Skip to main content
Erschienen in: Journal of Clinical Immunology 8/2021

Open Access 24.08.2021 | Original Article

A Novel STK4 Mutation Impairs T Cell Immunity Through Dysregulation of Cytokine-Induced Adhesion and Chemotaxis Genes

verfasst von: Andrea Guennoun, Salim Bougarn, Taushif Khan, Rafah Mackeh, Mahbuba Rahman, Fatima Al-Ali, Manar Ata, Waleed Aamer, Debra Prosser, Tanwir Habib, Evonne Chin-Smith, Khawla Al-Darwish, Qian Zhang, Alya Al-Shakaki, Amal Robay, Ronald G. Crystal, Khalid Fakhro, Amal Al-Naimi, Eman Al Maslamani, Amjad Tuffaha, Ibrahim Janahi, Mohammad Janahi, Donald R. Love, Mohammed Yousuf Karim, Bernice Lo, Amel Hassan, Mehdi Adeli, Nico Marr

Erschienen in: Journal of Clinical Immunology | Ausgabe 8/2021

Abstract

Purpose

Human serine/threonine kinase 4 (STK4) deficiency is a rare, autosomal recessive genetic disorder leading to combined immunodeficiency; however, the extent to which immune signaling and host defense are impaired is unclear. We assessed the functional consequences of a novel, homozygous nonsense STK4 mutation (NM_006282.2:c.871C > T, p.Arg291*) identified in a pediatric patient by comparing his innate and adaptive cell-mediated and humoral immune responses with those of three heterozygous relatives and unrelated controls.

Methods

The genetic etiology was verified by whole genome and Sanger sequencing. STK4 gene and protein expression was measured by quantitative RT-PCR and immunoblotting, respectively. Cellular abnormalities were assessed by high-throughput RT-RCR, RNA-Seq, ELISA, and flow cytometry. Antibody responses were assessed by ELISA and phage immunoprecipitation-sequencing.

Results

The patient exhibited partial loss of STK4 expression and complete loss of STK4 function combined with recurrent viral and bacterial infections, notably persistent Epstein–Barr virus viremia and pulmonary tuberculosis. Cellular and molecular analyses revealed abnormal fractions of T cell subsets, plasmacytoid dendritic cells, and NK cells. The transcriptional responses of the patient’s whole blood and PBMC samples indicated dysregulated interferon signaling, impaired T cell immunity, and increased T cell apoptosis as well as impaired regulation of cytokine-induced adhesion and leukocyte chemotaxis genes. Nonetheless, the patient had detectable vaccine-specific antibodies and IgG responses to various pathogens, consistent with a normal CD19 + B cell fraction, albeit with a distinctive antibody repertoire, largely driven by herpes virus antigens.

Conclusion

Patients with STK4 deficiency can exhibit broad impairment of immune function extending beyond lymphoid cells.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10875-021-01115-2.
Andrea Guennoun, Salim Bougarn and Taushif Khan contributed equally.
Donald R. Love, Mohammed Yousuf Karim, Bernice Lo and Amel Hassan contributed equally. Mehdi Adeli and Nico Marr contributed equally.

Publisher's Note

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

Introduction

Human serine/threonine kinase 4 (STK4) deficiency is a rare autosomal recessive (AR) genetic disorder leading to combined immunodeficiency with severe T cell lymphopenia. This condition is characterized by a predisposition to a wide range of bacterial and viral infectious diseases, mucocutaneous candidiasis, lymphomas, and congenital heart disease [1]. To date, STK4 deficiency has been reported in relatively few patients; therefore, the extent to which immune signaling and host defense mechanisms are impaired or dysregulated in affected individuals remains incompletely understood. However, the spectrum of clinical manifestations associated with STK4 deficiency has been steadily increasing with each new case report.
STK4 deficiency was first reported by Nehme et al. in two patients from unrelated Turkish families harboring a homozygous nonsense mutation in the STK4 gene [2]. The patients experienced complications due to recurrent bacterial and viral infections, most notably persistent Epstein–Barr virus (EBV) viremia, which ultimately resulted in Hodgkin B cell lymphoma. Due to weak expression of the homing receptors CCR7 and CD62L, the authors attributed the underlying mechanism of STK4 deficiency to increased death of naïve and proliferating T cells, and impaired homing of CD8+ T cells to secondary lymphoid organs [2]. Abdollahpour et al. reported the cases of three siblings of Iranian descent with a homozygous premature stop codon in the STK4 gene [3]. These patients suffered from T and B cell lymphopenia, intermittent neutropenia, and atrial septal defects, as well as recurrent bacterial and viral infections, mucocutaneous candidiasis, cutaneous warts, and skin abscesses. Interestingly, Schipp et al. reported a Turkish patient with STK4 deficiency who developed a highly malignant B cell lymphoma at the age of 10 years and a second, independent Hodgkin lymphoma 5 years later. However, no detectable EBV or other common virus infection was detected in this patient. The authors speculated that the lymphoma may have developed due to the lack of the tumor suppressive function of STK4 or perturbed immune surveillance due to the diminished CD4+ T cell compartment [4]. In contrast, most malignancies reported in patients with STK4 deficiency are associated with prolonged EBV viremia, ultimately leading to the development of B cell lymphomas [2, 57]. More specifically, patients present with Hodgkin B cell lymphoma [2], extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue [8], Burkitt’s lymphoma [7], or maxillary sinus diffuse large B cell lymphoma [9]. Additional clinical features in patients with STK4 deficiency include salt-losing tubulopathy, suggestive of an acquired Gitelman syndrome, immune complex glomerulonephritis, and Castleman-like disease [10], juvenile idiopathic arthritis [11], human beta-papillomavirus-associated epidermodysplasia verruciformis [11, 12], primary cardiac T cell lymphoma [6], and short stature [13].
Studies in mice and humans have shown that STK4 plays a pivotal role in lymphocyte function by regulating integrin-dependent T lymphocyte trafficking, proliferation, and differentiation [14, 15]. Of note, the STK4 protein is broadly expressed in various human hemopoietic cells, most notably monocytes, and is not restricted to lymphocytes (https://​www.​proteinatlas.​org/​ENSG00000101109-STK4). However, its role in T cell-independent functions is less well understood. Recently, Jørgensen et al. studied innate immune signaling in the context of STK4 deficiency by in vitro stimulation or infection of PBMCs obtained from an 11-year-old female STK4−/− patient of a consanguineous Syrian family. These studies revealed defective type I/II and III interferon (IFN) responses to a variety of purified Toll-like receptor (TLR) agonists, live viruses and bacterial lysates due to impaired phosphorylation of the kinase TBK1 and the transcription factor IRF3 [13]. The results also revealed increased apoptosis in STK4-deficient T cells and neutrophil granulocytes, possibly linked to defective FOXO signaling in STK4-deficient T cells as shown in earlier studies [2, 3], further supporting the important role of STK4 in T cell survival.
In this study, we identified an AR STK4 deficiency in a child from consanguineous parents, which was due to a novel homozygous stop-gain mutation in a region encoding a coiled-coil domain located downstream of the kinase domain. We investigated the functional consequences of the new variant on innate and adaptive cell-mediated and humoral immune responses.

Results

Clinical Description of the Case and Family Members
The patient was the third child (male) of consanguineous parents (first-degree cousins). The patient had a younger sibling who was healthy, as were both parents. Of note, the parents reported the death of the patient’s two elder siblings; one died with a history of chronic headache, coughing, and lymphoma, while the other died with a history of chronic coughing (Fig. 1A). However, no detailed medical records or genetic data were available for the two deceased siblings.
The patient suffered from recurrent skin rashes starting from infancy, recurrent chest infections since early childhood, and an overall failure to thrive with low weight gain and short stature (data not shown), consistent with previous reports of other patients with STK4 deficiency [13]. The patient’s early medical history included a productive cough of yellow/white mucoid sputum associated with intermittent fever, which was more common at bedtime; however, early medical records were very limited and likely to be incomplete. After closer clinical monitoring starting at elementary school age, the patient was diagnosed with bronchiectasis and asthma, and started on asthma control medication, including Ventolin. He also experienced complications due to recurrent viral and bacterial infections, chronic suppurative otitis media, and recurrent pneumonia. The patient was also diagnosed with tuberculosis (TB), which was confirmed by Mycobacteria tuberculosis complex-positive culture, while the result of a QuantiFERON assay performed in parallel was “indeterminate.” The patient was treated for pulmonary TB for approximately 1 year. However, a year after stopping treatment, the patient suffered from TB reactivation and was put on anti-TB medication (cycloserine, linezolid, moxifloxacin, and pyrazinamide) for a further 2 years. As a teenager, he also presented with a lower chest infection, and a chest X-ray confirmed lower left consolidation. A sputum culture revealed abundant growth of Haemophilus influenzae and Streptococcus pneumoniae, as well as modest growth of methicillin-resistant Staphylococcus aureus. About a year later, the patient was hospitalized with a second episode of lower chest infection by H. influenzae and multiple-drug-resistant Klebsiella pneumoniae. EBV viremia was also detected during the early teenage years and persisted to the most recent follow-up (Supplementary Table S1). During his teenage years, the patient also suffered from intermittent neutropenia and severe lymphopenia (Supplementary Table S2) with markedly decreased naïve CD45RA+ cells (11.1%; normal range 46–77%); the onset may have been earlier but was not detected due to the late diagnosis. The patient had received BCG vaccination at birth, as well as OPV, MMR, varicella, and meningococcal vaccines at school age. Antibody responses to childhood vaccination were within the normal range (Supplementary Table S3).
Homozygosity for a Stop-Gain Mutation in the STK4 Gene
Whole genome sequencing revealed a rare, homozygous nonsense mutation in the STK4 gene (NM_006282.2:c.871C > T, p.Arg291*) in the patient, whereas both parents and the younger sibling were identified as heterozygous carriers, suggesting an AR inheritance pattern (Fig. 1A). The STK4 genotypes of the patient and his family members were confirmed by clinical Sanger sequencing (Fig. 1B). The combined annotation-dependent depletion (CADD) score of the variant was 42, providing further evidence of its deleteriousness (Fig. 1C and D).
The mutant STK4 Allele is a LOF Variant
The mutant STK4 protein was not detected in PBMC-derived T cells from the patient by Western blot analysis using a monoclonal antibody directed against the N-terminus of the protein, whereas intermediate STK4 protein levels were detected in the parents compared to two unrelated healthy controls (Fig. 1E). A STK4 transcript was detected by mRNA-Seq and RT-qPCR in PBMC-derived T cells of the patient, albeit at reduced levels compared to controls with a wild-type genotype and the heterozygous parents (Supplementary Figure S1A and B).
Reduced Fractions of naïve T Helper, and Dendritic Cell Subsets, as well as Increased Effector Memory and Apoptotic T Helper and Precursor NK Cells in the PBMCs of the Patient
We performed polychromatic flow cytometric analyses of PBMCs obtained from the patient at middle-school-age to compare the lymphocyte subset distribution with that of his parents, his younger sibling, and one unrelated control (Fig. 2). As expected, we found a lower fraction of T cells in the patient compared to the controls (Fig. 2A, B and C), which was mainly attributed to selective CD4+ T cell lymphopenia (Fig. 2C). The CD19+ B cell population was not affected in the patient (11.7% vs. control range 10.9–20.8%) (Fig. 2A). Similarly, the patient’s CD56+CD3 NK cells were within the normal range (4.59% vs. control range 1.87–6.82%) (Fig. 2B). However, we noticed a significant increase in the CD56bright NK cell subset in the patient (1.78% vs. control range 0.058–0.42%) (Fig. 2B). We then assessed CD4+ T cell subsets and found an increase in programmed death-1 (PD-1)-expressing T helper cells in the patient (Fig. 2D). Further analysis of PBMC expression of CD45RA and CCR7 revealed low frequencies of CD45RA+CCR7+ double-positive naïve T cells in the patient, while his CD45RACCR7 effector memory population was increased (Fig. 2E). Similarly, the CD27+CD28+ T cell subset, which consists mainly of naïve T cells, was also slightly decreased in the patient (Fig. 2F). Finally, assessment of the dendritic cell (DC) subsets showed a decrease in the CD11cCD123+ plasmacytoid DC (pDC) population in the patient, while his CD11c+CD123 myeloid DCs (mDCs) population remained normal (Fig. 2G).
The Patient has a Distinct Antiviral Antibody Repertoire
To further assess the humoral immunity status of the patient, we performed large-scale serum antibody profiling by phage immunoprecipitation-sequencing (PhIP-Seq). The patient was seropositive for antibodies specific to a variety of common viruses and bacteria, including human herpesviruses (HHV)-4 (EBV), -5 (CMV) and -8; enterovirus (EV)-B; and human respiratory syncytial virus, human rhinoviruses A and B, S. pneumoniae and S. aureus (Fig. 3A). The antibody repertoire breadth in the patient was similar to that of the controls (Fig. 3B). Nevertheless, PCA of the enriched antibody-antigen interactions showed an overall distinct antibody repertoire in the patient compared to those of his family members and unrelated controls (Fig. 3C). These differences were largely driven by antibodies directed against structural proteins of human herpesviruses (HHV)-4 and -5, which was consistent with the patient’s active EBV viremia (Fig. 3D).
Gene Expression Signatures Suggest Dysregulated Interferon Signaling and Impaired T Cell Activation, Inhibition of T Cell Proliferation, and Increased T Cell Apoptosis
To further elucidate the functional consequences of STK4 deficiency at the molecular level, we performed gene expression analyses using either whole blood (WB) samples or PBMCs isolated from the patient and control subjects following in vitro stimulation with a variety of immune activators. First, we stimulated WB of the patient, his family members, and an unrelated control subject with purified pattern recognition receptor (PRR) agonists (lipopolysaccharides of Escherichia coli K12 (LPSK12) (TLR4 agonist), muramyl dipeptide (MDP) (NOD2 agonist), Poly(I:C) (TLR3 agonist), Poly(dA:dT) (multiple-PRR agonist), resiquimod (R848) (TLR7/8 agonist), cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) (STING agonist), and 5′ triphosphate double-stranded RNA (5'ppp-dsRNA) (RIG-I agonist)), cytokines (IFN-a/IFN-b (an interferon-α/β receptor IFNAR agonist), IFN-g, IL-1b, and TNF-a), a potent mitogen (phorbol 12-myristate 13-acetate (PMA)/ionomycin), and BCR or TCR activators. We measured the expression of 180 functionally well-annotated target genes selected a priori from a larger set of genes responsive to WB stimulation with purified PRR agonists, recombinant cytokines, and pyogenic bacteria [16] (see “Methods” and Supplementary Table S5, Online Resource 1); mock stimulations served as controls. Among these target genes, we then filtered for differentially expressed genes (DEGs) for which the transcriptional responses of the patient’s WB samples to any of the in vitro stimulation conditions showed high variance compared with those of the other family members and the unrelated control (Supplementary Figure S2). The identified DEGs were associated with caspases and apoptosis, type I and II interferon signaling, inflammation, cell signaling, and ubiquitination, as well as cell movement and phagocytosis (Fig. 4 and Supplementary Table S6).
Finally, we performed mRNA-Seq of PBMCs isolated from the patient and stimulated or not with either IFN-a/IFN-b or PMA/ionomycin. Both parents and three unrelated, healthy controls were assessed for comparison. We filtered for genes responsive to in vitro stimulation among the unrelated control subjects and found a marked dysregulation, but not complete abrogation, of IFN-regulated gene expression in the patient (Supplementary Figure S3A and S3B). After an additional filtering step (see Methods, Online Resource 1), we performed gene enrichment analyses on the subsets of either IFN-a/IFN-b- or PMA/ionomycin-responsive DEGs that were dysregulated in the patient using ClueGO [17] and Cytoscape [18], thereby taking advantage of functionally grouped GO and KEGG/BioCarta pathway annotation networks. To gain a better mechanistic understanding of the molecular consequences of STK4 deficiency, we focused our analyses on gene networks that either encompass STK4 or that include genes encoding products that interact directly with the STK4 protein (Supplementary Figure S3C). This analysis revealed several gene networks involved in the regulation of cell adhesion and leukocyte chemotaxis encompassing STK4, as well as gene networks involved in IFN-a/IFN-b- or mitogen-induced regulation of gene expression and biosynthesis processes that are typically associated with cell chemotaxis and adhesion-mediated cell signaling (Fig. 5A). Several of the genes that belong to these GO and pathway annotation networks are also previously reported type I interferon-responsive genes [19] (Fig. 5B). We also assessed regulatory effects on the subsets of IFN-a/IFN-b- or PMA/ionomycin-responsive DEGs using IPA and examined which genes or their upstream regulators are known to bind to, or are regulated by, STK4. This analysis revealed two regulatory networks of IFN-a/IFN-b-responsive genes encompassing several cytokine-, chemokine-, and adhesion factor- or receptor genes that are indirectly regulated by STK4, which was consistent with our ClueGO analysis. These genes are also involved in cytotoxicity and death of immune cells, as well as adhesion and migration of lymphocytes and mononuclear leukocytes (Fig. 5C, D, Supplementary Figure S3D-E). However, no regulatory effects networks were identified among PMA/ionomycin-responsive genes through the use of IPA (data not shown). Finally, we performed a canonical pathway comparison analysis of the entire sets of either IFN-a/IFN-b- or PMA/ionomycin-responsive genes using IPA to identify pathways that are differentially activated or inhibited in the patient’s cells compared to those of the controls. In response to IFNAR activation, nine pathways were differentially regulated in the patient compared to the controls, most of which are linked to T cell signaling and apoptosis, cell proliferation, oxidative stress, and, interestingly, IL-23 signaling (Fig. 5E). Similarly, we observed several pathways that are normally repressed following mitogen activation, but instead were highly activated or dysregulated in the patient’s cells. These included pathways primarily involved in T cell effector functions, T and B cell activation, cell cycle arrest, and apoptosis (Fig. 5E).
Given the apparent dysregulation of IL-23 signaling in the patient’s cells, we also examined absolute IL-23 and IFNG gene expression, either at baseline or following stimulation with either PMA/ionomycin or IFN-a/IFN-b. In comparison to the control subjects, IL-23 gene expression in the patient was highly impaired at baseline and largely unresponsive to PMA/ionomycin stimulation, whereas IFNG gene expression appeared normal (Supplementary Figure S1C and D).

Discussion

In this study, we identified a novel stop-gain mutation in a patient with AR complete STK4 deficiency. This mutation was found to be located in a genomic region that encodes the coiled-coil domain of STK4, downstream of its protein kinase domain. We were unable to detect even a truncated STK4 protein in the patient using a monoclonal antibody to the N-terminal region of STK4, suggesting that protein expression of the mutated allele is completely abrogated due to nonsense-mediated decay.
In accordance with earlier case reports [5, 8, 20], we found that the PBMCs isolated from our patient had reduced fractions of CD4+ naïve, but increased effector memory, T helper cell subsets compared with those in the STK4wt/mut family members and an unrelated STK4wt/wt control. Furthermore, flow cytometric analysis showed a considerable proportion of the remaining T helper cell subset in the patient expressed higher levels of PD-1, and our RNA-Seq analyses revealed dysregulation of several pathways in the patient, suggesting elevated T cell exhaustion and impaired effector functions of the residual T cells. Whether this is a consequence of persistent EBV viremia [2123] or an intrinsic feature of STK4 deficiency, or perhaps both, remains to be established. Previous studies have shown that EBV reactivation correlates with the expression of PD-1/PD-L1 antigens in patients with proliferative glomerulonephritis [24]. On the other hand, CD4+ T cell lymphopenia has also been reported in STK4-deficient patients in the absence of detectable EBV infection [4]. In addition, the patient presented with episodes of intermittent neutropenia, which is consistent with previous observations [2, 3, 6, 9, 13].
Our results also highlight that STK4 deficiency can lead to the impairment of a variety of T cell-independent and innate immune responses. Indeed, we detected a considerable proportion of CD56bright NK cells in the PBMCs isolated from the patient. While these cells constitute only a small fraction of NK cells in the peripheral blood of healthy individuals, they represent the majority of NK cells in secondary lymphoid tissues. CD56bright NK cells are thought to be NK cell precursors [25] and may have immunoregulatory properties [26]. We also observed a decreased fraction of pDCs in the patient’s peripheral blood. Whether this is an indirect consequence of active EBV infection, as shown in mouse studies [27], or whether low levels of pDCs contribute directly to a lack of EBV control, remains unclear. As reported by Jørgensen et al. [13], we also observed dysregulated type I and II IFN signaling in the patient’s cells. Interestingly, transcriptomic analysis of the patient’s PBMCs in response to IFNAR activation in vitro revealed a marked dysregulation of IFN-regulated gene expression, affecting interferon-stimulated genes (ISGs). Our enrichment analyses of either IFN-a/IFN-b- or PMA/ionomycin-responsive genes that showed differential expression between the patient and controls revealed several gene networks reminiscent of dysregulated cytokine-stimulated cell adhesion, leukocyte chemotaxis, and impaired T cell activation, likely resulting in T cell exhaustion and enhanced immune cell death. Dysregulation of these proinflammatory cytokines and chemokines has also been implicated in cancer pathogenesis [28]. Moreover, in accordance with our findings, Dang et al. demonstrated that leukocytes of patients with AR STK4 deficiency exhibited impaired chemotaxis after stimulation with CXCL11 and did not bind to ICAM-1 [5].
It cannot be ruled out that the dysregulated type I IFN-induced gene expression signature in the patient’s PBMCs is, in part, also a consequence of abnormal proportions of some leukocyte subsets. Indeed, we observed decreased fractions of pDCs, which are potent producers of type I IFN under in vitro and in vivo conditions [29]. Nonetheless, the short duration of the in vitro stimulation and gene expression experiments (2 h) and the low proportions of pDCs, CD56bright NK cells, and effector memory T cell subsets relative to the total PBMC population make it less likely that abnormal proportions of some leukocytes subsets in the patient have a major effect on their PBMC transcriptome. Overall, the suboptimal IFN signaling may contribute to the T cell immunodeficiency and the vulnerability of STK4-deficient patients to viral infection and cancer development. However, overall fractions of CD19+ B cells (Fig. 2A) and IgG antibody responses to childhood vaccination (Supplementary Table S3) or common microbial infection (Fig. 3) did not appear to be diminished in our patient, apart from our observation that the antibodies were predominantly specific for HHV-4 and -5 antigens. Of note, a recent study in STK4−/− mice and nine patients from five unrelated families with STK4 deficiency suggested that STK4 is required for normal humoral immunity since knockout mice and patients had reduced marginal zone B (MZB) cells as well as reduced numbers of innate-like B-1b cell subsets, while the overall fractions of circulating CD19+ B cells were normal, as in our patient [30]. This raises the possibility that patients with STK4 deficiency may also have a selective impairment in the ability to mount robust T cell-independent, polysaccharide-specific antibody responses to control natural infection with encapsulated bacteria, such as H. influenzae, K. pneumoniae and S. pneumoniae, which is consistent with the clinical history of our patient. Polysaccharide-specific antibody responses (or the lack of) are undetectable using the PhIP-Seq assay as it exclusively detects antibodies that target protein antigens and is limited in its capacity to detect conformational and post-translationally modified epitopes [31]. High efficacy of plain polysaccharide-based vaccines also depends on the maturation of MZB cells, which usually does not occur until the second year of life [32]. In our patient, the specific antibody responses were at the lower end of our laboratory reference range (Supplementary Table S3). However, anti-pneumococcal polysaccharide antibodies cannot be used as definitive markers of MZB cell-mediated immunity due to the introduction of the conjugate pneumococcal vaccine into the local routine immunization schedule. The literature shows variability in the specific antibody responses in STK4-deficient patients, ranging from normal to absent [30]. The history of infection with H. influenzae, K. pneumonia, and S. pneumoniae in our patient could have also interfered with the utility and interpretation of tests of responses to plain polysaccharide vaccines. Therefore, humoral immunity of patients with STK4 deficiency toward encapsulated bacteria requires further investigation.
We also demonstrated a profound impairment of IL-23 gene expression in the patient’s PBMCs, both at baseline and following in vitro stimulation. IL-23 is produced by innate lymphoid cells, gamma-delta T cells, DCs, and macrophages, and it has been shown that IL-23-dependent IFN- immunity plays a pivotal role in controlling Mycobacterium tuberculosis (Mtb) infection [33]. It is therefore tempting to speculate that impaired IL-23 gene expression contributed to patient’s susceptibility to pulmonary TB. Despite the clinical evidence of pulmonary TB, the patient’s QuantiFERON test result was indeterminate, which is likely to reflect a combination of cellular dysfunction and profound lymphopenia. Of note, Radwan et al. [7] also speculated that complications in a 9-year-old Egyptian boy with STK4 deficiency were associated with mycobacterial infection, although tuberculin skin-test results were negative, and the results from QuantiFERON tests were inconclusive.
It remains unclear whether malignancies in STK4-deficient patients are a secondary consequence of persistent EBV viremia, or whether such patients are inherently prone to malignancies due to dysregulation of oncogenes, even in the absence of EBV infection [4]. Interestingly, our RNA-Seq experiments revealed upregulation of mitogen-induced B cell-activating factor (BAFF) receptor gene (TNFRSF13C) expression in the patient, suggesting activation of BAFF signaling, in contrast to the controls where this pathway was inhibited following PMA/ionomycin stimulation (Fig. 5 and Supplementary Table S4). Studies in vitro and in mice have shown that EBV drives autonomous B cell proliferation [34], which also depends on T cell-independent survival signals provided by the BAFF receptor. Excessive BAFF levels have been implicated in several B-lineage malignancies [3538], which have also been reported in the context of STK4 deficiency, with or without EBV viremia [2, 58, 10]. Our observations provide further mechanistic insights into the susceptibility of STK4-deficient patients to malignancies, although they do not allow firm conclusions to be drawn about the role of EBV in this process. Nonetheless, it is tempting to speculate that STK4-deficient patients, particularly those with persistent EBV viremia, may benefit from treatment with immune checkpoint inhibitors. Using a humanized mouse model, Ma et al. [39] demonstrated a direct beneficial effect of PD-1/CTLA-4 blockade mediated by monoclonal antibodies against PD-1 or CTLA-4 alone, or in combination, on EBV-associated B cell lymphomas, thereby providing further evidence in support of this hypothesis. However, TB reactivation or primary Mtb infections have also been reported in cancer patients who received checkpoint inhibitors [4042]. Therefore, the potential therapeutic benefits of checkpoint inhibitors in patients with STK4 deficiency require further investigation.

Methods

Detailed methods are provided in the Online Resources (Online Resource 1).

Acknowledgements

The authors would like to thank the patient and his family for participating in this study. The authors also thank Stephen Elledge (Brigham and Women’s Hospital, Harvard University Medical School) for kindly providing the VirScan phage library; the Integrated Genomics Core and Deep Phenotyping Services team of Sidra Medicine for their assistance with the high-throughput PCR experiments, RNA sequencing, and flow cytometry; and Jessica Tamanini (Insight Editing London) for proofreading and editing the manuscript.

Declarations

Ethics Approval

The study protocol was approved by the institutional review board (IRB) of Sidra Medicine (protocol no. 1601002512) and Weill Cornell Medicine, Qatar (protocol no. 13–00065).
Written informed consent was obtained from each participant in accordance with local regulations governing human subject research.
All the authors have seen and approved the manuscript, which has not been accepted or published elsewhere.

Competing Interests

The authors declare no competing interests.
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.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, et al. Correction to: Human Inborn Errors of Immunity: 2019 Update on the Classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol. 2020;40(1):65.PubMedPubMedCentral Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, et al. Correction to: Human Inborn Errors of Immunity: 2019 Update on the Classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol. 2020;40(1):65.PubMedPubMedCentral
2.
Zurück zum Zitat Nehme NT, Schmid JP, Debeurme F, Andre-Schmutz I, Lim A, Nitschke P, et al. MST1 mutations in autosomal recessive primary immunodeficiency characterized by defective naive T-cell survival. Blood. 2012;119(15):3458–68.PubMed Nehme NT, Schmid JP, Debeurme F, Andre-Schmutz I, Lim A, Nitschke P, et al. MST1 mutations in autosomal recessive primary immunodeficiency characterized by defective naive T-cell survival. Blood. 2012;119(15):3458–68.PubMed
3.
Zurück zum Zitat Abdollahpour H, Appaswamy G, Kotlarz D, Diestelhorst J, Beier R, Schaffer AA, et al. The phenotype of human STK4 deficiency. Blood. 2012;119(15):3450–7.PubMedPubMedCentral Abdollahpour H, Appaswamy G, Kotlarz D, Diestelhorst J, Beier R, Schaffer AA, et al. The phenotype of human STK4 deficiency. Blood. 2012;119(15):3450–7.PubMedPubMedCentral
4.
Zurück zum Zitat Schipp C, Schlutermann D, Honscheid A, Nabhani S, Holl J, Oommen PT, et al. EBV negative lymphoma and autoimmune lymphoproliferative syndrome like phenotype extend the clinical spectrum of primary immunodeficiency caused by STK4 deficiency. Front Immunol. 2018;9:2400.PubMedPubMedCentral Schipp C, Schlutermann D, Honscheid A, Nabhani S, Holl J, Oommen PT, et al. EBV negative lymphoma and autoimmune lymphoproliferative syndrome like phenotype extend the clinical spectrum of primary immunodeficiency caused by STK4 deficiency. Front Immunol. 2018;9:2400.PubMedPubMedCentral
5.
Zurück zum Zitat Dang TS, Willet JD, Griffin HR, Morgan NV, O’Boyle G, Arkwright PD, et al. Defective leukocyte adhesion and chemotaxis contributes to combined immunodeficiency in humans with autosomal recessive MST1 deficiency. J Clin Immunol. 2016;36(2):117–22.PubMedPubMedCentral Dang TS, Willet JD, Griffin HR, Morgan NV, O’Boyle G, Arkwright PD, et al. Defective leukocyte adhesion and chemotaxis contributes to combined immunodeficiency in humans with autosomal recessive MST1 deficiency. J Clin Immunol. 2016;36(2):117–22.PubMedPubMedCentral
6.
Zurück zum Zitat Sherkat R, Sabri MR, Dehghan B, Bigdelian H, Reisi N, Afsharmoghadam N, et al. EBV lymphoproliferative-associated disease and primary cardiac T-cell lymphoma in a STK4 deficient patient: a case report. Medicine (Baltimore). 2017;96(48):e8852. Sherkat R, Sabri MR, Dehghan B, Bigdelian H, Reisi N, Afsharmoghadam N, et al. EBV lymphoproliferative-associated disease and primary cardiac T-cell lymphoma in a STK4 deficient patient: a case report. Medicine (Baltimore). 2017;96(48):e8852.
7.
Zurück zum Zitat Radwan N, El-Owaidy R, El-Sayed ZA, Abdel-Baky A, El-Haddad A, Rashad H, et al. A case of STK4 deficiency with complications evoking mycobacterial infection. J Clin Immunol. 2020;40(4):665–9.PubMed Radwan N, El-Owaidy R, El-Sayed ZA, Abdel-Baky A, El-Haddad A, Rashad H, et al. A case of STK4 deficiency with complications evoking mycobacterial infection. J Clin Immunol. 2020;40(4):665–9.PubMed
8.
Zurück zum Zitat Lum SH, Bonney D, Cheesman E, Wrignt NB, Hughes S, Wynn R. Successful curative therapy with rituximab and allogeneic haematopoietic stem cell transplantation for MALT lymphoma associated with STK4-mutated CD4+ lymphocytopenia. Pediatr Blood Cancer. 2016;63(9):1657–9.PubMed Lum SH, Bonney D, Cheesman E, Wrignt NB, Hughes S, Wynn R. Successful curative therapy with rituximab and allogeneic haematopoietic stem cell transplantation for MALT lymphoma associated with STK4-mutated CD4+ lymphocytopenia. Pediatr Blood Cancer. 2016;63(9):1657–9.PubMed
9.
Zurück zum Zitat Ashrafi F, Klein C, Poorpooneh M, Sherkat R, Khoshnevisan R. A case report of sinusoidal diffuse large B-cell lymphoma in a STK4 deficient patient. Medicine (Baltimore). 2020;99(9):e18601. Ashrafi F, Klein C, Poorpooneh M, Sherkat R, Khoshnevisan R. A case report of sinusoidal diffuse large B-cell lymphoma in a STK4 deficient patient. Medicine (Baltimore). 2020;99(9):e18601.
10.
Zurück zum Zitat Al-Saud B, Alajlan H, Sabar H, Anwar S, Alruwaili H, Al-Hussain T, et al. STK4 deficiency in a patient with immune complex glomerulonephritis, salt-losing tubulopathy, and Castleman’s-like disease. J Clin Immunol. 2019;39(8):823–6.PubMed Al-Saud B, Alajlan H, Sabar H, Anwar S, Alruwaili H, Al-Hussain T, et al. STK4 deficiency in a patient with immune complex glomerulonephritis, salt-losing tubulopathy, and Castleman’s-like disease. J Clin Immunol. 2019;39(8):823–6.PubMed
11.
Zurück zum Zitat Sharafian S, Ziaee V, Shahrooei M, Ahadi M, Parvaneh N. A Novel STK4 Mutation presenting with juvenile idiopathic arthritis and epidermodysplasia verruciformis. J Clin Immunol. 2019;39(1):11–4.PubMed Sharafian S, Ziaee V, Shahrooei M, Ahadi M, Parvaneh N. A Novel STK4 Mutation presenting with juvenile idiopathic arthritis and epidermodysplasia verruciformis. J Clin Immunol. 2019;39(1):11–4.PubMed
12.
Zurück zum Zitat Crequer A, Picard C, Patin E, D’Amico A, Abhyankar A, Munzer M, et al. Inherited MST1 deficiency underlies susceptibility to EV-HPV infections. PLoS One. 2012;7(8):e44010.PubMedPubMedCentral Crequer A, Picard C, Patin E, D’Amico A, Abhyankar A, Munzer M, et al. Inherited MST1 deficiency underlies susceptibility to EV-HPV infections. PLoS One. 2012;7(8):e44010.PubMedPubMedCentral
14.
Zurück zum Zitat Choi J, Oh S, Lee D, Oh HJ, Park JY, Lee SB, et al. Mst1-FoxO signaling protects Naive T lymphocytes from cellular oxidative stress in mice. PLoS One. 2009;4(11):e8011.PubMedPubMedCentral Choi J, Oh S, Lee D, Oh HJ, Park JY, Lee SB, et al. Mst1-FoxO signaling protects Naive T lymphocytes from cellular oxidative stress in mice. PLoS One. 2009;4(11):e8011.PubMedPubMedCentral
15.
Zurück zum Zitat Ueda Y, Kondo N, Kinashi T. MST1/2 balance immune activation and tolerance by orchestrating adhesion, transcription, and organelle dynamics in lymphocytes. Front Immunol. 2020;11:733.PubMedPubMedCentral Ueda Y, Kondo N, Kinashi T. MST1/2 balance immune activation and tolerance by orchestrating adhesion, transcription, and organelle dynamics in lymphocytes. Front Immunol. 2020;11:733.PubMedPubMedCentral
16.
Zurück zum Zitat Alsina L, Israelsson E, Altman MC, Dang KK, Ghandil P, Israel L, et al. A narrow repertoire of transcriptional modules responsive to pyogenic bacteria is impaired in patients carrying loss-of-function mutations in MYD88 or IRAK4. Nat Immunol. 2014;15(12):1134–42.PubMedPubMedCentral Alsina L, Israelsson E, Altman MC, Dang KK, Ghandil P, Israel L, et al. A narrow repertoire of transcriptional modules responsive to pyogenic bacteria is impaired in patients carrying loss-of-function mutations in MYD88 or IRAK4. Nat Immunol. 2014;15(12):1134–42.PubMedPubMedCentral
17.
Zurück zum Zitat Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25(8):1091–3.PubMedPubMedCentral Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25(8):1091–3.PubMedPubMedCentral
18.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentral Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentral
19.
Zurück zum Zitat Shaw AE, Hughes J, Gu Q, Behdenna A, Singer JB, Dennis T, et al. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017;15(12):e2004086.PubMedPubMedCentral Shaw AE, Hughes J, Gu Q, Behdenna A, Singer JB, Dennis T, et al. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017;15(12):e2004086.PubMedPubMedCentral
20.
Zurück zum Zitat Halacli SO, Ayvaz DC, Sun-Tan C, Erman B, Uz E, Yilmaz DY, et al. STK4 (MST1) deficiency in two siblings with autoimmune cytopenias: A novel mutation. Clin Immunol. 2015;161(2):316–23.PubMed Halacli SO, Ayvaz DC, Sun-Tan C, Erman B, Uz E, Yilmaz DY, et al. STK4 (MST1) deficiency in two siblings with autoimmune cytopenias: A novel mutation. Clin Immunol. 2015;161(2):316–23.PubMed
21.
Zurück zum Zitat Hong JJ, Amancha PK, Rogers K, Ansari AA, Villinger F. Re-evaluation of PD-1 expression by T cells as a marker for immune exhaustion during SIV infection. PLoS One. 2013;8(3):e60186.PubMedPubMedCentral Hong JJ, Amancha PK, Rogers K, Ansari AA, Villinger F. Re-evaluation of PD-1 expression by T cells as a marker for immune exhaustion during SIV infection. PLoS One. 2013;8(3):e60186.PubMedPubMedCentral
22.
Zurück zum Zitat Hofmeyer KA, Jeon H, Zang X. The PD-1/PD-L1 (B7–H1) pathway in chronic infection-induced cytotoxic T lymphocyte exhaustion. J Biomed Biotechnol. 2011;2011:451694.PubMedPubMedCentral Hofmeyer KA, Jeon H, Zang X. The PD-1/PD-L1 (B7–H1) pathway in chronic infection-induced cytotoxic T lymphocyte exhaustion. J Biomed Biotechnol. 2011;2011:451694.PubMedPubMedCentral
23.
Zurück zum Zitat Dong Y, Li X, Zhang L, Zhu Q, Chen C, Bao J, et al. CD4(+) T cell exhaustion revealed by high PD-1 and LAG-3 expression and the loss of helper T cell function in chronic hepatitis B. BMC Immunol. 2019;20(1):27.PubMedPubMedCentral Dong Y, Li X, Zhang L, Zhu Q, Chen C, Bao J, et al. CD4(+) T cell exhaustion revealed by high PD-1 and LAG-3 expression and the loss of helper T cell function in chronic hepatitis B. BMC Immunol. 2019;20(1):27.PubMedPubMedCentral
24.
Zurück zum Zitat Grywalska E, Smarz-Widelska I, Korona-Glowniak I, Mertowski S, Gosik K, Hymos A, et al. PD-1 and PD-L1 expression on circulating lymphocytes as a marker of Epstein-Barr virus reactivation-associated proliferative glomerulonephritis. Int J Mol Sci. 2020;21(21):8001. Grywalska E, Smarz-Widelska I, Korona-Glowniak I, Mertowski S, Gosik K, Hymos A, et al. PD-1 and PD-L1 expression on circulating lymphocytes as a marker of Epstein-Barr virus reactivation-associated proliferative glomerulonephritis. Int J Mol Sci. 2020;21(21):8001.
25.
26.
Zurück zum Zitat Poli A, Michel T, Theresine M, Andres E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology. 2009;126(4):458–65.PubMedPubMedCentral Poli A, Michel T, Theresine M, Andres E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology. 2009;126(4):458–65.PubMedPubMedCentral
27.
Zurück zum Zitat Gujer C, Murer A, Muller A, Vanoaica D, Sutter K, Jacque E, et al. Plasmacytoid dendritic cells respond to Epstein-Barr virus infection with a distinct type I interferon subtype profile. Blood Adv. 2019;3(7):1129–44.PubMedPubMedCentral Gujer C, Murer A, Muller A, Vanoaica D, Sutter K, Jacque E, et al. Plasmacytoid dendritic cells respond to Epstein-Barr virus infection with a distinct type I interferon subtype profile. Blood Adv. 2019;3(7):1129–44.PubMedPubMedCentral
28.
Zurück zum Zitat MollicaPoeta V, Massara M, Capucetti A, Bonecchi R. Chemokines and chemokine receptors: new targets for cancer immunotherapy. Front Immunol. 2019;10:379. MollicaPoeta V, Massara M, Capucetti A, Bonecchi R. Chemokines and chemokine receptors: new targets for cancer immunotherapy. Front Immunol. 2019;10:379.
29.
30.
Zurück zum Zitat Moran I, Avery DT, Payne K, Lenthall H, Davies EG, Burns S, et al. B cell-intrinsic requirement for STK4 in humoral immunity in mice and human subjects. J Allergy Clin Immunol. 2019;143(6):2302–5.PubMed Moran I, Avery DT, Payne K, Lenthall H, Davies EG, Burns S, et al. B cell-intrinsic requirement for STK4 in humoral immunity in mice and human subjects. J Allergy Clin Immunol. 2019;143(6):2302–5.PubMed
31.
Zurück zum Zitat Mohan D, Wansley DL, Sie BM, Noon MS, Baer AN, Laserson U, et al. Publisher correction: PhIP-Seq characterization of serum antibodies using oligonucleotide-encoded peptidomes. Nat Protoc. 2019;14(8):2596.PubMed Mohan D, Wansley DL, Sie BM, Noon MS, Baer AN, Laserson U, et al. Publisher correction: PhIP-Seq characterization of serum antibodies using oligonucleotide-encoded peptidomes. Nat Protoc. 2019;14(8):2596.PubMed
32.
Zurück zum Zitat Pollard AJ, Bijker EM. Publisher correction: a guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21(2):129.PubMedPubMedCentral Pollard AJ, Bijker EM. Publisher correction: a guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21(2):129.PubMedPubMedCentral
33.
Zurück zum Zitat Boisson-Dupuis S, Ramirez-Alejo N, Li Z, Patin E, Rao G, Kerner G, et al. Tuberculosis and impaired IL-23-dependent IFN-gamma immunity in humans homozygous for a common TYK2 missense variant. Sci Immunol. 2018;3(30):eaau8714. Boisson-Dupuis S, Ramirez-Alejo N, Li Z, Patin E, Rao G, Kerner G, et al. Tuberculosis and impaired IL-23-dependent IFN-gamma immunity in humans homozygous for a common TYK2 missense variant. Sci Immunol. 2018;3(30):eaau8714.
34.
Zurück zum Zitat Frederico B, May JS, Efstathiou S, Stevenson PG. BAFF receptor deficiency limits gammaherpesvirus infection. J Virol. 2014;88(8):3965–75.PubMedPubMedCentral Frederico B, May JS, Efstathiou S, Stevenson PG. BAFF receptor deficiency limits gammaherpesvirus infection. J Virol. 2014;88(8):3965–75.PubMedPubMedCentral
35.
Zurück zum Zitat Kuo SH, Yeh PY, Chen LT, Wu MS, Lin CW, Yeh KH, et al. Overexpression of B cell-activating factor of TNF family (BAFF) is associated with Helicobacter pylori-independent growth of gastric diffuse large B-cell lymphoma with histologic evidence of MALT lymphoma. Blood. 2008;112(7):2927–34.PubMed Kuo SH, Yeh PY, Chen LT, Wu MS, Lin CW, Yeh KH, et al. Overexpression of B cell-activating factor of TNF family (BAFF) is associated with Helicobacter pylori-independent growth of gastric diffuse large B-cell lymphoma with histologic evidence of MALT lymphoma. Blood. 2008;112(7):2927–34.PubMed
36.
Zurück zum Zitat Novak AJ, Grote DM, Stenson M, Ziesmer SC, Witzig TE, Habermann TM, et al. Expression of BLyS and its receptors in B-cell non-Hodgkin lymphoma: correlation with disease activity and patient outcome. Blood. 2004;104(8):2247–53.PubMed Novak AJ, Grote DM, Stenson M, Ziesmer SC, Witzig TE, Habermann TM, et al. Expression of BLyS and its receptors in B-cell non-Hodgkin lymphoma: correlation with disease activity and patient outcome. Blood. 2004;104(8):2247–53.PubMed
37.
Zurück zum Zitat Oki Y, Georgakis GV, Migone TS, Kwak LW, Younes A. Prognostic significance of serum B-lymphocyte stimulator in Hodgkin’s lymphoma. Haematologica. 2007;92(2):269–70.PubMed Oki Y, Georgakis GV, Migone TS, Kwak LW, Younes A. Prognostic significance of serum B-lymphocyte stimulator in Hodgkin’s lymphoma. Haematologica. 2007;92(2):269–70.PubMed
38.
Zurück zum Zitat Onda K, Iijima K, Katagiri YU, Okita H, Saito M, Shimizu T, et al. Differential effects of BAFF on B cell precursor acute lymphoblastic leukemia and Burkitt lymphoma. Int J Hematol. 2010;91(5):808–19.PubMed Onda K, Iijima K, Katagiri YU, Okita H, Saito M, Shimizu T, et al. Differential effects of BAFF on B cell precursor acute lymphoblastic leukemia and Burkitt lymphoma. Int J Hematol. 2010;91(5):808–19.PubMed
39.
Zurück zum Zitat Ma SD, Xu X, Jones R, Delecluse HJ, Zumwalde NA, Sharma A, et al. PD-1/CTLA-4 blockade inhibits Epstein-Barr virus-induced lymphoma growth in a cord blood humanized-mouse model. PLoS Pathog. 2016;12(5):e1005642.PubMedPubMedCentral Ma SD, Xu X, Jones R, Delecluse HJ, Zumwalde NA, Sharma A, et al. PD-1/CTLA-4 blockade inhibits Epstein-Barr virus-induced lymphoma growth in a cord blood humanized-mouse model. PLoS Pathog. 2016;12(5):e1005642.PubMedPubMedCentral
40.
Zurück zum Zitat Inthasot V, Bruyneel M, Muylle I, Ninane V. Severe pulmonary infections complicating nivolumab treatment for lung cancer: a report of two cases. Acta Clin Belg. 2020;75(4):308–10.PubMed Inthasot V, Bruyneel M, Muylle I, Ninane V. Severe pulmonary infections complicating nivolumab treatment for lung cancer: a report of two cases. Acta Clin Belg. 2020;75(4):308–10.PubMed
41.
Zurück zum Zitat Barber DL, Sakai S, Kudchadkar RR, Fling SP, Day TA, Vergara JA, et al. Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci Transl Med. 2019;11(475):eaat2702. Barber DL, Sakai S, Kudchadkar RR, Fling SP, Day TA, Vergara JA, et al. Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci Transl Med. 2019;11(475):eaat2702.
42.
Zurück zum Zitat Anastasopoulou A, Ziogas DC, Samarkos M, Kirkwood JM, Gogas H. Reactivation of tuberculosis in cancer patients following administration of immune checkpoint inhibitors: current evidence and clinical practice recommendations. J Immunother Cancer. 2019;7(1):239.PubMedPubMedCentral Anastasopoulou A, Ziogas DC, Samarkos M, Kirkwood JM, Gogas H. Reactivation of tuberculosis in cancer patients following administration of immune checkpoint inhibitors: current evidence and clinical practice recommendations. J Immunother Cancer. 2019;7(1):239.PubMedPubMedCentral
43.
Zurück zum Zitat Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60.PubMedPubMedCentral Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60.PubMedPubMedCentral
44.
Zurück zum Zitat DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491–8.PubMedPubMedCentral DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491–8.PubMedPubMedCentral
45.
Zurück zum Zitat Cingolani P, Platts A, le Wang L, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6(2):80–92. Cingolani P, Platts A, le Wang L, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6(2):80–92.
46.
Zurück zum Zitat Love JM, Prosser D, Love DR, Chintakindi KP, Dalal AB, Aggarwal S. A novel glycine decarboxylase gene mutation in an Indian family with nonketotic hyperglycinemia. J Child Neurol. 2014;29(1):122–7.PubMed Love JM, Prosser D, Love DR, Chintakindi KP, Dalal AB, Aggarwal S. A novel glycine decarboxylase gene mutation in an Indian family with nonketotic hyperglycinemia. J Child Neurol. 2014;29(1):122–7.PubMed
47.
Zurück zum Zitat de Jonge HJ, Fehrmann RS, de Bont ES, Hofstra RM, Gerbens F, Kamps WA, et al. Evidence based selection of housekeeping genes. PLoS One. 2007;2(9):e898.PubMedPubMedCentral de Jonge HJ, Fehrmann RS, de Bont ES, Hofstra RM, Gerbens F, Kamps WA, et al. Evidence based selection of housekeeping genes. PLoS One. 2007;2(9):e898.PubMedPubMedCentral
48.
Zurück zum Zitat Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.PubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.PubMed
49.
Zurück zum Zitat Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMed Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMed
50.
Zurück zum Zitat Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9.PubMed Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9.PubMed
51.
Zurück zum Zitat Zhang Y, Parmigiani G, Johnson WE. ComBat-seq: batch effect adjustment for RNA-seq count data. NAR Genom Bioinform. 2020;2(3):lqaa078.PubMedPubMedCentral Zhang Y, Parmigiani G, Johnson WE. ComBat-seq: batch effect adjustment for RNA-seq count data. NAR Genom Bioinform. 2020;2(3):lqaa078.PubMedPubMedCentral
52.
Zurück zum Zitat Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40.PubMed Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40.PubMed
54.
Zurück zum Zitat Luck K, Kim DK, Lambourne L, Spirohn K, Begg BE, Bian W, et al. A reference map of the human binary protein interactome. Nature. 2020;580(7803):402–8.PubMedPubMedCentral Luck K, Kim DK, Lambourne L, Spirohn K, Begg BE, Bian W, et al. A reference map of the human binary protein interactome. Nature. 2020;580(7803):402–8.PubMedPubMedCentral
55.
Zurück zum Zitat Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–13.PubMed Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–13.PubMed
56.
Zurück zum Zitat Kramer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523–30.PubMed Kramer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523–30.PubMed
57.
Zurück zum Zitat Xu GJ, Kula T, Xu Q, Li MZ, Vernon SD, Ndung’u T, et al. Viral immunology. Comprehensive serological profiling of human populations using a synthetic human virome. Science. 2015;348(6239):aaa0698.PubMedPubMedCentral Xu GJ, Kula T, Xu Q, Li MZ, Vernon SD, Ndung’u T, et al. Viral immunology. Comprehensive serological profiling of human populations using a synthetic human virome. Science. 2015;348(6239):aaa0698.PubMedPubMedCentral
58.
Zurück zum Zitat Khan T, Rahman M, Ali FA, Huang SSY, Ata M, Zhang Q, et al. Distinct antibody repertoires against endemic human coronaviruses in children and adults. JCI Insight. 2021;6(4):e144499. Khan T, Rahman M, Ali FA, Huang SSY, Ata M, Zhang Q, et al. Distinct antibody repertoires against endemic human coronaviruses in children and adults. JCI Insight. 2021;6(4):e144499.
59.
Zurück zum Zitat Mina MJ, Kula T, Leng Y, Li M, de Vries RD, Knip M, et al. Measles virus infection diminishes preexisting antibodies that offer protection from other pathogens. Science. 2019;366(6465):599–606.PubMedPubMedCentral Mina MJ, Kula T, Leng Y, Li M, de Vries RD, Knip M, et al. Measles virus infection diminishes preexisting antibodies that offer protection from other pathogens. Science. 2019;366(6465):599–606.PubMedPubMedCentral
60.
Zurück zum Zitat Hasan MR, Rahman M, Khan T, Saeed A, Sundararaju S, Flores A, et al. Virome-wide serological profiling reveals association of herpesviruses with obesity. Sci Rep. 2021;11(1):2562.PubMedPubMedCentral Hasan MR, Rahman M, Khan T, Saeed A, Sundararaju S, Flores A, et al. Virome-wide serological profiling reveals association of herpesviruses with obesity. Sci Rep. 2021;11(1):2562.PubMedPubMedCentral
Metadaten
Titel
A Novel STK4 Mutation Impairs T Cell Immunity Through Dysregulation of Cytokine-Induced Adhesion and Chemotaxis Genes
verfasst von
Andrea Guennoun
Salim Bougarn
Taushif Khan
Rafah Mackeh
Mahbuba Rahman
Fatima Al-Ali
Manar Ata
Waleed Aamer
Debra Prosser
Tanwir Habib
Evonne Chin-Smith
Khawla Al-Darwish
Qian Zhang
Alya Al-Shakaki
Amal Robay
Ronald G. Crystal
Khalid Fakhro
Amal Al-Naimi
Eman Al Maslamani
Amjad Tuffaha
Ibrahim Janahi
Mohammad Janahi
Donald R. Love
Mohammed Yousuf Karim
Bernice Lo
Amel Hassan
Mehdi Adeli
Nico Marr
Publikationsdatum
24.08.2021
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 8/2021
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-021-01115-2

Weitere Artikel der Ausgabe 8/2021

Journal of Clinical Immunology 8/2021 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.