Abstract
The most recent updated classification of inborn errors of immunity/primary immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee, was published in January 2020. Within days of completing this report, it was already out of date, evidenced by the frequent publication of genetic variants proposed to cause novel inborn errors of immunity. As the next formal report from the IUIS Expert Committee will not be published until 2022, we felt it important to provide the community with a brief update of recent contributions to the field of inborn errors of immunity. Herein, we highlight studies that have identified 26 additional monogenic gene defects that reach the threshold to represent novel causes of immune defects.
Keywords: Inborn errors of immunity, immune dysregulation, primary immunodeficiencies, autoinflammatory disorders
Introduction
Inborn errors of immunity (IEI) are generally considered to result from monogenic germline defects that manifest as increased susceptibility to severe and/or recurrent infectious diseases, autoimmune or autoinflammatory conditions, atopic manifestations, and hematopoietic or solid tissue malignancies [1]. Over the past decade, the discovery of new IEIs has been occurring at an impressive rate. Indeed, the 2011 biennial update published by the IUIS Committee update listed 191 IEIs; this number increased to 430 in the 2019 update [2, 3]. This near-exponential increase in gene discovery is being driven by the accessibility and affordability of next-generation sequencing, and the efficient application of these technologies to elucidate the molecular etiology of unsolved cases of IEIs that are likely to result from single-gene defects [4].
Over the last 12 months, we have witnessed the ongoing rapid identification, and occasionally detailed molecular, biochemical, and cellular characterization, of genetic variants that cause, or are at least associated with, human diseases impacting host defense or immune regulation. Here, we will summarize reports on variants detected in 26 genes which we consider represent novel IEI (Table 1). Many additional genetic variants have been reported recently. However, those listed here have been adjudicated by the IUIS Committee to meet the strict criteria to be considered disease-causing [57]. These criteria include:
The patient’s candidate genotype is monogenic and must not occur in individuals without the clinical phenotype;
Experimental studies must indicate the genetic variant impairs, destroys, or alters expression or function of the gene product;
The causal relationship between the candidate genotype and the clinical phenotype must be confirmed via a relevant cellular phenotype, including—where possible—rescue of a functional defect by reconstitution with the wild-type gene, or via a relevant animal phenotype [57].
Table 1.
Novel gene defects underlying IEI—2020 | ||||||||
Genetic defect | Inheritance/mechanisms | T/NK cells | B cells | Ig levels | Clinical features, cellular defects, and evidence of variant pathogenicity | Table (for classication)[2, 3] | Refs | |
PAX1 (8 patients, 3 families; 2 papers with overlapping patients) | AR (LOF) |
• T−B+NK+ SCID • Severe T lymphopenia, low TRECs, |
• Normal | • ~ Normal IgM, low IgA, normal to ↑ IgE, |
• Omenn’s-like syndrome (erythroderma, lymphocytosis, eosinophilia, ↓ proliferation to PHA, severe/recurrent infections), • No thymus, T cell deficiency not corrected by HSCT despite donor chimerism • Also: otofaciocervical syndrome type 2 (OTFCS2) Validation • Reporter assays established hypomorphic LOF of mutant alleles • Patients’ derived iPSCs differentiated to thymic epithelial cells demonstrated abnormalities in thymic epithelial progenitors, consistent with failure of HSCT to reconstitute T cells • Similar to mouse model |
Table 1 Subtable 1 | [5, 6] | |
SLP76 (1 patient) | AR (LOF) |
• T cells reduced, • ↓↓ CD4+, ↑ CD8+ T cell proportions • Low naïve, ↑ TCM CD4+ T cells; • CD8+ T cell mostly clonally expanded TEMRA • Low TRECs • NK normal numbers |
• Normal numbers but ↓ class-switched memory and transitional B cells, • ↑ naïve and immature B cells |
• High IgM, low IgA |
• Combined immunodeficiency, • Early-onset skin abscesses, rash recurrent infections, autoimmunity, • Neutrophil dysfunction, platelets dysfunction • ↓ T cell proliferation to PHA, anti-CD3/CD28 stimulation, partially restored by IL-2 • ↓ NK cell degranulation • ↓ Actin polymerization Validation • Generation of SLP76-deficient Jurkat T cells; expressing WT or mutant SLP76 allele, mimicking patient cellular phenotypes; • Partial rescue of some of the functional defects by expression of WT SLP76 |
Table 1 Subtable 1 | [7] | |
MCM10 (1 patient) | AR (LOF) |
• Mild lymphopenia • ↓ TCM, TEM cells • ↓↓↓ NK cells (↑ CD56bright, nearly absent CD56dim mature NK) |
• ↓ B cells | • slightly ↓ IgG, normal IgM/A |
• Severe (fatal) CMV infection • HLH-like (based on biomarkers, not clinical features • Phenocopies GINS1 and MCM4 deficiencies • ↓ NK function Validation • Detailed functional analysis of MCM10 variant in primary fibroblasts, transfectants, iPSC, and CRISPR/Cas9-gene-edited NK92 human NK cell line • assessed mutant allele on NK cell development in humanized mice reconstituted with CD34+ HSC generated from the patients or healthy donor IPSCs |
Table 2 Subtable 2 | [8] | |
IL6ST (gp130; 12 patients, 8 families) | AD (DN) |
• Normal T cell numbers • ↑ naïve CD4 and CD8 T cell proportions • ↓ TCM CD4+ and CD8+ T cells and TEM CD8 T cells; • ↓ MAIT, Tfh cells, • ↑ Th2, • Low to normal NK cell counts |
Normal numbers of B cells, low memory B cell proportions |
• Normal/low IgG, A, • Normal IgM, • Hyper IgE. • vaccine IgG normal |
• HIES – STAT3-like; • Dermatitis/eczema, eosinophilia, recurrent skin infections, pneumonia, bronchiectasis, pneumatoceles with severe secondary pulmonary aspergillosis, connective tissue defects (scoliosis, face, joints, fractures, palate, tooth retention) • Phenocopies aspects of IL6R and IL11R deficiencies (due to unresponsiveness to these cytokines) Validation • LOF and DN alleles shown by overexpression in GP130 deficient HEK293T cells; • Impaired GP130/STAT3 signaling (mostly downstream of IL-6) in patients’ fibroblasts and leukocytes |
Table 2 Subtable 5 | [9] | |
IL6ST (complete deficiency; 6 patients, 4 families) | AR (LOF) | ND (death in utero or in neonatal period occurred for most affected individuals) |
• Fatal Stuve-Wiedemann-like syndrome; skeletal dysplasia, lung dysfunction, renal abnormalities, thrombocytopenia, dermatitis, eczema • Defective acute phase response • Complete unresponsiveness to IL-6 family cytokines Validation • Complete LOE (for one allele) and LOF for two alleles tested by overexpression in GP130 deficient HEK293T to all cytokines tested of the IL-6 family. • Effects of variants well-characterized, including a partial rescue of patient amniocytes |
Table 2 Subtable 5 |
[10, 11] | |||
FNIP1 (6 patients; 5 families) | AR (LOF) | Mild T cell lymphocytosis | B cell lymphopenia (absent/low; BM block [few immature B cells]) | agamma/hypogammaglobulinemia |
• Early onset recurrent infections (sinopulmonary) • Bronchiectasis • Congenital heart defects (e.g., hypertrophic cardiomyopathy) • Variable neutropenia (severe or intermittent) Crohn disease (one patient) • Developmental delay • Increased AMPK activity Validation |
Table 3 Subtable 1 |
[14, 15] | |
PIK3CG (2 patients; 2 families) | AR (LOF) |
• Normal CD4, • ↓ Treg, ↓ CD8 |
• Normal but ↓ memory B cells |
• Hypogamma • Intact vaccine responses |
• Cytopenia/lymphopenia, eosinophilia, lymphadenopathy, splenomegaly, • recurrent infections • HLH-like; ↑ inflammatory markers Validation • ↓ T cell proliferation, activation in vitro • Cellular defects recapitulated in PIK3CG targeted Jurkat T cell line and Pik3cg ko mice |
Table 3 Subtable 2 |
[16, 17] | |
CTNNBL1 (1 patient) | AR (LOF) | ↓ T cells |
• Reduced memory B cells; • Impaired CSR, SHM |
• Progressive severe hypogamma |
CVID, autoimmune cytopenias, hypogamma, recurrent infections, hyperplastic GC’s; (mutation reduces binding of CTNNBL1 to AID, resulting in less nuclear translocation of AID) Validation • detailed functional analysis of CTNNBL1 variant in EBV B lines, and engineered RAMOS cell lines; • mutant allele reduces the binding of CTNNBL1 to AID, with impaired nuclear translocation of AID; • defective SHM rescued in mutant Ramos cells by WT CTNNBL1 |
Table 3 Subtable 3 |
[18] | |
TNFSF13 (APRIL; 1 patient) | AR (LOF) | • Normal T/NK cells |
Normal total B cell counts, ↑ IgM+ marginal zone /↓ sw memory B cells, ↓ blood plasmablasts |
• Hypogamma |
CVID, chronic but mild infections Validation • LOE and LOF allele, • Functional analysis of the variant in PBMCs and overexpression; • Impaired function of iPSC-moDC in promoting B cell differentiation could be rescued with exogenous APRIL |
Table 3 Subtable 3 | [19] | |
SOCS1 (15 patients; 10 families) | AD (by haploinsufficiency) | • ↓CD4, CD8 T cells |
• Predom naïve B cells • ↓ sw memory, ↑ CD21lo B |
• ↓ IgM, G, A but protective specific Abs; • ANA’s, autoAbs • ↑ serum BAFF |
• Recurrent bacterial infections, • Severe multisystemic autoimmunity (flared in context of infection-induced inflammation), ITP, AIHA, SLE, GN, hepatosplenomegaly, psoriasis, arthritis, thyroiditis, hepatitis • Evan’s syndrome • 1 patient developed COVID19/MIS-C • neutropenia, lymphopenia • incomplete penetrance Validation • ↑ pSTAT1, ↑ type I/II IFN signature • Reporter assays confirmed impaired inhibition of mutant SOCS • Jakinib effective in vitro and in vivo. |
Table 4 Subtable 4 | [20–22] | |
TET2 (3 patients, 2 families) | AR (LOF) |
• ↑ DN T cells • ↓ Th1, Th17, Tfh cells |
• ↓ Memory B cells • Impaired B cell diff in vitro to plasma cells |
• Variable (hyper−/ hypogamma, ↓ response to pneumococcal vaccine |
• ALPS-like (↑ sCD25, sFasL, IL10) • Recurrent viral infections, lymphadenopathy, hepatosplenomegaly, autoimmunity (cytopenias, B cell lymphoma (EBV+ HL-like) • Failure to thrive, developmental delay • EBV viremia • DNA hypermethylation • Defective FAS-mediated apoptosis |
Table 4 Subtable 7 | [23] | |
CEBPE (3 patients, 1 family) | AR (GOF) |
• Mild lymphopenia •↓ naive, ↑ TEMRA cells |
ND | ND |
• Recurrent abdominal pain, aseptic fever, systemic inflammation; abscesses, ulceration, infections; mild bleeding diathesis • Autoinflammasome activation/↑ IFN gene expression (autoinflammation, immunodeficiency, neutrophil dysfunction) Validation • homozygous CEBPE variant affecting DNA binding domain, associated with autoinflammation/immunodeficiency • showed altered chromatin occupancy of mutant CEBPE, and transcriptional changes, in patient cells targeting inflammasome and IFN-related genes |
Table 5 Subtable 2 | [24] | |
TBX21 (T-bet; 1 patient) | AR (LOF) |
• Normal %’s total, CD4 and CD8 T cells, naïve and memory subsets, low frequencies of NK cells • ↓ CXCR3+ CCR6− Th1 cells, CXCR3+ Tfh and CXCR3+ Treg CD4+T cells • ↑ immature NK • ↓ iNKT, MAIT, Vδ2+ γδ T cells |
• Normal | • Normal |
• MSMD • ↓IFN-γ and TNF-α production by T cell subsets (γδ T cells, MAIT cells, iNKT cells, NK cells,, Vδ2+γδ, Vδ1+γδ, and CD4+ T cells Validation • Biochemical and molecular analysis established the impact of this variant on Tbet function; • Impaired production of IFNγ, TNFα by TBX21-mutant naïve CD4+ T cells under Th1 polarizing cultured in vitro • Tbet-dependent functions restored in patient cells and cell lines by WT TBX21 |
Table 6 Subtable 1 | [25] | |
IFNG (2 patients [cousins]) | AR (LOF) |
• Normal frequencies of T and NK cells; • ↑ proportions of naive CD4+ and CD8+ T cells; • ↓ frequency of invariant iNKT |
• Normal B cell frequencies; • ↓ memory B cells (↓ IgA+ / IgM+, ↑ IgG+ memory B cells) |
• Normal |
MSMD/BCG-osis • No IFN-γ producing cells Validation • LOE and LOF allele • Biochemical and molecular analysis established impact of this variant on IFN-γ production from patients’ cells • No IFN-γ production by T and NK cells; • Impaired IFN-γ production by patient-derived Herpesvirus saimiri–immortalized T cells restored by introduction of WT IFNG |
Table 6 Subtable 1 | [26] | |
NOS2 (1 patient) | AR |
• ↓ CD4+ T cells; • ↓ NK cells (mostly all immature cells) • normal CD8+ T |
↓ B cells | ND (specific Ab levels normal) |
• Severe susceptibility to CMV-induced disease; fatal • Pneumocystis pneumonia secondary to CMV • Apparent intact responses to infection with other herpes viruses (EBV, VZV, HSV) Validation: • Confirmed functional defect in transfected cells; truncated NOS2 failed to induce nitrous oxide • Recapitulates susceptibility of Nos2 deficient mice to murine CMV infection [27] (these mice are also susceptible to numerous other pathogens) |
Table 6 Subtable 3 | [28] | |
SNORA31 (5 patients, unrelated) | AD | • Normal | • Normal | • Seropositive for IgG against many viruses |
• Forebrain herpes simplex virus-1 (HSV-1) encephalitis Validation: susceptibility of human pluripotent stem cell (hPSC)–derived cortical neurons from patients or hPSC-derived neurons from healthy donors but engineered to express variant SNORA31 to HSV1 infection, corrected by exogenous IFN-β • Incomplete penetrance |
Table 6 Subtable 4 | [29] | |
ATG4A (1 patient) | AD | Normal | Normal | Normal |
• Mollaret’s meningitis (recurrent lymphocytic meningitis) due to HSV2 • History of multiple episodes of meningitis; HSV2+ Validation • Impaired HSV2-induced autophagy → increased viral replication and apoptosis of patient fibroblasts • These defects were rescued by introduction of WT ATG4 or LC3B2 into patient fibroblasts |
Table 6 Subtable 4 | [30] | |
MAP1LC3B2 (1 patient) | ||||||||
MAPK8 (3 patients, 1 family) | AD (haploinsufficiency) |
• Normal total T cells, CD4+ and CD8+ T subsets, NK cells • ↓ Th17 cells |
• Normal total B cells and subsets | • Normal |
• Chronic mucocutaneous candidiasis (CMC) • Connective tissue disorder (similar to Ehlers-Danlos syndrome) • ↓ Th17 cells ex vivo, in vitro • ↓ Responses of fibroblasts to IL-17A, IL-17F • ↓ c-Jun/ATF-2-dependant TGF β signaling Validation • MAKP8 variant LOE in HEK293 T, heterozygous patients’ cells (↓ Th17 cells ex vivo, in vitro; ↓ fibroblast responses to IL-17A, IL-17F; ↓ TGFβ signaling) • Defective responses of fibroblasts restored by WT MAPK8 |
Table 6 Subtable 6 |
[31] | |
LSM11 (2 siblings, 1 family) | AR (LOF) | Not reported | • Aicardi-Goutieres syndrome (type 1 IFN-opathy) | Table 7 Subtable 1 | [32] | |||
RNU7–1 (16 patients, 11 kindreds) | ||||||||
CDC42 (15 patients; numerous kindreds reported in 7 papers) | AD | • Normal/decreased T cell numbers, • normal %CD4/CD8 but skewed differentiation | • Normal/B-lymphopenia | • Variable (↑↓) IgM, G, A, E |
• Neonatal onset: pancytopenia, fever, rash, hepatosplenomegaly, systemic inflammation, myelofibrosis/proliferation, HLH, multisystemic inflammatory disease. • ↑ serum levels of IL1, IL18, IFN-γ, ferritin, sCD25, CRP etc. • Recurrent GIT/RT infection; • Neurodevelopmental delay, FTT • Mutation affects actin function; • Treated with Anakinra/ IFN-γ mAb ↓NK function (cytox), |
Table 7 Subtable 1 |
[33–39] | |
STAT2 (GOF, 3 patients; all deceased; 2 additional deceased sibs but not genotyped; 2 unrelated families) | AR (GOF) | • Low frequency of NK, ↑ frequency of T cells (esp naïve), normal NK degranulation |
• Total B cell frequencies within age-matched controls’ ranges • slight ↑ transitional and naïve B cells %‘s |
• Low/normal |
• Severe fatal early-onset autoinflammation (skin ulceration, fever, seizures, intracranial calcification, multiorgan dysfunction, abnormal neurodevelopment; phenocopy of USP18 deficiency) • ↑ serum IFN-α, IL6, TNFα • IFN-opathy gene signature (impaired regulation of late cellular responses to type 1 IFN), Validation • Study of the mutant STAT2 alleles in STAT2 deficient human cell line, and patient’s immortalized fibroblasts • Patient cells hyper-sensitive to IFN-α → prolonged JAK/STAT signaling/transcriptional activation • Biochemical confirmation that mutant allele is GOF in homozygous, but not heterozygous, combination • Impaired interaction of GOF STAT2 protein with USP18, a negative regulator of type 1 IFN responses |
Table 7 Subtable 1 |
[40, 41] | |
RIPK1 (12 patients; 5 families, 2 papers) | AD |
• Normal T and NK cell numbers • Low/normal CD4+ T cells • Normal/hi CD8+ T cells • ↑ DN T cells |
• Normal B cells | ND |
• Autoinflamm disorder: regular/prolonged fevers, lymphadenopathy, spleno/hepatomegaly, ulcers, arthralgia, GI features, • ↑ inflamm markers, ↑ pro-inflamm cytokines/gene signature; • Responsive to Tocilizumab (not IL1/TNF blockade) |
Table 7 Subtable 2 |
[42, 43] | |
NCKAP1L (9 patients; 7 families, 3 papers) |
AR (LOF) |
• Normal T cell numbers • ↑TCM, exhausted cells; • Possibly immature NK cells but intact function |
• Normal B cells and naïve/memory subsets • ↑ CD21lo cells |
• Normal/ ↑ Ig levels • autoAbs |
• Recurrent URTI, skin rashes/abscesses, ulcers, • Anti dsDNA Abs, SLE-like, lymphadenopathy, fever, HLH-like • FTT • Immunodeficiency coupled with atopy, lymphoproliferation, hyperinflammation ,and cytokine overproduction (↑ Th1) • ↓ T cell proliferation, cytoskeletal defects; |
Table 7 Subtable 3 |
[44–46] | |
UBA1 (25 patients) | XL (somatic LOF mutations) |
• ↓ Peripheral lymphocyte counts • Loss of immature B cells, nonclassical and intermediate monocyte populations |
• Late adulthood onset treatment-refractory inflammatory syndrome • Fevers, cytopenias, dysplastic bone marrow, • Neutrophilic cutaneous and pulmonary inflammation, chondritis and vasculitis • Most patients have an inflammatory syndrome (relapsing polychondritis, Sweet’s syndrome, polyarteritis nodosa, giant-cell arteritis) or a hematologic condition (MDS, multiple myeloma) • Often fatal Dysregulated proinflammatory neutrophil activation, high inflammatory markers in patients’ serum Validation: • Overexpression of mutant allele favored the production of a catalytically deficient UBA1 • Defective ubiquitylation of patients’ mutant monocytes • CRISPR-Cas9 Uba1–deficient zebrafish model recapitulates the phenotype of systemic inflammation |
Table 7 | [47] | |||
Novel phenocopies of inborn errors of immunity | ||||||||
Disease | Mechanisms of disease pathogenesis | Associated/clinical features | Table | Refs | ||||
Severe COVID-19 | • High levels of neutralizing autoAbs against type 1 IFNs (primarily IFNα, IFNω) | • Severe, life-threatening infection with SARS-CoV-2 | Table 10 | [48] |
Abbreviations: AR, autosomal recessive; AD, autosomal dominant; AID, activation-induced cytidine deaminase; CSR, class switch recombination; SHM, somatic hypermutation; MDS, myelodysplastic syndrome; LOE, loss of expression; LOF, loss of function; GOF, gain of function; DN, dominant negative; MSMD, Mendelian susceptibility to mycobacterial disease; HLH, hemophagocytic lymphohistiocytosis; FTT, failure to thrive; hPSC, human pluripotent stem cells; iPSC, induced pluripotent stem cells; CMV, cytomegalovirus
• Not all Tables in the 2020 classifications [2, 3] are listed in the “Table” column because not all Tables are represented by the new variants detailed above
• Mutations in PAX1 previously reported to cause OTFCS2 (but CID/SCID not reported) [49]
• Variants in IL6ST have been previously listed to cause an IEI due to recessive partial LOF alleles [50]
• Dominant-negative mutations in IL6ST all target the intracellular domain of gp130, truncating the STAT3 binding sites as well as recycling motif, leading to sustained expression of a dead receptor (as opposed to recessive alleles, when detected in heterozygous carriers are benign)
• Fnip1 ko mice also previously reported [12, 13]; humans very similar
• Variants in CEBPE have been previously listed to cause an IEI due to recessive LOF alleles [51]
• Mutations in CDC42 previously identified in individuals with neurodevelopmental delay (Takenouchi-Kosaki syndrome) [52]; in these patients with autoinflammation and CDC42 mutations no such features were noted, except mild facial dysmorphism in some [33–39]
.• Variants in STAT2 have been previously listed to cause an IEI due to recessive LOF alleles [53, 54]
• The same amino acid was found to be affected (R148Q/W) for both families affected by STAT2 GOF [53, 54]
• Variants in RIPK1 have been previously listed to cause an IEI due to recessive LOF alleles [55, 56]; the heterozygous dominant mutations in RIPK1 reported here all affect the D324 amino acid residue that is important for cleavage
We also considered (i) the numbers of individuals affected by the novel variants, (ii) sufficient justification for excluding alternative candidate gene variants identified in single cases especially in situations of consanguinity with recessive disease, (iii) the depth of the clinical descriptions of affected individuals, and (iv) the level of immune and mechanistic characterization.
Novel Causes of Inborn Errors of Immunity
Currently, inborn errors of immunity are listed in 10 tables: Immunodeficiencies affecting cellular and humoral immunity (Table I), Combined immunodeficiencies (CID) with syndromic features (Table II), Predominantly antibody deficiencies (Table III), Diseases of immune dysregulation (Table IV), Congenital defects of phagocytes (Table V), Defects in intrinsic and innate immunity (Table VI), Autoinflammatory diseases (Table VII), Complement deficiencies (Table VIII), Bone Marrow failure (Table IX), and Phenocopies of inborn errors of immunity (Table X). Several of these tables are further partitioned into various subtables (e.g., Table I is split into Subtable 1 [T−B+ Severe Combined Immune Deficiency (SCID)], Subtable 2 [T−B− SCID] and Subtable 3 [CID, generally less profound than SCID]) [2, 3].
Recently-reported gene defects have been found for most categories of inborn errors of immunity, including novel causes of:
Predominantly antibody deficiencies (FNIP1 [14, 15], PIK3CG [16, 17], CTNNBL1 [18], TNFSF13 [19]);
Autoinflammatory diseases (SOCS1 [20–22], TET2 [23], CEBPE [24], CDC42 [33–39], LSM11, RNU7–1 [32], STAT2 [40, 41], RIPK1 [42, 43], NCKAP1L [44–46]), UBA1 (somatic mutations) [47]; and
Susceptibility to infection with specific pathogens (MAPK8 [31]; TBX21 [25], IFNG [26], NOS2 [28], SNORA31 [29], ATG4A, MAP1LC3B2 [30]) (Table 1).
Notably, several of these genes are already included in previous IUIS updates, namely IL6ST, STAT2, CEBPE, and RIPK1 [2, 3]. However, they are listed here because the variant identified is pathogenic via a distinct mechanism and/or different mode of inheritance; i.e., autosomal recessive (AR) vs autosomal dominant for IL6ST [9] or RIPK1 [42, 43], partial deficiency vs complete deficiency for IL6ST [10, 11], or AR loss of function vs AR gain of function for CEBPE [24] or STAT2 [40, 41]. Furthermore, the GOF variants reported for CEBPE appear to represent the first described germline neomorphic mutation in inborn errors of immunity where the variant allele has completely novel functions not seen for the wild type gene [24]. Thus, these findings underscore the importance of appropriately interpreting genetic variants identified by next-generation sequencing, not discarding variants of unknown significance simply because they do not match the expected zygosity or clinical phenotype of previously reported studies, and to rigorously validate the impact of novel variants on the function of the encoded protein.
Joining the Dots with Discoveries of Novel Inborn Errors of Immunity
Many known inborn errors of immunity impact a defined signaling pathway such that mutations in components of these same pathways can represent clinical phenocopies of diseases causes by distinct genetic variants (genetic heterogeneity). In other words, physiological homogeneity can be identified for many genotypes underlying a given phenotype. Classic examples of this are Mendelian susceptibility to mycobacterial disease (MSMD), which results from impaired IFNγ-mediated immunity following exposure to mycobacterial species [58], and herpes simplex virus encephalitis (HSE) resulting from impaired TLR3-mediated anti-HSV1 immunity [59, 60]. Thus, variants in genes affecting the production of IFNγ (e.g., IL12RB1, IL12RB2, IL23R, TYK2, IKBKG, SPPL2A, IRF8) or cellular responses to IFNγ (e.g., IFNGR1, IFNGR2, STAT1, JAK1) result in MSMD in otherwise healthy individuals [58]. Similarly, inactivating mutations in signaling components of the TLR3 signaling pathway (TLR3, UNC93B, TRIF, TRAF3, TBK1, IRF3) underlie HSE due to impaired type 1 IFN-mediated central nervous system (CNS) intrinsic immunity against HSV1 [59, 60].
Recent discoveries have further linked common clinical phenotypes with unique genotypes that converge in a shared pathway. Thus, the non-redundant role of IFNγ-mediated immunity in host defense against mycobacterial infection [58] has been definitively established by the identification of individuals with inactivating bi-allelic mutations in not only IFNG itself [26] but also TBX21 [25], the transcription factor that regulates expression and production of IFNγ.
Interestingly, variants in the small nucleolar RNA SNORA31 predispose affected individuals to HSE. Mechanistically, patient’s iPSC-derived cortical neurons were found to be highly susceptible to HSV-1 infection in vitro, and this could be restored by exogenous IFNβ [29, 60]. However, responses of these cells to TLR3 and IFNβ, but not HSV1, are intact, revealing that SNORA31 functions to regulate cell-intrinsic immunity to HSV-1 by a mechanism independent of TLR3 signaling [29, 60]. The discovery of individuals with SNORA31 variants will facilitate further understanding of CNS-intrinsic host defense.
The discoveries of individuals with complete gp130-deficiency due to null/nonsense bi-allelic mutations of IL6ST [11], or pathogenic dominant-negative heterozygous variants of IL6ST [9], and a phenotype of eczema, hyper-IgE, and eosinophilia, likely explain these features of autosomal dominant hyper-IgE syndrome due to STAT3 negative dominance [61] and further highlight the role of IL-6 signaling in restraining atopic and allergic responses. Furthermore, the lack of mucocutaneous candidiasis in patients with impaired signaling via receptors for IL-6 (IL6R, IL6ST mutations [9, 11, 50, 62, 63]; anti-IL-6 autoantibodies [64]), IL-23 (biallelic IL23R variants) [65] or IL-21 (biallelic IL21 or IL21R variants) [66] argues that individually these cytokines are not required for the STAT3-mediated generation of human Th17 cells and host defense against fungal infections. Rather, the combinatorial defect of impaired STAT3 signaling downstream of these receptors explains chronic mucocutaneous candidiasis in an individual with dominant-negative STAT3 mutations. These findings again reveal the capacity for inborn errors of immunity to provide convincing evidence for basic immunological concepts. Indeed, this is further exemplified by the discovery that variants of ATG4A or MAP1LC3B2 cause recurrent HSV2 infection of the CNS, thereby establishing hitherto non-redundant functions of the autophagy pathway in non-hematopoietic cell-mediated intrinsic anti-viral immune responses [30].
SARS-CoV2 and Inborn Errors of Immunity
The COVID19 pandemic of 2020 has clearly changed the world in many ways. It has also yielded opportunities to understand host requirements for immunity against SARS-CoV2 infection. A recent study of ~ 650 individuals who developed severe COVID-19 found that ~ 3.5% of patients harbored germline loss-of-function variants in genes previously found to be important for host defense against influenza or other viral infections (e.g., bi-allelic loss of function mutations of IRF7 or IFNAR1, heterozygous mutations in TLR3, TICAM1, TBK1, or IRF3) [67] due to the key role of these genes in the type 1 IFN signaling pathway [59, 68]. An accompanying study found that, strikingly, ~ 10% of patients with severe COVID-19 have high levels of neutralizing autoantibodies (autoAbs) against type 1 IFNs in their serum [48]. The impact of these autoAbs was evidenced by the inability to detect IFN in serum from these patients, and their capacity to prevent anti-viral immune responses in vitro [48] (Table 1). These studies defined a crucial and non-redundant role for type 1 IFNs in immune control of SARS-CoV2 infection, and thus prevention of severe COVID-19. Furthermore, they also established that autoAbs against type 1 IFN phenocopy an inborn error of immunity, as previously determined for autoAbs against IFNγ and susceptibility to mycobacterial disease, anti-Th17 cytokine (IL-17A, IL-17F, IL-22) autoAbs in individuals with chronic mucocutaneous candidiasis, or pyogenic infections due to anti-IL-6 autoAbs [64, 69].
Conclusions
Discoveries over the past 12 months in the field of inborn errors of immunity have further identified non-redundant functions of key genes in human immune cell development, host defense, and immune regulation. In some cases, these functions go well beyond what may have been expected or anticipated based on animal models (e.g., TBX21 [25]). They have also already highlighted the heterogenous phenotypes that can result from variants in the same gene (e.g., CDC42 [33–39, 52]), indicated that significant diseases can arise from mono-allelic or bi-allelic loss of function (IL6ST [9], RIPK1 [42, 43]) or bi-allelic loss- or gain-of-function (CEBPE [24], STAT2 [40, 41]) variants in the same gene, or from autoAb phenocopies of monogenic lesions (e.g., COVID19 and anti-IFN Abs) [48], and identified novel somatic mutations as pathogenic causes of immune disorders (UBA1) [47]. Importantly, they have also provided opportunities for therapeutic interventions, such as JAK inhibitors to treat STAT2 gain of function [40, 41] or SOCS1 deficiency [22], IFNγ to treat mycobacterial disease [25, 26], or early IFN-β or IFN-α2a treatment of SARS-CoV2 infection in COVID-19 patients with autoantibodies against IFN-α or IFN-ω [67] or impaired type 1 IFN responses [70]. This snapshot of genetic discoveries underpinning human immune disorders further highlights the critical contributions of inborn errors of immunity to our broader understanding of basic, translational, and clinical immunology.
Authors Contribution
SGT wrote the drafts of the manuscript, prepared the table, and revised the original manuscripts for resubmission. All co-authors contributed to and edited drafts of the original and revised manuscripts and table, and approved the final submitted version.
Funding
The members of the Inborn Errors of Immunity committee would like to thank the International Union of Immunological Societies for funding, as well as CSL Behring, Baxalta, and Shire/Takeda for providing educational grants to enable us to compile this interim update to novel causes of immune diseases. This work was also supported in part by the Intramural Research Program of the NIAID, NIH. SGT is supported by an Investigator Grant (Level 3) awarded by the National Health and Medical Research Council of Australia. IM is a senior clinical investigator of FWO Vlaanderen (EBD-D8974-FKM).
Data Availability
Not applicable.
Declarations
Ethics Approval
This work is a review of recently-reported genetic variants that represent novel inborn errors of immunity. No human research studies were performed in order to produce this review. Thus, no approvals by appropriate institutional review boards or human research ethics committees were required to undertake the preparation of this report.
Consent to Participate
Not applicable as this is a review of recently-reported genetic variants.
Consent for Publication
The authors consent to publish the content of this review. However, as noted above, as this is a review of recently-reported genetic variants that represent novel inborn errors of immunity, we did not require consent to publish from participants.
Conflict of Interest
The authors declare that they have no conflict of interest.
Footnotes
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Notarangelo LD, Bacchetta R, Casanova JL, Su HC. Human inborn errors of immunity: an expanding universe. Sci Immunol. 2020;5(49):eabb1662. doi: 10.1126/sciimmunol.abb1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bousfiha A, Jeddane L, Picard C, Al-Herz W, Ailal F, Chatila T, et al. Human inborn errors of immunity: 2019 update of the IUIS phenotypical classification. J Clin Immunol. 2020;40(1):66–81. doi: 10.1007/s10875-020-00758-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tangye SG, Al-Herz W, Bousfiha A, Chatila T, Cunningham-Rundles C, Etzioni A, et al. 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):24–64. doi: 10.1007/s10875-019-00737-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Meyts I, Bosch B, Bolze A, Boisson B, Itan Y, Belkadi A, Pedergnana V, Moens L, Picard C, Cobat A, Bossuyt X, Abel L, Casanova JL. Exome and genome sequencing for inborn errors of immunity. J Allergy Clin Immunol. 2016;138(4):957–969. doi: 10.1016/j.jaci.2016.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Yamazaki Y, Urrutia R, Franco LM, Giliani S, Zhang K, Alazami AM, et al. PAX1 is essential for development and function of the human thymus. Sci Immunol. 2020;5(44):eaax1036. doi: 10.1126/sciimmunol.aax1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Paganini I, Sestini R, Capone GL, Putignano AL, Contini E, Giotti I, Gensini F, Marozza A, Barilaro A, Porfirio B, Papi L. A novel PAX1 null homozygous mutation in autosomal recessive otofaciocervical syndrome associated with severe combined immunodeficiency. Clin Genet. 2017;92(6):664–668. doi: 10.1111/cge.13085. [DOI] [PubMed] [Google Scholar]
- 7.Lev A, Lee YN, Sun G, Hallumi E, Simon AJ, Zrihen KS, et al. Inherited SLP76 deficiency in humans causes severe combined immunodeficiency, neutrophil and platelet defects. J Exp Med. 2021;218(3):e20201062. doi: 10.1084/jem.20201062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mace EM, Paust S, Conte MI, Baxley RM, Schmit MM, Patil SL, Guilz NC, Mukherjee M, Pezzi AE, Chmielowiec J, Tatineni S, Chinn IK, Akdemir ZC, Jhangiani SN, Muzny DM, Stray-Pedersen A, Bradley RE, Moody M, Connor PP, Heaps AG, Steward C, Banerjee PP, Gibbs RA, Borowiak M, Lupski JR, Jolles S, Bielinsky AK, Orange JS. Human NK cell deficiency as a result of biallelic mutations in MCM10. J Clin Invest. 2020;130:5272–5286. doi: 10.1172/JCI134966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Beziat V, Tavernier SJ, Chen YH, Ma CS, Materna M, Laurence A, et al. Dominant-negative mutations in human IL6ST underlie hyper-IgE syndrome. J Exp Med. 2020;217(6):e20191804. doi: 10.1084/jem.20191804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Monies D, Abouelhoda M, Assoum M, Moghrabi N, Rafiullah R, Almontashiri N, Alowain M, Alzaidan H, Alsayed M, Subhani S, Cupler E, Faden M, Alhashem A, Qari A, Chedrawi A, Aldhalaan H, Kurdi W, Khan S, Rahbeeni Z, Alotaibi M, Goljan E, Elbardisy H, ElKalioby M, Shah Z, Alruwaili H, Jaafar A, Albar R, Akilan A, Tayeb H, Tahir A, Fawzy M, Nasr M, Makki S, Alfaifi A, Akleh H, Yamani S, Bubshait D, Mahnashi M, Basha T, Alsagheir A, Abu Khaled M, Alsaleem K, Almugbel M, Badawi M, Bashiri F, Bohlega S, Sulaiman R, Tous E, Ahmed S, Algoufi T, al-Mousa H, Alaki E, Alhumaidi S, Alghamdi H, Alghamdi M, Sahly A, Nahrir S, al-Ahmari A, Alkuraya H, Almehaidib A, Abanemai M, Alsohaibaini F, Alsaud B, Arnaout R, Abdel-Salam GMH, Aldhekri H, AlKhater S, Alqadi K, Alsabban E, Alshareef T, Awartani K, Banjar H, Alsahan N, Abosoudah I, Alashwal A, Aldekhail W, Alhajjar S, al-Mayouf S, Alsemari A, Alshuaibi W, Altala S, Altalhi A, Baz S, Hamad M, Abalkhail T, Alenazi B, Alkaff A, Almohareb F, al Mutairi F, Alsaleh M, Alsonbul A, Alzelaye S, Bahzad S, Manee AB, Jarrad O, Meriki N, Albeirouti B, Alqasmi A, AlBalwi M, Makhseed N, Hassan S, Salih I, Salih MA, Shaheen M, Sermin S, Shahrukh S, Hashmi S, Shawli A, Tajuddin A, Tamim A, Alnahari A, Ghemlas I, Hussein M, Wali S, Murad H, Meyer BF, Alkuraya FS. Lessons learned from large-scale, first-tier clinical exome sequencing in a highly consanguineous population. Am J Hum Genet. 2019;104(6):1182–1201. doi: 10.1016/j.ajhg.2019.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen YH, Grigelioniene G, Newton PT, Gullander J, Elfving M, Hammarsjo A, et al. Absence of GP130 cytokine receptor signaling causes extended Stuve-Wiedemann syndrome. J Exp Med. 2020;217(3):e20191306. doi: 10.1084/jem.20191306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Park H, Staehling K, Tsang M, Appleby MW, Brunkow ME, Margineantu D, Hockenbery DM, Habib T, Liggitt HD, Carlson G, Iritani BM. Disruption of Fnip1 reveals a metabolic checkpoint controlling B lymphocyte development. Immunity. 2012;36(5):769–781. doi: 10.1016/j.immuni.2012.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Siggs OM, Stockenhuber A, Deobagkar-Lele M, Bull KR, Crockford TL, Kingston BL, Crawford G, Anzilotti C, Steeples V, Ghaffari S, Czibik G, Bellahcene M, Watkins H, Ashrafian H, Davies B, Woods A, Carling D, Yavari A, Beutler B, Cornall RJ. Mutation of Fnip1 is associated with B-cell deficiency, cardiomyopathy, and elevated AMPK activity. Proc Natl Acad Sci U S A. 2016;113(26):E3706–E3715. doi: 10.1073/pnas.1607592113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Niehues T, Ozgur TT, Bickes M, Waldmann R, Schoning J, Brasen J, et al. Mutations of the gene FNIP1 associated with a syndromic autosomal recessive immunodeficiency with cardiomyopathy and pre-excitation syndrome. Eur J Immunol. 2020;50(7):1078–1080. doi: 10.1002/eji.201948504. [DOI] [PubMed] [Google Scholar]
- 15.Saettini F, Poli C, Vengoechea J, Bonanomi S, Orellana JC, Fazio G, et al. Absent B cells, agammaglobulinemia, and hypertrophic cardiomyopathy in folliculin interacting protein 1 deficiency. Blood. 2021;137:493–9 10.1182/blood.2020006441. [DOI] [PMC free article] [PubMed]
- 16.Takeda AJ, Maher TJ, Zhang Y, Lanahan SM, Bucklin ML, Compton SR, Tyler PM, Comrie WA, Matsuda M, Olivier KN, Pittaluga S, McElwee JJ, Long Priel DA, Kuhns DB, Williams RL, Mustillo PJ, Wymann MP, Koneti Rao V, Lucas CL. Human PI3Kgamma deficiency and its microbiota-dependent mouse model reveal immunodeficiency and tissue immunopathology. Nat Commun. 2019;10(1):4364. doi: 10.1038/s41467-019-12311-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Thian M, Hoeger B, Kamnev A, Poyer F, Kostel Bal S, Caldera M, et al. Germline biallelic PIK3CG mutations in a multifaceted immunodeficiency with immune dysregulation. Haematologica. 2020;105:e488. doi: 10.3324/haematol.2019.231399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kuhny M, Forbes LR, Cakan E, Vega-Loza A, Kostiuk V, Dinesh RK, et al. Disease-associated CTNNBL1 mutation impairs somatic hypermutation by decreasing nuclear AID. J Clin Invest. 2020;130(8):4411-4422. doi: 10.1172/JCI131297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yeh TW, Okano T, Naruto T, Yamashita M, Okamura M, Tanita K, du L, Pan-Hammarström Q, Mitsuiki N, Okada S, Kanegane H, Imai K, Morio T. APRIL-dependent life-long plasmacyte maintenance and immunoglobulin production in humans. J Allergy Clin Immunol. 2020;146:1109–1120.e4. doi: 10.1016/j.jaci.2020.03.025. [DOI] [PubMed] [Google Scholar]
- 20.Lee PY, Platt CD, Weeks S, Grace RF, Maher G, Gauthier K, Devana S, Vitali S, Randolph AG, McDonald DR, Geha RS, Chou J. Immune dysregulation and multisystem inflammatory syndrome in children (MIS-C) in individuals with haploinsufficiency of SOCS1. J Allergy Clin Immunol. 2020;146(5):1194–1200.e1. doi: 10.1016/j.jaci.2020.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Thaventhiran JED, Lango Allen H, Burren OS, Rae W, Greene D, Staples E, et al. Whole-genome sequencing of a sporadic primary immunodeficiency cohort. Nature. 2020;583(7814):90–95. doi: 10.1038/s41586-020-2265-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hadjadj J, Castro CN, Tusseau M, Stolzenberg MC, Mazerolles F, Aladjidi N, Armstrong M, Ashrafian H, Cutcutache I, Ebetsberger-Dachs G, Elliott KS, Durieu I, Fabien N, Fusaro M, Heeg M, Schmitt Y, Bras M, Knight JC, Lega JC, Lesca G, Mathieu AL, Moreews M, Moreira B, Nosbaum A, Page M, Picard C, Ronan Leahy T, Rouvet I, Ryan E, Sanlaville D, Schwarz K, Skelton A, Viallard JF, Viel S, Villard M, Callebaut I, Picard C, Walzer T, Ehl S, Fischer A, Neven B, Belot A, Rieux-Laucat F. Early-onset autoimmunity associated with SOCS1 haploinsufficiency. Nat Commun. 2020;11(1):5341. doi: 10.1038/s41467-020-18925-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Stremenova Spegarova J, Lawless D, Mohamad SMB, Engelhardt KR, Doody G, Shrimpton J, Rensing-Ehl A, Ehl S, Rieux-Laucat F, Cargo C, Griffin H, Mikulasova A, Acres M, Morgan NV, Poulter JA, Sheridan EG, Chetcuti P, O'Riordan S, Anwar R, Carter CR, Przyborski S, Windebank K, Cant AJ, Lako M, Bacon CM, Savic S, Hambleton S. Germline TET2 loss of function causes childhood immunodeficiency and lymphoma. Blood. 2020;136(9):1055–1066. doi: 10.1182/blood.2020005844. [DOI] [PubMed] [Google Scholar]
- 24.Goos H, Fogarty CL, Sahu B, Plagnol V, Rajamaki K, Nurmi K, et al. Gain-of-function CEBPE mutation causes noncanonical autoinflammatory inflammasomopathy. J Allergy Clin Immunol. 2019;144(5):1364–1376. doi: 10.1016/j.jaci.2019.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yang R, Mele F, Worley L, Langlais D, Rosain J, Benhsaien I, Elarabi H, Croft CA, Doisne JM, Zhang P, Weisshaar M, Jarrossay D, Latorre D, Shen Y, Han J, Ogishi M, Gruber C, Markle J, al Ali F, Rahman M, Khan T, Seeleuthner Y, Kerner G, Husquin LT, Maclsaac JL, Jeljeli M, Errami A, Ailal F, Kobor MS, Oleaga-Quintas C, Roynard M, Bourgey M, el Baghdadi J, Boisson-Dupuis S, Puel A, Batteux F, Rozenberg F, Marr N, Pan-Hammarström Q, Bogunovic D, Quintana-Murci L, Carroll T, Ma CS, Abel L, Bousfiha A, di Santo JP, Glimcher LH, Gros P, Tangye SG, Sallusto F, Bustamante J, Casanova JL. Human T-bet governs innate and innate-like adaptive IFN-gamma immunity against mycobacteria. Cell. 2020;183(7):1826–1847.e31. doi: 10.1016/j.cell.2020.10.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kerner G, Rosain J, Guerin A, AlKhabaz A, Oleaga-Quintas C, Rapaport F, et al. Inherited human IFNgamma deficiency underlies mycobacterial disease. J Clin Invest. 2020;130(6):3158–3171. doi: 10.1172/JCI135460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Noda S, Tanaka K, Sawamura S, Sasaki M, Matsumoto T, Mikami K, Aiba Y, Hasegawa H, Kawabe N, Koga Y. Role of nitric oxide synthase type 2 in acute infection with murine cytomegalovirus. J Immunol. 2001;166(5):3533–3541. doi: 10.4049/jimmunol.166.5.3533. [DOI] [PubMed] [Google Scholar]
- 28.Drutman SB, Mansouri D, Mahdaviani SA, Neehus AL, Hum D, Bryk R, Hernandez N, Belkaya S, Rapaport F, Bigio B, Fisch R, Rahman M, Khan T, al Ali F, Marjani M, Mansouri N, Lorenzo-Diaz L, Emile JF, Marr N, Jouanguy E, Bustamante J, Abel L, Boisson-Dupuis S, Béziat V, Nathan C, Casanova JL. Fatal Cytomegalovirus infection in an adult with inherited NOS2 deficiency. N Engl J Med. 2020;382(5):437–445. doi: 10.1056/NEJMoa1910640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lafaille FG, Harschnitz O, Lee YS, Zhang P, Hasek ML, Kerner G, Itan Y, Ewaleifoh O, Rapaport F, Carlile TM, Carter-Timofte ME, Paquet D, Dobbs K, Zimmer B, Gao D, Rojas-Duran MF, Kwart D, Rattina V, Ciancanelli MJ, McAlpine JL, Lorenzo L, Boucherit S, Rozenberg F, Halwani R, Henry B, Amenzoui N, Alsum Z, Marques L, Church JA, al-Muhsen S, Tardieu M, Bousfiha AA, Paludan SR, Mogensen TH, Quintana-Murci L, Tessier-Lavigne M, Smith GA, Notarangelo LD, Studer L, Gilbert W, Abel L, Casanova JL, Zhang SY. Human SNORA31 variations impair cortical neuron-intrinsic immunity to HSV-1 and underlie herpes simplex encephalitis. Nat Med. 2019;25(12):1873–1884. doi: 10.1038/s41591-019-0672-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hait AS, Olagnier D, Sancho-Shimizu V, Skipper KA, Helleberg M, Larsen SM, et al. Defects in LC3B2 and ATG4A underlie HSV2 meningitis and reveal a critical role for autophagy in antiviral defense in humans. Sci Immunol. 2020;5(54):eabc2691. doi: 10.1126/sciimmunol.abc2691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li J, Ritelli M, Ma CS, Rao G, Habib T, Corvilain E, et al. Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-beta. Sci Immunol. 2019;4(41):eabc2691. doi: 10.1126/sciimmunol.aax7965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Uggenti C, Lepelley A, Depp M, Badrock AP, Rodero MP, El-Daher MT, et al. cGAS-mediated induction of type I interferon due to inborn errors of histone pre-mRNA processing. Nat Genet. 2020;52(12):1364–1372. doi: 10.1038/s41588-020-00737-3. [DOI] [PubMed] [Google Scholar]
- 33.Verboon JM, Mahmut D, Kim AR, Nakamura M, Abdulhay NJ, Nandakumar SK, Gupta N, Akie TE, Geddis AE, Manes B, Kapp ME, Hofmann I, Gabriel SB, Klein DE, Williams DA, Frangoul HA, Parkhurst SM, Crane GM, Cantor AB, Sankaran VG. Infantile myelofibrosis and myeloproliferation with CDC42 dysfunction. J Clin Immunol. 2020;40:554–566. doi: 10.1007/s10875-020-00778-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lam MT, Coppola S, Krumbach OHF, Prencipe G, Insalaco A, Cifaldi C, Brigida I, Zara E, Scala S, di Cesare S, Martinelli S, di Rocco M, Pascarella A, Niceta M, Pantaleoni F, Ciolfi A, Netter P, Carisey AF, Diehl M, Akbarzadeh M, Conti F, Merli P, Pastore A, Levi Mortera S, Camerini S, Farina L, Buchholzer M, Pannone L, Cao TN, Coban-Akdemir ZH, Jhangiani SN, Muzny DM, Gibbs RA, Basso-Ricci L, Chiriaco M, Dvorsky R, Putignani L, Carsetti R, Janning P, Stray-Pedersen A, Erichsen HC, Horne AC, Bryceson YT, Torralba-Raga L, Ramme K, Rosti V, Bracaglia C, Messia V, Palma P, Finocchi A, Locatelli F, Chinn IK, Lupski JR, Mace EM, Cancrini C, Aiuti A, Ahmadian MR, Orange JS, de Benedetti F, Tartaglia M. A novel disorder involving dyshematopoiesis, inflammation, and HLH due to aberrant CDC42 function. J Exp Med. 2019;216(12):2778–2799. doi: 10.1084/jem.20190147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gernez Y, de Jesus AA, Alsaleem H, Macaubas C, Roy A, Lovell D, Jagadeesh KA, Alehashemi S, Erdman L, Grimley M, Talarico S, Bacchetta R, Lewis DB, Canna SW, Laxer RM, Mellins ED, Goldbach-Mansky R, Weinacht KG. Severe autoinflammation in 4 patients with C-terminal variants in cell division control protein 42 homolog (CDC42) successfully treated with IL-1beta inhibition. J Allergy Clin Immunol. 2019;144(4):1122–1125. doi: 10.1016/j.jaci.2019.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bucciol G, Pillay B, Casas-Martin J, Delafontaine S, Proesmans M, Lorent N, Coolen J, Tousseyn T, Bossuyt X, Ma CS, Schrijvers R, Tangye SG, Moens L, Meyts I. Systemic inflammation and myelofibrosis in a patient with Takenouchi-Kosaki syndrome due to CDC42 Tyr64Cys mutation. J Clin Immunol. 2020;40:567–570. doi: 10.1007/s10875-020-00742-5. [DOI] [PubMed] [Google Scholar]
- 37.Bekhouche B, Tourville A, Ravichandran Y, Tacine R, Abrami L, Dussiot M, Khau-Dancasius A, Boccara O, Khirat M, Mangeney M, Dingli F, Loew D, Boëda B, Jordan P, Molina TJ, Bellon N, Fraitag S, Hadj-Rabia S, Blanche S, Puel A, Etienne-Manneville S, van der Goot FG, Cherfils J, Hermine O, Casanova JL, Bodemer C, Smahi A, Delon J. A toxic palmitoylation of Cdc42 enhances NF-kappaB signaling and drives a severe autoinflammatory syndrome. J Allergy Clin Immunol. 2020;146(5):1201–1204.e8. doi: 10.1016/j.jaci.2020.03.020. [DOI] [PubMed] [Google Scholar]
- 38.He T, Huang Y, Ling J, Yang J. A new patient with NOCARH syndrome due to CDC42 defect. J Clin Immunol. 2020;40(4):571–575. doi: 10.1007/s10875-020-00786-7. [DOI] [PubMed] [Google Scholar]
- 39.Szczawinska-Poplonyk A, Ploski R, Bernatowska E, Pac M. A novel CDC42 mutation in an 11-year old child manifesting as syndromic immunodeficiency, autoinflammation, hemophagocytic lymphohistiocytosis, and malignancy: a case report. Front Immunol. 2020;11:318. doi: 10.3389/fimmu.2020.00318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gruber C, Martin-Fernandez M, Ailal F, Qiu X, Taft J, Altman J, et al. Homozygous STAT2 gain-of-function mutation by loss of USP18 activity in a patient with type I interferonopathy. J Exp Med. 2020;217(5):e20192319. doi: 10.1084/jem.20192319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Duncan CJA, Thompson BJ, Chen R, Rice GI, Gothe F, Young DF, et al. Severe type I interferonopathy and unrestrained interferon signaling due to a homozygous germline mutation in STAT2. Sci Immunol. 2019;4(42):eaav7501. doi: 10.1126/sciimmunol.aav7501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tao P, Sun J, Wu Z, Wang S, Wang J, Li W, Pan H, Bai R, Zhang J, Wang Y, Lee PY, Ying W, Zhou Q, Hou J, Wang W, Sun B, Yang M, Liu D, Fang R, Han H, Yang Z, Huang X, Li H, Deuitch N, Zhang Y, Dissanayake D, Haude K, McWalter K, Roadhouse C, MacKenzie JJ, Laxer RM, Aksentijevich I, Yu X, Wang X, Yuan J, Zhou Q. A dominant autoinflammatory disease caused by non-cleavable variants of RIPK1. Nature. 2020;577(7788):109–114. doi: 10.1038/s41586-019-1830-y. [DOI] [PubMed] [Google Scholar]
- 43.Lalaoui N, Boyden SE, Oda H, Wood GM, Stone DL, Chau D, Liu L, Stoffels M, Kratina T, Lawlor KE, Zaal KJM, Hoffmann PM, Etemadi N, Shield-Artin K, Biben C, Tsai WL, Blake MD, Kuehn HS, Yang D, Anderton H, Silke N, Wachsmuth L, Zheng L, Moura NS, Beck DB, Gutierrez-Cruz G, Ombrello AK, Pinto-Patarroyo GP, Kueh AJ, Herold MJ, Hall C, Wang H, Chae JJ, Dmitrieva NI, McKenzie M, Light A, Barham BK, Jones A, Romeo TM, Zhou Q, Aksentijevich I, Mullikin JC, Gross AJ, Shum AK, Hawkins ED, Masters SL, Lenardo MJ, Boehm M, Rosenzweig SD, Pasparakis M, Voss AK, Gadina M, Kastner DL, Silke J. Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease. Nature. 2020;577(7788):103–108. doi: 10.1038/s41586-019-1828-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cook SA, Comrie WA, Poli MC, Similuk M, Oler AJ, Faruqi AJ, Kuhns DB, Yang S, Vargas-Hernández A, Carisey AF, Fournier B, Anderson DE, Price S, Smelkinson M, Abou Chahla W, Forbes LR, Mace EM, Cao TN, Coban-Akdemir ZH, Jhangiani SN, Muzny DM, Gibbs RA, Lupski JR, Orange JS, Cuvelier GDE, al Hassani M, al Kaabi N, al Yafei Z, Jyonouchi S, Raje N, Caldwell JW, Huang Y, Burkhardt JK, Latour S, Chen B, ElGhazali G, Rao VK, Chinn IK, Lenardo MJ. HEM1 deficiency disrupts mTORC2 and F-actin control in inherited immunodysregulatory disease. Science. 2020;369(6500):202–207. doi: 10.1126/science.aay5663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Salzer E, Zoghi S, Kiss MG, Kage F, Rashkova C, Stahnke S, et al. The cytoskeletal regulator HEM1 governs B cell development and prevents autoimmunity. Sci Immunol. 2020;5(49):eabc3979. doi: 10.1126/sciimmunol.abc3979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Castro CN, Rosenzwajg M, Carapito R, Shahrooei M, Konantz M, Khan A, et al. NCKAP1L defects lead to a novel syndrome combining immunodeficiency, lymphoproliferation, and hyperinflammation. J Exp Med. 2020;217(12):e20192275. doi: 10.1084/jem.20192275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Beck DB, Ferrada MA, Sikora KA, Ombrello AK, Collins JC, Pei W, Balanda N, Ross DL, Ospina Cardona D, Wu Z, Patel B, Manthiram K, Groarke EM, Gutierrez-Rodrigues F, Hoffmann P, Rosenzweig S, Nakabo S, Dillon LW, Hourigan CS, Tsai WL, Gupta S, Carmona-Rivera C, Asmar AJ, Xu L, Oda H, Goodspeed W, Barron KS, Nehrebecky M, Jones A, Laird RS, Deuitch N, Rowczenio D, Rominger E, Wells KV, Lee CCR, Wang W, Trick M, Mullikin J, Wigerblad G, Brooks S, Dell’Orso S, Deng Z, Chae JJ, Dulau-Florea A, Malicdan MCV, Novacic D, Colbert RA, Kaplan MJ, Gadina M, Savic S, Lachmann HJ, Abu-Asab M, Solomon BD, Retterer K, Gahl WA, Burgess SM, Aksentijevich I, Young NS, Calvo KR, Werner A, Kastner DL, Grayson PC. Somatic mutations in UBA1 and severe adult-onset autoinflammatory disease. N Engl J Med. 2020;383:2628–2638. doi: 10.1056/NEJMoa2026834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585. doi: 10.1126/science.abd4585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pohl E, Aykut A, Beleggia F, Karaca E, Durmaz B, Keupp K, Arslan E, Onay MP, Yigit G, Özkinay F, Wollnik B. A hypofunctional PAX1 mutation causes autosomal recessively inherited otofaciocervical syndrome. Hum Genet. 2013;132(11):1311–1320. doi: 10.1007/s00439-013-1337-9. [DOI] [PubMed] [Google Scholar]
- 50.Spencer S, Kostel Bal S, Egner W, Lango Allen H, Raza SI, Ma CA, et al. Loss of the interleukin-6 receptor causes immunodeficiency, atopy, and abnormal inflammatory responses. J Exp Med. 2019;216(9):1986–1998. doi: 10.1084/jem.20190344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gombart AF, Koeffler HP. Neutrophil specific granule deficiency and mutations in the gene encoding transcription factor C/EBP(epsilon) Curr Opin Hematol. 2002;9(1):36–42. doi: 10.1097/00062752-200201000-00007. [DOI] [PubMed] [Google Scholar]
- 52.Su HC, Orange JS. The growing spectrum of human diseases caused by inherited CDC42 mutations. J Clin Immunol. 2020;40(4):551–553. doi: 10.1007/s10875-020-00785-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hambleton S, Goodbourn S, Young DF, Dickinson P, Mohamad SM, Valappil M, et al. STAT2 deficiency and susceptibility to viral illness in humans. Proc Natl Acad Sci U S A. 2013;110(8):3053–3058. doi: 10.1073/pnas.1220098110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Moens L, Van Eyck L, Jochmans D, Mitera T, Frans G, Bossuyt X, et al. A novel kindred with inherited STAT2 deficiency and severe viral illness. J Allergy Clin Immunol. 2017;139(6):1995–1997. doi: 10.1016/j.jaci.2016.10.033. [DOI] [PubMed] [Google Scholar]
- 55.Cuchet-Lourenco D, Eletto D, Wu C, Plagnol V, Papapietro O, Curtis J, et al. Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation. Science. 2018;361(6404):810–813. doi: 10.1126/science.aar2641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li Y, Fuhrer M, Bahrami E, Socha P, Klaudel-Dreszler M, Bouzidi A, et al. Human RIPK1 deficiency causes combined immunodeficiency and inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2019;116(3):970–975. doi: 10.1073/pnas.1813582116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Casanova JL, Conley ME, Seligman SJ, Abel L, Notarangelo LD. Guidelines for genetic studies in single patients: lessons from primary immunodeficiencies. J Exp Med. 2014;211(11):2137–2149. doi: 10.1084/jem.20140520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bustamante J. Mendelian susceptibility to mycobacterial disease: recent discoveries. Hum Genet. 2020;139(6–7):993–1000. doi: 10.1007/s00439-020-02120-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Moens L, Meyts I. Recent human genetic errors of innate immunity leading to increased susceptibility to infection. Curr Opin Immunol. 2020;62:79–90. doi: 10.1016/j.coi.2019.12.002. [DOI] [PubMed] [Google Scholar]
- 60.Zhang SY. Herpes simplex virus encephalitis of childhood: inborn errors of central nervous system cell-intrinsic immunity. Hum Genet. 2020;139(6–7):911–918. doi: 10.1007/s00439-020-02127-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Casanova JL, Holland SM, Notarangelo LD. Inborn errors of human JAKs and STATs. Immunity. 2012;36(4):515–528. doi: 10.1016/j.immuni.2012.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shahin T, Aschenbrenner D, Cagdas D, Bal SK, Conde CD, Garncarz W, Medgyesi D, Schwerd T, Karaatmaca B, Cetinkaya PG, Esenboga S, Twigg SRF, Cant A, Wilkie AOM, Tezcan I, Uhlig HH, Boztug K. Selective loss of function variants in IL6ST cause hyper-IgE syndrome with distinct impairments of T-cell phenotype and function. Haematologica. 2019;104(3):609–621. doi: 10.3324/haematol.2018.194233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Nahum A, Sharfe N, Broides A, Dadi H, Naghdi Z, Mandola AB, Vong L, Arbiv A, Dalal I, Brami I, Wormser O, Levy J, Roifman CM. Defining the biological responses of IL-6 by the study of a novel IL-6 receptor chain immunodeficiency. J Allergy Clin Immunol. 2020;145(3):1011–1015. doi: 10.1016/j.jaci.2019.11.015. [DOI] [PubMed] [Google Scholar]
- 64.Puel A, Picard C, Lorrot M, Pons C, Chrabieh M, Lorenzo L, Mamani-Matsuda M, Jouanguy E, Gendrel D, Casanova JL. Recurrent staphylococcal cellulitis and subcutaneous abscesses in a child with autoantibodies against IL-6. J Immunol. 2008;180(1):647–654. doi: 10.4049/jimmunol.180.1.647. [DOI] [PubMed] [Google Scholar]
- 65.Martinez-Barricarte R, Markle JG, Ma CS, Deenick EK, Ramirez-Alejo N, Mele F, et al. Human IFN-gamma immunity to mycobacteria is governed by both IL-12 and IL-23. Sci Immunol. 2018;3(30):eaau6759. doi: 10.1126/sciimmunol.aau6759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kotlarz D, Zietara N, Milner JD, Klein C. Human IL-21 and IL-21R deficiencies: two novel entities of primary immunodeficiency. Curr Opin Pediatr. 2014;26(6):704–712. doi: 10.1097/MOP.0000000000000160. [DOI] [PubMed] [Google Scholar]
- 67.Zhang Q, Bastard P, Liu Z, Le Pen J, Moncada-Velez M, Chen J, et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4570. doi: 10.1126/science.abd4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhang Q. Human genetics of life-threatening influenza pneumonitis. Hum Genet. 2020;139(6–7):941–948. doi: 10.1007/s00439-019-02108-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ku CL, Chi CY, von Bernuth H, Doffinger R. Autoantibodies against cytokines: phenocopies of primary immunodeficiencies? Hum Genet. 2020;139(6–7):783–794. doi: 10.1007/s00439-020-02180-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Levy R, Bastard P, Lanternier F, Lecuit M, Zhang SY, Casanova JL. IFN-alpha2a therapy in two patients with inborn errors of TLR3 and IRF3 infected with SARS-CoV-2. J Clin Immunol. 2021;41:26–7. 10.1007/s10875-020-00933-0. [DOI] [PMC free article] [PubMed]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.