution between 2004 and 2017 (Table 1). P8 and P9 exhibited mosaic DOCK8 expression in CD3+, CD19+, and CD56+ cells compatible with somatic reversions (Fig. 1a and b). P1 suffered from various co-morbidities before HSCT, including a corrected cardiac defect (Shone complex), corrected cleft palate with residual dysphagia and seizures, with resulting dystrophia and delayed psychomotor development. P2 was the first patient ever reported with successful HSCT for DOCK8 deficiency; however, transplantation in 2004 preceded the first description of the underlying genetic cause [20]. HSCT details are presented in Table 1. Notable long-term morbidity was a presumably total-body irradiation (TBI)-related thyroid cancer with pulmonary metastasis in P2 treated surgically with ongoing remission. We report on a cumulative follow-up of 727 patient months post-HSCT and a median follow-up of 78 months (33–187).
Table 1
Patient and HSCT characteristics
https://static-content.springer.com/image/art%3A10.1007%2Fs10875-021-01069-5/MediaObjects/10875_2021_1069_Tab1_HTML.png
Gray background highlights patients with mixed chimerism. Alem alemtuzumab (in mg/kg), BM bone marrow, BW body weight, CSA cyclosporine, Cy cyclophosphamide (in mg/kg), Flu fludarabine (in mg/m2), HLA human leucocyte antigen, LoTF loss to follow-up, mMUD mismatched unrelated donor, MRD matched related donor, MUD matched unrelated donor, MTX methotrexate, NEU neutrophils, PBSC peripheral blood stem cells, PLT platelets, TBI total-body irradiation, Treo treosulfan (in g/m2), WBC white blood cells. § and # denote siblings; “*” (asterisk) = P7 received a CD34 + stem cell boost on day + 170; “/” (slash) = no thrombocytopenia < 50.000/µl for P2 during HSCT

Hematologic and Immunologic Reconstitution

Complete donor chimerism was present in four patients (P6–P9; Fig. 1a and c, Table 2). Kinetics of lineage-specific peripheral blood chimerism are shown in Fig. 1c. In patients with mixed chimerism, median whole blood donor chimerism was 50% (16–75%), whereas median donor CD3+ T-cell chimerism was 97% (range: 87–99%) and median CD19+ B-cell chimerism was 41% (30–85%). Post-HSCT DOCK8 expression by flow cytometry was assessed for six patients and corresponded well with lineage-specific chimerism measured by XY-FISH or PCR (Fig. 1a and c, Table 2). We also measured CD16+CD56+ NK-cell chimerism by flow cytometry, revealing a median of 52% (5–99%), seemingly independent from the degree of T-cell chimerism.
Table 2
Outcome post-HSCT
https://static-content.springer.com/image/art%3A10.1007%2Fs10875-021-01069-5/MediaObjects/10875_2021_1069_Tab2_HTML.png
Most recent results for donor chimerism, blood count, lymphocyte subpopulations, immunoglobulin levels, and vaccination titers are shown. Reported allergies and challenged prior allergens are subject to patient or guardian reports. Time of analysis is given in months post-HSCT. Gray background highlights patients with mixed chimerism. Bold denotes values outside age-adjusted norm
EOS eosinophils, HB hemoglobin, IVIG intravenous immunoglobulin substitution, LoTF loss to follow-up, LYM lymphocytes, NA not available, NV no vaccination performed, PLT platelets, RTE recent thymic emigrants, SCIG subcutaneous immunogloblin substitution, WBC white blood cells. “*” (asterisk) = weekly subcutaneous immunoglobulin substitution
No patient experienced primary graft failure (Table 1). P7 received a CD34+ stem cell boost from the original donor on day + 170 for poor graft function following CMV-, BK-, and JC-viremia and ganciclovir treatment. At last follow-up, all patients had (near) complete hematologic reconstitution with no significant differences between patients with mixed or complete donor chimerism. Median hemoglobin value was 12.8 g/dl (mixed chimerism: 12.9, complete donor chimerism: 12.7; P = 0.42) with mild anemia in P4 and P6 (P6 has iron deficiency anemia). Median platelet count was 171 g/l (mixed: 179, complete: 159; P = 0.61) with mild thrombocytopenia in four patients and median leucocyte count was 4.8 g/l (mixed: 4.8, complete: 5.2; P0.52) with mild lymphopenia in P1, whose Shone complex was corrected by cardiac surgery with partial thymectomy pre-HSCT (Table 2).
Complete immunologic reconstitution as defined by normal values for lymphocyte subsets was seen in six patients with no significant differences between patients with mixed or complete donor chimerism (Table 2). Median cell counts were 1055/µl CD3+ T-cells (mixed: 1055, complete: 1239; P = 0.88), 534/µl CD4+ helper T-cells (mixed: 534, complete: 592; P = 0.83), and 439/µl CD8+ cytotoxic T-cells (mixed: 427, complete: 586; P = 0.61). Moderate CD4+- and severe CD8+-T-lymphopenia were present in P1. Median CD19+ B-cell count was 289/µl (mixed: 289, complete: 300; P = 0.66). Median CD16+CD56+ NK-cell count was 78/µl (mixed: 76, complete: 101; P = 0.52) with mild NK-lymphopenia in P1 and P2. Kinetics of immunologic reconstitution are shown in figure S1.
Thymopoiesis, as assessed by recent thymic emigrants (RTE; CD3+CD45RA+CD31+), was reduced in P1 (after thymectomy), with overall median RTE counts of 148/µl (mixed: 122, complete: 261; P = 0.93; Table 2). Naïve (CD45RA+) and memory (CD45R0+) T-cell subsets were available for eight patients for CD4+ and six patients for CD8+ T-cell-subsets, showing a reduction in memory CD8+ cells in patients with mixed chimerism (median overall: 158/µl, mixed: 136, complete: 261; P = 0.016). However, this may be skewed by partial thymectomy in P1, limited data availability, and biased ratios of naïve versus memory T-cells in P3 post-HSCT. T-cell proliferation to CD3/CD28 stimulation was normal in seven patients (Fig. 1d), missing for P4 and P9, who had normal T-cell proliferation to antigen stimulation (tetanus and diphtheria; data not shown). P6 had a pathologic reaction to antigen stimulation, but she was not vaccinated, maybe accounting for that. P8 has a reduced proliferation to candida stimulation (not shown).
Analysis of B-cell subsets was available for eight patients (Table 2), showing no significant differences between patients with complete or mixed chimerism. Median CD27+-memory-B-cell count was 27/µl (mixed: 18, complete: 29; P = 0.45), median IgM-memory-B-cells (CD19+CD27+IgD+IgM+) count was 12/µl (mixed: 8, complete: 14; P = 0.37), and median switched-memory-B-cell (CD19+CD27 + IgDIgM) count was 9/µl (mixed: 9, complete: 13; P = 0.29). A reduction in switched-memory-B cells was observed in three patients. Hereof, P7 had a complete donor chimerism and P1 and P5 had mixed chimerism. P1, P4, and P5 present with persistent hypogammaglobinemia at last follow-up, defined as IgG values below the age-adjusted normal range (Table 2). P1 receives weekly subcutaneous immunoglobulin (SCIG) substitution. P5 received intravenous immunoglobulin (IVIG) substitution every 4 to 8 weeks per patient preference, but discontinued substitution after 6 years without subsequent infectious complications. Excluding P1, median serum IgG level was significantly reduced in patients with mixed chimerism (overall: 10.1 g/l, mixed: 7,7, complete: 10.8; P = 0.047). IgG kinetics post-HSCT are shown in figure S2; no IgG-subclass analyses were performed. Overall median serum IgM level was 0.64 g/l including P1 (mixed: 0.60, complete: 1.03, P = 0.16) and median serum IgA level was 0.91 g/l (mixed: 0.89, complete: 0.98; P = 0.67). Pre-HSCT IgG levels were not analyzed because all patients except P4 were on Ig substitution.
Production of specific antibodies in response to vaccinations is shown in Table 2. Two patients were not vaccinated post-HSCT. P7, who also has reduced switched-memory-B-cells, generally showed poor humoral responses to vaccinations. Vaccination with lived-attenuated viruses (mumps, measles, rubella, varicella) was declined by seven patients, but serological testing suggests protective titers in those vaccinated.
In summary, these results show satisfactory immune reconstitution post-HSCT in all patients except P1, who had partial thymectomy pre-HSCT. A tendency to hypogammaglobulinemia in patients with mixed chimerism was observed. No further significant correlation between mixed chimerism and incomplete hematologic or immunologic reconstitution could be detected in this cohort.

Infections

Viremia was frequently diagnosed during pharmacological immunosuppression post-HSCT and treated with virostatic pharmacotherapy (Table 1). Notably, P9 experienced an aciclovir-resistant HSV infection (genetically proven) post-HSCT, which was successfully treated with foscarnet. At last follow-up, only P8 reported continued susceptibility to infection. She has recurrent respiratory infections presumably on the grounds of advanced structural lung damage pre-HSCT and requires regular prophylactic antibiotic treatment. P1 is free from severe infections under immunoglobulin replacement. Further noteworthy infections post-HSCT are shown in Table 1. Overall, infection frequency notably decreased after more than 1 year post-HSCT. No correlation between infectious complications and mixed chimerism was apparent.

Allergies

Eczema or atopic dermatitis resolved quickly post-HSCT in all patients and the allergic diathesis abated substantially. Two patients reported no allergies at last follow-up and the remainder reported mainly food allergies, specifically to dairy, egg, nut, or wheat products (Table 2). P8 reported severe allergic reactions to Gouda cheese and cefepime. In general, reported allergies post-HSCT were consistent with pre-HSCT conditions. Challenges of few food allergens showed tolerance post-HSCT, but exposure was mainly avoided per patient preference. Semi-quantitative multiparameter immunoblots were available for six patients for pre- and post-HSCT comparison (Fig. 2a), showing no obvious correlation between allergic tendency on immunoblot and donor chimerism. Skin prick testing was not performed. Overall, patients with high reactivity to food allergens on immunoblot pre-HSCT retained this post-HSCT without good correlation with clinical symptoms.
Total serum IgE was slowly down-trending post-HSCT (median IgE 17,438 IU/ml pre-HSCT), but continues to be elevated in five patients with a median level of 203 IU/ml (mixed: 203, complete: 246; P = 0.39). Eosinophilia resolved post-HSCT with a median eosinophil count of 0.11G/l (mixed: 0.11, complete: 0.12; P = 0.32), compared to 0.82G/l pre-HSCT (Fig. 2b, Table 2).
Bronchial hyperreactivity to aerosol allergens was documented in five patients pre-HSCT (P2, P5, P6, P7, and P9; Fig. 3). At last follow-up, four patients reported frequent use of inhaled β2 agonists, of which P5 and P8 require daily treatment. Routine spirometric testing showed persistent light or moderate airway obstruction in P3, P6, and P8 (Fig. 3). P8 reports limited physical capacities due to frequent respiratory infections with the remainder reporting no constraints.

Discussion

The IEI resulting from bi-allelic deficiency of DOCK8 presents with combined T- and B-cell immunodeficiency. Hallmarks include frequent respiratory and skin infections, food allergies, eczema, eosinophilia, and elevated IgE. Subsequent organ damage, susceptibility to malignancies, and vascular complications entail high mortality and morbidity, justifying HSCT as a curative treatment option. Limited data on long-term outcomes for patients with DOCK8 deficiency and mixed chimerism post-HSCT are available, warranting this report on nine patients with a median follow-up of 78 months (total 727 patient months) post-HSCT and in-depth analysis of cell lineage-specific chimerism. Overall, we report successful HSCT after reduced toxicity conditioning in all patients with improvement of susceptibility to infections and allergies and resolution of eczema. Specifically, patients with a mixed chimerism present nearly complete T-cell chimerism, suggesting a selective advantage for wild-type donor T-cells, but lower donor B-cell chimerism with a possible tendency to lower IgG values and immunoglobulin substitution. No increased allergic, respiratory, or infectious complications were observed in patients with mixed chimerism. Secondary thyroid cancer in P2 was attributed to TBI conditioning and was not interpreted as an increased risk for oncogenesis in the context of mixed chimerism.
Aydin et al. reported on a large cohort of 81 patients with DOCK8 deficiency and HSCT on behalf of the inborn errors working party of EBMT and ESID with promising overall outcomes of HSCT [23]. More recently, Haskologlu et al. reported on a Turkish cohort of 20 patients with DOCK8 deficiency of whom 11 patients underwent HSCT [28]. However, these and other reports have focused on survival, conditioning regimens, and transplant-related outcomes, rather than improvement of clinical aspects and immunologic reconstitution, particularly in patients with mixed chimerism. Al-Herz et al. reported a retrospective review of 11 patients with ameliorated infectious and atopic symptoms post-HSCT, including an analysis of mixed lineage-specific chimerism in three patients, albeit with significantly shorter follow-up than our cohort [19]. Overall survival was 100% in our study, compared to 91% (Haskologlu et al.), 91% (Al-Herz et al.), and 84% (Aydin et al.), and none of our patients suffered from cGvHD. In patients with mixed chimerism, lineage-specific analysis showed a median of 97% (87–99%) of donor T-cells and 41% (30–85%) of donor B-cells, comparable to results from Al-Herz et al. (donor T-cells 82–97%, donor B-cells 0–46%, donor myeloid 0–7%) [19]. In line with our findings, Al-Herz et al. conclude that donor-derived T cells have a selective advantage. The role of DOCK8 in T-cell survival may provide a selective advantage for DOCK8 wild-type donor T-cells after HSCT, resulting in more complete T-cell donor chimerism [30, 31]. The extensive lymphocyte phenotype of DOCK8 deficiency and its correction post-HSCT have been catalogued by Pillay et al., although only in patients with complete donor T-cell chimerism [29]. Due to the retrospective nature of our study and lacking consent for biobanking, no further T-cell phenotyping or cytokine profiles are available for our patients for further assessment of T-cell reconstitution in DOCK8 deficiency with mixed chimerism. P2–P5 present with near complete T-cell chimerism with proper overall T-cell subset reconstitution, and therefore, we expect comparable T-cell phenotypes to patients with complete chimerism as shown by Pillay et al. [29]. P1, however, with 87% CD3+ donor T-cells has reduced thymopoiesis after partial thymectomy, weakening overall interpretation of T-cell subsets for patients with mixed chimerism. Further analysis of T-cell subsets for P1 would therefore not add meaningful knowledge to T-cell reconstitution in mixed chimerism for DOCK8 deficiency. Therefore, we believe our cohort may add additional information for T-cell reconstitution post-HSCT in patients with mixed chimerism.
Other studies found lower proliferative rates of DOCK8-deficient B-cells [8, 11]. Conversely, we observed no relevant selective advantage for wild-type B-cells in vivo as evidenced by an overall lower but stable B-cell chimerism over time after HSCT, but subtle effects cannot be excluded due to the low number of patients. Myeloid chimerism was not available for our patients, but a selective advantage for wild-type DOCK8 in lymphoid cells, rather than myeloid cells, was previously shown [32].
DOCK8 deficiency has been shown to impair immunological synapses through ICAM-1 in mouse models and reduced persistence in germinal centers and affinity maturation or differentiation into marginal zone cells in DOCK8-deficient B-cells [33]. Additionally, decreased generation of memory B-cells with diminished long-lasting antibody production has been related to impaired B-cell signaling [2, 11] and reduced B-cell proliferation was seen in patients with DOCK8 deficiency [11, 29]. DOCK8 deficiency has been shown to disrupt B-cell responses to signals via TLR, BCR, CD40, and cytokines, especially IL-21 [29]. In one patient, Al-Herz et al. found increased donor chimerism in the switched memory B-cell compartment, in agreement with recent findings of DOCK8 enrichment in the memory B-cells [32]. However, we did not analyze this in our patients but reduced switched memory B-cell numbers were not clearly associated with mixed chimerism in our cohort. The latter is exemplified by P5 with hypogammaglobulinemia and a higher CD19+ donor B-cell chimerism, but the lowest CD19+ B-cell count and reduced memory B-cells as compared to other patients with mixed chimerism. B-cell reconstitution post-HSCT is variable and influenced by many factors in pediatric patients [34]. Additionally, DOCK8-mutant B cells are unable to form marginal zone B cells or to persist in germinal centers and to undergo affinity maturation [33]. Split chimerism, with allogeneic T-cells but persistent autologous B-cells, impairs reconstitution of humoral immunity and has been implied with defective IL-21 receptor signaling in T-cell-dependent B-cell differentiation post-HSCT in patients with X-SCID and JAK3-SCID [35]. Our data show a possible tendency to hypogammaglobulinemia and necessity for immunoglobulin substitution in patients with mixed chimerism, but with the limited data available, no causal claim for DOCK8-specific effects can be made. Further research is required to elucidate the effect of split chimerism on B-cell function and adaptive humoral immunity reconstitution in general and particularly in DOCK8 deficiency.
We observed no correlation between persistent airway obstruction post-HSCT and mixed chimerism. Persistent food allergies post-HSCT are commonly seen in DOCK8-deficient patients without correlation with chimerism and are slow to improve post-HSCT [12], presumably due to persistence of long-lived IgE-producing plasma cells [36] and tissue-resident memory B-cells. Happel et al. report similar results for 12 patients and detected allergen-specific mast cells through skin prick tests after HSCT [36]. Only few of our patients reported challenging prior food allergies post-HSCT; however, reintroduction of a diversified diet post-HSCT has been successfully reported for numerous other patients [28, 3638]. The low incidence of allergic events post-HSCT, despite overall stable allergen reactivity on immunoblot, may be attributed to higher T-cell chimerism and a speculative role of regulatory T-cell (Treg) functions. Janssen et al. demonstrated reduced suppressive activity of Treg in DOCK8 deficiency [39]. Treg activity may contribute to improvement of eczema in our patients and previous reports [23, 37, 38]. Unfortunately, no evaluation of Treg was available for our patients. A possible role for Treg suppression of TH2-cells in atopic dermatitis is reviewed by Agrawal et al. [40]. Interestingly, Treg express an affinity towards skin-homing through CCR4, CCR6, and cutaneous lymphocyte-associated antigen (CLA), a similar mechanism as seen in IPEX syndrome, caused by a Treg defect [40]. Further studies are necessary to elucidate the role of Treg in DOCK8 deficiency. Unfortunately, the importance of NK-cell immunity in defense against virus infections cannot be properly addressed by this report, due to missing appropriate functional assays [14].
We believe that early diagnosis and timely curative management are crucial in DOCK8 deficiency as non-reversible organ damage may occur due to infectious or auto-inflammatory complications, but may be delayed by varying phenotypes or near-normal DOCK8 expression caused by hypomorphic variants or somatic reversions [5]. Somatic reversions were present in two of our patients and both present with complete donor chimerism post-HSCT. The importance of early detection is exemplified by P8, who was diagnosed at a later age than her sister P3 and suffers from structural pre-HSCT lung damage with frequent respiratory infections despite complete donor chimerism. In comparison, P3 presents as a healthy and active young girl, despite mixed chimerism. Somatic reversion in general may ameliorate disease course, but patients still do experience fatal complications [8]. Rare constellations such as somatic reversion of a hypomorphic DOCK8 allele leading to an atypical and relatively milder phenotype have been reported [9] and Pillay et al. recently reported in-depth analysis of partial somatic reversion in three patients with compound heterozygous mutations in DOCK8 with clinical improvement over time [32]. Overall, T-cells showed highest levels of somatic reversion, in agreement with our findings regarding selective advantages for donor T-cells post-HSCT. Therefore, indication for HSCT in patients with somatic mosaicism should be evaluated individually, but we currently recommend HSCT with reduced conditioning for those with infectious complications to prevent organ damage and fatal complications. Hypothetically there could be long-term complications of clonal selection by somatic reversion, but Pillay et al. have not observed that [32]. Metabolomic biomarkers may support clinicians in future differential diagnosis of atopic dermatitis in order to achieve early diagnosis of DOCK8 deficiency [41], but currently, the diagnosis demands experienced clinicians and intracellular flow cytometry for DOCK8, most successfully detected in B-cells, which show minimal reversion [8], and confirmation by Sanger and/or exome sequencing.
With this report, we hope to increase our understanding of long-term immunologic outcomes post-HSCT for DOCK8 deficiency and support clinicians, patients, and families facing this debilitating disease. We highlight the importance of intracellular flow cytometry for DOCK8 for diagnosis and chimerism monitoring at the single cell level. A review of conditioning regimens is beyond the scope of this report, but overall, promising outcomes for reduced toxicity conditioning reported here and elsewhere [23] support the notion that such regimens are preferable for this disease, despite a higher frequency of mixed chimerism. Aware of the relatively small cohort analyzed, we could not demonstrate a consistent detrimental effect of mixed chimerism on immunological and clinical outcomes, especially the frequency of infections. Still, further research is needed to investigate the effect of mixed chimerism in DOCK8 deficiency, especially on cell lineages not covered in this report. Given this uncertainty, we advocate aiming for complete donor chimerism in treating DOCK8 deficiency, but suggest aiming at reduced toxicity over myeloablation, especially for patients with pre-existing organ damage.

Acknowledgements

The authors wish to thank patients and guardians consenting to this report and all staff involved in patient care and diagnostics.

Declarations

Ethics Approval

All caretakers consented to data collection via the German pediatric registry for stem cell transplantation (“Pädiatrisches Register für Stammzelltransplantation”, PRST), which was approved by the ethics committee of the MHH, Hannover, Germany and by the local ethics committee of LMU, Munich.
Informed written consent about this data analysis and publication was obtained from all individual participants or respective legal guardians included in the study, except for patient 4 who was lost to follow-up and could not be reached.
See above.

Conflict of Interest

MA declares travel support by Medac. All other authors have no conflict of interest to declare.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361(21):2046–55.CrossRef Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361(21):2046–55.CrossRef
2.
Zurück zum Zitat Aydin SE, Kilic SS, Aytekin C, Kumar A, Porras O, Kainulainen L, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options - a review of 136 patients. J Clin Immunol. 2015;35(2):189–98.CrossRef Aydin SE, Kilic SS, Aytekin C, Kumar A, Porras O, Kainulainen L, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options - a review of 136 patients. J Clin Immunol. 2015;35(2):189–98.CrossRef
3.
Zurück zum Zitat Engelhardt KR, Gertz ME, Keles S, Schäffer AA, Sigmund EC, Glocker C, et al. The extended clinical phenotype of 64 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2015;136(2):402–12.CrossRef Engelhardt KR, Gertz ME, Keles S, Schäffer AA, Sigmund EC, Glocker C, et al. The extended clinical phenotype of 64 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2015;136(2):402–12.CrossRef
4.
Zurück zum Zitat Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol. 2009;124(6):1289-302.e4.CrossRef Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol. 2009;124(6):1289-302.e4.CrossRef
5.
Zurück zum Zitat Zhang Q, Jing H, Su HC. Recent advances in DOCK8 immunodeficiency syndrome. J Clin Immunol. 2016;36(5):441–9.CrossRef Zhang Q, Jing H, Su HC. Recent advances in DOCK8 immunodeficiency syndrome. J Clin Immunol. 2016;36(5):441–9.CrossRef
6.
Zurück zum Zitat Biggs CM, Keles S, Chatila TA. DOCK8 deficiency: insights into pathophysiology, clinical features and management. Clin Immunol. 2017;181:75–82.CrossRef Biggs CM, Keles S, Chatila TA. DOCK8 deficiency: insights into pathophysiology, clinical features and management. Clin Immunol. 2017;181:75–82.CrossRef
7.
Zurück zum Zitat Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, et al. Dedicator of cytokinesis 8-deficient CD4(+) T cells are biased to a T(H)2 effector fate at the expense of T(H)1 and T(H)17 cells. J Allergy Clin Immunol. 2017;139(3):933–49.CrossRef Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, et al. Dedicator of cytokinesis 8-deficient CD4(+) T cells are biased to a T(H)2 effector fate at the expense of T(H)1 and T(H)17 cells. J Allergy Clin Immunol. 2017;139(3):933–49.CrossRef
8.
Zurück zum Zitat Jing H, Zhang Q, Zhang Y, Hill BJ, Dove CG, Gelfand EW, et al. Somatic reversion in dedicator of cytokinesis 8 immunodeficiency modulates disease phenotype. J Allergy Clin Immunol. 2014;133(6):1667–75.CrossRef Jing H, Zhang Q, Zhang Y, Hill BJ, Dove CG, Gelfand EW, et al. Somatic reversion in dedicator of cytokinesis 8 immunodeficiency modulates disease phenotype. J Allergy Clin Immunol. 2014;133(6):1667–75.CrossRef
9.
Zurück zum Zitat Kienzler AK, van Schouwenburg PA, Taylor J, Marwah I, Sharma RU, Noakes C, et al. Hypomorphic function and somatic reversion of DOCK8 cause combined immunodeficiency without hyper-IgE. Clin Immunol. 2016;163:17–21.CrossRef Kienzler AK, van Schouwenburg PA, Taylor J, Marwah I, Sharma RU, Noakes C, et al. Hypomorphic function and somatic reversion of DOCK8 cause combined immunodeficiency without hyper-IgE. Clin Immunol. 2016;163:17–21.CrossRef
10.
Zurück zum Zitat Zhang Q, Davis JC, Dove CG, Su HC. Genetic, clinical, and laboratory markers for DOCK8 immunodeficiency syndrome. Dis Markers. 2010;29(3–4):131–9.PubMed Zhang Q, Davis JC, Dove CG, Su HC. Genetic, clinical, and laboratory markers for DOCK8 immunodeficiency syndrome. Dis Markers. 2010;29(3–4):131–9.PubMed
11.
Zurück zum Zitat Jabara HH, McDonald DR, Janssen E, Massaad MJ, Ramesh N, Borzutzky A, et al. DOCK8 functions as an adaptor that links TLR-MyD88 signaling to B cell activation. Nat Immunol. 2012;13(6):612–20.CrossRef Jabara HH, McDonald DR, Janssen E, Massaad MJ, Ramesh N, Borzutzky A, et al. DOCK8 functions as an adaptor that links TLR-MyD88 signaling to B cell activation. Nat Immunol. 2012;13(6):612–20.CrossRef
12.
Zurück zum Zitat Albert MH, Freeman AF. Wiskott-Aldrich syndrome (WAS) and dedicator of Cytokinesis 8- (DOCK8) Deficiency. Front Pediatr. 2019;7:451.CrossRef Albert MH, Freeman AF. Wiskott-Aldrich syndrome (WAS) and dedicator of Cytokinesis 8- (DOCK8) Deficiency. Front Pediatr. 2019;7:451.CrossRef
13.
Zurück zum Zitat Schober T, Magg T, Laschinger M, Rohlfs M, Linhares ND, Puchalka J, et al. A human immunodeficiency syndrome caused by mutations in CARMIL2. Nat Commun. 2017;8:14209.CrossRef Schober T, Magg T, Laschinger M, Rohlfs M, Linhares ND, Puchalka J, et al. A human immunodeficiency syndrome caused by mutations in CARMIL2. Nat Commun. 2017;8:14209.CrossRef
14.
Zurück zum Zitat Ham H, Guerrier S, Kim J, Schoon RA, Anderson EL, Hamann MJ, et al. Dedicator of cytokinesis 8 interacts with talin and Wiskott-Aldrich syndrome protein to regulate NK cell cytotoxicity. J Immunol (Baltimore, Md: 1950). 2013;190(7):3661–9.CrossRef Ham H, Guerrier S, Kim J, Schoon RA, Anderson EL, Hamann MJ, et al. Dedicator of cytokinesis 8 interacts with talin and Wiskott-Aldrich syndrome protein to regulate NK cell cytotoxicity. J Immunol (Baltimore, Md: 1950). 2013;190(7):3661–9.CrossRef
15.
Zurück zum Zitat Kim D, Uner A, Saglam A, Chadburn A, Crane GM. Peripheral eosinophilia in primary immunodeficiencies of actin dysregulation: a case series of Wiskott-Aldrich syndrome, CARMIL2 and DOCK8 deficiency and review of the literature. Ann Diagn Pathol. 2019;43:151413.CrossRef Kim D, Uner A, Saglam A, Chadburn A, Crane GM. Peripheral eosinophilia in primary immunodeficiencies of actin dysregulation: a case series of Wiskott-Aldrich syndrome, CARMIL2 and DOCK8 deficiency and review of the literature. Ann Diagn Pathol. 2019;43:151413.CrossRef
16.
Zurück zum Zitat Kane A, Deenick EK, Ma CS, Cook MC, Uzel G, Tangye SG. STAT3 is a central regulator of lymphocyte differentiation and function. Curr Opin Immunol. 2014;28:49–57.CrossRef Kane A, Deenick EK, Ma CS, Cook MC, Uzel G, Tangye SG. STAT3 is a central regulator of lymphocyte differentiation and function. Curr Opin Immunol. 2014;28:49–57.CrossRef
17.
Zurück zum Zitat Aryan Z, Nabavi M, Shabani M, Keles S, Zoghi S, Chatila T, et al. Hypomorphic DOCK8 deletion causes hypereosinophilic syndrome. Pediatr Blood Cancer. 2020;67(2):e28084.CrossRef Aryan Z, Nabavi M, Shabani M, Keles S, Zoghi S, Chatila T, et al. Hypomorphic DOCK8 deletion causes hypereosinophilic syndrome. Pediatr Blood Cancer. 2020;67(2):e28084.CrossRef
18.
Zurück zum Zitat Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357(16):1608–19.CrossRef Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357(16):1608–19.CrossRef
19.
Zurück zum Zitat Al-Herz W, Chu JI, van der Spek J, Raghupathy R, Massaad MJ, Keles S, et al. Hematopoietic stem cell transplantation outcomes for 11 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2016;138(3):852-9 e3.CrossRef Al-Herz W, Chu JI, van der Spek J, Raghupathy R, Massaad MJ, Keles S, et al. Hematopoietic stem cell transplantation outcomes for 11 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2016;138(3):852-9 e3.CrossRef
20.
Zurück zum Zitat Bittner TC, Pannicke U, Renner ED, Notheis G, Hoffmann F, Belohradsky BH, et al. Successful long-term correction of autosomal recessive hyper-IgE syndrome due to DOCK8 deficiency by hematopoietic stem cell transplantation. Klin Padiatr. 2010;222(6):351–5.CrossRef Bittner TC, Pannicke U, Renner ED, Notheis G, Hoffmann F, Belohradsky BH, et al. Successful long-term correction of autosomal recessive hyper-IgE syndrome due to DOCK8 deficiency by hematopoietic stem cell transplantation. Klin Padiatr. 2010;222(6):351–5.CrossRef
21.
Zurück zum Zitat McDonald DR, Massaad MJ, Johnston A, Keles S, Chatila T, Geha RS, et al. Successful engraftment of donor marrow after allogeneic hematopoietic cell transplantation in autosomal-recessive hyper-IgE syndrome caused by dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2010;126(6):1304-5.e3.CrossRef McDonald DR, Massaad MJ, Johnston A, Keles S, Chatila T, Geha RS, et al. Successful engraftment of donor marrow after allogeneic hematopoietic cell transplantation in autosomal-recessive hyper-IgE syndrome caused by dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol. 2010;126(6):1304-5.e3.CrossRef
22.
Zurück zum Zitat Shah NN, Freeman AF, Su H, Cole K, Parta M, Moutsopoulos NM, et al. Haploidentical related donor hematopoietic stem cell transplantation for dedicator-of-cytokinesis 8 deficiency using post-transplantation cyclophosphamide. Biol Blood Marrow Transplant. 2017;23(6):980–90.CrossRef Shah NN, Freeman AF, Su H, Cole K, Parta M, Moutsopoulos NM, et al. Haploidentical related donor hematopoietic stem cell transplantation for dedicator-of-cytokinesis 8 deficiency using post-transplantation cyclophosphamide. Biol Blood Marrow Transplant. 2017;23(6):980–90.CrossRef
23.
Zurück zum Zitat Aydin SE, Freeman AF, Al-Herz W, Al-Mousa HA, Arnaout RK, Aydin RC, et al. Hematopoietic stem cell transplantation as treatment for patients with DOCK8 deficiency. J Allergy Clin Immunol Pract. 2019;7(3):848–55.CrossRef Aydin SE, Freeman AF, Al-Herz W, Al-Mousa HA, Arnaout RK, Aydin RC, et al. Hematopoietic stem cell transplantation as treatment for patients with DOCK8 deficiency. J Allergy Clin Immunol Pract. 2019;7(3):848–55.CrossRef
24.
Zurück zum Zitat Slatter MA, Boztug H, Pötschger U, Sykora KW, Lankester A, Yaniv I, et al. Treosulfan-based conditioning regimens for allogeneic haematopoietic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transplant. 2015;50(12):1536–41.CrossRef Slatter MA, Boztug H, Pötschger U, Sykora KW, Lankester A, Yaniv I, et al. Treosulfan-based conditioning regimens for allogeneic haematopoietic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transplant. 2015;50(12):1536–41.CrossRef
25.
Zurück zum Zitat Slatter MA, Rao K, Abd Hamid IJ, Nademi Z, Chiesa R, Elfeky R, et al. Treosulfan and fludarabine conditioning for hematopoietic stem cell transplantation in children with primary immunodeficiency: UK experience. Biol Blood Marrow Transplant. 2018;24(3):529–36.CrossRef Slatter MA, Rao K, Abd Hamid IJ, Nademi Z, Chiesa R, Elfeky R, et al. Treosulfan and fludarabine conditioning for hematopoietic stem cell transplantation in children with primary immunodeficiency: UK experience. Biol Blood Marrow Transplant. 2018;24(3):529–36.CrossRef
26.
Zurück zum Zitat Hartz B, Marsh R, Rao K, Henter JI, Jordan M, Filipovich L, et al. The minimum required level of donor chimerism in hereditary hemophagocytic lymphohistiocytosis. Blood. 2016;127(25):3281–90.CrossRef Hartz B, Marsh R, Rao K, Henter JI, Jordan M, Filipovich L, et al. The minimum required level of donor chimerism in hereditary hemophagocytic lymphohistiocytosis. Blood. 2016;127(25):3281–90.CrossRef
27.
Zurück zum Zitat Moratto D, Giliani S, Bonfim C, Mazzolari E, Fischer A, Ochs HD, et al. Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980–2009: an international collaborative study. Blood. 2011;118(6):1675–84.CrossRef Moratto D, Giliani S, Bonfim C, Mazzolari E, Fischer A, Ochs HD, et al. Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980–2009: an international collaborative study. Blood. 2011;118(6):1675–84.CrossRef
28.
Zurück zum Zitat Haskologlu S, Kostel Bal S, Islamoglu C, Aytekin C, Guner S, Sevinc S, et al. Clinical, immunological features and follow up of 20 patients with dedicator of cytokinesis 8 (DOCK8) deficiency. Pediatr Allergy Immunol. 2020;31(5):515–27. Haskologlu S, Kostel Bal S, Islamoglu C, Aytekin C, Guner S, Sevinc S, et al. Clinical, immunological features and follow up of 20 patients with dedicator of cytokinesis 8 (DOCK8) deficiency. Pediatr Allergy Immunol. 2020;31(5):515–27.
29.
Zurück zum Zitat Pillay BA, Avery DT, Smart JM, Cole T, Choo S, Chan D, et al. Hematopoietic stem cell transplant effectively rescues lymphocyte differentiation and function in DOCK8-deficient patients. JCI Insight. 2019;5(11):e127527. Pillay BA, Avery DT, Smart JM, Cole T, Choo S, Chan D, et al. Hematopoietic stem cell transplant effectively rescues lymphocyte differentiation and function in DOCK8-deficient patients. JCI Insight. 2019;5(11):e127527.
30.
Zurück zum Zitat Randall KL, Chan SS, Ma CS, Fung I, Mei Y, Yabas M, et al. DOCK8 deficiency impairs CD8 T cell survival and function in humans and mice. J Exp Med. 2011;208(11):2305–20.CrossRef Randall KL, Chan SS, Ma CS, Fung I, Mei Y, Yabas M, et al. DOCK8 deficiency impairs CD8 T cell survival and function in humans and mice. J Exp Med. 2011;208(11):2305–20.CrossRef
31.
Zurück zum Zitat Lambe T, Crawford G, Johnson AL, Crockford TL, Bouriez-Jones T, Smyth AM, et al. DOCK8 is essential for T-cell survival and the maintenance of CD8+ T-cell memory. Eur J Immunol. 2011;41(12):3423–35.CrossRef Lambe T, Crawford G, Johnson AL, Crockford TL, Bouriez-Jones T, Smyth AM, et al. DOCK8 is essential for T-cell survival and the maintenance of CD8+ T-cell memory. Eur J Immunol. 2011;41(12):3423–35.CrossRef
32.
Zurück zum Zitat Pillay BA, Fusaro M, Gray PE, Statham AL, Burnett L, Bezrodnik L, et al. Somatic reversion of pathogenic DOCK8 variants alters lymphocyte differentiation and function to effectively cure DOCK8 deficiency. J Clin Invest. 2021;131(3):e142434. Pillay BA, Fusaro M, Gray PE, Statham AL, Burnett L, Bezrodnik L, et al. Somatic reversion of pathogenic DOCK8 variants alters lymphocyte differentiation and function to effectively cure DOCK8 deficiency. J Clin Invest. 2021;131(3):e142434.
33.
Zurück zum Zitat Randall KL, Lambe T, Johnson AL, Treanor B, Kucharska E, Domaschenz H, et al. Dock8 mutations cripple B cell immunological synapses, germinal centers and long-lived antibody production. Nat Immunol. 2009;10(12):1283–91.CrossRef Randall KL, Lambe T, Johnson AL, Treanor B, Kucharska E, Domaschenz H, et al. Dock8 mutations cripple B cell immunological synapses, germinal centers and long-lived antibody production. Nat Immunol. 2009;10(12):1283–91.CrossRef
34.
Zurück zum Zitat van der Maas NG, Berghuis D, van der Burg M, Lankester AC. B cell reconstitution and influencing factors after hematopoietic stem cell transplantation in children. Front Immunol. 2019;10:782.CrossRef van der Maas NG, Berghuis D, van der Burg M, Lankester AC. B cell reconstitution and influencing factors after hematopoietic stem cell transplantation in children. Front Immunol. 2019;10:782.CrossRef
35.
Zurück zum Zitat Recher M, Berglund LJ, Avery DT, Cowan MJ, Gennery AR, Smart J, et al. IL-21 is the primary common γ chain-binding cytokine required for human B-cell differentiation in vivo. Blood. 2011;118(26):6824–35.CrossRef Recher M, Berglund LJ, Avery DT, Cowan MJ, Gennery AR, Smart J, et al. IL-21 is the primary common γ chain-binding cytokine required for human B-cell differentiation in vivo. Blood. 2011;118(26):6824–35.CrossRef
36.
Zurück zum Zitat Happel CS, Stone KD, Freeman AF, Shah NN, Wang A, Lyons JJ, et al. Food allergies can persist after myeloablative hematopoietic stem cell transplantation in dedicator of cytokinesis 8-deficient patients. J Allergy Clin Immunol. 2016;137(6):1895-8.e5.CrossRef Happel CS, Stone KD, Freeman AF, Shah NN, Wang A, Lyons JJ, et al. Food allergies can persist after myeloablative hematopoietic stem cell transplantation in dedicator of cytokinesis 8-deficient patients. J Allergy Clin Immunol. 2016;137(6):1895-8.e5.CrossRef
37.
Zurück zum Zitat Barlogis V, Galambrun C, Chambost H, Lamoureux-Toth S, Petit P, Stephan JL, et al. Successful allogeneic hematopoietic stem cell transplantation for DOCK8 deficiency. J Allergy Clin Immunol. 2011;128(2):420-22.e2.CrossRef Barlogis V, Galambrun C, Chambost H, Lamoureux-Toth S, Petit P, Stephan JL, et al. Successful allogeneic hematopoietic stem cell transplantation for DOCK8 deficiency. J Allergy Clin Immunol. 2011;128(2):420-22.e2.CrossRef
38.
Zurück zum Zitat Metin A, Tavil B, Azik F, Azkur D, Ok-Bozkaya I, Kocabas C, et al. Successful bone marrow transplantation for DOCK8 deficient hyper IgE syndrome. Pediatr Transplant. 2012;16(4):398–9.CrossRef Metin A, Tavil B, Azik F, Azkur D, Ok-Bozkaya I, Kocabas C, et al. Successful bone marrow transplantation for DOCK8 deficient hyper IgE syndrome. Pediatr Transplant. 2012;16(4):398–9.CrossRef
39.
Zurück zum Zitat Janssen E, Morbach H, Ullas S, Bannock JM, Massad C, Menard L, et al. Dedicator of cytokinesis 8–deficient patients have a breakdown in peripheral B-cell tolerance and defective regulatory T cells. J Allergy Clin Immunol. 2014;134(6):1365–74.CrossRef Janssen E, Morbach H, Ullas S, Bannock JM, Massad C, Menard L, et al. Dedicator of cytokinesis 8–deficient patients have a breakdown in peripheral B-cell tolerance and defective regulatory T cells. J Allergy Clin Immunol. 2014;134(6):1365–74.CrossRef
40.
Zurück zum Zitat Agrawal R, Wisniewski JA, Woodfolk JA. The role of regulatory T cells in atopic dermatitis. Curr Probl Dermatol. 2011;41:112–24.CrossRef Agrawal R, Wisniewski JA, Woodfolk JA. The role of regulatory T cells in atopic dermatitis. Curr Probl Dermatol. 2011;41:112–24.CrossRef
41.
Zurück zum Zitat Jacob M, Gu X, Luo X, Al-Mousa H, Arnaout R, Al-Saud B, et al. Metabolomics distinguishes DOCK8 deficiency from atopic dermatitis: towards a biomarker discovery. Metabolites. 2019;9(11):274. Jacob M, Gu X, Luo X, Al-Mousa H, Arnaout R, Al-Saud B, et al. Metabolomics distinguishes DOCK8 deficiency from atopic dermatitis: towards a biomarker discovery. Metabolites. 2019;9(11):274.
Metadaten
Titel
Lineage-Specific Chimerism and Outcome After Hematopoietic Stem Cell Transplantation for DOCK8 Deficiency
verfasst von
Johannes Raedler
Thomas Magg
Meino Rohlfs
Christoph Klein
Tanja Vallée
Fabian Hauck
Michael H. Albert
Publikationsdatum
02.06.2021
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 7/2021
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-021-01069-5

Weitere Artikel der Ausgabe 7/2021

Journal of Clinical Immunology 7/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.