Skip to main content
Springer logoLink to Springer
. 2020 Jan 17;40(1):24–64. doi: 10.1007/s10875-019-00737-x

Human Inborn Errors of Immunity: 2019 Update on the Classification from the International Union of Immunological Societies Expert Committee

Stuart G Tangye 1,2,, Waleed Al-Herz 3, Aziz Bousfiha 4, Talal Chatila 5, Charlotte Cunningham-Rundles 6, Amos Etzioni 7, Jose Luis Franco 8, Steven M Holland 9, Christoph Klein 10, Tomohiro Morio 11, Hans D Ochs 12, Eric Oksenhendler 13, Capucine Picard 14,15, Jennifer Puck 16, Troy R Torgerson 12, Jean-Laurent Casanova 17,18,19,20, Kathleen E Sullivan 21
PMCID: PMC7082301  PMID: 31953710

Abstract

We report the updated classification of Inborn Errors of Immunity/Primary Immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee. This report documents the key clinical and laboratory features of 430 inborn errors of immunity, including 64 gene defects that have either been discovered in the past 2 years since the previous update (published January 2018) or were characterized earlier but have since been confirmed or expanded upon in subsequent studies. The application of next-generation sequencing continues to expedite the rapid identification of novel gene defects, rare or common; broaden the immunological and clinical phenotypes of conditions arising from known gene defects and even known variants; and implement gene-specific therapies. These advances are contributing to greater understanding of the molecular, cellular, and immunological mechanisms of disease, thereby enhancing immunological knowledge while improving the management of patients and their families. This report serves as a valuable resource for the molecular diagnosis of individuals with heritable immunological disorders and also for the scientific dissection of cellular and molecular mechanisms underlying inborn errors of immunity and related human diseases.

Keywords: IUIS, primary immune deficiency, inborn errors of immunity, immune dysregulation, autoinflammatory disorders, next-generation sequencing


Inborn errors of immunity, also referred to as primary immunodeficiencies, manifest as increased susceptibility to infectious diseases, autoimmunity, autoinflammatory diseases, allergy, and/or malignancy. These conditions are caused by monogenic germline mutations that result in loss of expression, loss-of-function (LOF; amorphic/hypomorphic), or gain-of-function (GOF; hypermorphic) of the encoded protein [1, 2]. Heterozygous lesions may underlie autosomal dominant traits by GOF, haploinsufficiency, or negative dominance. Biallelic lesions typically cause autosomal recessive traits by LOF of the encoded protein (rarely GOF), while X-linked recessive traits arise from LOF of genes on the X chromosome, either in the hemizygous state in males or in the homozygous state in females. Rare X-linked dominant traits can also arise from LOF or GOF variants. This results in aberrant immunity due to the critical roles of these proteins in the development, maintenance and function of cells of the immune system, or cells other than leukocytes that contribute to immunity, during homeostasis and in response to external (e.g., infectious agents or environmental antigens) and internal (e.g., cytokines, self-antigens and cancer cells) stimuli [35]. Inborn errors of immunity were traditionally considered to be rare diseases, affecting ~ 1 in 10,000 to 1 in 50,000 births. However, with ongoing discovery of novel inborn errors of immunity (Fig. 1a) and improved definition of clinical phenotypes [68], the collective prevalence of these conditions is more likely to be at least 1/1000–1/5000 [9]. Indeed, more common inborn errors have recently been described [10]. Regardless of their exact incidence and prevalence, inborn errors of immunity represent an unprecedented model to link defined monogenic defects with clinical phenotypes of immune dysregulation, in a broad sense of the term. As a committee, we are aware that human immunity involves cells other than circulating or tissue leukocytes and that it can be scaled up from the immune system to the whole organism. Inborn errors of immunity have unequivocally revealed non-redundant roles of single genes and their products in immune function [3, 4, 68], formed the basis of improved mechanism-based therapies for the immunopathology underlying many diseases [8, 11], established immunological paradigms representing the foundations of basic, clinical and translational immunology [35, 9, 1214], and provided insights into the molecular pathogenesis of more common diseases [9, 15]. Clear examples of these include:

  • The initial description by Bruton of X-linked agammaglobulinemia (XLA) and the ability to treat this condition with antibody replacement therapy (the mainstay treatment for antibody deficiency diseases such as CVID) [16]

  • The discovery of mutations in BTK [12] and the subsequent development of BTK-inhibitors such as ibrutinib for the treatment of B cell malignancies [14]

  • Progressive CD4 T cell deficiency explains opportunistic infections secondary to HIV infection [9].

Fig. 1.

Fig. 1

Rate of discovery of novel inborn errors of immunity: 1983–2019. a The number of genetic defects underlying monogenic immune disorders as reported by the IUIS/WHO committee in the indicated year. b The number of pathogenic gene variants listed in each table by the IUIS committee. Report published in 2017, and the number of new genes for each table contained in this report (red bars). The numbers in each column correspond to the number of genes reported in the 2017 IUIS update (blue bars) [1, 2], the number of new genes for each table contained in this report (red bars), and the total number of genes for each table. Note: only data for Tables 1, 2, 3, 4, 5, 6, 7, and 8 are shown, because Table 9 (bone marrow failure) is a new addition to the current report.

Thus, the study of inborn errors of immunity has provided profound advances in the practice of precision molecular medicine.

Since the early 1950s, when XLA was one of the first primary immune deficiencies to be described [16], clinical immunology has leveraged advances in the development of new methods to expedite the identification of defects of the immune system and the cellular, molecular, and genetic aberrations underlying these conditions. Indeed, the completion of the Human Genome Project in the early 2000s, coupled with rapid developments in next generation DNA sequencing (NGS) technologies, enabled the application of cost-effective and time-efficient sequencing of targeted gene panels, whole exomes, or whole genomes to cohorts of patients suspected of having a monogenic explanation for their disease. These platforms have led to a quantum leap in the identification and diagnosis of previously undefined genetically determined defects of the immune system (Fig. 1a, b; [68]).

The International Union of Immunological Societies Expert Committee of Inborn Errors of Immunity comprises pediatric and adult clinical immunologists, clinician/scientists and researchers in basic immunology from across the globe (https://iuis.org/committees/iei/). A major objective and responsibility of the committee is to provide the clinical and research communities with an update of genetic causes of immune deficiency and dysregulation. The committee has existed since 1970 and has published an updated report approximately every 2 years to inform the field of these advances (Fig. 1a). In March 2019, the committee met in New York to discuss and debate the inclusion of genetic variants published over the preceding 2 years (since June 2017) [1, 2], as well as gene mutations that had appeared in the literature earlier but, based on newly available evidence, were now substantiated (Fig. 1b).

Rather than simply including every gene variant reported, the committee applies very stringent criteria such that only those genes with convincing evidence of disease pathogenicity are classified as causes of novel inborn errors of immunity [17]. The Committee makes informed judgments for including new genetic causes of immunological conditions based on what we believe is most useful for practitioners caring for patients. Our current, and continuously evolving, practice is that criteria for inclusion can be met by several ways, for instance peer-reviewed publication of (1) multiple cases from unrelated kindreds, including detailed immunologic data, or (2) very few cases, or even a single case (see below), for whom compelling mechanistic/pathogenic data is also provided, generally from parallel studies in an animal or cell culture model.

Herein, we provide this latest update. The inborn errors of immunity are listed in 10 tables: Combined immunodeficiencies (Table 1), Combined immunodeficiencies with syndromic features (Table 2), Predominantly antibody deficiencies (Table 3), Diseases of immune dysregulation (Table 4), Congenital defects of phagocytes (Table 5), Defects in intrinsic and innate immunity (Table 6), Autoinflammatory diseases (Table 7), Complement deficiencies (Table 8), and Phenocopies of inborn errors of immunity (Table 10) (Fig. 1b). Since the last update (published January 2018) [1, 2], we have added a new table to consolidate genes that cause bone marrow failure (Table 9). Our division into phenotypes does not imply that the presentation is homogeneous. Rather, we recognize that substantial phenotypic and clinical heterogeneity exists within groups of patients with mutations in the same gene and even between individuals from the same pedigree with the identical gene mutation. To simplify the classification, each disorder has been listed only once, although distinct disorders due to mutations in the same gene, but with different modes of inheritance and pathogenic mechanisms are listed individually. Thus, several genes appear more than once in this update (some examples are listed below). Sub-divisions within each table segregate groups of disorders into coherent phenotypic sets. OMIM numbers are also provided within each table. If a OMIM number has not yet been issued for a particular genetic condition, then the number provided generally refers to the OMIM for that gene. Beneath each table, the new disorders added to this update are highlighted for easy reference.

Table 1.

Immunodeficiencies affecting cellular and humoral immunity

Disease Genetic defect Inheritance OMIM T cells B cells Ig Associated features
1. T-B+ severe combined immune deficiency (SCID)
  γc deficiency (common gamma chain SCID, CD132 deficiency) IL2RG XL 308380 Very low Normal to high Low Low NK
  JAK3 deficiency JAK3 AR 600173 Very low Normal to high Low Low NK
  IL7Rα deficiency IL7R AR 146661 Very low Normal to high Low Normal NK
  CD45 deficiency PTPRC AR 151460 Very low Normal Low Normal γ/δ Τ cells
  CD3δ deficiency CD3D AR 186790 Very low Normal Low Normal NK, no γ/δ T cells
  CD3ε deficiency CD3E AR 186830 Very low Normal Low Normal NK, no γ/δ T cells
  CD3ζ deficiency CD3Z AR 186780 Very low Normal Low Normal NK, no γ/δ T cells
  Coronin-1A deficiency CORO1A AR 605000 Very low Normal Low Detectable thymus
  LAT deficiency LAT AR 602354 Normal to low Normal to low High Typical SCID or combined immunodeficiency, the latter with adenopathy, splenomegaly, recurrent infections, autoimmunity
2. T-B- SCID
  RAG deficiency

RAG1

RAG2

AR

179615

179616

Very low Very low Decreased Normal NK cell number, but increased risk of graft rejection, possibly due to activated NK cells
  DCLRE1C (Artemis) deficiency DCLRE1C AR 605988 Very low Very low Decreased Normal NK cell number, but increased risk of graft rejection, possibly due to activated NK cells, radiation sensitivity
  DNA PKcs deficiency PRKDC AR 615966 Very low Very low Variable Normal NK, radiation sensitivity, microcephaly
  Cernunnos/XLF deficiency NHEJ1 AR 611290 Very low Very low Decreased Normal NK, radiation sensitivity, microcephaly
  DNA ligase IV deficiency LIG4 AR 601837 Very low Very low Decreased Normal NK, radiation sensitivity, microcephaly
  Adenosine deaminase (ADA) deficiency ADA AR 608958 Very low Low, decreasing Low, decreasing Low NK, bone defects, may have pulmonary alveolar proteinosis, cognitive defects
  AK2 defect AK2 AR 103020 Very low Very Low Decreased Reticular dysgenesis with neutropenia; deafness
  Activated RAC2 defect RAC2 AD GOF 602049 Very low Very Low Low, poor specific antibody responses Recurrent bacterial and viral infections, lymphoproliferation; neutropenia
3. Combined immunodeficiency (CID), generally less profound than SCID
  CD40 ligand (CD154) deficiency CD40LG XL 308230 Normal to low sIgM+IgD+ naïve B cells present; IgG+, IgA+, IgE+ memory B cells absent IgM normal or high, other Ig isotypes low Severe and opportunistic infections, idiopathic neutropenia; hepatitis and cholangitis, Cryptosporidium infections, cholangiocarcinoma; autoimmune blood cytopenias; peripheral neuroectodermal tumors
  CD40 deficiency CD40 AR 606843 Normal Neutropenia, opportunistic infections, gastrointestinal and biliary tract and liver disease, Cryptosporidium infections
  ICOS deficiency ICOS AR 604558 Normal Normal Low Recurrent infections, autoimmunity, gastroenteritis, granulomas
  ICOSL deficiency ICOSLG AR 605717 Low Low Low Recurrent bacterial and viral infections, neutropenia
  CD3γ deficiency CD3G AR 186740 Normal number, but low TCR expression Normal Normal Immune deficiency and autoimmunity of variable severity
  CD8 deficiency CD8A AR 186910 Absent CD8, Normal CD4 Normal Normal Recurrent infections, may be asymptomatic
  ZAP-70 deficiency (ZAP70 LOF) ZAP70 AR 269840 Low CD8 number, normal CD4 number but with poor function Normal Normal May have immune dysregulation, autoimmunity
  ZAP-70 combined hypomorphic and activating mutations ZAP70 AR (LOF/GOF) 617006 Decreased CD8, normal or decreased CD4 cells Normal or decreased Normal IgA, low IgM, low/normal IgG; protective Ab responses to vaccines Severe autoimmunity (bullous pemphigoid, inflammatory colitis
  MHC class I deficiency TAP1 AR 170260 Low CD8, normal CD4, absent MHC I on lymphocytes Normal Normal Vasculitis, pyoderma gangrenosum
TAP2 AR 170261
TAPBP AR 601962
B2M AR 109700 Sinopulmonary infections, cutaneous granulomas. Absent β2m associated proteins MHC-I, CD1a, CD1b, and CD1c
  MHC class II deficiency group A, B, C, D CIITA AR 600005 Low CD4+ T cells, reduced MHC II expression on lymphocytes Normal Normal to low Failure to thrive, respiratory and gastrointestinal infections, liver/biliary tract disease
RFXANK AR 603200
RFX5 AR 601863
RFXAP AR 601861
  IKAROS deficiency IKZF1 AD DN 603023 no memory T cells no memory B cells Low Ig, recurrent sinopulmonary infections, pneumocystis early CID onset
  DOCK8 deficiency DOCK8 AR 243700 T cell lymphopenia, reduced naïve CD8 T cells, increased exhausted CD8+ TEM cells, reduced MAIT, NKT cells, increased γδ T cells; poor proliferation; few Treg with poor function increased total B cells, reduced memory B cells Poor peripheral B cell tolerance. Low IgM, normal/high IgG and IgA, very high IgE, poor antibody responses Low NK cells with poor function. Eosinophilia, recurrent infections, cutaneous viral, fungal and staphylococcal infections, severe atopy/allergic disease, cancer diathesis
  DOCK2 deficiency DOCK2 AR 603122 Low Normal IgG normal or low, poor antibody responses Early invasive herpes viral, bacterial infections, Normal NK cell number, but defective function. Poor interferon responses in hematopoietic and non-hematopoietic cells
  Polymerase and deficiency

POLD1

POLD2

AR

174761

600815

Low CD4 T cells Low B cells but normal maturation Low igG Recurrent respiratory tract infections, skin infections, warts and molluscum, short stature, intellectual disability
  RHOH deficiency RHOH AR 602037 Normal, few naïve T cells, restricted repertoire, poor proliferation to CD3 Normal Normal HPV infection, lung granulomas, molluscum contagiosum, lymphoma
  STK4 deficiency STK4 AR 614868 CD4 lymphopenia, reduced naïve T cells, increased TEM and TEMRA cells, poor proliferation Reduced memory B cells Reduced IgM, increased IgG, IgA, IgE; impaired Ab responses Intermittent neutropenia, bacterial, viral (HPV, EBV, molluscum), candidal infections, lymphoproliferation, autoimmune cytopenias, lymphoma, congenital heart disease
  TCRα deficiency TRAC AR 615387 Absent TCRαβ except for a minor CD3-dim TCRαβ population; most T cells γδ; poor proliferation Normal Normal Recurrent viral, bacterial, fungal infections, immune dysregulation and autoimmunity, diarrhea
  LCK deficiency LCK AR 615758 Low CD4+, low Treg, restricted T cell repertoire, poor TCR signaling Normal Normal IgG and IgA, high IgM Recurrent infections, immune dysregulation, autoimmunity
  ITK deficiency ITK AR 186973 Progressive CD4 T cell lymphopenia; reduced T cell activation Normal Normal to low serum Ig EBV associated B cell lymphoproliferation, lymphoma, immune dysregulation
  MALT1 deficiency MALT1 AR 615468 Normal number, poor proliferation Normal Normal levels, poor specific antibody response Bacterial, fungal and viral infections
  CARD11 deficiency CARD11 AR LOF 615206 Normal number, predominantly naïve T cells, poor proliferation Normal, transitional B cell predominance Absent/low Pneumocystis jirovecii pneumonia, bacterial and viral infections
  BCL10 deficiency BCL10 AR 616098 Normal number, few memory T and Treg cells, poor antigen and anti-CD3 proliferation Normal number, decreased memory and switched B cells Low Recurrent bacterial and viral infections, candidiasis, gastroenteritis
  IL-21 deficiency IL21 AR 615767 Normal number, normal/low function Low, decreased memory and switched B cells Hypogammaglobulinemia, poor specific antibody responses; increased IgE Severe early onset colitis, recurrent sinopulmonary infections
  IL-21R deficiency IL21R AR 615207 Normal number, low cytokine production, poor antigen proliferation Normal, decreased memory and switched B cells Recurrent infections, Pneumocystis jiroveci, Cryptosporidium infections, liver disease
  OX40 deficiency TNFRSF4 AR 615593 Normal numbers, low antigen specific memory CD4+ Normal numbers, low memory B cells Normal Impaired immunity to HHV8, Kaposi’s sarcoma
  IKBKB deficiency IKBKB AR 615592 Normal number, absent Treg and γ/δ T cells, impaired TCR activation Normal number, poor function Low Recurrent bacterial, viral, fungal infections, opportunistic infections
  NIK deficiency MAP 3 K14 AR 604655 Normal number, poor proliferation to antigen Low, low switched memory B cells Low Ig’s Low NK number and function, recurrent bacterial, viral and Cryptosporidium infections
  RelB deficiency RELB AR 604758 Normal number, poor diversity, reduced proliferation to mitogens; no response to Ag Marked increase in B cell number Normal Ig levels but Impaired specific antibody responses Recurrent infections
  RelA haploinsufficiency RELA AD 618287 Normal/increased Normal Normal Chronic mucocutaneous ulceration, Impaired NFkB activation; reduced production of inflammatory cytokines
  Moesin deficiency MSN XL 300988 Normal number, defective migration, proliferation Low number Low Ig’s over time Recurrent infections with bacteria, varicella, neutropenia
  TFRC deficiency TFRC AR 616740 Normal number, poor proliferation Normal number, low memory B cells Low Recurrent infections, neutropenia, thrombocytopenia
  c-Rel deficiency REL AR 164910 Normal, decreased memory CD4, poor proliferation Low, mostly naïve; few switched memory B cells, impaired proliferation Low, poor specific antibody responses Recurrent infections with bacteria, mycobacteria, salmonella and opportunistic organisms. Defective innate immunity
  FCHO1 deficiency FCHO1 AR 613437 Low, poor proliferation Normal number Normal Recurrent infections (viral, mycobacteria, bacterial, fungal), lymphoproliferation, failure to thrive, increased activation-induced T cell death, defective clathrin-mediated endocytosis

SCID/CID spectrum: Infants with SCID who have maternal T cell engraftment may have T cells in normal numbers that do not function normally; these cells may cause autoimmune cytopenias or graft versus host disease. Hypomorphic mutations in several of the genes that cause SCID may result in Omenn syndrome (OS), or “leaky” SCID, or still less profound combined immunodeficiency (CID) phenotypes. Both OS and leaky SCID can be associated with > 300 autologous T cells/μL of peripheral blood and reduced, rather than absent, proliferative responses when compared with typical SCID caused by null mutations. A spectrum of clinical findings including typical SCID, OS, leaky SCID, CID, granulomas with T lymphopenia, autoimmunity and CD4 T lymphopenia can be found in an allelic series of RAG1/2 and other SCID-associated genes. There can be clinical overlap between some genes listed here and those listed in Table 7

Total number of disorders in Table 1: 50

Total number of mutant genes: 58

New inborn errors of immunity: 8; New inborn errors of immunity: 8; RAC2 GOF [1821]; ICOSLG [22]; AD DN IKZF1 [23]; POLD1 [24, 25]; POLD2 [24]; RELA [26, 27]; REL [28]; FCHO1 [29]

SCID severe combined immunodeficiency, CID combined immunodeficiency, EBV Epstein-Barr virus, MHC major histocompatibility complex, HPV human papillomavirus, Treg T regulatory cell, XL X-linked inheritance, AR autosomal recessive inheritance, AD autosomal dominant inheritance, LOF loss-of-function, GOF gain-of-function

Table 2.

Combined immunodeficiencies with associated or syndromic features

Disease Genetic defect Inheritance OMIM T cells B cells Ig Associated features
1. Immunodeficiency with congenital thrombocytopenia
  Wiskott-Aldrich syndrome (WAS LOF) WAS XL 300392 Progressive decrease in numbers, abnormal lymphocyte responses to anti-CD3 Normal numbers Low IgM and antibody responses to polysaccharides, often high IgA and IgE Thrombocytopenia with small platelets, eczema, recurrent bacterial/viral infections, bloody diarrhea, lymphoma, autoimmune disease, IgA- nephropathy. Patients with XL-thrombocytopenia have later onset of complications and more favourable life expectancy but eventually develop similar complications as observed in WAS
  WIP deficiency WIPF1 AR 602357 Reduced, defective lymphocyte responses to anti-CD3 Normal or low Normal, except for high IgE Thrombocytopenia with or without small platelets, recurrent bacterial and viral infections, eczema, bloody diarrhea; WAS protein absent
  Arp2/3-mediated filament branching defect ARPC1B AR 604223 Normal Normal numbers Normal except for high IgA and IgE Mild thrombocytopenia with normal sized platelets, recurrent invasive infections; colitis, vasculitis, autoantibodies (ANA, ANCA), eosinophilia; defective Arp2/3 filament branching
2. DNA repair defects other than those listed in Table 1
  Ataxia-telangiectasia ATM AR 607585 Progressive decrease, poor proliferation to mitogens; may have low TRECs and T cells by newborn screening (NBS) Normal Often low IgA, IgE and IgG subclasses, increased IgM monomers; antibodies variably decreased Ataxia, telangiectasia especially of sclerae; pulmonary infections; lymphoreticular and other malignancies; increased alpha fetoprotein; increased radiosensitivity, chromosomal instability and chromosomal translocations
  Nijmegen breakage syndrome NBS1 AR 602667 Progressive decrease; may have low TRECs and T cells by NBS Variably reduced Often low IgA, IgE, and IgG subclasses, increased IgM; antibodies variably decreased Microcephaly, dysmorphic facies; lymphomas and solid tumors; increased radiosensitivity;, chromosomal instability
  Bloom syndrome BLM AR 604610 Normal Normal Low Short stature, dysmorphic facies sun-sensitive erythema; marrow failure; leukemia, lymphoma; chromosomal instability
  Immunodeficiency with centromeric instability and facial anomalies (ICF types 1, 2, 3, 4) DNMT3B AR 602900 Decreased or normal, responses to PHA may be decreased Decreased or normal Hypogammaglobulinemia or agammaglobulinemia, variable antibody deficiency Facial dysmorphic features, developmental delay, macroglossia; bacterial/opportunistic infections; malabsorption; cytopenias; malignancies; multiradial configurations of chromosomes 1, 9, 16
ZBTB24 AR 614064 Decreased or normal Facial dysmorphic features, macroglossia; bacterial/opportunistic infections; malabsorption; cytopenias; malignancies; multiradial configurations of chromosomes 1, 9, 16
CDCA7 AR 609937 Decreased or normal; responses to PHA may be decreased
HELLS AR 603946 Decreased or normal
  PMS2 deficiency PMS2 AR 600259 Normal Low B cells, switched and non-switched Low IgG and IgA, high IgM, abnormal antibody responses Recurrent infections; café-au-lait spots; lymphoma, colorectal carcinoma, brain tumors
  RNF168 deficiency (Radiosensitivity, Immune Deficiency, Dysmorphic features, Learning difficulties [RIDDLE] syndrome) RNF168 AR 612688 Normal Normal Low IgG or IgA Short stature, mild defect of motor control to ataxia; normal intelligence to learning difficulties; mild facial dysmorphism to microcephaly; increased radiosensitivity
  MCM4 deficiency MCM4 AR 602638 Normal Normal Normal NK cells: low number and function; viral infections (EBV, HSV, VZV); short stature; B cell lymphoma; adrenal failure
  X-linked reticulate pigmentary disorder (POLA1 deficiency) POLA1 XL 301220 Not assessed Not assessed Not assessed Hyperpigmentation, characteristic facies, lung and GI involvement
  POLE1 (Polymerase ε subunit 1) deficiency (FILS syndrome) POLE1 AR 174762 Normal; decreased T cell proliferation Low memory B cells Low IgG2 and IgM, lack of antibody to PPS Recurrent respiratory infections, meningitis; facial dysmorphism, livido, short stature
  POLE2 (Polymerase ε subunit 2) deficiency POLE2 AR 602670 Lymphopenia, lack of TRECS at NBS, absent proliferation in response to antigens Very low Hypogammaglobulinemia Recurrent infections, disseminated BCG infections; autoimmunity (type 1 diabetes), hypothyroidism, facial dysmorphism
  Ligase I deficiency LIG1 AR 126391 Lymphopenia, increased γδ T cells, decreased mitogen response Normal Hypogammaglobulinemia, Reduced antibody responses Recurrent bacterial and viral infections; growth retardation; sun sensitivity, radiation sensitivity; macrocytic red blood cells
  NSMCE3 deficiency NSMCE3 AR 608243 Decreased number, poor responses to mitogens and antigens Normal Normal IgG, IgA, normal to elevated IgM; decreased antibody responses to PPS Severe lung disease (possibly viral); thymic hypoplasia; chromosomal breakage, radiation sensitivity
  ERCC6L2 (Hebo deficiency) ERCC6L2 AR 615667 Lymphopenia Low Normal Facial dysmorphism, microcephaly; bone marrow failure
  GINS1 deficiency GINS1 AR 610608 Low or normal Low or normal High IgA, low IgM and IgG Neutropenia; IUGR; NK cells very low
3. Thymic defects with additional congenital anomalies

  DiGeorge/velocardio-facial syndrome

Chromosome 22q11.2 deletion syndrome (22q11.2DS)

Large deletion (3 Mb) typically in chromosome 22 (TBX1) AD 602054 Decreased or normal, 5% have low TRECs at NBS and < 1500 CD3T cells/μL in neonatal period Normal Normal or decreased Hypoparathyroidism; conotruncal cardiac malformation, velopalatal insufficiency; abnormal facies; intellectual disability
  DiGeorge/velocardio-facial syndrome Unknown Sporadic Decreased or normal
  TBX1 deficiency TBX1 AD 602054 Decreased or normal, may have low TRECs at NBS
  CHARGE syndrome CHD7 AD 608892 Decreased or normal, may have low TRECs at NBS; response to PHA may be decreased Normal Normal or decreased Coloboma of eye; heart anomaly; choanal atresia; intellectual disability; genital and ear anomalies, CNS malformation; some are SCID-like
SEMA3E AD 608166
Unknown
  Winged helix nude FOXN1 deficiency FOXN1 AR 601705 Very low Normal Decreased Severe infections; abnormal thymic epithelium, immunodeficiency; congenital alopecia, nail dystrophy; neural tube defect
  FOXN1 haploinsufficiency FOXN1 AD 600838 Severe T cell lymphopenia at birth, normalised by adulthood Normal/low Not assessed Recurrent, viral and bacterial respiratory tract infections; skin involvement (eczema, dermatitis), nail dystrophy
  Chromosome 10p13-p14 deletion syndrome (10p13-p14DS) Del10p13-p14 AD 601362 Normal, rarely lymphopenia and decreased lymphoproliferation to mitogens and antigens; hypoplastic thymus may be present Normal Normal Hypoparathyroidism; renal disease; deafness; growth retardation; facial dysmorphism; cardiac defects may be present; recurrent infections ±
  Chromosome 11q deletion syndrome (Jacobsen syndrome) 11q23del AD 147791 Lymphopenia; low NK cells Decreased B cells and switched memory B cells Hypogammaglobulinemia, decreased antibody responses Recurrent respiratory infections; multiple warts; facial dysmorphism, growth retardation
4. Immuno-osseous dysplasias
  Cartilage hair hypoplasia (CHH) RMRP AR 157660 Varies from severely decreased (SCID) to normal; impaired lymphocyte proliferation Normal Normal or reduced, antibodies variably decreased Short-limbed dwarfism with metaphyseal dysostosis; sparse hair; bone marrow failure; autoimmunity; susceptibility to lymphoma and other cancers; impaired spermatogenesis; neuronal dysplasia of the intestine
  Schimke immuno-osseous dysplasia SMARCAL1 AR 606622 Decreased Normal Normal Short stature, spondiloepiphyseal dysplasia, intrauterine growth retardation; nephropathy; bacterial, viral, fungal infections; may present as SCID; bone marrow failure
  MYSM1 deficiency MYSM1 AR 612176 T cell lymphopenia, reduced naïve T cells, low NK cells B cell deficiency Hypogammaglobulinemia Short stature; recurrent infections; congenital bone marrow failure, myelodysplasia; immunodeficiency affecting B cells and granulocytes; skeletal anomalies; cataracts; developmental delay
  MOPD1 deficiency (Roifman syndrome) RNU4ATAC AR 601428 Decreased NK cell function Decreased total and memory B cells Hypogammaglobulinemia, variably decreased specific antibodies Recurrent bacterial infections; lymphadenopathy; spondyloepiphyseal dysplasia, extreme intrauterine growth retardation; retinal dystrophy; facial dysmorphism; may present with microcephaly; short stature
  Immunoskeletal dysplasia with neurodevelopmental abnormalities (EXTL3 deficiency) EXTL3 AR 617425 Decreased Normal Decreased to normal Short stature; cervical spinal stenosis, neurodevelopmental impairment; eosinophilia; may have early infant mortality
5. Hyper IgE syndromes (HIES)
  AD-HIES STAT3 deficiency (Job syndrome) STAT3 AD LOF (dominant negative) 147060 Normal overall; Th17, T follicular helper, MAIT, NKT cells decreased, Tregs may be increased; impaired responses to STAT3-activatng cytokines Normal, reduced memory B cells, BAFF expression increased, impaired responses to STAT3-activating cytokines Very high IgE, specific antibody production decreased Distinctive facial features (broad nasal bridge); bacterial infections (boils, pulmonary abscesses, pneumatoceles) due to S. aureus, pulmonary aspergillus, Pneumocystis jirovecii; eczema, mucocutaneous candidiasis; hyperextensible joints, osteoporosis and bone fractures, scoliosis, retained primary teeth; coronary and cerebral aneurysms
  IL6 receptor deficiency IL6R AR 147880 Normal/increased; normal responses to mitogens Normal total and memory B; reduced switched memory B Normal/low serum IgM, G, A. Very high IgE; specific antibody production low Recurrent pyogenic infections, cold abscesses; high circulating IL-6 levels
  IL6 signal transducer (IL6ST) deficiency IL6ST AR 618523 Decreased Th17 cells Reduced switched and non-switched memory B cells High IgE, specific antibody production variably affected Bacterial infections, boils, eczema, pulmonary abscesses, pneumatoceles; bone fractures; scoliosis; retention of primary teeth; craniosynostosis

  ZNF341 deficiency

AR-HIES

ZNF341 AR 618282 Decreased Th17 and NK cells Normal, reduced memory B cells, impaired responses to STAT3-activaitng cytokines High IgE and IgG, specific antibody production decreased Phenocopy of AD-HIES; mild facial dysmorphism; early onset eczema, MCC, bacterial skin infections, abscesses, recurrent bacterial respiratory infections (S. aureus), lung abscesses and pneumatoceles; hyperextensible joints; bone fractures and retention of primary teeth
ERBIN deficiency ERBB2IP AD 606944 Increased circulating Treg Normal Moderately increased IgE Recurrent respiratory infections, susceptibility to S. aureus, eczema; hyperextensible joints, scoliosis; arterial dilatation in some patients
Loeys-Dietz syndrome (TGFBR deficiency) TGFBR1 AD 609192 Normal Normal Elevated IgE Recurrent respiratory infectons; eczema, food allergies; hyper-extensible joints, scoliosis, retention of primary teeths; aortic aneurisms.
TGFBR2 610168
Comel-Netherton syndrome SPINK5 AR 605010 Normal Low switched and non-switched B cells High IgE and IgA, Antibody variably decreased Congenital ichthyosis, bamboo hair, atopic diathesis; increased bacterial infections; failure to thrive
PGM3 deficiency PGM3 AR 172100 CD8 and CD4 T cells may be decreased Low B and memory B cells Normal or elevated IgG and IgA, most with high IgE, eosinophilia Severe atopy; autoimmunity; bacterial and viral infections; skeletal anomalies/dysplasia: short stature, brachydactyly, dysmorphic facial features; intellectual disability and cognitive impairment, delayed CNS myelination in some affected individuals
  CARD11 deficiency (heterozygous DN) CARD11 AD LOF 617638 Normal overall, but defective T cell activation and proliferation; skewing toward Th2 Normal to low High IgE, poor specific antibody production; impaired activation of both NF-κB and mTORC1 pathways Variable atopy, eczema, food allergies, eosinophilia; cutaneous viral infections, recurrent respiratory infections; lymphoma; CID
6. Defects of vitamin B12 and folate metabolism
  Transcobalamin 2 deficiency TCN2 AR 613441 Normal Variable Decreased Megaloblastic anemia, pancytopenia; if untreated (B12) for prolonged periods results in intellectual disability
  SLC46A1/PCFT deficiency causing hereditary folate malabsorption SLC46A1 AR 229050 Variable numbers and activation profile Variable Decreased Megaloblastic anemia, failure to thrive; if untreated for prolonged periods results in intellectual disability
  Methylene-tetrahydrofolate dehydrogenase 1 (MTHFD1) deficiency MTHFD1 AR 172460 Low thymic output, normal in vitro proliferation Low Decreased/poor antibody responses to conjugated polysaccharide antigens Recurrent bacterial infection, Pneumocystis jirovecii; megaloblastic anemia; failure to thrive; neutropenia; seizures, intellectual disability; folate-responsive
7. Anhidrotic ectodermodysplasia with immunodeficiency (EDA-ID)
  EDA-ID due to NEMO/IKBKG deficiency (ectodermal dysplasia, immune deficiency) IKBKG XL 300248 Normal or decreased, TCR activation impaired Normal; Low memory and isotype switched B cells Decreased, some with elevated IgA, IgM, poor specific antibody responses, absent antibodies to polysaccharide antigens Anhidrotic ectodermal dysplasia (in some); various infections (bacteria, mycobacteria, viruses, fungi); colitis; conical teeth, variable defects of skin, hair and teeth; monocyte dysfunction
  EDA-ID due to IKBA GOF mutation NFKBIA AD GOF 164008 Normal total T cells, TCR activation impaired Normal B cell numbers, impaired BCR activation, low memory and isotype switched B cells Decreased IgG and IgA, elevated IgM, poor specific antibody responses, absent antibody to polysaccharide antigens Anhidrotic ectodermal dysplasia; various infections (bacteria, mycobacteria, viruses, fungi); colitis; variable defects of skin, hair and teeth; T cell and monocyte dysfunction
  EDA-ID due to IKBKB GOF mutation IKBKB AD GOF 618204 Decreased T cells, impaired TCR activation Normal number, poor function Reduced Recurrent bacterial, viral, fungal infections; variable ectodermal defects
8. Calcium channel defects
ORAI-1 deficiency ORAI1 AR 610277 Normal, defective TCR mediated activation Normal Normal Autoimmunity; EDA; non-progressive myopathy
STIM1 deficiency STIM1 AR 605921
9. Other defects
  Purine nucleoside phosphorylase (PNP) deficiency PNP AR 164050 Progressive decrease Normal Normal or low Autoimmune hemolytic anemia; neurological impairment
  Immunodeficiency with multiple intestinal atresias TTC7A AR 609332 Variable, but sometimes absent or low TRECs at NBS; may have SCID phenotype at birth Normal or low Markedly decreased IgG, IgM, IgA Bacterial (sepsis), fungal, viral infections; multiple intestinal atresias, often with intrauterine polyhydramnios and early demise
  Tricho-Hepato-Enteric Syndrome (THES) TTC37 AR 222470 Impaired IFNγ production Variably low numbers of switched memory B cells Hypogammaglobulinemia, may have low antibody responses Respiratory infections; IUGR; facial dysmorphic features, wooly hair; early onset intractable diarrhea, liver cirrhosis; platelet abnormalities
SKIV2L 614602
  Hepatic veno-occlusive disease with immunodeficiency (VODI) SP110 AR 604457 Normal (decreased memory T cells) Normal (decreased memory B cells) Decreased IgG, IgA, IgM, absent germinal center and tissue plasma cells Hepatic veno-occlusive disease; susceptibility to Pneumocystis jirovecii pneumonia, CMV, candida; thrombocytopenia; hepatosplenomegaly; cerebrospinal leukodystrophy
  BCL11B deficiency BCL11B AD 617237 Low, poor proliferation Normal Normal Congenital abnormalities, neonatal teeth, dysmorphic facies; absent corpus callosum, neurocognitive deficits
  EPG5 deficiency (Vici syndrome) EPG5 AR 615068 Profound depletion of CD4+ cells Defective Decreased (particularly IgG2) Agenesis of the corpus callosum; cataracts; cardiomyopathy; skin hypopigmentation; intellectual disability; microcephaly; recurrent infections, chronic mucocutaneous candidiasis
  HOIL1 deficiency RBCK1 AR 610924 Normal numbers Normal, decreased memory B cells Poor antibody responses to polysaccharides Bacterial infections; autoinflammation; amylopectinosis
  HOIP deficiency RNF31 AR 612487 Normal numbers Normal, decreased memory B cells decreased Bacterial infections; autoinflammation; amylopectinosis; lymphangiectasia
  Hennekam-lymphangiectasia-lymphedema syndrome CCBE1 AR 612753 Low/variable Low/variable decreased Lymphangiectasia and lymphedema with facial abnormalities and other dysmorphic features
FAT4 AR 612411 Low/variable Low/variable decreased Lymphangiectasia and lymphedema with facial abnormalities and other dysmorphic features
  Activating de novo mutations in nuclear factor, erythroid 2- like (NFE2L2) NFE2L2 AD 617744 Not reported Decreased switched memory B cells Hypogammaglobulinemia, decreased antibody responses Recurrent respiratory and skin infections; growth retardation, developmental delay; white matter cerebral lesions; increased level of homocysteine; increased expression of stress response genes
  STAT5b deficiency STAT5B AR 245590 Modestly decreased, reduced Treg number and function Normal hypergammaglobulinemia, increased IgE Growth-hormone insensitive dwarfism; dysmorphic features; eczema; lymphocytic interstitial pneumonitis; prominent autoimmunity
  STAT5b deficiency STAT5B AD (dominant negative) 604260 Normal Normal Increased IgE Growth-failure; eczema (no immune defects compared to AR STAT5 deficiency)
  Kabuki syndrome (type 1 and 2) KMT2D AD 602113 Normal Normal Low IgA and occasionally low IgG Typical facial abnormalities, cleft or high arched palate, skeletal abnormalities, short stature; intellectual disability; congenital heart defects; recurrent infections (otitis media, pneumonia) in 50% of patients; autoimmunity may be present
KDM6A XL (females may be affected) 300128
  KMT2A deficiency (Wiedemann-Steiner syndrome) KMT2A AD 605130 Normal Decreased switched and non-switched memory B cells Hypogammaglobulinemia, decreased antibody responses Respiratory infections; short stature; hypertelorism; hairy elbows; developmental delay, intellectual disability

Total number of disorders in Table 2: 58

Total number of mutant genes in Table 2: 62

New inborn errors of immunity: 12; LIG1 [30]; FOXN1 haploinsufficiency [31]; IL6R [32, 33]; IL6ST [34, 35]; ZNF341 [36, 37]; ERBB2IP [38]; TGFBR1 [39]; TGFBR2 [39]; AD LOF CARD11 [40, 41]; AD GOF IKBKB [42]; SKIV2L [43]; NFE2L2 [44]

Unknown cause of DiGeorge syndrome, unknown cause of CHARGE syndrome, unknown gene(s) within 10p13–14 deletion responsible for phenotype

EDA ectodermal dysplasia anhydrotic, HSV herpes simplex virus, VZV varicella zoster virus, BCG Bacillus Calmette-Guerin, NBS newborn screen, TREC T cell receptor excision circle (biomarker for low T cells used in NBS), IUGR interuterine growth retardation

Table 3.

Predominantly antibody deficiencies

Disease Genetic defect Inheritance OMIM Ig Associated features
1. Severe reduction in all serum immunoglobulin isotypes with profoundly decreased or absent B cells, agammaglobulinemia
  BTK deficiency, X-linked agammaglobulinemia (XLA) BTK XL 300300 All isotypes decreased in majority of patients, some patients have detectable immunoglobulins Severe bacterial infections, normal numbers of pro-B cells
  μ heavy chain deficiency IGHM AR 147020 All isotypes decreased Severe bacterial infections, normal numbers of pro-B cells
  λ5 deficiency IGLL1 AR 146770
  Igα deficiency CD79A AR 112205
  Igβ deficiency CD79B AR 147245
  BLNK deficiency BLNK AR 604515
  p110δ deficiency PIK3CD AR 602839 Severe bacterial infections; autoimmune complications (IBD)
  p85 deficiency PIK3R1 AR 615214 Severe bacterial infections, cytopenias, decreased or absent pro-B cells
  E47 transcription factor deficiency TCF3 AD 616941 Recurrent bacterial infections
TCF3 AR 147141 Severe, recurrent bacterial infections, failure to thrive
  SLC39A7 (ZIP7) deficiency SLC39A7 AR 601416 Early onset infections, blistering dermatosis, failure to thrive, thrombocytopenia
  Hoffman syndrome/TOP2B deficiency TOP2B AD 126431 Recurrent infections, facial dysmorphism, limb anomalies
2. Severe reduction in at least 2 serum immunoglobulin isotypes with normal or low number of B cells, CVID phenotype
  Common variable immune deficiency with no gene defect specified (CVID) Unknown Variable Low IgG and IgA and/or IgM Clinical phenotypes vary: most have recurrent infections, some have polyclonal lymphoproliferation, autoimmune cytopenias and/or granulomatous disease
  Activated p110δ syndrome (APDS) PIK3CD GOF AD 615513 (APDS1) Normal/increased IgM, reduced IgG and IgA Severe bacterial infections; reduced memory B cells and increased transitional B cells, EBV ± CMV viremia, lymphadenopathy/splenomegaly, autoimmunity, lymphoproliferation, lymphoma
PIK3R1 AD 616005 (APDS2) Severe bacterial infections, reduced memory B cells and increased transitional B cells, lymphadenopathy/splenomegaly, lymphoproliferation, lymphoma; developmental delay
  PTEN deficiency (LOF) PTEN AD 158350 Normal/Decreased Recurrent infections, Lymphoproliferation, Autoimmunity; developmental delay
  CD19 deficiency CD19 AR 107265 Low IgG and IgA and/or IgM Recurrent infections, may have glomerulonephritis (CD81 mutation abolishes expression of CD19, thereby phenocopying CD19 mutations)
  CD81 deficiency CD81 AR 186845 Low IgG, low or normal IgA and IgM
  CD20 deficiency CD20 AR 112210 Low IgG, normal or elevated IgM and IgA Recurrent infections
  CD21 deficiency CD21 AR 120650 Low IgG, impaired anti-pneumococcal response Recurrent infections
  TACI deficiency# TNFRSF13B AR or AD 604907 Low IgG and IgA and/or IgM Variable clinical expression and penetrance for monoallelic variants
  BAFF receptor deficiency TNFRSF13C AR 606269 Low IgG and IgM, Variable clinical expression
  TWEAK deficiency TNFSF12 AD 602695 Low IgM and A, lack of anti-pneumococcal antibody Pneumonia, bacterial infections, warts, thrombocytopenia. Neutropenia
  TRNT1 deficiency TRNT1 AR 612907 B cell deficiency and hypogammaglobulinemia congenital sideroblastic anemia, deafness, developmental delay
  NFKB1 deficiency NFKB1 AD 164011 Normal or low IgG, IgA, IgM, low or normal B cells, low memory B cells Recurrent sinopulmonary infections, COPD, EBV proliferation, autoimmune cytopenias, alopecia and autoimmune thyroiditis
  NFKB2 deficiency NFKB2 AD 615577 Low serum IgG, A and M; low B cell numbers Recurrent sinopulmonary infections, alopecia and endocrinopathies
  IKAROS deficiency IKZF1 AD (haploinsufficiency) 603023 Low IgG, IgA, IgM, low or normal B cells; B cells and Ig levels reduce with age Decreased pro-B cells, recurrent sinopulmonary infections; increased risk of ALL, autoimmunity, CVID phenotype
  IRF2BP2 deficiency IRF2BP2 AD 615332 Hypogammaglobulinemia, absent IgA Recurrent infections, possible autoimmunity and inflammatory disease
  ATP6AP1 deficiency ATP6AP1 XL 300972 Variable immunoglobulin findings Hepatopathy, leukopenia, low copper
  ARHGEF1 deficiency ARHGEF1 AR 618459 Hypogammaglobulinemia; lack of antibody Recurrent infections, bronchiectasis
  SH3KBP1 (CIN85) deficiency SH3KBP1 XL 300310 IgM, IgG deficiency; loss of antibody Severe bacterial infections
  SEC61A1 deficiency SEC61A1 AD 609213 Hypogammaglobulinemia Severe recurrent respiratory tract infections
  RAC2 deficiency RAC2 AR 602049 Low IgG, IgA, IgM, low or normal B cells; reduced Ab responses following vaccination Recurrent sinopulmonary infections, selective IgA deficiency; poststreptococcal glomerulonephritis; urticaria
  Mannosyl-oligosaccharide glucosidase deficiency MOGS AR 601336 Low IgG, IgA, IgM, increased B cells; poor Ab responses following vaccination Bacterial and viral infections; severe neurologic disease; also known as congenital disorder of glycosylation type IIb (CDG-IIb)
3. Severe reduction in serum IgG and IgA with normal/elevated IgM and normal numbers of B cells, hyper IgM
  AID deficiency AICDA AR 6055258 IgG and IgA decreased, IgM increased; normal memory B cells but lacking somatic hypermutation Bacterial infections, enlarged lymph nodes and germinal centers; autoimmunity
AD 605257 IgG absent or decreased, IgA undetected, IgM increased; normal memory B cells with intact somatic hypermutation Bacterial infections, enlarged lymph nodes and germinal centers. Mutations uniquely localize to the nuclear export signal.
  UNG deficiency UNG AR 191525 IgG and IgA decreased, IgM increased Enlarged lymph nodes and germinal centers
  INO80 deficiency INO80 AR 610169 IgG and IgA decreased, IgM increased Severe bacterial infections
  MSH6 deficiency MSH6 AR 600678 Variable IgG, defects, increased IgM in some, normal B cells, low switched memory B cells, Ig class switch recombination and somatic hypermutation defects Family or personal history of cancer
4. Isotype, light chain, or functional deficiencies with generally normal numbers of B cells
  Ig heavy chain mutations and deletions Mutation or chromosomal deletion at 14q32 AR One or more IgG and/or IgA subclasses as well as IgE may be absent May be asymptomatic
  Kappa chain deficiency IGKC AR 147200 All immunoglobulins have lambda light chain Asymptomatic
  Isolated IgG subclass deficiency Unknown ? Reduction in one or more IgG subclass Usually asymptomatic, a minority may have poor antibody response to specific antigens and recurrent viral/bacterial infections
  IgG subclass deficiency with IgA deficiency Unknown ? Reduced IgA with decrease in one or more IgG subclass Recurrent bacterial infections
May be asymptomatic
  Selective IgA deficiency Unknown ? Absent IgA with other isotypes normal, normal subclasses and specific antibodies May be asymptomatic Bacterial infections, autoimmunity mildly increased
  Specific antibody deficiency with normal Ig levels and normal B cells Unknown ? Normal Reduced ability to produce antibodies to specific antigens
  Transient hypogammaglobulinemia of infancy Unknown ? IgG and IgA decreased Normal ability to produce antibodies to vaccine antigens, usually not associated with significant infections
  CARD11 GOF CARD11 AD GOF 616452 Polyclonal B cell lymphocytosis due to constitutive NF-κB activation Splenomegaly, lymphadenopathy, poor vaccine response
  Selective IgM deficiency Unknown ? Absent serum IgM Pneumococcal/bacterial

Common variable immunodeficiency disorders (CVID) include several clinical and laboratory phenotypes that may be caused by distinct genetic and/or environmental factors. Some patients with CVID and no known genetic defect have markedly reduced numbers of B cells as well as hypogammaglobulinemia. Identification of causal variants can assist in defining treatment. In addition to monogenic causes on this table, a small minority of patients with XLP (Table 4), WHIM syndrome (Table 6), ICF (Table 2), VODI (Table 2), thymoma with immunodeficiency (Good syndrome), or myelodysplasia are first seen by an immunologist because of recurrent infections, hypogammaglobulinemia, and normal or reduced numbers of B cells

Total number of disorders in Table 3: 46

Total number of mutant genes in Table 3: 39

New disorders: 9: AR PIK3CD [4648]; AR TCF3 [49, 50]; SLC39A7 [51]; TOP2B [52]; ARHGEF1 [53]; SH3KBP1 [54]; SEC61A1 [55]; AR LOF RAC2 [56]; AD AICDA

EBV Epstein-Barr virus, COPD chronic obstructive pulmonary disease

#Heterozygous variants in TNFRSF13B have been detected in healthy individuals, thus such variants are likely to be disease-modifying rather than disease-causing

Table 4.

Diseases of immune dysregulation

Disease Genetic defect Inheritance OMIM Circulating T cells Circulating B cells Functional defect Associated features
1. Familial hemophagocytic lymphohistiocytosis (FHL syndromes)
  Perforin deficiency (FHL2) PRF1 AR 170280 Increased activated T cells Normal Decreased to absent NK and CTL activities cytotoxicity Fever, HSM, hemophagocytic lymphohistiocytosis (HLH), cytopenias
  UNC13D/Munc13–4 deficiency (FHL3) UNC13D AR 608897 Increased activated T cells Normal Decreased to absent NK and CTL activities (cytotoxicity and/or degranulation) Fever, HSM, HLH, cytopenias,
  Syntaxin 11 deficiency (FHL4) STX11 AR 605014
  STXBP2/Munc18–2 deficiency (FHL5) STXBP2 AR or AD 601717
  FAAP24 deficiency FAAP24 AR 610884 Increased activated T cells Normal Failure to kill autologous EBV transformed B cells. Normal NK cell function EBV-driven lymphoproliferative disease
  SLC7A7 deficiency SLC7A7 AR 222700 Normal Normal

Hyper-inflammatory response of macrophages

Normal NK cell function

Lysinuric protein intolerance, bleeding tendency, alveolar proteinosis
2. FHL syndromes with hypopigmentation
  Chediak-Higashi syndrome LYST AR 606897 Increased activated T cells Normal Decreased NK and CTL activities (cytotoxicity and/or degranulation) Partial albinism, recurrent infections, fever, HSM, HLH, giant lysosomes, neutropenia, cytopenias, bleeding tendency, progressive neurological dysfunction
  Griscelli syndrome, type 2 RAB27A AR 603868 Normal Normal Decreased NK and CTL activities (cytotoxicity and/or degranulation) Partial albinism, fever, HSM, HLH, cytopenias
  Hermansky-Pudlak syndrome, type 2 AP3B1 AR 603401 Normal Normal Decreased NK and CTL activities (cytotoxicity and/or degranulation) Partial albinism, recurrent infections, pulmonary fibrosis, increased bleeding, neutropenia, HLH
  Hermansky-Pudlak syndrome, type 10 AP3D1 AR 617050 Normal Normal Decreased NK and CTL activities (cytotoxicity and/or degranulation) Oculocutaneous albinism, severe neutropenia, recurrent infections, seizures, hearing loss and neurodevelopmental delay
3. Regulatory T cell defects
  IPEX, immune dysregulation, polyendocrinopathy, enteropathy X-linked FOXP3 XL 300292 Normal Normal Lack of (and/or impaired function of) CD4+ CD25+ FOXP3+ regulatory T cells (Tregs) Autoimmune enteropathy, early onset diabetes, thyroiditis hemolytic anemia, thrombocytopenia, eczema, elevated IgE and IgA
  CD25 deficiency IL2RA AR 147730 Normal to decreased Normal No CD4 + C25+ cells with impaired function of Tregs cells Lymphoproliferation, autoimmunity, impaired T cell proliferation in vitro
  CD122 deficiency IL2RB AR 618495 Increased memory CD8 T cells, decreased Tregs Increased memory B cells Diminished IL2Rβ expression, dysregulated signaling in response to IL-2/IL-15; increased immature NK cells Lymphoproliferation, lymphadenopathy, hepatosplenomegaly, autoimmune hemolytic anemia, dermatitis, enteropathy, hypergammaglobulinemia, recurrent viral (EBV, CMV) infections
  CTLA4 haploinsufficiency (ALPS-V) CTLA4 AD 123890 Decreased Decreased Impaired function of Tregs. Autoimmune cytopenias, enteropathy, interstitial lung disease, extra-lymphoid lymphocytic infiltration, recurrent infections
  LRBA deficiency LRBA AR 606453 Normal or decreased CD4 numbers T cell dysregulation Low or normal numbers of B cells Reduced IgG and IgA in most Recurrent infections, inflammatory bowel disease, autoimmunity
  DEF6 deficiency DEF6 AR 610094 Mild CD4 and CD8 lymphopenia Low or normal numbers of B cells Impaired Treg function Enteropathy, hepatosplenomegaly, cardiomyopathy, recurrent infections
  STAT3 GOF mutation STAT3 AD GOF 102582 Decreased Decreased Enhanced STAT3 signaling, leading to increased Th17 cell differentiation, lymphoproliferation and autoimmunity. Decreased Tregs and impaired function Lymphoproliferation, solid organ autoimmunity, recurrent infections
  BACH2 deficiency BACH2 AD 605394 Progressive T cell lymphopenia Impaired memory B cell development Haploinsufficiency for a critical lineage specification transcription factor Lymphocytic colitis, sinopulmonary infections
  FERMT1 deficiency FERMT1 AR 173650 Normal Normal Intracellular accumulation of IgG, IgM, IgA, and C3 in colloid bodies under the basement membrane Dermatosis characterized by congenital blistering, skin atrophy, photosensitivity, skin fragility, and scaling
4. Autoimmunity with or without lymphoproliferation
  APECED (APS-1), autoimmune polyendocrinopathy with candidiasis and ectodermal dystrophy AIRE AR or AD 240300 Normal Normal AIRE serves as check-point in the thymus for negative selection of autoreactive T cells and for generation of Tregs Autoimmunity: hypoparathyroidism, hypothyroidism, adrenal insufficiency, diabetes, gonadal dysfunction and other endocrine abnormalities; dental enamel hypoplasia, alopecia areata enteropathy, pernicious anemia; chronic mucocutaneous candidiasis
  ITCH deficiency ITCH AR 606409 Not assessed Not assessed Itch deficiency may cause immune dysregulation by affecting both anergy induction in auto-reactive effector T cells and generation of Tregs Early-onset chronic lung disease (interstitial pneumonitis), autoimmunity (thyroiditis, type I diabetes, chronic diarrhea/enteropathy, and hepatitis), failure to thrive, developmental delay, dysmorphic facial features
  Tripeptidyl-peptidase II deficiency TPP2 AR 190470 Decreased Decreased TPP2 deficiency results in premature immunosenescence and immune dysregulation Variable lymphoproliferation, severe autoimmune cytopenias, hypergammaglobulinemia, recurrent infections
  JAK1 GOF JAK1 AD GOF 147795 Not assessed Not assessed Hyperactive JAK1 HSM, eosinophilia, eosinophilic enteritis, thyroid disease, poor growth, viral infections
  Prolidase deficiency PEPD AR 613230 Normal Normal Peptidase D Autoantibodies common, chronic skin ulcers, eczema, infections
5. Immune dysregulation with colitis
  IL-10 deficiency IL10 AR 124092 Normal Normal No functional IL-10 secretion Inflammatory bowel disease (IBD), folliculitis, recurrent respiratory diseases, arthritis,
  IL-10R deficiency IL10RA AR 146933 Normal Normal Leukocytes unresponsive to IL-10 IBD, folliculitis, recurrent respiratory diseases, arthritis, lymphoma
IL10RB AR 123889 Normal Normal Leukocytes unresponsive to IL-10, and IL-22, IL-26, IL-28A, IL-28B and IL-29
  NFAT5 haploinsufficiency NFAT5 AD 604708 Normal Normal Decreased memory B cells and plasmablasts IBD, recurrent sinopulmonary infections
  TGFB1 deficiency TGFB1 AR 618213 Normal Normal Decreased T cell proliferation in response to anti-CD3 IBD, immunodeficiency, recurrent viral infections, microcephaly, and encephalopathy
  RIPK1 RIPK1 AR 618108 Reduced Normal/reduced Reduced activation of MAPK, NFkB pathways to Recurrent infections, early-onset IBD, progressive polyarthritis
6. Autoimmune lymphoproliferative syndrome (ALPS, Canale-Smith syndrome)
  ALPS-FAS TNFRSF6

AD

AR

134637 Increased TCR α/β+CD4CD8 double negative (DN) T cells Normal, low memory B cells Apoptosis defect FAS mediated Splenomegaly, adenopathies, autoimmune cytopenias, increased lymphoma risk, IgG and A normal or increased, elevated serum FasL, IL-10, vitamin B12
  ALPS-FASLG TNFSF6 AR 134638 Increased DN T cells Normal Apoptosis defect FASL mediated Splenomegaly, adenopathies, autoimmune cytopenias, SLE, soluble FasL is not elevated
  ALPS-Caspase10 CASP10 AD 601762 Increased DN T cells Normal Defective lymphocyte apoptosis Adenopathies, splenomegaly, autoimmunity
  ALPS-Caspase 8 CASP8 AR 601763 Slightly increased DN T cells Normal Defective lymphocyte apoptosis and activation Adenopathies, splenomegaly, bacterial and viral infections, hypogammaglobulinemia
  FADD deficiency FADD AR 602457 Increased DN T cells Normal Defective lymphocyte apoptosis Functional hyposplenism, bacterial and viral infections, recurrent episodes of encephalopathy and liver dysfunction
7. Susceptibility to EBV and lymphoproliferative conditions
  SAP deficiency (XLP1) SH2D1A XL 300490 Normal or Increased activated T cells Reduced Memory B cells Reduced NK cell and CTL cytotoxic activity

Clinical and immunologic features triggered by EBV infection: HLH, Lymphoproliferation, Aplastic anemia, Lymphoma.

Hypogammaglobulinemia, Absent iNKT cells

  XIAP deficiency (XLP2) XIAP XL 300079 Normal or Increased activated T cells; low/normal iNK T cells Normal or reduced Memory B cells Increased T cells susceptibility to apoptosis to CD95 and enhanced activation-induced cell death (AICD)

EBV infection, Splenomegaly, lymphoproliferation

HLH, Colitis, IBD, hepatitis

Low iNKT cells

  CD27 deficiency CD27 AR 615122 Normal No memory B cells hypogammaglobulinemia; poor Ab responses to some vaccines/infections Features triggered by EBV infection, HLH, aplastic anemia, low iNKT cells, B-lymphoma
  CD70 deficiency CD70 AR 602840 Normal number, low Treg, poor activation and function Decreased memory B cells hypogammaglobulinemia; poor Ab responses to some vaccines/infections EBV susceptibility, Hodgkin lymphoma; autoimmunity in some patients
  CTPS1 deficiency CTPS1 AR 615897 Normal to low, but reduced activation, proliferation Decreased memory B cells Normal/high IgG poor proliferation to antigen Recurrent/chronic bacterial and viral infections (EBV, VZV), EBV lymphoproliferation, B cell non-Hodgkin lymphoma
  CD137 deficiency (41BB) TNFRSF9 AR 602250 Normal Normal Low IgG, low IgA, poor responses to T cell-dependent and T cell independent antigens, decreased T cell proliferation, IFNγ secretion, cytotoxicity EBV lymphoproliferation, B cell lymphoma, chronic active EBV infection
  RASGRP1 deficiency RASGRP1 AR 603962 Poor activation, proliferation, motility. Reduced naïve T cells Poor activation, proliferation, motility Normal IgM, IgG, increased IgA

Recurrent pneumonia, herpesvirus infections, EBV associated lymphoma

Decreased NK cell function

  RLTPR deficiency CARMIL2 AR 610859 Normal number, high CD4, increased naïve CD4+ and CD8+ T cells, low Treg and MAIT, poor CD28-induced function Normal B cell numbers, reduced memory B cells Normal to low, poor T dependent antibody response Recurrent bacterial, fungal and mycobacterial infections, viral warts, molluscum and EBV lymphoproliferative and other malignancy, atopy
  X-linked magnesium EBV and neoplasia (XMEN) MAGT1 XL 300853 Low CD4 Low recent thymic emigrant cels, inverted CD4/CD8 ratio, reduced MAIT cells, poor proliferation to CD3 Normal but decreased memory B cells

Progressive hypogammaglobulinemia

Reduced NK cell and CTL cytotoxic activity due to impaired expression of NKG2D

EBV infection, lymphoma, viral infections, respiratory and GI infections

Glycosylation defects

  PRKCD deficiency PRKCD AR 615559 Normal Low memory B cells, high CD5 B cells Apoptotic defect in B cells Recurrent infections, EBV chronic infection, lymphoproliferation, SLE-like autoimmunity (nephrotic and antiphospholipid syndromes), low IgG

Total number of disorders in Table 4: 44

Total number of mutant genes in Table 4: 45

New disorders: 8; SLC7A7 [57]; IL2RB [58, 59]; DEF6 [60]; FERMT1 [61]; TGFB1 [62]; RIPK1 [63, 64]; TNFRSF9 [46, 65, 66]; STAT5B AD DN []

FHL familial hemophagocytic lymphohistiocytosis, HLH hemophagocytic lymphohistiocytosis, HSM hepatosplenomegaly, DN double-negative, SLE systemic lupus erythematous, IBD Inflammatory bowel disease

Table 5.

Congenital defects of phagocyte number or function

Disease Genetic defect Inheritance OMIM Affected cells Affected function Associated features
1. Congenital neutropenias
  Elastase deficiency (Severe congential neutropenia [SCN] 1) ELANE AD 130130 N Myeloid differentiation

Susceptibility to MDS/leukemia

Severe congenital neutropenia or cyclic neutropenia

  GFI 1 deficiency (SCN2) GFI1 AD 600871 N Myeloid differentiation B/T lymphopenia
  HAX1 deficiency (Kostmann Disease) (SCN3) HAX1 AR 605998 N Myeloid differentiation Cognitive and neurological defects in patients with defects in both HAX1 isoforms, susceptibility to MDS/leukemia
  G6PC3 deficiency (SCN4) G6PC3 AR 611045 N Myeloid differentiation, chemotaxis, O2 production Structural heart defects, urogenital abnormalities, inner ear deafness, and venous angiectasias of trunks and limbs
  VPS45 deficiency (SCN5) VPS45 AR 610035 N Myeloid differentiation, migration Extramedullary hematopoiesis, bone marrow fibrosis, nephromegaly
  Glycogen storage disease type 1b G6PT1 AR 602671 N + M Myeloid differentiation, chemotaxis, O2 production Fasting hypoglycemia, lactic acidosis, hyperlipidemia, hepatomegaly
  X-linked neutropenia/myelodysplasia WAS XL GOF 300299 N Differentiation, mitosis. Results from GOF mutations in GTPase binding domain of WASp Neutropenia, myeloid maturation arrest, monocytopenia, variable lymphoid anomalies
  P14/LAMTOR2 deficiency LAMTOR2 AR 610389 N + M Endosomal biogenesis

Neutropenia

Hypogammaglobulinemia ↓CD8 cytotoxicity, partial albinism, growth failure

  Barth Syndrome (3-Methylglutaconic aciduria type II) TAZ XL 300394

N + L

Mel

Mitochondrial function Cardiomyopathy, myopathy, growth retardation, neutropenia
  Cohen syndrome VPS13B AR 607817 N Myeloid differentiation Dysmorphism, mental retardation, obesity, deafness, neutropenia
  Clericuzio syndrome (Poikiloderma with neutropenia) USB1 AR 613276 N Myeloid differentiation Retinopathy, developmental delay, facial dysmorphisms, poikiloderma
  JAGN1 deficiency JAGN1 AR 616012 N Myeloid differentiation Myeloid maturation arrest, osteopenia
  3-Methylglutaconic aciduria CLPB AR 616254 N

Myeloid differentiation

Mitochondrial protein

Neurocognitive developmental aberrations, microcephaly, hypoglycemia, hypotonia, ataxia, seizures, cataracts, IUGR
  G-CSF receptor deficiency CSF3R AR 138971 N Stress granulopoiesis disturbed
  SMARCD2 deficiency SMARCD2 AR 601736 N Chromatin remodeling, Myeloid differentiation and neutrophil functional defect Neutropenia, developmental aberrations, bones, hematopoietic stem cells, myelodysplasia
  Specific granule deficiency CEBPE AR 189965 N Terminal maturation and global dysfunction Neutropenia, Neutrophils with bilobed nuclei
  Shwachman-Diamond Syndrome SBDS AR 607444 N Neutrophil maturation, chemotaxis, ribosomal biogenesis Pancytopenia, exocrine pancreatic insufficiency, chondrodysplasia
DNAJC21 AR 617052 N + HSC Pancytopenia, exocrine pancreatic insufficiency
EFL1 AR 617941 N + HSC
  HYOU1 deficiency HYOU1 AR 601746 N Unfolded protein response Hypoglycemia, inflammatory complications
  SRP54 deficiency SRP54 AD 604857 N Protein translocation to ER, myeloid differentiation and neutrophil functional defect Neutropenia, exocrine pancreatic insufficiency
2. Defects of motility
  Leukocyte adhesion deficiency type 1 (LAD1) ITGB2 AR 600065

N + M +

L + NK

Adherence, chemotaxis, endocytosis, T/NK cytotoxicity Delayed cord separation, skin ulcers, periodontitis, leukocytosis
  Leukocyte adhesion deficiency type 2 (LAD2) SLC35C1 AR 605881 N + M Rolling, chemotaxis Mild LAD type 1 features with hh-blood group, growth retardation, developmental delay
  Leukocyte adhesion deficiency type 3 (LAD3) FERMT3 AR 607901

N + M +

L + NK

Adherence, chemotaxis LAD type 1 plus bleeding tendency
  Rac2 deficiency RAC2 AD LOF 608203 N

Adherence, chemotaxis

O2− production

Poor wound healing, leukocytosis
  β actin deficiency ACTB AD 102630 N + M Motility Mental retardation, short stature
  Localized juvenile periodontitis FPR1 AR 136537 N Formylpeptide induced chemotaxis Periodontitis only
  Papillon-Lefèvre syndrome CTSC AR 602365 N + M Chemotaxis Periodontitis, palmoplantar hyperkeratosis in some patients
  WDR1 deficiency WDR1 AR 604734 N Spreading, survival, chemotaxis Mild neutropenia, poor wound healing, severe stomatitis, neutrophil nuclei herniate
  Cystic fibrosis CFTR AR 602421 M only Chemotaxis Respiratory infections, pancreatic insufficiency, elevated sweat chloride
  Neutropenia with combined immune deficiency due to MKL1 deficiency MKL1 AR 606078 N + M + L + NK Impaired expression of cytoskeletal genes Mild thrombocytopenia
3. Defects of respiratory burst
  X-linked chronic granulomatous disease (CGD), gp91phox CYBB XL 306400 N + M Killing (faulty O2− production)

Infections, autoinflammatory phenotype, IBD

McLeod phenotype in patients with deletions extending into the contiguous Kell locus

  Autosomal recessive CGD CYBA AR 608508 Infections, autoinflammatory phenotype
CYBC1 618334
NCF1 608512
NCF2 608515
NCF4 613960
  G6PD deficiency class I G6PD XL 305900 N Reduced O2− production Infections
4. Other non-lymphoid defects
GATA2 deficiency GATA2 AD 137295 Monocytes + peripheral DC Multi lineage cytopenias Susceptibility to mycobacteria, HPV, histoplasmosis, alveolar proteinosis, MDS/AML/CMML, lymphedema
Pulmonary alveolar proteinosis CSF2RA XL (Biallelic mutations in pseudo-autosomal gene) 300770 Alveolar macrophages GM-CSF signaling Alveolar proteinosis
CSFR2B AR 614370

Total number of disorders in Table 5: 34

Total number of mutant genes in Table 5: 41

New disorders: 3; SRP54 [67, 68]; DNAJC21 [69]; CYBC1 [70, 71]

Removed: Cyclic neutropenia was merged with elastase deficiency

MDS myelodysplastic syndrome, IUGR intrauterine growth retardation, LAD leukocyte adhesion deficiency, AML acute myelogenous leukemia, CMML chronic myelomonocytic leukemia, N neutrophil, M monocyte, MEL melanocyte, L lymphocyte, NK natural killer

Table 6.

Defects in intrinsic and innate immunity

Disease Genetic defect Inheritance OMIM Affected cells Affected function Associated features
1. Mendelian susceptibility to mycobacterial disease (MSMD)
  IL-12 and IL-23 receptor β1 chain deficiency IL12RB1 AR 601604 L + NK IFN-γ secretion Susceptibility to mycobacteria and Salmonella
  IL-12p40 (IL-12 and IL-23) deficiency IL12B AR 161561 M
  IL-12Rβ2 deficiency IL12RB2 AR 601642 L + NK
  IL-23R deficiency IL23R AR 607562 L + NK
  IFN-γ receptor 1 deficiency IFNGR1 AR 209950 M + L IFN-γ binding and signaling
AD 615978 M + L
  IFN-γ receptor 2 deficiency IFNGR2 AR 147569 M + L IFN-γ signaling
  STAT1 deficiency STAT1 AD LOF 614892 M + L
  Macrophage gp91 phox deficiency CYBB XL 300645 Macrophage only Killing (faulty O2− production) Isolated susceptibility to mycobacteria
  IRF8 deficiency IRF8 AD 614893 M + L Impaired development of cDCs and Th1* cells Susceptibility to mycobacteria
AR 226990 M Lack of circulating monocytes and DCs, reduced NK cell numbers and function reported in some patients Susceptibility to mycobacteria and multiple other infectious agents including EBV
  SPPL2a deficiency SPPL2A AR 608238 M + L Impaired development of cDCs and Th1* cells Susceptibility to mycobacteria and Salmonella
  Tyk2 deficiency TYK2 AR 611521 M + L Impaired cellular responses to IL-10, IL-12, IL-23, and type I IFNs Susceptibility to intracellular bacteria (mycobacteria, Salmonella), and viruses
  P1104A TYK2 homozygosity TYK2 AR 176941 L Impaired cellular responses to IL-23 MSMD or tuberculosis
  ISG15 deficiency ISG15 AR 147571 IFNγ production defect Susceptibility to mycobacteria (BCG), brain calcification
  RORγt deficiency RORC AR 602943 L + NK Lack of functional RORγT protein, IFNγ production defect, complete absence of IL-17A/F-producing T cells Susceptibility to mycobacteria and candida
  JAK1 deficiency JAK1 AR LOF 147795 N + L Reduced JAK1 activation to cytokines, Reduced IFNγ production Susceptibility to mycobacteria and viruses, urothelial carcinoma
2. Epidermodysplasia verruciformis (HPV)
  EVER1 deficiency TMC6 AR 605828 Keratinocytes EVER1, EVER2 and CIB1 form a complex in keratinocytes Human papillomavirus (HPV) (group B1) infections and cancer of the skin (typical EV)
  EVER2 deficiency TMC8 605829
  CIB1 deficiency CIB1 618267
  WHIM (warts, hypogammaglobulinemia, infections, myelokathexis) syndrome CXCR4 AD GOF 162643 Leukocytes Increased response of the CXCR4 chemokine receptor to its ligand CXCL12 (SDF-1) Warts (HPV) infection, neutropenia, low B cell number, hypogammaglobulinemia
3. Predisposition to severe viral infection
  STAT1 deficiency STAT1 AR LOF 600555 Leukocytes and other cells

STAT1-dependent

IFN-α/β, γ and λ responses

Severe viral infections, mycobacterial infection
  STAT2 deficiency STAT2 AR 600556 Leukocytes and other cells

STAT2-dependent

IFN-α/β and λ response

Severe viral infections (disseminated vaccine-strain measles)
  IRF9 deficiency IRF9 AR 147574* Leukocytes and other cells IRF9- and ISGF3-dependent IFN-α/β and λ responses Severe influenza disease
  IRF7 deficiency IRF7 AR 605047 Leukocytes, plasmacytoid dendritic cells, non-hematopoietic cells IFN-α, β and γ production and IFN-λ production
  IFNAR1 deficiency IFNAR1 AR 107450* Leukocytes and other cells IFNAR1-dependent responses to IFN-α/β Severe disease caused by Yellow Fever vaccine and Measles vaccine
  IFNAR2 deficiency IFNAR2 AR 602376 Broadly expressed IFNAR2-dependent responses to IFN-α/β Severe viral infections (disseminated vaccine-strain measles, HHV6)
  CD16 deficiency FCGR3A AR 146740 NK cells Altered NK cells function Severe herpes viral infections, particularly VZV, Epstein-Barr virus (EBV), and (HPV)
  MDA5 deficiency IFIH1 AR LOF 606951 Broadly expressed Viral recognition and IFN induction Rhinovirus and other RNA viruses
  RNA polymerase III deficiency POLR3A AD 614258 Leukocytes and other cells Impaired viral recognition and IFN induction in response to VZV or poly I:C Severe VZV infection
POLR3C AD 617454
POLR3F AD 617455
4. Herpes simplex encephalitis (HSE)
  TLR3 deficiency TLR3 AD 613002 Central nervous system (CNS) resident cells and fibroblasts TLR3-dependent IFN-α, β and γ response Herpes simplex virus 1 encephalitis (incomplete clinical penetrance for all etiologies listed here); severe pulmonary influenza; VZV
AR
  UNC93B1 deficiency UNC93B1 AR 608204 UNC-93B-dependent IFN-α, β and γ response Herpes simplex virus 1 encephalitis
  TRAF3 deficiency TRAF3 AD 601896 TRAF3-dependent IFN-α, β and γ response
  TRIF deficiency TICAM1 AD 607601 TRIF-dependent IFN-α, β and γ response
AR
  TBK1 deficiency TBK1 AD 604834

TBK1-dependent

IFN-α, β and γ response

  IRF3 deficiency IRF3 AD 616532 Low IFN-α/β production in response to HSV1 and decreased IRF3 phosphorylation
  DBR1 deficiency DBR1 AR 607024 Impaired production of anti-viral IFNs HSE of the brainstem. Other viral infections of the brainstem.
5. Predisposition to invasive fungal diseases
CARD9 deficiency CARD9 AR 607212 Mononuclear phagocytes CARD9 signaling pathway Invasive candidiasis infection, deep dermatophytoses, other invasive fungal infections
6. Predisposition to mucocutaneous candidiasis
  IL-17RA deficiency IL17RA AR 605461 Epithelial cells, fibroblasts, mononuclear phagocytes IL-17RA signaling pathway CMC, folliculitis
  IL-17RC deficiency IL17RC AR 610925 IL-17RC signaling pathway CMC
  IL-17F deficiency IL17F AD 606496 T cells IL-17F-containing dimers CMC, folliculitis
  STAT1 GOF STAT1 AD GOF 600555 T cells, B cells, monocytes Gain-of-function STAT1 mutations that impair the development of IL-17-producing T cells CMC, various fungal, bacterial and viral (HSV) infections, auto-immunity (thyroiditis, diabetes, cytopenias), enteropathy
  ACT1 deficiency TRAF3IP2 AR 607043 T cells, fibroblasts Fibroblasts fail to respond to IL-17A and IL-17F, and their T cells to IL-17E CMC, blepharitis, folliculitis, and macroglossia
7. TLR signaling pathway deficiency with bacterial susceptibility
  IRAK4 deficiency IRAK4 AR 606883 Lymphocytes + granulocytes+ monocytes TIR-IRAK4 signaling pathway Bacterial infections (pyogens)
  MyD88 deficiency MYD88 AR 602170 Lymphocytes + granulocytes + monocytes TIR-MyD88 signaling pathway
  IRAK1 deficiency IRAK1 XL 300283 Lymphocytes + granulocytes + monocytes TIR-IRAK1 signaling pathway Bacterial infections, X-linked MECP2 deficiency-related syndrome due to a large de novo Xq28 chromosomal deletion encompassing both MECP2 and IRAK1
  TIRAP deficiency TIRAP AR 614382 Lymphocytes + granulocytes + monocytes TIRAP- signaling pathway, TLR1/2, TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes Staphylococcal disease during childhood
8. Other inborn errors of immunity related to non-hematopoietic tissues
  Isolated congenital asplenia (ICA) RPSA AD 271400 No spleen RPSA encodes ribosomal protein SA, a component of the small subunit of the ribosome Bacteremia (encapsulated bacteria)
HMOX AR 141250 Macrophages HO-1 regulates iron recycling and heme-dependent damage occurs Hemolysis, nephritis, inflammation
  Trypanosomiasis APOL1 AD 603743 Somatic Pore forming serum protein Trypanosomiasis
  Acute liver failure due to NBAS deficiency NBAS AR 608025 Somatic and hematopoietic ER stress Fever induces liver failure

  Acute necrotizing encephalopathy

  Osteopetrosis

RANBP2 AR 601181 Ubiquitous expression Nuclear pore Fever induces acute encephalopathy
CLCN7 AR 602727 Osteoclasts Secretory lysosomes Osteopetrosis with hypocalcemia, neurologic features
SNX10 AR 614780 Osteopetrosis with visual impairment
OSTM1 AR 607649 Osteopetrosis with hypocalcemia, neurologic features
PLEKHM1 AR 611466 Osteopetrosis
TCIRG1 AR 604592 Osteopetrosis with hypocalcemia
TNFRSF11A AR 603499 Osteoclastogenesis Osteopetrosis
TNFSF11 AR 602642 Stromal Osteoclastogenesis Osteopetrosis with severe growth retardation
  Hidradenitis suppurativa NCSTN AD 605254 Epidermis Notch signaling/gamma-secretase in hair follicle regulates keratinization Verneuil’s disease/Hidradenitis suppurativa with acne
PSEN AD 613737 Verneuil’s disease/Hidradenitis suppurative with cutaneous hyperpigmentation
PSENEN AD 613736 Verneuil’s disease/Hidradenitis suppurativa
9. Other inborn errors of immunity related to leukocytes
  IRF4 haploinsufficiency IRF4 AD 601900 L + M IRF4 is a pleiotropic transcription factor Whipple’s disease
  IL-18BP deficiency IL18BP AR 604113 Leukocytes and other cells IL-18BP neutralizes secreted IL-18 Fulminant viral hepatitis

Total number of disorders in Table 6: 53

Total number of mutant genes in Table 6: 64

New genes: 13, IL12RB2 [72]; IL23R [72]; SPPL2A [73]; TYK2 P1104A allele [10]; CIB1 [74]; IRF9 [75]; IFNAR1 [76]; POLR3A [77]; POLR3C [77]; POLR3F [78]; DBR1 [79]; IRF4 [80]; IL18BP [81]

NF-κB nuclear factor kappa B, TIR Toll and Interleukin 1 receptor, IFN interferon, TLR Toll-like receptor, MDC myeloid dendritic cell, CNS central nervous system, CMC chronic mucocutaneous candidiasis, HPV human papillomavirus, VZV varicella zoster virus, EBV, Epstein-Barr virus

Table 7.

Autoinflammatory disorders

Disease Genetic defect Inheritance OMIM T cells B cells Functional defect Associated features
1. Type 1 interferonopathies
  STING-associated vasculopathy, infantile-onset (SAVI) TMEM173 AR 612374 Not assessed Not assessed STING activates both the NF-kappa-B and IRF3 transcription pathways to induce expression of IFN Skin vasculopathy, inflammatory lung disease, systemic autoinflammation and ICC, FCL
  ADA2 deficiency ADA2 AR 607575 Not assessed Not assessed ADAs deactivate extracellular adenosine and terminate signaling through adenosine receptors Polyarteritis nodosa, childhood-onset, early-onset recurrent ischemic stroke and fever; some patients develop hypogammaglobulinemia
  TREX1 deficiency, Aicardi-Goutieres syndrome 1 (AGS1) TREX1 AR 606609 Not assessed Not assessed Intracellular accumulation of abnormal ss DNA species leading to increased type I IFN production Classical AGS, SLE, FCL
  RNASEH2B deficiency, AGS2 RNASEH2B AR 610326 Not assessed Not assessed Intracellular accumulation of abnormal RNA-DNA hybrid species leading to increased type I IFN production Classical AGS, SP
  RNASEH2C deficiency, AGS3 RNASEH2C AR 610330 Not assessed Not assessed Classical AGS
  RNASEH2A deficiency, AGS4 RNASEH2A AR 606034 Not assessed Not assessed Classical AGS
  SAMHD1 deficiency, AGS5 SAMHD1 AR 606754 Not assessed Not assessed Controls dNTPs in the cytosol, failure of which leads to increased type I IFN production Classical AGS, FCL
  ADAR1 deficiency, AGS6 ADAR1 AR 146920 Not assessed Not assessed Catalyzes the deamination of adenosine to inosine in dsRNA substrates, failure of which leads to increased type I IFN production Classical AGS, BSN, SP
  Aicardi-Goutieres syndrome 7 (AGS7) IFIH1 AD GOF 615846 Not assessed Not assessed IFIH1 gene encodes a cytoplasmic viral RNA receptor that activates type I interferon signaling through the MAVS adaptor molecule Classical AGS, SLE, SP, SMS
  DNAse II deficiency DNASE2 AR 126350 Not assessed Not assessed DNAse II degrades and eliminates DNA. Loss of DNase II activity induces type I interferon signaling AGS
  Pediatric systemic lupus erythematosus due to DNASE1L3 deficiency DNASE1L3 AR 614420 DNASE1L3 is an endonuclease that degrades extracellular DNA. DNASE1L3 deficiency decreases clearance of apoptotic cells Very early onset SLE, reduced complement levels, autoantibodies (dsDNA, ANCA), lupus nephritis, hypocomplementemic urticarial vasculitis syndrome
  Spondyloenchondro-dysplasia with immune dysregulation (SPENCD) ACP5 AR 171640 Not assessed Not assessed Upregulation of IFN through mechanism possibly relating to pDCS Short stature, SP, ICC, SLE, thrombocytopenia and autoimmune hemolytic anemia, possibly recurrent bacterial and viral infections
  X-linked reticulate pigmentary disorder POLA1 XL 301220 Not assessed Not assessed POLA1 is required for synthesis of cytosolic RNA:DNA and its deficiency leads to increase production of type I interferon Hyperpigmentation, characteristic facies, lung and GI involvement
  USP18 deficiency USP18 AR 607057 Not assessed Not assessed Defective negative regulation of ISG15 leading to increased IFN TORCH-like syndrome
  OAS1 deficiency OAS1 AD GOF 164350 Low Increased interferon from recognition of RNA Pulmonary alveolar proteinosis, skin rash
2. Defects affecting the inflammasome
  Familial Mediterranean fever MEFV AR LOF 249100 Mature granulocytes, cytokine-activated monocytes. Increased inflammasome-mediated induction of IL1β. Recurrent fever, serositis and inflammation responsive to colchicine. Predisposes to vasculitis and inflammatory bowel disease.
AD 134610 Mature granulocytes, cytokine-activated monocytes. Usually M694del variant.
  Mevalonate kinase deficiency (Hyper IgD syndrome) MVK AR 260920 Somatic and hemaotpoietic affecting cholesterol synthesis, pathogenesis of disease unclear Periodic fever and leukocytosis with high IgD levels
  Muckle-Wells syndrome NLRP3 AD GOF 191900 PMNs Monocytes Defect in cryopyrin, involved in leukocyte apoptosis and NFkB signaling and IL-1 processing Urticaria, SNHL, amyloidosis.
  Familial cold autoinflammatory syndrome 1 AD GOF 120100 PMNs, monocytes Non-pruritic urticaria, arthritis, chills, fever and leukocytosis after cold exposure.
  Neonatal onset multisystem inflammatory disease (NOMID) or chronic infantile neurologic cutaneous and articular syndrome (CINCA) AD GOF 607115 PMNs, chondrocytes Neonatal onset rash, chronic meningitis, and arthropathy with fever and inflammation.
  Familial cold autoinflammatory syndrome 2 NLRP12 AD GOF 611762 PMNs, monocytes Non-pruritic urticaria, arthritis, chills, fever and leukocytosis after cold exposure.
  NLRC4-MAS (macrophage activating syndrome) NLRC4 AD GOF 616050 PMNs monocytes macrophages Gain of function mutation in NLRC4 results in elevated secretion of IL-1β and IL-18 as well as macrophage activation Severe enterocolitis and macrophage activation syndrome
  Familial cold autoinflammatory syndrome 4 616115
  PLAID (PLCγ2 associated antibody deficiency and immune dysregulation) PLCG2 AD GOF 614878 B cells, NK, Mast cells Mutations activate IL-1 pathways Cold urticaria hypogammaglobulinemia, impaired humoral immunity, autoinflammation
  Familial cold autoinflammatory syndrome 3 or APLAID (c2120A > C) 614468
  NLRP1 deficiency NLRP1 AR 617388 leukocytes Systemic elevation of IL-18 and caspase 1, suggesting involvement of NLRP1 inflammasome Dyskeratosis, autoimmunity and arthritis
  NLRP1 GOF NLRP1 AD GOF 615225 Keratinocytes Increased IL1β Palmoplantar carcinoma, corneal scarring; recurrent respiratory papillomatosis
3. Non-inflammasome-related conditions
  TNF receptor-associated periodic syndrome (TRAPS) TNFRSF1A AD 142680 PMNs, monocytes Mutations of 55-kD TNF receptor leading to intracellular receptor retention or diminished soluble cytokine receptor available to bind TNF Recurrent fever, serositis, rash, and ocular or joint inflammation
  Pyogenic sterile arthritis, pyoderma gangrenosum, acne (PAPA) syndrome, hyperzincemia and hypercalprotectinemia PSTPIP1 AD 604416 Hematopoietic tissues, upregulated in activated T cells Disordered actin reorganization leading to compromised physiologic signaling during inflammatory response Destructive arthritis, inflammatory skin rash, myositis
  Blau syndrome NOD2 AD 186580 Monocytes Mutations in nucleotide binding site of CARD15, possibly disrupting interactions with lipopolysaccharides and NF-kB signaling Uveitis, granulomatous synovitis, camptodactyly, rash and cranial neuropathies, 30% develop Crohn colitis
  ADAM17 deficiency ADAM17 AR 614328 Leukocytes and epithelial cells Defective TNFα production Early onset diarrhea and skin lesions
  Chronic recurrent multifocal osteomyelitis and congenital dyserythropoietic anemia (Majeed syndrome) LPIN2 AR 609628 Neutrophils, bone marrow cells Undefined Chronic recurrent multifocal osteomyelitis, transfusion-dependent anemia, cutaneous inflammatory disorders
  DIRA (Deficiency of the Interleukin 1 Receptor Antagonist) IL1RN AR 612852 PMNs, Monocytes Mutations in the IL1 receptor antagonist allow unopposed action of Interleukin 1 Neonatal onset of sterile multifocal osteomyelitis, periostitis and pustulosis.
  DITRA (Deficiency of IL-36 receptor antagonist) IL36RN AR 614204 Keratinocytes, leukocytes Mutations in IL-36RN leads to increase IL-8 production Pustular psoriasis
  SLC29A3 mutation SLC29A3 AR 602782 Leukocytes, bone cells Hyperpigmentation hypertrichosis, histiocytosis-lymphadenopathy plus syndrome
  CAMPS (CARD14 mediated psoriasis) CARD14 AD 602723 Mainly in keratinocytes Mutations in CARD14 activate the NF-kB pathway and production of IL-8 Psoriasis
  Cherubism SH3BP2 AD 118400 Stroma cells, bone cells Hyperactived macrophage and increase NF-kB Bone degeneration in jaws
  CANDLE (chronic atypical neutrophilic dermatitis with lipodystrophy) PSMB8* AR and AD 256040 Keratinocytes, B cell adipose cells Mutations cause increased IFN signaling through an undefined mechanism Contractures, panniculitis, ICC, fevers
PSMG2 AR 609702 Lymphocytes Panniculitis, lipodystrophy, autoimmune hemolytic anemia
  COPA defect COPA AD 6011924 PMN and tissue specific cells Defective intracellular transport via the coat protein complex I (COPI) Autoimmune inflammatory arthritis and interstitial lung disease with Th17 dysregulation and autoantibody production
  Otulipenia/ORAS OTULIN AR 615712 Leukocytes Increase LUBAC induction of NF-KB activation leading to high proinflamatory cytokines levels. Fever, diarrhea, dermatitis
  A20 deficiency TNFAIP3 AD 616744 Lymphocytes Defective inhibition of NF-KB signaling pathway Arthralgia, mucosal ulcers, ocular inflammation
  AP1S3 deficiency AP1S3 AR 615781 Keratinocytes Disrupted TLR3 translocation Pustular psoriasis
  ALPI deficiency ALPI AR 171740 Intestinal epithelial cells Deficient inhibition of LPS in intestine Inflammatory bowel disease
  TRIM22 TRIM22 AR 606559 Macrophages, intestinal epithelial cells Granulomatous colitis Inflammatory bowel disease
  T cell lymphoma subcutaneous panniculitis-like (TIM3 deficiency) HAVCR2 AR 618398 Leukocytes Increased inflammasome activity due to defective checkpoint signaling Panniculitis, HLH, polyclonal cutaneous T cell infiltrates or T cell lymphoma

Total number of disorders in Table 7: 45

Total number of mutant genes in Table 7: 42

New disorders: 9; DNASE2 [82]; DNASE1L3 [8385]; OAS1 [86]; AD MEFV; NLRP1 GOF [87, 88]; ALPI [89]; TRIM22 [90]; PSMG2 [91]; HAVCR2 [92, 93]

IFN interferon, HSM hepatosplenomegaly, CSF cerebrospinal fluid, SLE systemic lupus erythematosus, TORCH toxoplasmosis, other, rubella, cytomegalovirus, and herpes infections, SNHL sensorineural hearing loss, AGS Aicardi-Goutières syndrome, BSN bilateral striatal necrosis, FCL familial chilblain lupus, ICC intracranial calcification, IFN interferon type I, pDCs plasmacytoid dendritic cells, SP spastic paraparesis, SMS Singleton-Merten syndrome, ss single-stranded DNA

*Variants in PSMB4, PSMB9, PSMA3, and POMP have been proposed to cause a similar CANDLE phenotype in compound heterozygous monogenic (PSMB4), digenic (PSMA3/PSMB8, PSMB9/PSMB4, PSMB4/PSMB8) and AD monogenic (POMP) models [94]

Table 8.

Complement deficiencies

Disease Genetic defect Inheritance Gene OMIM Laboratory features Associated features
Complement deficiencies
  C1q deficiency due to defects C1QA AR 120550 Absent CH50 hemolytic activity, defective activation of the classical pathway, diminished clearance of apoptotic cells SLE, infections with encapsulated organisms
C1QB AR 120570
C1QC AR 120575
  C1r deficiency C1R AR 613785 Absent CH50 hemolytic activity, defective activation of the classical pathway SLE, infections with encapsulated organisms, Ehlers-Danlos phenotype
  C1r Periodontal Ehlers-Danlos C1R AD GOF 613785 Normal CH50 Hyperpigmentation, skin fragility
  C1s deficiency C1S AR 613785 Absent CH50 hemolytic activity, defective activation of the classical pathway SLE, infections with encapsulated organisms, Ehlers-Danlos phenotype
  C1s Periodontal Ehlers-Danlos C1S AD GOF 613785 Normal CH50 Hyperpigmentation, skin fragility
  Complete C4 deficiency C4A + C4B AR 120810 Absent CH50 hemolytic activity, defective activation of the classical pathway, complete deficiency requires biallelic mutations/deletions/conversions of both C4A and C4B SLE, infections with encapsulated organisms, partial deficiency is common (either C4A or C4B) and appears to have a modest effect on host defense
  C2 deficiency C2 AR 217000 Absent CH50 hemolytic activity, defective activation of the classical pathway SLE, infections with encapsulated organisms, atherosclerosis
  C3 deficiency (LOF) C3 AR 120700 Absent CH50 and AH50 hemolytic activity, defective opsonization, defective humoral immune response Infections, glomerulonephritis, atypical hemolytic-uremic syndrome with GOF mutations.
  C3 GOF C3 AD GOF 120700 Increased activation of complement Atypical hemolytic-uremic syndrome
  C5 deficiency C5 AR 120900

Absent CH50 and AH50 hemolytic activity

Defective bactericidal activity

Disseminated neisserial infections
  C6 deficiency C6 AR 217050 Absent CH50 and AH50 hemolytic activity, defective bactericidal activity
  C7 deficiency C7 AR 217070
  C8α deficiency C8A AR 120950
  C8 γ deficiency C8G AR 120930
  C8 β deficiency C8B AR 120960
  C9 deficiency C9 AR 120940 Reduced CH50 and AP50 hemolytic activity, deficient bactericidal activity Mild susceptibility to disseminated neisserial infections
  MASP2 deficiency MASP2 AR 605102 Deficient activation of the lectin activation pathway Pyogenic infections, inflammatory lung disease, autoimmunity
  Ficolin 3 deficiency FCN3 AR 604973 Absence of complement activation by the Ficolin 3 pathway. Respiratory infections, abscesses
  C1 inhibitor deficiency SERPING1 AD 606860 Spontaneous activation of the complement pathway with consumption of C4/C2, spontaneous activation of the contact system with generation of bradykinin from high molecular weight kininogen Hereditary angioedema
  Factor B GOF CFB AD GOF 612924 Gain-of-function mutation with increased spontaneous AH50 Atypical hemolytic-uremic syndrome
  Factor B deficiency CFB AR 615561 Deficient activation of the alternative pathway Infections with encapsulated organisms
  Factor D deficiency CFD AR 134350 Absent AH50 hemolytic activity Neisserial infections
  Properdin deficiency CFP XL 300383 Absent AH50 hemolytic activity Neisserial infections
  Factor I deficiency CFI AR 217030 Spontaneous activation of the alternative complement pathway with consumption of C3 Infections, disseminated neisserial infections, atypical Hemolytic-uremic syndrome, preeclampsia
  Factor H deficiency CFH AR or AD 134370 Spontaneous activation of the alternative complement pathway with consumption of C3
  Factor H-related protein deficiencies CFHR1 AR or AD 134371, Normal CH50, AH50, autoantibodies to Factor H., linked deletions of one or more CFHR genes leads to susceptibility autoantibody-mediated aHUS Older onset atypical hemolytic-uremic syndrome, disseminated neisserial infections
CFHR2 600889,
CFHR3 605336,
CFHR4 605337,
CFHR5 608593
  Thrombomodulin deficiency THBD AD 188040 Normal CH50, AH50 Atypical hemolytic-uremic syndrome
  Membrane Cofactor Protein (CD46) deficiency CD46 AD 120920 Inhibitor of complement alternate pathway, decreased C3b binding Atypical hemolytic-uremic syndrome, infections, preeclampsia
  Membrane Attack Complex Inhibitor (CD59) deficiency CD59 AR 107271 Erythrocytes highly susceptible to complement-mediated lysis Hemolytic anemia, polyneuropathy
  CD55 deficiency (CHAPEL disease) CD55 AR 125240 Hyperactivation of complement on endothelium Protein losing enteropathy, thrombosis

Total number of disorders in Table 8: 30

Total number of mutant genes in Table 8: 36

New disorders: 2; C1S AD GOF [95], C1R AD GOF [95]

MAC membrane attack complex, SLE systemic lupus erythematosus

Table 10.

Phenocopies of inborn errors of immunity

Disease Genetic defect/presumed pathogenesis Circulating T cells Circulating B cells Serum Ig Associated features/similar PID
1. Phenocopies of inborn errors of immunity
Associated with somatic mutations
  Autoimmune lymphoproliferative syndrome (ALPS–SFAS) Somatic mutation in TNFRSF6 Increased CD4CD8double negative (DN) αβ T cells Normal, but increased number of CD5+ B cells Normal or increased Splenomegaly, lymphadenopathy, autoimmune cytopenias, Defective lymphocyte apoptosis/ALPS–FAS (=ALPS type Im)
  RAS-associated autoimmune leukoproliferative disease (RALD) Somatic mutation in KRAS (GOF) Normal B cell lymphocytosis Normal or increased Splenomegaly, lymphadenopathy, autoimmune cytopenias, granulocytosis, monocytosis/ALPS-like
  RAS-associated autoimmune leukoproliferative disease (RALD) Somatic mutation in NRAS (GOF) Increased CD4−CD8− double negative (DN) T alpha/beta cells Lymphocytosis Normal or increased Splenomegaly, lymphadenopathy, autoantibodies/ALPS-like
  Cryopyrinopathy, (Muckle-Wells/CINCA/NOMID-like syndrome) Somatic mutation in NLRP3 Normal Normal Normal Urticaria-like rash, arthropathy, neurological signs
  Hypereosinophilic syndrome due to somatic mutations in STAT5b Somatic mutation in STAT5B (GOF) Normal Normal Normal Eosinophilia, atopic dermatitis, urticarial rash, diarrhea
Associated with autoantibodies
  Chronic mucocutaneous candidiasis AutoAb to IL-17 and/or IL-22 Normal Normal Normal Endocrinopathy, chronic mucocutaneous candidiasis/CMC
  Adult-onset immunodeficiency with susceptibility to mycobacteria AutoAb to IFNγ Decreased naive T cells Normal Normal Mycobacterial, fungal, Salmonella VZV infections/MSMD, or CID
  Recurrent skin infection AutoAb to IL-6 Normal Normal Normal Staphylococcal infections/STAT3 deficiency
  Pulmonary alveolar proteinosis AutoAb to GM-CSF Normal Normal Normal Pulmonary alveolar proteinosis, cryptococcal meningitis, disseminated nocardiosis/CSF2RA deficiency
  Acquired angioedema AutoAb to CI inhibitor Normal Normal Normal Angioedema/C1 INH deficiency (hereditary angioedema)
  Atypical hemolytic uremic syndrome AutoAb to Complement Factor H Normal Normal Normal aHUS = Spontaneous activation of the alternative complement pathway
  Thymoma with hypogammaglobulinemia (Good syndrome) AutoAb to various cytokines Increased CD8+ T cells No B cells Decreased Invasive bacterial, viral or opportunistic infections, autoimmunity, PRCA, lichen planus, cytopenia, colitis, chronic diarrhea

aHUS atypical hemolytic uremic syndrome, XL X-linked inheritance, AR autosomal recessive inheritance, AD autosomal dominant inheritance, LOF loss-of-function, GOF gain-of-function, PRCA pure red cell aplasia

Total number of conditions for Table 10: 12

Table 9.

Bone marrow failure

Disease Genetic defect Inheritance Gene OMIM T cells B cells Other affected cells Associated features Major Category Subcategory
Bone marrow failure
Fanconi anemia type A FANCA AR 227650 Normal to low Normal to low HSC Normal to low NK, CNS, skeletal, skin, cardiac, GI, urogenital anomalies, increased chromosomal breakage Bone marrow failure with immune deficiency Fanconi Anemia
Fanconi anemia type B FANCB XLR 300514
Fanconi anemia type C FANCC AR 227645
Fanconi anemia type D1 BRCA2 AR 605724
Fanconi anemia type D2 FANCD2 AR 227646
Fanconi anemia type E FANCE AR 600901
Fanconi anemia type F FANCF AR 603467
Fanconi anemia type G XRCC9 AR 614082
Fanconi anemia type I FANCI AR 609053
Fanconi anemia type J BRIP1 AR 609054
Fanconi anemia type L FANCL AR 614083
Fanconi anemia type M FANCM AR 618096
Fanconi anemia type N PALB2 AR 610832
Fanconi anemia type O RAD51C AR 613390
Fanconi anemia type P SLX4 AR 613951
Fanconi anemia type Q ERCC4 AR 615272
Fanconi anemia type R RAD51 AR 617244
Fanconi anemia type S BRCA1 AR 617883
Fanconi anemia type T UBE2T AR 616435
Fanconi anemia type U XRCC2 AR 617247
Fanconi anemia type V MAD2L2 AR 617243
Fanconi anemia type W RFWD3 AR 617784
MIRAGE (myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, enteropathy) SAMD9 AD GOF 617053 Not reported Not reported HSC, myeloid cells Intrauterine growth retardation, gonadal abnormalities, adrenal failure, MDS with chromosome 7 aberrations, predisposition to infections, enteropathy, absent spleen
Ataxia pancytopenia syndrome SAMD9L AD GOF 611170 Normal Low HSC, myeloid cells MDS, neurological features
DKCX1 DKC1 XL 305000 Normal to low Normal to low HSC Bone marrow failure, pulmonary and hepatic fibrosis, nail dystrophy, leukoplakia, reticulate skin pigmentation; microcephaly, neurodevelopmental delay Dyskeratosis Congenita
DKCA1 TERC AD 127550
DKCA2 TERT AD 187270
DKCA3 TINF2 AD 604319
DKCA4 RTEL1 AD 616373
DKCA5 TINF2 AD 268130
DKCA6 ACD AD 616553
DKCB1 NOLA3 AR 224230
DKCB2 NOLA2 AR 613987
DKCB3 WRAP53 AR 613988
DKCB4 TERT AR 613989
DKCB5 RTEL1 AR 615190 Low Nail dystrophy, leukoplakia, bone marrow failure, severe B cell immunodeficiency, intrauterine growth retardation, growth retardation, microcephaly, cerebellar hypoplasia, and esophageal dysfunction
DKCB6 PARN AR 616353 Normal to low Developmental delay, microcephaly, and cerebellar hypoplasia
DKCB7 ACD AR 616553 Normal to low Bone marrow failure, pulmonary and hepatic fibrosis, nail dystrophy, leukoplakia, reticulate skin pigmentation; microcephaly, neurodevelopmental delay
BMFS1 (SRP72-deficiency) SRP72 AD 602122 NA NA Bone marrow failure and congenital nerve deafness
BMFS2 ERCC6L2 AR 615667 NA NA Bone marrow failure, learning difficulties, microcephaly
BMFS5 TP53 AD 618165 NA Low B Erythroid hypoplasia, B cell deficiency
Coats plus syndrome STN1 AR 613129 Normal Normal Intrauterine growth retardation, premature aging, pancytopenia, hypocellular bone marrow, gastrointestinal hemorrhage due to vascular ectasia, intracranial calcification, abnormal telomeres
CTC1 AR 617053 Not reported Not reported

Total number of disorders in Table 9: 43

Total number of mutant genes in Table 9: 43

HSC hematopoietic stem cell, NK natural killer, CNS central nervous system, GI gastrointestinal, MDS myelodysplastic syndrome, DKCX X-inked dyskeratosis congenital, DKCA autosomal dominant dyskeratosis congenita, DKCB autosomal recessive dyskeratosis congenita, BMFS bone marrow failure syndrome

The advances in our understanding of clinical immunology continue to expand at a vast and remarkable rate, with the addition in this update of many—64, distributed across all tables (Fig. 1b)—novel genetic defects underlying inborn errors of immunity. Perhaps not surprisingly, most if not all of these new variants were identified by NGS, thus highlighting that whole exome/whole genome sequencing has become the gold standard for identifying novel pathogenic gene variants [68]. Indeed, since the first application of NGS to identify novel inborn errors of immunity was published in 2010 [18], ~ 45% of all currently known disease-causing variants have been discovered by whole exome/genome sequencing. Thus, a typical approach to identifying a pathogenic variant in a new patient might now consist of first sequencing a phenotype-driven panel of genes and advancing to whole exome/genome sequencing if the cause of disease remains elusive.

In this update, we increase the list of immunological diseases to 404, with 430 known genetic defects identified as causing these conditions. The unbiased application of NGS to the discovery and characterization of novel inborn errors of immunity continues to inform clinical and basic immunology. Thus, additional phenotypes have been identified for conditions resulting from variants in known and novel genes; the penetrance of genetic variants on clinical phenotypes has been shown to be highly variable; and clinical entities sharing common phenotypes have been discovered. For example, this update includes the findings that bi-allelic mutations in ZNF341 [19, 20], IL6ST (encoding gp130, a common component of the receptors for IL-6, IL-11, IL-27, LIF, OSM, CNTF) [21, 22], or IL6R [23, 24] all cause conditions that resemble autosomal dominant hyper-IgE syndrome due to dominant negative mutations in STAT3 [15]. Detailed analyses of these patients revealed a novel mechanism of regulating STAT3 signaling (via the transcription factor ZNF341) and defined the exact consequences of impaired IL-6/IL-6R/gp130 and putatively IL-11/IL-11R/gp130 signaling to the phenotype of AD-HIES.

Furthermore, key findings over the past 2 years continue to reveal that distinct mechanisms of disease (GOF, LOF, dominant negative, haploinsufficient), as well as different modes of inheritance (autosomal recessive, autosomal dominant) of variants in the same gene can cause disparate clinical conditions. This is a fascinating aspect of the genetics of human disease, and a salient reminder to be cognizant of the nature of the genetic variants identified from NGS. It is these genes that have several entries in this update. A few recent examples include:

  1. Heterozygous variants in CARD11 [25, 26] or STAT5B [27] can be pathogenic due to negative dominance. This was potentially unexpected because autosomal recessive LOF variants in both of these genes were previously reported to cause combined immunodeficiency and severe immune dysregulation, respectively, yet heterozygous relatives of these affected individuals were healthy [28, 29].

  2. While heterozygous dominant negative mutations in TCF3, encoding the transcription factor E47, cause B cell deficiency and agammaglobulinemia [30], nonsense mutations in TCF3 have now been identified that are pathogenic only in an autosomal recessive state, as heterozygous carriers of these particular allelic variants remained healthy [31, 32].

  3. A heterozygous hypermorphic variant in IKBKB was found to cause a combined immunodeficiency [33] not too dissimilar to the original description of bi-allelic, recessive variants in IKBKB [34]. Similarly, bi-allelic LOF mutations in PIK3CD are now known to cause B cell deficiency and agammaglobulinemia [3537], which is quite distinct from the immune dysregulated state of individuals with monoallelic activating PIK3CD mutations [1, 37]. This observation nicely parallels the earlier findings of either homozygous or heterozygous mutations in PIK3R1 that clinically phenocopy recessive or activating mutations in PIK3CD respectively [1, 37].

  4. Distinct diseases can result from heterozygous mutations in IKZF1 (Ikaros): combined immunodeficiency due to dominant negative alleles [38] or CVID due to haploinsufficiency [39].

  5. Similar to STAT1 [40], variants in RAC2 [4145] or CARD11 [25, 26, 28] can be pathogenic either as monoallelic GOF or LOF or bi-allelic recessive LOF.

Thus, these findings have revealed the fundamental importance of elucidating the impact of a novel variant on the function of the encoded protein and thus the mechanism of pathogenicity. Furthermore, these new entries are an important reminder not to overlook the potential significance of identifying heterozygous variants in genes previously believed to cause disease only in a biallelic manner or to result in a previously defined specific clinical entity. Indeed, there are now at least 35 genes that have multiple entries in the current update, reflecting the distinct mechanisms by which variants result in or cause disease (e.g., STAT1, STAT3, NLRP1, RAC2, ZAP70, CARD11, IKBKB, WAS, JAK1, IFIH1, C3, C1R, C1S–GOF or LOF; STAT5, STAT1, CARD11, ACD, CFH, CFHR1–5, FOXN1, RAC2, TCF3, AICDA, PIK3R1, IFNGR1, TREX1, TICAM1, IRF8–AD or AR; PIK3CD–AD GOF, AR LOF; IKZF1–AD, or haploinsufficient; NLRP3—distinct disease phenotypes despite all resulting from GOF alleles).

As noted above, genetic, biochemical, and functional analyses of putative novel pathogenic variants need to meet stringent criteria to be considered for inclusion in this update [17]. These criteria can make reporting genetic findings from single cases challenging, as often the best evidence that a novel variant is disease-causing is to identify additional, similarly affected but unrelated individuals with the same variants, or functionally similar variants in the same gene. While this can be challenging, particularly in light of the rarity of individual inborn errors of immunity, robust mechanistic laboratory investigations continue to provide compelling data from single patients, with or without evidence from animal models. Specifically, homozygous LOF mutations in IRF9 [46] and IL18BP [47] were identified and rigorously characterized in single patients and found to be the molecular cause of life-threatening influenza and fulminant viral hepatitis, respectively.

The study and discovery of novel inborn errors of immunity can also enable improved patient management by implementing gene-specific targeted therapies. Thus, JAK inhibitors are being used to treat disorders of immune dysregulation resulting from GOF mutations in JAK1, STAT1 or STAT3 [11], while mTOR inhibitors such as rapamycin or PI3K p110δ-specific inhibitors have been reported for the treatment of individuals with PIK3CD GOF or PIK3R1 LOF mutations [37]. Regarding novel gene defects, immune dysregulation due to DEF6 deficiency was successfully treated with abatacept (CTLA4-Ig) [48]. This correlated with impaired CTLA4 expression and function in DEF6-deficient T cells [48] and parallels the therapeutic use of abatacept and belatacept for LRBA-deficiency and CTLA4 haploinsufficiency, both of which are characterized by reduced CTLA4 expression in affected regulatory T cells [49, 50]. From a theoretical perspective, the finding that MSMD can be caused by mutations in IL12RB2, IL23R or SPPL2A and that these mutations are associated with impaired production of IFNγ—a requisite of anti-mycobacterial immunity—implies that IFNγ administration could be therapeutically beneficial in these clinical settings [51, 52]. Similarly, recombinant IL18BP could potentially ameliorate viral-induced liver toxicity due to IL18BP deficiency [47].

The goals of the IUIS Expert Committee on Inborn Errors of Immunity are to increase awareness, facilitate recognition, promote optimal treatment, and support research in the field of disorders of immunity. Thus, this 2019 Update and the accompanying “Phenotypical IUIS Classification” publications are intended as resources for clinicians and researchers. Importantly, these tables underpin the design of panels used for targeted gene sequencing to facilitate genetic diagnoses or inborn errors. In the past 5 years, the number of gene defects underlying inborn errors of immunity has nearly doubled from ~ 250 to 430 (Fig. 1a). The human genome contains 1800–2000 genes that are known to be involved in immune responses [13]. Thus, the discovery and study of inborn errors of immunity has elegantly illustrated that > 20% of these immune genes play non-redundant roles in host defense and immune regulation. With the improved identification and phenotyping of patients with rare diseases, combined with high throughput genome sequencing, the number of genes fundamentally required for immunity will no doubt continue to increase, further revealing critical and novel roles for specific genes, molecules, pathways and cell types in immune responses, as well as mechanisms of disease pathogenesis and targets for immunotherapies. The field of inborn errors of immunity, and the global clinical and research communities, will therefore continue to provide key insights into basic and clinical immunology.

Acknowledgments

The members of the Inborn Errors of Immunity committee would like to thanks the International Union of Immunological Societies (IUIS) for funding, as well as CSL Behring, Baxalta and Shire/Takeda for providing educational grants to enable us to compile this classification update.

Compliance with Ethical Standards

Conflict of Interest

The authors declare that they have no conflict of interest.

Footnotes

The original version of this article was revised: The in-text citations and the references were mismatched.

Publisher’s Note

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

Change history

2/22/2020

The original version of this article unfortunately contained mistakes in reference numbers. The in-text citations and the references were mismatched. The original article has been corrected.

References

  • 1.Picard C, Bobby Gaspar H, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, et al. International Union of Immunological Societies: 2017 primary immunodeficiency diseases committee report on inborn errors of immunity. J Clin Immunol. 2018;38(1):96–128. doi: 10.1007/s10875-017-0464-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bousfiha A, Jeddane L, Picard C, Ailal F, Bobby Gaspar H, Al-Herz W, et al. The 2017 IUIS phenotypic classification for primary Immunodeficiencies. J Clin Immunol. 2018;38(1):129–143. doi: 10.1007/s10875-017-0465-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Casanova JL, Abel L. Human genetics of infectious diseases: unique insights into immunological redundancy. Semin Immunol. 2018;36:1–12. doi: 10.1016/j.smim.2017.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Fischer A, Rausell A. What do primary immunodeficiencies tell us about the essentiality/redundancy of immune responses? Semin Immunol. 2018;36:13–16. doi: 10.1016/j.smim.2017.12.001. [DOI] [PubMed] [Google Scholar]
  • 5.Zhang SY, Jouanguy E, Zhang Q, Abel L, Puel A, Casanova JL. Human inborn errors of immunity to infection affecting cells other than leukocytes: from the immune system to the whole organism. Curr Opin Immunol. 2019;59:88–100. doi: 10.1016/j.coi.2019.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bucciol G, Moens L, Bosch B, Bossuyt X, Casanova JL, Puel A, et al. Lessons learned from the study of human inborn errors of innate immunity. J Allergy Clin Immunol. 2019;143(2):507–527. doi: 10.1016/j.jaci.2018.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Meyts I, Bosch B, Bolze A, Boisson B, Itan Y, Belkadi A, et al. 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]
  • 8.Picard C, Fischer A. Contribution of high-throughput DNA sequencing to the study of primary immunodeficiencies. Eur J Immunol. 2014;44(10):2854–2861. doi: 10.1002/eji.201444669. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang Q, Frange P, Blanche S, Casanova JL. Pathogenesis of infections in HIV-infected individuals: insights from primary immunodeficiencies. Curr Opin Immunol. 2017;48:122–133. doi: 10.1016/j.coi.2017.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kerner G, Ramirez-Alejo N, Seeleuthner Y, Yang R, Ogishi M, Cobat A, et al. Homozygosity for TYK2 P1104A underlies tuberculosis in about 1% of patients in a cohort of European ancestry. Proc Natl Acad Sci U S A. 2019;116(21):10430–10434. doi: 10.1073/pnas.1903561116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Leiding JW, Forbes LR. Mechanism-based precision therapy for the treatment of primary immunodeficiency and primary Immunodysregulatory diseases. J Allergy Clin Immunol Pract. 2019;7(3):761–773. doi: 10.1016/j.jaip.2018.12.017. [DOI] [PubMed] [Google Scholar]
  • 12.Conley ME, Dobbs AK, Farmer DM, Kilic S, Paris K, Grigoriadou S, et al. Primary B cell immunodeficiencies: comparisons and contrasts. Annu Rev Immunol. 2009;27:199–227. doi: 10.1146/annurev.immunol.021908.132649. [DOI] [PubMed] [Google Scholar]
  • 13.Fischer A, Rausell A. Primary immunodeficiencies suggest redundancy within the human immune system. Sci Immunol. 2016;1(6). 10.1126/sciimmunol.aah5861. [DOI] [PubMed]
  • 14.Gayko U, Fung M, Clow F, Sun S, Faust E, Price S, et al. Development of the Bruton's tyrosine kinase inhibitor ibrutinib for B cell malignancies. Ann N Y Acad Sci. 2015;1358:82–94. doi: 10.1111/nyas.12878. [DOI] [PubMed] [Google Scholar]
  • 15.Ma CS, Tangye SG. Flow Cytometric-based analysis of defects in lymphocyte differentiation and function due to inborn errors of immunity. Front Immunol. 2019;10:2108. doi: 10.3389/fimmu.2019.02108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bruton OC. Agammaglobulinemia Pediatrics. 1952;9(6):722–728. [PubMed] [Google Scholar]
  • 17.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]
  • 18.Byun M, Abhyankar A, Lelarge V, Plancoulaine S, Palanduz A, Telhan L, et al. Whole-exome sequencing-based discovery of STIM1 deficiency in a child with fatal classic Kaposi sarcoma. J Exp Med. 2010;207(11):2307–2312. doi: 10.1084/jem.20101597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Beziat V, Li J, Lin JX, Ma CS, Li P, Bousfiha A, et al. A recessive form of hyper-IgE syndrome by disruption of ZNF341-dependent STAT3 transcription and activity. Sci Immunol. 2018;3(24). 10.1126/sciimmunol.aat4956. [DOI] [PMC free article] [PubMed]
  • 20.Frey-Jakobs S, Hartberger JM, Fliegauf M, Bossen C, Wehmeyer ML, Neubauer JC, et al. ZNF341 controls STAT3 expression and thereby immunocompetence. Sci Immunol. 2018;3(24). 10.1126/sciimmunol.aat4941. [DOI] [PMC free article] [PubMed]
  • 21.Shahin T, Aschenbrenner D, Cagdas D, Bal SK, Conde CD, Garncarz W, et al. 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]
  • 22.Schwerd T, Twigg SRF, Aschenbrenner D, Manrique S, Miller KA, Taylor IB, et al. A biallelic mutation in IL6ST encoding the GP130 co-receptor causes immunodeficiency and craniosynostosis. J Exp Med. 2017;214(9):2547–2562. doi: 10.1084/jem.20161810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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]
  • 24.Nahum A, Sharfe N, Broides A, Dadi H, Naghdi Z, Mandola AB, et al. Defining the biological responses of IL-6 by the study of a novel IL-6 receptor chain (IL6R) immunodeficiency. J Allergy Clin Immunol. 2019. 10.1016/j.jaci.2019.11.015. [DOI] [PubMed]
  • 25.Ma CA, Stinson JR, Zhang Y, Abbott JK, Weinreich MA, Hauk PJ, et al. Germline hypomorphic CARD11 mutations in severe atopic disease. Nat Genet. 2017;49(8):1192–1201. doi: 10.1038/ng.3898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dorjbal B, Stinson JR, Ma CA, Weinreich MA, Miraghazadeh B, Hartberger JM, et al. Hypomorphic caspase activation and recruitment domain 11 (CARD11) mutations associated with diverse immunologic phenotypes with or without atopic disease. J Allergy Clin Immunol. 2019;143(4):1482–1495. doi: 10.1016/j.jaci.2018.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Klammt J, Neumann D, Gevers EF, Andrew SF, Schwartz ID, Rockstroh D, et al. Dominant-negative STAT5B mutations cause growth hormone insensitivity with short stature and mild immune dysregulation. Nat Commun. 2018;9(1):2105. doi: 10.1038/s41467-018-04521-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lu HY, Bauman BM, Arjunaraja S, Dorjbal B, Milner JD, Snow AL, et al. The CBM-opathies-A rapidly expanding Spectrum of human inborn errors of immunity caused by mutations in the CARD11-BCL10-MALT1 complex. Front Immunol. 2018;9:2078. doi: 10.3389/fimmu.2018.02078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nadeau K, Hwa V, Rosenfeld RG. STAT5b deficiency: an unsuspected cause of growth failure, immunodeficiency, and severe pulmonary disease. J Pediatr. 2011;158(5):701–708. doi: 10.1016/j.jpeds.2010.12.042. [DOI] [PubMed] [Google Scholar]
  • 30.Boisson B, Wang YD, Bosompem A, Ma CS, Lim A, Kochetkov T, et al. A recurrent dominant negative E47 mutation causes agammaglobulinemia and BCR(−) B cells. J Clin Invest. 2013;123(11):4781–4785. doi: 10.1172/JCI71927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ben-Ali M, Yang J, Chan KW, Ben-Mustapha I, Mekki N, Benabdesselem C, et al. Homozygous transcription factor 3 gene (TCF3) mutation is associated with severe hypogammaglobulinemia and B-cell acute lymphoblastic leukemia. J Allergy Clin Immunol. 2017;140(4):1191–4 e4. doi: 10.1016/j.jaci.2017.04.037. [DOI] [PubMed] [Google Scholar]
  • 32.Qureshi S, Sheikh MDA, Qamar FN. Autosomal recessive Agammaglobulinemia - first case with a novel TCF3 mutation from Pakistan. Clin Immunol. 2019;198:100–101. doi: 10.1016/j.clim.2018.07.016. [DOI] [PubMed] [Google Scholar]
  • 33.Cardinez C, Miraghazadeh B, Tanita K, da Silva E, Hoshino A, Okada S, et al. Gain-of-function IKBKB mutation causes human combined immune deficiency. J Exp Med. 2018;215(11):2715–2724. doi: 10.1084/jem.20180639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pannicke U, Baumann B, Fuchs S, Henneke P, Rensing-Ehl A, Rizzi M, et al. Deficiency of innate and acquired immunity caused by an IKBKB mutation. N Engl J Med. 2013;369(26):2504–2514. doi: 10.1056/NEJMoa1309199. [DOI] [PubMed] [Google Scholar]
  • 35.Sogkas G, Fedchenko M, Dhingra A, Jablonka A, Schmidt RE, Atschekzei F. Primary immunodeficiency disorder caused by phosphoinositide 3-kinase delta deficiency. J Allergy Clin Immunol. 2018;142(5):1650–1653. doi: 10.1016/j.jaci.2018.06.039. [DOI] [PubMed] [Google Scholar]
  • 36.Cohen SB, Bainter W, Johnson JL, Lin TY, Wong JCY, Wallace JG, et al. Human primary immunodeficiency caused by expression of a kinase-dead p110delta mutant. J Allergy Clin Immunol. 2019;143(2):797–9 e2. doi: 10.1016/j.jaci.2018.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tangye SG, Bier J, Lau A, Nguyen T, Uzel G, Deenick EK. Immune Dysregulation and disease pathogenesis due to activating mutations in PIK3CD-the Goldilocks' effect. J Clin Immunol. 2019;39(2):148–158. doi: 10.1007/s10875-019-00612-9. [DOI] [PubMed] [Google Scholar]
  • 38.Boutboul D, Kuehn HS, Van de Wyngaert Z, Niemela JE, Callebaut I, Stoddard J, et al. Dominant-negative IKZF1 mutations cause a T, B, and myeloid cell combined immunodeficiency. J Clin Invest. 2018;128(7):3071–3087. doi: 10.1172/JCI98164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kuehn HS, Boisson B, Cunningham-Rundles C, Reichenbach J, Stray-Pedersen A, Gelfand EW, et al. Loss of B cells in patients with heterozygous mutations in IKAROS. N Engl J Med. 2016;374(11):1032–1043. doi: 10.1056/NEJMoa1512234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Toubiana J, Okada S, Hiller J, Oleastro M, Lagos Gomez M, Aldave Becerra JC, et al. Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood. 2016;127(25):3154–3164. doi: 10.1182/blood-2015-11-679902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Alkhairy OK, Rezaei N, Graham RR, Abolhassani H, Borte S, Hultenby K, et al. RAC2 loss-of-function mutation in 2 siblings with characteristics of common variable immunodeficiency. J Allergy Clin Immunol. 2015;135(5):1380–4 e1-5. doi: 10.1016/j.jaci.2014.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hsu AP, Donko A, Arrington ME, Swamydas M, Fink D, Das A, et al. Dominant activating RAC2 mutation with lymphopenia, immunodeficiency, and cytoskeletal defects. Blood. 2019;133(18):1977–1988. doi: 10.1182/blood-2018-11-886028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lougaris V, Chou J, Beano A, Wallace JG, Baronio M, Gazzurelli L, et al. A monoallelic activating mutation in RAC2 resulting in a combined immunodeficiency. J Allergy Clin Immunol. 2019;143(4):1649–1653. doi: 10.1016/j.jaci.2019.01.001. [DOI] [PubMed] [Google Scholar]
  • 44.Sharapova SO, Haapaniemi E, Sakovich IS, Kostyuchenko LV, Donko A, Dulau-Florea A, et al. Heterozygous activating mutation in RAC2 causes infantile-onset combined immunodeficiency with susceptibility to viral infections. Clin Immunol. 2019;205:1–5. doi: 10.1016/j.clim.2019.05.003. [DOI] [PubMed] [Google Scholar]
  • 45.Smits BM, Lelieveld PHC, Ververs FA, Turkenburg M, de Koning C, van Dijk M, et al. A dominant activating RAC2 variant associated with immunodeficiency and pulmonary disease. Clin Immunol. 2019;108248. 10.1016/j.clim.2019.108248. [DOI] [PubMed]
  • 46.Hernandez N, Melki I, Jing H, Habib T, Huang SSY, Danielson J, et al. Life-threatening influenza pneumonitis in a child with inherited IRF9 deficiency. J Exp Med. 2018;215(10):2567–2585. doi: 10.1084/jem.20180628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Belkaya S, Michailidis E, Korol CB, Kabbani M, Cobat A, Bastard P, et al. Inherited IL-18BP deficiency in human fulminant viral hepatitis. J Exp Med. 2019;216(8):1777–1790. doi: 10.1084/jem.20190669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Serwas NK, Hoeger B, Ardy RC, Stulz SV, Sui Z, Memaran N, et al. Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nat Commun. 2019;10(1):3106. doi: 10.1038/s41467-019-10812-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, et al. AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science. 2015;349(6246):436–440. doi: 10.1126/science.aaa1663. [DOI] [PubMed] [Google Scholar]
  • 50.Schwab C, Gabrysch A, Olbrich P, Patino V, Warnatz K, Wolff D, et al. Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol. 2018;142(6):1932–1946. doi: 10.1016/j.jaci.2018.02.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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). 10.1126/sciimmunol.aau6759. [DOI] [PMC free article] [PubMed]
  • 52.Kong XF, Martinez-Barricarte R, Kennedy J, Mele F, Lazarov T, Deenick EK, et al. Disruption of an antimycobacterial circuit between dendritic and helper T cells in human SPPL2a deficiency. Nat Immunol. 2018;19(9):973–985. doi: 10.1038/s41590-018-0178-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Roussel L, Landekic M, Golizeh M, Gavino C, Zhong MC, Chen J, et al. Loss of human ICOSL results in combined immunodeficiency. J Exp Med. 2018;215(12):3151–3164. doi: 10.1084/jem.20180668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Conde CD, Petronczki OY, Baris S, Willmann KL, Girardi E, Salzer E, et al. Polymerase delta deficiency causes syndromic immunodeficiency with replicative stress. J Clin Invest. 2019;129(10):4194–4206. doi: 10.1172/JCI128903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cui Y, Keles S, Charbonnier LM, Jule AM, Henderson L, Celik SC, et al. Combined immunodeficiency due to a loss of function mutation in DNA Polymerase Delta 1. J Allergy Clin Immunol. 2019. 10.1016/j.jaci.2019.10.004. [DOI] [PMC free article] [PubMed]
  • 56.Badran YR, Dedeoglu F, Leyva Castillo JM, Bainter W, Ohsumi TK, Bousvaros A, et al. Human RELA haploinsufficiency results in autosomal-dominant chronic mucocutaneous ulceration. J Exp Med. 2017;214(7):1937–1947. doi: 10.1084/jem.20160724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Comrie WA, Faruqi AJ, Price S, Zhang Y, Rao VK, Su HC, et al. RELA haploinsufficiency in CD4 lymphoproliferative disease with autoimmune cytopenias. J Allergy Clin Immunol. 2018;141(4):1507–1510. doi: 10.1016/j.jaci.2017.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Beaussant-Cohen S, Jaber F, Massaad MJ, Weeks S, Jones J, Alosaimi MF, et al. Combined immunodeficiency in a patient with c-Rel deficiency. J Allergy Clin Immunol. 2019;144(2):606–608. doi: 10.1016/j.jaci.2019.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Calzoni E, Platt CD, Keles S, Kuehn HS, Beaussant-Cohen S, Zhang Y, et al. F-BAR domain only protein 1 (FCHO1) deficiency is a novel cause of combined immune deficiency in human subjects. J Allergy Clin Immunol. 2019;143(6):2317–2321. doi: 10.1016/j.jaci.2019.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maffucci P, Chavez J, Jurkiw TJ, O'Brien PJ, Abbott JK, Reynolds PR, et al. Biallelic mutations in DNA ligase 1 underlie a spectrum of immune deficiencies. J Clin Invest. 2018;128(12):5489–5504. doi: 10.1172/JCI99629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bosticardo M, Yamazaki Y, Cowan J, Giardino G, Corsino C, Scalia G, et al. Heterozygous FOXN1 variants cause low TRECs and severe T cell Lymphopenia, revealing a crucial role of FOXN1 in supporting early Thymopoiesis. Am J Hum Genet. 2019;105(3):549–561. doi: 10.1016/j.ajhg.2019.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lyons JJ, Liu Y, Ma CA, Yu X, O'Connell MP, Lawrence MG, et al. ERBIN deficiency links STAT3 and TGF-beta pathway defects with atopy in humans. J Exp Med. 2017;214(3):669–680. doi: 10.1084/jem.20161435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schepers D, Tortora G, Morisaki H, MacCarrick G, Lindsay M, Liang D, et al. A mutation update on the LDS-associated genes TGFB2/3 and SMAD2/3. Hum Mutat. 2018;39(5):621–634. doi: 10.1002/humu.23407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fabre A, Charroux B, Martinez-Vinson C, Roquelaure B, Odul E, Sayar E, et al. SKIV2L mutations cause syndromic diarrhea, or trichohepatoenteric syndrome. Am J Hum Genet. 2012;90(4):689–692. doi: 10.1016/j.ajhg.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Huppke P, Weissbach S, Church JA, Schnur R, Krusen M, Dreha-Kulaczewski S, et al. Activating de novo mutations in NFE2L2 encoding NRF2 cause a multisystem disorder. Nat Commun. 2017;8(1):818. doi: 10.1038/s41467-017-00932-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Rodriguez R, Fournier B, Cordeiro DJ, Winter S, Izawa K, Martin E, et al. Concomitant PIK3CD and TNFRSF9 deficiencies cause chronic active Epstein-Barr virus infection of T cells. J Exp Med. 2019. 10.1084/jem.20190678. [DOI] [PMC free article] [PubMed]
  • 67.Anzilotti C, Swan DJ, Boisson B, Deobagkar-Lele M, Oliveira C, Chabosseau P, et al. An essential role for the Zn(2+) transporter ZIP7 in B cell development. Nat Immunol. 2019;20(3):350–361. doi: 10.1038/s41590-018-0295-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Broderick L, Yost S, Li D, McGeough MD, Booshehri LM, Guaderrama M, et al. Mutations in topoisomerase IIbeta result in a B cell immunodeficiency. Nat Commun. 2019;10(1):3644. doi: 10.1038/s41467-019-11570-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bouafia A, Lofek S, Bruneau J, Chentout L, Lamrini H, Trinquand A, et al. Loss of ARHGEF1 causes a human primary antibody deficiency. J Clin Invest. 2019;129(3):1047–1060. doi: 10.1172/JCI120572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Keller B, Shoukier M, Schulz K, Bhatt A, Heine I, Strohmeier V, et al. Germline deletion of CIN85 in humans with X chromosome-linked antibody deficiency. J Exp Med. 2018;215(5):1327–1336. doi: 10.1084/jem.20170534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Schubert D, Klein MC, Hassdenteufel S, Caballero-Oteyza A, Yang L, Proietti M, et al. Plasma cell deficiency in human subjects with heterozygous mutations in Sec61 translocon alpha 1 subunit (SEC61A1) J Allergy Clin Immunol. 2018;141(4):1427–1438. doi: 10.1016/j.jaci.2017.06.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mauhin W, Habarou F, Gobin S, Servais A, Brassier A, Grisel C, et al. Update on Lysinuric protein intolerance, a multi-faceted disease retrospective cohort analysis from birth to adulthood. Orphanet J Rare Dis. 2017;12(1):3. doi: 10.1186/s13023-016-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fernandez IZ, Baxter RM, Garcia-Perez JE, Vendrame E, Ranganath T, Kong DS, et al. A novel human IL2RB mutation results in T and NK cell-driven immune dysregulation. J Exp Med. 2019;216(6):1255–1267. doi: 10.1084/jem.20182015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zhang Z, Gothe F, Pennamen P, James JR, McDonald D, Mata CP, et al. Human interleukin-2 receptor beta mutations associated with defects in immunity and peripheral tolerance. J Exp Med. 2019;216(6):1311–1327. doi: 10.1084/jem.20182304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Has C, Castiglia D, del Rio M, Diez MG, Piccinni E, Kiritsi D, et al. Kindler syndrome: extension of FERMT1 mutational spectrum and natural history. Hum Mutat. 2011;32(11):1204–1212. doi: 10.1002/humu.21576. [DOI] [PubMed] [Google Scholar]
  • 76.Kotlarz D, Marquardt B, Baroy T, Lee WS, Konnikova L, Hollizeck S, et al. Human TGF-beta1 deficiency causes severe inflammatory bowel disease and encephalopathy. Nat Genet. 2018;50(3):344–348. doi: 10.1038/s41588-018-0063-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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]
  • 78.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]
  • 79.Alosaimi MF, Hoenig M, Jaber F, Platt CD, Jones J, Wallace J, et al. Immunodeficiency and EBV-induced lymphoproliferation caused by 4-1BB deficiency. J Allergy Clin Immunol. 2019;144(2):574–83 e5. doi: 10.1016/j.jaci.2019.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Somekh I, Thian M, Medgyesi D, Gulez N, Magg T, Gallon Duque A, et al. CD137 deficiency causes immune dysregulation with predisposition to lymphomagenesis. Blood. 2019. 10.1182/blood.2019000644. [DOI] [PMC free article] [PubMed]
  • 81.Carapito R, Konantz M, Paillard C, Miao Z, Pichot A, Leduc MS, et al. Mutations in signal recognition particle SRP54 cause syndromic neutropenia with Shwachman-diamond-like features. J Clin Invest. 2017;127(11):4090–4103. doi: 10.1172/JCI92876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Bellanne-Chantelot C, Schmaltz-Panneau B, Marty C, Fenneteau O, Callebaut I, Clauin S, et al. Mutations in the SRP54 gene cause severe congenital neutropenia as well as Shwachman-diamond-like syndrome. Blood. 2018;132(12):1318–1331. doi: 10.1182/blood-2017-12-820308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dhanraj S, Matveev A, Li H, Lauhasurayotin S, Jardine L, Cada M, et al. Biallelic mutations in DNAJC21 cause Shwachman-diamond syndrome. Blood. 2017;129(11):1557–1562. doi: 10.1182/blood-2016-08-735431. [DOI] [PubMed] [Google Scholar]
  • 84.Arnadottir GA, Norddahl GL, Gudmundsdottir S, Agustsdottir AB, Sigurdsson S, Jensson BO, et al. A homozygous loss-of-function mutation leading to CYBC1 deficiency causes chronic granulomatous disease. Nat Commun. 2018;9(1):4447. doi: 10.1038/s41467-018-06964-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Thomas DC, Charbonnier LM, Schejtman A, Aldhekri H, Coomber EL, Dufficy ER, et al. EROS/CYBC1 mutations: decreased NADPH oxidase function and chronic granulomatous disease. J Allergy Clin Immunol. 2019;143(2):782–785. doi: 10.1016/j.jaci.2018.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.de Jong SJ, Crequer A, Matos I, Hum D, Gunasekharan V, Lorenzo L, et al. The human CIB1-EVER1-EVER2 complex governs keratinocyte-intrinsic immunity to beta-papillomaviruses. J Exp Med. 2018;215(9):2289–2310. doi: 10.1084/jem.20170308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hernandez N, Bucciol G, Moens L, Le Pen J, Shahrooei M, Goudouris E, et al. Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J Exp Med. 2019;216(9):2057–2070. doi: 10.1084/jem.20182295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ogunjimi B, Zhang SY, Sorensen KB, Skipper KA, Carter-Timofte M, Kerner G, et al. Inborn errors in RNA polymerase III underlie severe varicella zoster virus infections. J Clin Invest. 2017;127(9):3543–3556. doi: 10.1172/JCI92280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Carter-Timofte ME, Hansen AF, Mardahl M, Fribourg S, Rapaport F, Zhang SY, et al. Varicella-zoster virus CNS vasculitis and RNA polymerase III gene mutation in identical twins. Neurol Neuroimmunol Neuroinflamm. 2018;5(6):e500. doi: 10.1212/NXI.0000000000000500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhang SY, Clark NE, Freije CA, Pauwels E, Taggart AJ, Okada S, et al. Inborn errors of RNA lariat metabolism in humans with brainstem viral infection. Cell. 2018;172(5):952–965. doi: 10.1016/j.cell.2018.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Guerin A, Kerner G, Marr N, Markle JG, Fenollar F, Wong N, et al. IRF4 haploinsufficiency in a family with Whipple's disease. Elife. 2018;7. 10.7554/eLife.32340. [DOI] [PMC free article] [PubMed]
  • 92.Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, et al. Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest. 2015;125(11):4196–4211. doi: 10.1172/JCI81260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Rodero MP, Tesser A, Bartok E, Rice GI, Della Mina E, Depp M, et al. Type I interferon-mediated autoinflammation due to DNase II deficiency. Nat Commun. 2017;8(1):2176. doi: 10.1038/s41467-017-01932-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Al-Mayouf SM, Sunker A, Abdwani R, Abrawi SA, Almurshedi F, Alhashmi N, et al. Loss-of-function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus. Nat Genet. 2011;43(12):1186–1188. doi: 10.1038/ng.975. [DOI] [PubMed] [Google Scholar]
  • 95.Ozcakar ZB, Foster J, 2nd, Diaz-Horta O, Kasapcopur O, Fan YS, Yalcinkaya F, et al. DNASE1L3 mutations in hypocomplementemic urticarial vasculitis syndrome. Arthritis Rheum. 2013;65(8):2183–2189. doi: 10.1002/art.38010. [DOI] [PubMed] [Google Scholar]
  • 96.Carbonella A, Mancano G, Gremese E, Alkuraya FS, Patel N, Gurrieri F, et al. An autosomal recessive DNASE1L3-related autoimmune disease with unusual clinical presentation mimicking systemic lupus erythematosus. Lupus. 2017;26(7):768–772. doi: 10.1177/0961203316676382. [DOI] [PubMed] [Google Scholar]
  • 97.Cho K, Yamada M, Agematsu K, Kanegane H, Miyake N, Ueki M, et al. Heterozygous mutations in OAS1 cause infantile-onset pulmonary alveolar Proteinosis with Hypogammaglobulinemia. Am J Hum Genet. 2018;102(3):480–486. doi: 10.1016/j.ajhg.2018.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhong FL, Mamai O, Sborgi L, Boussofara L, Hopkins R, Robinson K, et al. Germline NLRP1 mutations cause skin inflammatory and Cancer susceptibility syndromes via Inflammasome activation. Cell. 2016;167(1):187–202. doi: 10.1016/j.cell.2016.09.001. [DOI] [PubMed] [Google Scholar]
  • 99.Drutman SB, Haerynck F, Zhong FL, Hum D, Hernandez NJ, Belkaya S, et al. Homozygous NLRP1 gain-of-function mutation in siblings with a syndromic form of recurrent respiratory papillomatosis. Proc Natl Acad Sci U S A. 2019;116(38):19055–19063. doi: 10.1073/pnas.1906184116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Parlato M, Charbit-Henrion F, Pan J, Romano C, Duclaux-Loras R, Le Du MH, et al. Human ALPI deficiency causes inflammatory bowel disease and highlights a key mechanism of gut homeostasis. EMBO Mol Med. 2018;10(4). 10.15252/emmm.201708483. [DOI] [PMC free article] [PubMed]
  • 101.Li Q, Lee CH, Peters LA, Mastropaolo LA, Thoeni C, Elkadri A, et al. Variants in TRIM22 that affect NOD2 signaling are associated with very-early-onset inflammatory bowel disease. Gastroenterology. 2016;150(5):1196–1207. doi: 10.1053/j.gastro.2016.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.de Jesus AA, Brehm A, VanTries R, Pillet P, Parentelli AS, Montealegre Sanchez GA, et al. Novel proteasome assembly chaperone mutations in PSMG2/PAC2 cause the autoinflammatory interferonopathy CANDLE/PRAAS4. J Allergy Clin Immunol. 2019;143(5):1939–1943. doi: 10.1016/j.jaci.2018.12.1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gayden T, Sepulveda FE, Khuong-Quang DA, Pratt J, Valera ET, Garrigue A, et al. Germline HAVCR2 mutations altering TIM-3 characterize subcutaneous panniculitis-like T cell lymphomas with hemophagocytic lymphohistiocytic syndrome. Nat Genet. 2018;50(12):1650–1657. doi: 10.1038/s41588-018-0251-4. [DOI] [PubMed] [Google Scholar]
  • 104.Polprasert C, Takeuchi Y, Kakiuchi N, Yoshida K, Assanasen T, Sitthi W, et al. Frequent germline mutations of HAVCR2 in sporadic subcutaneous panniculitis-like T-cell lymphoma. Blood Adv. 2019;3(4):588–595. doi: 10.1182/bloodadvances.2018028340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kapferer-Seebacher I, Pepin M, Werner R, Aitman TJ, Nordgren A, Stoiber H, et al. Periodontal Ehlers-Danlos syndrome is caused by mutations in C1R and C1S, which encode subcomponents C1r and C1s of complement. Am J Hum Genet. 2016;99(5):1005–1014. doi: 10.1016/j.ajhg.2016.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical Immunology are provided here courtesy of Springer

RESOURCES