Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 1.
Published in final edited form as: Curr Opin Immunol. 2019 May 20;59:88–100. doi: 10.1016/j.coi.2019.03.008

Human inborn errors of immunity to infection affecting cells other than leukocytes: from the immune system to the whole organism

Shen-Ying Zhang 1,2,3, Emmanuelle Jouanguy 1,2,3, Qian Zhang 1, Laurent Abel 1,2,3, Anne Puel 1,2,3, Jean-Laurent Casanova 1,2,3,4,5,@
PMCID: PMC6774828  NIHMSID: NIHMS1529936  PMID: 31121434

Abstract

Studies of vertebrate immunity have traditionally focused on professional cells, including circulating and tissue-resident leukocytes. However, evidence to suggest that non-professional cells are also essential for protective immunity in natural conditions of infection has emerged from three lines of research in human genetics. First, studies of Mendelian resistance to infection have revealed an essential role of DARC-expressing erythrocytes in protection against Plasmodium vivax infection, and an essential role of FUT2-expressing intestinal epithelial cells for protection against norovirus and rotavirus infections. Second, studies of inborn errors of non-hematopoietic cell-extrinsic immunity have shown that APOL1 and complement cascade components secreted by hepatocytes are essential for protective immunity to trypanosome and pyogenic bacteria, respectively. Third, studies of inborn errors of non-hematopoietic cell-intrinsic immunity have suggested that keratinocytes, pulmonary epithelial cells, and cortical neurons are essential for tissue-specific protective immunity to human papillomaviruses, influenza virus, and herpes simplex virus, respectively. Various other types of genetic resistance or predisposition to infection in human populations are not readily explained by inborn variants of genes operating in leukocytes and may therefore involve defects in other cells. The probing of this unchartered territory by human genetics is reshaping immunology, by scaling immunity to infection up from the immune system to the whole organism.

Introduction

The human body comprises more than 400 discernable cell types [1]. All cell types are thought to contribute to host defense, because (i) their presence ensures the physical delineation and integrity of tissues and organs, (ii) they interact with professional leukocytes via soluble and membrane-bound molecules, and (iii) they are targets of microbes and their products. However, host defense in vertebrates has traditionally been attributed to the immune system, which was largely seen as restricted to immunoglobulins (Ig) and complement during the “immunochemistry” period in the history of immunology. This definition was expanded to include leukocytes during the more recent “immunobiology” period, which began in the 1960s [2]. This is understandable, as immunology has, as a discipline, mostly focused on antigens rather than pathogens, as a direct consequence of Pasteur’s discovery of the principle of specific vaccination in 1881 [3]. reinforced by Landsteiner’s discovery of specific antibody responses to haptens in 1917 [4]. Phagocytes have been known to contribute to protective immunity since Metchnikoff s work, but their role in host defense was rarely studied by immunologists, due to a lack of antigen specificity [5]. Only since the 1990s have immunologists turned their attention to the contributions of innate immunity to host defense at large, as illustrated by their rediscovery of granulocytes, which had traditionally been studied by hematologists [6]. Nevertheless, it is generally thought that immunity is ensured exclusively by the circulating and tissue-resident professional leukocytes, including myeloid and lymphoid cells, mostly of hematopoietic origin, together with certain tissue macrophages generated earlier in embryonic development [7,8].

Other cells, not included in this definition of the immune system, are rarely considered to play a role in host defense in humans or other vertebrates. The classic dichotomies of cellular and humoral immunity, and innate and adaptive immunity, typically refer to leukocytes [9,10]. Nevertheless, the development or function of certain leukocyte subsets can be strongly affected by deficiencies of non-hematopoietic cells that prevent the development of certain lymphoid organs. Indeed, a phenocopy of severe combined immunodeficiency can result from thymic stromal cell-intrinsic defects of thymus development in patients with autosomal recessive (AR) FOXN1 deficiency or autosomal dominant (AD) Di George syndrome due to the 22q11.2 deletion or mutations of the CHD7 gene [11,12]. Likewise, AD RPSA deficiency results in isolated congenital asplenia, abolishing the filtering function of splenic macrophages [13]. Moreover, sickle-cell disease (SCD) and cystic fibrosis (CF) are well-known AR disorders of cells other than leukocytes with life-threatening infectious phenotypes [1419]. The HBB gene is expressed in erythrocytes, whereas CFTR is expressed in various cell types and organs, including, in particular, the ionocytes of the pulmonary epithelium [20]. Patients with SCD are prone to invasive bacterial infections because of functional asplenia [15,21]. Patients with CF are probably prone to respiratory infections because of abnormal fluid composition within the airway [1619]. Variants of DCTN4 act as modifiers of chronic Pseudomonas aeruginosa infection in CF patients [22]. Both SCD and CF are inborn errors conferring a predisposition to infection, but the cellular basis of the infections associated with them remains incompletely understood. We review here the human genetic studies that have provided evidence for an essential role of specific cells other than leukocytes in host defense under natural conditions of infection, a hallmark of human studies [2325].

Monogenic resistance to infections: erythrocytes and the intestinal epithelium

The study of monogenic resistance to human infections has provided compelling evidence that non-professional cells play an essential role in protective immunity. Protection against infection with Plasmodium vivax is conferred by a lack of expression of the Duffy antigen receptor for chemokines (DARC), a coreceptor for the parasite on the surface of erythrocytes [26]. Duffy-negative erythrocytes were shown to resist invasion by P. vivax [27] and its simian homolog P. knowlesi [28] in vitro. The resistance trait is AR and the causal single-nucleotide mutation, also known as FY*O (Duffy null), disrupts a binding site for the GATA1 erythroid transcription factor in the DARC promoter, thereby selectively preventing gene transcription in erythroid cells [29]. The relevance of this erythrocyte-intrinsic mechanism of host defense is illustrated by evolutionary studies of the DARC locus. Indeed, natural selection in areas of endemic disease has increased the proportion of resistant individuals. The Duffy null mutation is not found in European populations, but has become fixed in some African populations in which P. vivax infection has been endemic [30]. A recent study confirmed the positive selection of an FY*O null mutation, by showing that this mutation swept to fixation in Africa from a very low initial frequency (0.1%), with a very strong selection coefficient [31]. This gene displays “beneficial redundancy” [32], although the protection it confers is not absolute, with rare observations of P. vivax infection in Duffy-negative subjects [33,34]. Understandably, the deficiency of this chemokine receptor does not seem to have deleterious effects on any other physiological pathways. Indeed, the expression defect is erythrocyte-specific, as the DARC chemokine receptor is expressed normally on other cell types [29].

Resistance to certain enteric viruses is provided by biallelic loss-of-function (LOF) mutations of the fucosyltransferase 2 (FUT2) gene [35], which encodes a protein that regulates the expression of histoblood group antigens (HBGAs) on the surface of mucosal epithelial cells of the gastrointestinal, genitourinary, and respiratory tracts, and is responsible for the “secretor” phenotype [36,37]. Several LOF FUT2 mutations underlie the “non-secretor” phenotype, which confers resistance to norovirus [38,39] and rotavirus [40,41] infections. Common strains of norovirus (particularly genogroups I and II) and rotavirus (particularly the P[4] and P[8] genotypes) bind to HBGA [37,4244]. LOF mutations affecting HBGA expression therefore lead to resistance to these strains [35,45]. However, a recent study showed that the binding of rotavirus to HBGA was not essential for the in vitro infection of transformed cells [46]. This finding requires confirmation in more relevant cellular models, such as human intestinal organoids [47], but it suggests that HBGAs may enhance viral replication and disease development without actually being essential for infection per se [46]. The FUT2 gene also displays “beneficial redundancy” [32], although protection against enteric viral diseases is not absolute [45]. Moreover, FUT2 non-secretor status has been associated with predisposition to Crohn’s disease [48], Behçet’s disease [49], and various bacterial infections [35] including otitis media [50]. Consistently, a complex pattern of natural selection has been described for FUT2, with most variants displaying a long history of balancing selection in Eurasian and African populations [51]. Overall, FUT2 may be considered an important genetic factor influencing epithelial-intrinsic mucosal immunity to various pathogens in the intestinal tract [50,52]. Thus, humans can develop protective immunity to life-threatening infections through the natural selection of genotypes at the DARC and FUT2 loci that block the infection of nonprofessional target cells by parasites and viruses, respectively. Allelic variants of genes expressed in erythrocytes and the intestinal epithelium can therefore be life-saving.

Inborn errors of complement and APOL1: hepatocytes

Since 1966, inherited defects of various components of the complement cascade have been shown to underlie one or more infections [5359]. About 50 proteins contribute to the three branches of complement and their fine regulation [60,61]. Most of these proteins are synthesized in the liver, principally by hepatocytes [6266], but a few are produced predominantly by other cells [67]. The classical complement pathway component C1q can be produced in vitro by epithelial cells, fibroblasts, and monocytes/macrophages [68,69]. The alternative pathway component factor D is produced mostly by adipocytes [70,71] and, to a lesser extent, by monocytes/macrophages, whereas properdin is produced principally by monocytes/macrophages [72] and, to a lesser extent, by granulocytes and lymphocytes [72]. The main sources of terminal component C7 are monocytes and tissue-resident macrophages, including Kupffer cells [73]. Nevertheless, at least 90% of the total amount of complement proteins in the plasma is synthesized by liver hepatocytes [74], as demonstrated by the infectious diseases occurring as a result of acquired complement deficiency in patients with liver failure [74]. Defects in the early components (C1, C4, C2) of the classical pathway (C1-C9)[75] usually underlie invasive childhood infections with encapsulated pyogenic bacteria, such as Streptococcus pneumoniae and Haemophilus influenzae [5459,7584]. Defects of C3, affecting both the classical and alternative pathways, and of factor H or factor I from the alternative pathway also underlie recurrent and severe pyogenic infections [8492]. By contrast, defects of the terminal components (C5-C9) of the membrane-attack complex, or of the activating proteins properdin or factor D of the alternative pathway underlie selective susceptibility to life-threatening, invasive Neisseria diseases, typically meningococcal meningitis, which frequently recur [56,93110], Numerous mutations of the corresponding genes have been described since 1966 [55,56,58,76,7880,83,84,86,87,89102,107110]. Overall, these findings demonstrate that immunity mediated by hepatocytes can be life-saving, through the secretion of complement components into the bloodstream.

Apolipoprotein LI (APOL1), encoded by the APOL1 gene, which is found exclusively in primates, is a component of serum high-density lipoproteins that is also produced mostly in the liver by hepatocytes, and, to a lesser extent, in other tissues such as the lung, placenta, spleen, and kidney [111113]. Surprisingly, it has been shown to play a key role in host defense against trypanosomes. Trypanosoma evansi (T. evansi) is a weakly virulent parasite unable to cause disease in the vast majority of humans. However, it was found to be the cause of disease in an Indian patient whose serum displayed no lytic activity with T. evansi [114]. This patient has been diagnosed with AR, complete APOL1 deficiency [114]. The addition of recombinant APOL1 to the patient’ serum rescued its trypanolytic activity [114]. Human APOL1 is, therefore, essential for protective immunity to T. evansi in vivo. APOL1 also plays a role in immunity to infection with the more virulent T. brucei in vitro [115,116]. Following its endocytosis by trypanosomes, the secreted APOL1 forms anion-selective pores in the lysosomal membrane of the trypanosome, inducing uncontrolled osmotic swelling of the lysosome, leading to lysis of the trypanosome [117,118]. This mechanism explains the natural resistance of humans to infection with weakly virulent strains of trypanosomes, such as T. evansi. By contrast, T. brucei rhodesiense and T. brucei gambiense, which are endemic in Eastern and Western Africa, respectively, are resistant to APOL1, and are therefore able to cause sleeping sickness (also known as human African trypanosomiasis, HAT) in most infected individuals [119121]. The serum resistance-associated (SRA) protein of T. brucei rhodesiense and the serum resistance glycoprotein of T. brucei gambiense (TgsGP) interact specifically with APOL1, and this interaction renders the trypanosome resistant [122,123]. Interestingly, two variants encoding APOL1 of enhanced trypanolytic activity against T. brucei rhodesiense are only present in African populations where they harbor signatures of positive selection while increasing the risk of kidney disease [124], and were found to protect against T. brucei rhodesiense infection in an Ugandan population [125]. Liver-produced APOL1 is thus essential for host defense against trypanosomes, providing another example of the life-saving role of hepatocytes in immunity to infection through their secretion of particular products.

Epidermodysplasia verruciformis: keratinocytes

Human papillomaviruses (HPVs) display strict tropism for keratinocytes, the major component of stratifying epithelia [126,127]. This tropism is dependent on the lifecycle of HPV, which is built around epithelial stratification. HPVs target self-renewing stem cells in the basal layer of the epidermis and the bulge of the hair follicles. HPV infections are widespread, usually asymptomatic and cleared up after a few months by the T-cell response. Epidermodysplasia verruciformis (EV) is a genetic vulnerability to specific HPVs and was the first of the five known “Mendelian infections” (OMIM 226400) to be described [128130]. Indeed, EV was first clinically described in 1922 as a congenital dermatosis characterized by lifelong disseminated and persistent flat warts and pityriasis versicolor-like (PV-like) lesions [131]. An AR mode of inheritance was documented in 1933 and a viral etiology in 1946 [132,133]. EV is associated with a higher risk of non-melanoma skin carcinomas (NMSC) [134,135], but there are no other clinical signs in most patients with typical “isolated EV”, which is apparently due to inborn errors of keratinocytes [136]. Other patients suffering from EV and other infectious or tumoral manifestations display atypical, “syndromic EV”, due to inborn errors of T cells [137,138]. In 1978, Orth et al. showed that EV skin lesions are caused by a very specific group of weakly virulent, E5- and E8-deficient skin- and keratinocyte-tropic human HP Vs from the β genus [139]. β-HPVs are very common and cause asymptomatic infections of the skin in the general population [140,141]. In 2002, Orth et al. identified the first two EV-causing genes through genome-wide linkage (GWL) analysis, with the discovery of homozygous null mutations in EVER1 (TMC6) or EVER2 (TMC8) in patients with isolated EV [142147]. These patients have no detectable leukocyte phenotype [148,149], suggesting that EVER proteins govern keratinocyte-intrinsic immunity to β-HPVs [143].

In 2004, the observation that patients with severe combined immunodeficiency due to mutations of the related genes IL2RG and JAK3, normally expressed in both leukocytes and keratinocytes, often developed isolated EV years after successful hematopoietic stem cell transplantation [150,151], further suggested an essential role for keratinocytes in immunity to β-HPVs [150]. Biochemical studies of EVER proteins proved very difficult, at least partly because no specific antibody could be raised against these proteins. The recent discovery of AR complete CIB1 deficiency in other patients with isolated EV has provided new insight into the pathogenesis of EV [152,153]. In mice, the CIB1 protein is ubiquitous and functionally pleiotropic [154]. CIB1-deficient mice have phenotypes relating to the heart, vasculature, hemostasis, and fertility [155158]. None of these phenotypes has been found in CIB1-deficient patients presenting with isolated EV, even in late adulthood [152,153]. In fact, human CIB1 is strongly expressed in keratinocytes, in which it forms a complex with the EVER1 and EVER2 proteins [152]. CIB1 levels are very low in keratinocytes with deficiencies of EVER1 or EVER2 [152]. The requirement of EVER1 and EVER2 for CIB1 expression in keratinocytes accounts for EVER1, EVER2 and CIB1 deficiencies being clinical and virological phenocopies. These findings also suggest that this complex governs keratinocyte-intrinsic immunity to β-HPVs. However, the previously reported functions of EVER proteins, such as their interaction with the zinc transporter ZnT-1 [159], their control of the activity of the transcription factor AP-1 [159,160], and their involvement in cellular responses to TNF-α [160,161], were shown not to require CIB1 [152,153]. The disruption of these functions is not, therefore, the core mechanism of typical EV. By contrast, CIB1 does interact with E5 and E8, the viral proteins not expressed by β-HPVs, suggesting that the EVER-CIB1 complex acts as a restriction factor for HPVs in human keratinocytes [152]. Interestingly, the EVER-CIB1 complex is apparently not induced by anti-viral IFNs, unlike many other mechanisms of cell-intrinsic immunity [162165]. Overall, studies of EV suggest that keratinocyte-intrinsic immunity to β-HPVs is governed by the EVER-CIB1 complex.

Inborn errors underlying influenza pneumonitis: pulmonary epithelial cells

Influenza viruses, especially the influenza A virus (IAV), cause yearly seasonal and, more rarely, pandemic infections [166]. However, even during the worst ever pandemic, the 1918 “Spanish Flu” pandemic, only a minority (fewer than 10%) of symptomatic patients died. The proportion of lethal infections in the course of seasonal influenza is smaller still, with case-fatality ratios of only about 0.04-0.4% [167,168]. IAV is a single-stranded, segmented RNA virus from the Orthomyxoviridae family of viruses [166]. It is a respiratory virus that infects the lung epithelial cells, including both type 1 and 2 pneumocytes in vitro, with different preferences according to the strain concerned. Severe infections can cause acute respiratory distress syndrome (ARDS), which is characterized by damage to the epithelial-endothelial barrier, fluid leakage into the alveolar lumen, and respiratory insufficiency, often leading to multiple organ failure and associated with mortality rates of up to 60% [169]. In children, unlike adults, most cases of influenza ARDS cannot be accounted for by pre-existing cardiac or pulmonary lesions [168]. Influenza has an incubation period of only about 1-2 days and no viremia is detected before ARDS [170], suggesting that infection is initially confined to the lung, where pulmonary epithelial cell-intrinsic and resident alveolar macrophages probably play an important role in controlling the infection before leukocyte extravasation. Indeed, influenza ARDS has not been reported in children with any of the more than 200 primary immunodeficiencies affecting T and B cells, including severe combined immunodeficiencies and agammaglobulinemia [167,171]. Adaptive immunity is required for protective responses to influenza vaccination, but does not seem to be required for host defense against influenza virus. Collectively, these findings suggested that influenza ARDS in otherwise healthy, unvaccinated children might be due to single-gene inborn errors of cell-intrinsic or innate immunity.

Consistent with the genetic hypothesis, forward genetics approaches identified inherited Mxl deficiency as a strong determinant of vulnerability to influenza in mice [167,172]. Mx1 is an IFN-stimulated gene (ISG) product [172], so this observation also indicated the involvement of type I and III anti-viral IFNs. Human GATA2 deficiency, a primary immunodeficiency in which counts of dendritic cells, monocytes, NK cells, and B cells are low, underlying multiple infections, was recently reported to underlie influenza ARDS in adult patients, suggesting that IFN-producing plasmacytoid dendritic cells (pDCs) are crucial for immunity to influenza virus [173,174]. In 2015, AR IRF7 deficiency was identified as the first monogenic etiology of isolated influenza ARDS, in an otherwise healthy child [175]. IRF7 is a transcription factor that amplifies the expression of type I (13 IFNA, IFNB, IFNE, IFNK, IFNW) and type III (IL29, IL28A, IL28B) IFN genes. IRF7 deficiency is an inborn error of innate immunity, as the patient’s pDCs do not amplify the production of type I and III IFNs [175]. It is also an inborn error of cell-intrinsic immunity, as pulmonary epithelial cells (PECs) do not control viral growth [175]. These findings were corroborated by the subsequent discovery of an AR deficiency of IRF9, which normally forms ISGF3 with STAT1 and STAT2 upon stimulation with type I and III IFNs, in another child [176]. The patient’s fibroblasts did not activate ISGF3-dependent ISGs in response to type I IFNs and did not control viral growth [176]. The patient’s pDCs and PECs were not tested, but would not be predicted to form ISGF3 either. These newly identified inborn errors highlighted the importance of type I and III IFNs in the control of IAV infection. The relative contributions of PECs and pDCs (and other cell types) to the pathogenesis of influenza ARDS in IRF7- and IRF9-deficient patients are unknown. Interestingly, human patients with other inborn errors of type I and III IFN immunity have not been reported to suffer from severe influenza, suggesting that penetrance may be incomplete, due to a lack of infection or other mechanisms [177184]. Although the cellular basis of viral infections other than influenza seen in these patients is unclear, it may involve cells other than leukocytes. At any rate, the search for new genetic etiologies of influenza ARDS should clarify the molecular and cellular basis of this disease, including the potential contribution of IFN-dependent mechanisms in PECs.

Inborn errors underlying viral encephalitis: neurons and oligodendrocytes

At least twenty viruses can cause devastating encephalitis in humans, reaching the brain by crossing the blood-brain barrier (e.g. cytomegalovirus), via the peripheral nervous system (PNS) (e.g. herpes simplex virus 1 (HSV-1)), or both (e.g. influenza virus (IV)) [185]. Typically, viral encephalitis is not followed by dissemination of the virus to other organs, suggesting that its pathogenesis may involve an impairment of central nervous system (CNS)-specific immunity to viruses. Human genetic studies of isolated HSV-1 encephalitis (HSE) of the forebrain led to the discovery of single-gene inborn errors of the Toll-like receptor 3 (TLR3)-interferon (IFN)-α/β and -λ pathway, due to mono- or biallelic mutations of six TLR3 pathway genes (TLR3, UNC93B1, TRIF, TRAF3, TBK1 or IRF3) [186196]. Moreover, AR complete STAT1 deficiency [178,197,198] and X-linked partial NEMO deficiency [199201] were observed in children with mycobacterial disease who died of HSE. These findings suggested that TLR3-dependent IFN-α/β and -λ immunity is critical for host defense against HSV-1 in the CNS. TLR3-mediated antiviral immunity seems to be redundant in most TLR3-expressing cell types, including leukocytes in particular, accounting for the lack of viral dissemination during the course of HSE [188,192]. The hypothesis that CNS-specific cell-intrinsic immunity rather than leukocyte-mediated immunity is critical for host defense against neurotropic viral infection was then tested experimentally, initially with dermal fibroblasts and induced pluripotent stem cell (iPSC)-derived CNS- and PNS-resident cells from patients with forebrain HSE and mutations of TLR3 pathway genes. TLR3 pathway-deficient fibroblasts [187193] and iPSC-derived cortical neurons and oligodendrocytes [194] were much more susceptible to HSV-1 infection than control cells, probably due to the lack of TLR3-dependent IFN-β and IFN-λ responses. By contrast, in vitro-differentiated UNC-93B-deficient astrocytes or neural stem cells, and TLR3-deficient peripheral trigeminal neurons had a susceptibility to infection similar to that of control cells [202]. Microglial cells, the CNS resident macrophages, were not tested. TLR3-dependent, IFN-mediated cortical neuron- and oligodendrocyte-autonomous anti-HSV-1 immunity thus seems to be critical for host defense against HSV-1 infection of the human forebrain. These data provided a plausible mechanism for the pathogenesis of forebrain HSE [187,192].

Children with brainstem infections caused by HSV-1 or other viruses, including influenza B virus (IBV) and norovirus, have recently been studied. These children present inborn errors of RNA lariat metabolism, due to biallelic hypomorphic mutations of DBR1, which encodes the only known RNA lariat-debranching enzyme [203,204]. The antiviral responses of leukocytes from DBR1-deficient patients have not been studied, because of the unusual expression profile of DBR1. Indeed, DBR1 protein levels are highest in the brainstem and spinal cord, strongly suggesting that DBR1 deficiency disrupts immunity in brainstem-resident cells [203]. Inherited DBR1 deficiency probably underlies viral infection of the brainstem through the disruption of brainstem-specific and cell-intrinsic immunity to viruses. DBR1-deficient fibroblasts from patients, whose TLR3 and IFN-α/β responsive pathways were intact, have been studied as a proxy. They were found to be highly susceptible to HSV-1 and VSV, like TLR3- and STAT1-deficient fibroblasts [203]. The cellular basis of brainstem infection in patients with DBR1 mutations remains unknown, as iPSC-derived brainstem cells have not been tested. The molecular mechanism also remains elusive. DBR1-deficient fibroblasts have higher RNA lariat levels than control cells. This accumulation of RNA lariats may impair virus recognition by host cells, thereby damaging cell-intrinsic defenses against viral invasion. DBR1 may also regulate the processing of some host protein-coding RNAs, noncoding RNAs (ncRNAs) [205209], or viral RNA lariats [210213], thereby controlling cell-intrinsic defense against intracellular virus replication. Human genetic studies of viral encephalitis have thus shown that TLR3 governs cell-intrinsic immunity to HSV1 in the forebrain, whereas DBR1 governs cell-intrinsic immunity to various viruses in the brainstem. The genetic dissection of HSE and other types of viral encephalitis by forward genetics will pave the way for delineating the contribution of CNS-specific resident cells to protective immunity to viruses.

Conclusion

This review provides an overview of known examples of cell-intrinsic and cell-extrinsic mechanisms by which human non-professional, tissue-resident cells other than leukocytes contribute to protective immunity to infection in natural conditions (Table 1). These findings are probably no more than the tip of the iceberg. The range of cells and mechanisms involved in host defense is almost certainly much broader than previously appreciated. Most of the genetic etiologies of EV, influenza ARDS, and viral encephalitis, remain unknown. Future studies of these three infections will probably reveal novel mechanisms of organ- or tissue-specific, cell-intrinsic immunity to viruses. The pathogenesis of most other tissue- or organ-specific infections striking otherwise healthy patients remains unexplained. In addition, genetic resistance to infections other than P. vivax and norovirus infection, including, in particular, viral infections, which make use of only a few receptors to infect target cells, or microbial toxins that target a specific protein, will probably be discovered. Surprisingly, individuals seronegative for many common viruses can be identified in human populations. Finally, inborn errors of liver-produced complement and APOL1 may not be isolated examples of cell-extrinsic immunity by cells other than leukocytes. The forward genetic study of resistance and predisposition to various human infections, in terms of both microbial and anatomical diversity, may identify new components secreted by cells other than leukocytes as essential for certain types of host defense. Overall, it appears that protective immunity to the many microbes in our environment probably involves a much broader range of cells and molecules than initially thought based on the results of studies preferentially focusing on innate and adaptive leukocytes. Protective immunity requires many more cell types than are found in the classically defined immune system. It requires the whole organism.

Table 1.

Human inborn errors of immunity to infection affecting cells other than leukocytesa

Gene Relevant cell type Infectious phenotype References
DARC Erythrocytes Resistance to Plasmodium vivax 2631
FUT2 Intestinal epithelial cells Resistance to norovirus, rotavirus 35, 3841, 4446
C1 Hepatocytes Susceptibility to encapsulated pyogenic bacteria 5459, 7590, 91
C4
C2
C3
CFH
CFI
C5 Hepatocytes Susceptibility to invasive Neisseria infection 56, 93110
C6
C7
C8
C9
CFD
CFP
APOL1 Hepatocytes Susceptibility to Trypanosoma evansi 114
EVER1 Keratinocytes Susceptibility to β-human papillomaviruses 142144
EVER2 142147
CIB1 152, 153
IRF7 Pulmonary epithelial cells Susceptibility to influenza virus 175
IRF9 176
TLR3 Cortical neurons and oligodendrocytes Susceptibility to herpes simplex virus 1 188, 192194
UNC93B1 187, 194
TRIF 191
TRAF3 190
TBK1 189
IRF3 195
DBR1 Brainstem resident cells Susceptibility to various viruses 203
a.

See text for explanations.

Highlights.

  • Cells other than leukocytes can be essential for protective immunity in humans.

  • Human inborn errors of immunity to infection can affect non-professional cells, other than professional leukocytes.

  • Immunity to infection is not restricted to the immune system and involves the whole organism.

Acknowledgments

We thank the patients and their families for their participation in our studies. We thank all members of both branches of the St. Giles Laboratory of Human Genetics of Infectious Diseases for helpful discussions, and Dominick Papandrea, Cécile Patissier, and Yelena Nemirovskaya for administrative assistance. This work was funded, in part, by the National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH) Clinical and Translational Science Award (CTSA) program (UL1TR001866), NIH (R01NS072381, R01AI088364, R21AI137371, R21AI107508, R21NS084255), National Institute of Allergy and Infectious Diseases (NIAID) for a Cooperative Center on Human Immunology (CCHI) pilot award (U19AI111825), the French National Research Agency (ANR) under the “Investments for the future” program (ANR-10-IAHU-01), Integrative Biology of Emerging Infectious Diseases Laboratoire d’Excellence (ANR-10-LABX-62-IBEID), an IEIHSEER grant (ANR-14-CE14-0008-01), a SEAe-HostFactors grant (ANR-18-CE15-0020-02) and a grant from INCA/Cancéropole Ile-de-France (2013-1-PL BIO-11-INSERM 5-1), the Rockefeller University, Institut National de la Santé et de la Recherche Médicale (INSERM), the HHMI, Paris Descartes University, and the St. Giles Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declarations of interest: none.

References

  • 1.Vickaryous MK, Hall BK: Human cell type diversity, evolution, development, and classification with special reference to cells derived from the neural crest. Biol Rev Camb Philos Soc 2006, 81:425–455. [DOI] [PubMed] [Google Scholar]
  • 2.Silverstein A: A history of Immunology, 2nd Edition 2009. [Google Scholar]
  • 3.Pasteur L: Oeuvres complètes de Louis Pasteur, réunies par Pasteur Vallery-Radot. Paris: Masson et Cie; 1922-1939. [Google Scholar]
  • 4.Landsteiner K: The Specificity of Serological Reactions The Rockefeller Institute For Medical Research New York; Thomas Charles C Springfield Illinois; Baltimore Maryland: 1932. [Google Scholar]
  • 5.PMH M: Species and Specificity An Interpretation of the History of Immunology. Cambridge Univ Press; Cambridge, UK: 1995. [Google Scholar]
  • 6.Nathan C: Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 2006, 6:173–182. [DOI] [PubMed] [Google Scholar]
  • 7.Mass E, Ballesteros I, Farlik M, Halbritter F, Gunther P, Crozet L, Jacome-Galarza CE, Handler K, Klughammer J, Kobayashi Y, et al. : Specification of tissue-resident macrophages during organogenesis. Science 2016, 353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Perdiguero EG, Geissmann F: The development and maintenance of resident macrophages. Nat Immunol 2016, 17:2–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Fearon DT, Locksley RM: The instructive role of innate immunity in the acquired immune response. Science 1996, 272:50–53. [DOI] [PubMed] [Google Scholar]
  • 10.Silverstein AM: Cellular versus humoral immunology: a century-long dispute. Nat Immunol 2003, 4:425–428. [DOI] [PubMed] [Google Scholar]
  • 11.Rota IA, Dhalla F: FOXN1 deficient nude severe combined immunodeficiency. Orphanet J Rare Dis 2017, 12:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mehr S, Hsu P, Campbell D: Immunodeficiency in CHARGE syndrome. Am J Med Genet C Semin Med Genet 2017, 175:516–523. [DOI] [PubMed] [Google Scholar]
  • 13.Bolze A, Boisson B, Bosch B, Antipenko A, Bouaziz M, Sackstein P, Chaker-Margot M, Barlogis V, Briggs T, Colino E, et al. : Incomplete penetrance for isolated congenital asplenia in humans with mutations in translated and untranslated RPSA exons. Proc Natl Acad Sci U S A 2018, 115:E8007–E8016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Piel FB, Steinberg MH, Rees DC: Sickle Cell Disease. N Engl J Med 2017, 377:305. [DOI] [PubMed] [Google Scholar]
  • 15.Booth C, Inusa B, Obaro SK: Infection in sickle cell disease: a review. Int J Infect Dis 2010, 14:e2–e12. [DOI] [PubMed] [Google Scholar]
  • 16.Massip-Copiz MM, Santa-Coloma TA: Extracellular pH and lung infections in cystic fibrosis. Eur J Cell Biol 2018, 97:402–410. [DOI] [PubMed] [Google Scholar]
  • 17.Kiedrowski MR, Bomberger JM: Viral-Bacterial Co-infections in the Cystic Fibrosis Respiratory Tract. Front Immunol 2018, 9:3067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lu M, Saddi V, Britton PN, Selvadurai H, Robinson PD, Pandit C, Marais BJ, Fitzgerald DA: Disease caused by non-tuberculous mycobacteria in children with cystic fibrosis. Paediatr Respir Rev 2018. [DOI] [PubMed] [Google Scholar]
  • 19.Schwarz C, Hartl D, Eickmeier O, Hector A, Benden C, Durieu I, Sole A, Gartner S, Milla CE, Barry PJ: Progress in Definition, Prevention and Treatment of Fungal Infections in Cystic Fibrosis. Mycopathologia 2018, 183:21–32. [DOI] [PubMed] [Google Scholar]
  • 20.Plasschaert LW, Zilionis R, Choo-Wing R, Savova V, Knehr J, Roma G, Klein AM, Jaffe AB: A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 2018, 560:377–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Brousse V, Buffet P, Rees D: The spleen and sickle cell disease: the sick(led) spleen. Br J Haematol 2014, 166:165–176. [DOI] [PubMed] [Google Scholar]
  • 22.Emond MJ, Louie T, Emerson J, Zhao W, Mathias RA, Knowles MR, Wright FA, Rieder MJ, Tabor HK, Nickerson DA, et al. : Exome sequencing of extreme phenotypes identifies DCTN4 as a modifier of chronic Pseudomonas aeruginosa infection in cystic fibrosis. Nat Genet 2012, 44:886–889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Casanova JL, Abel L: The human model: a genetic dissection of immunity to infection in natural conditions. Nat Rev Immunol 2004, 4:55–66. [DOI] [PubMed] [Google Scholar]
  • 24.Quintana-Murci L, Alcais A, Abel L, Casanova JL: Immunology in natura: clinical, epidemiological and evolutionary genetics of infectious diseases. Nat Immunol 2007, 8:1165–1171. [DOI] [PubMed] [Google Scholar]
  • 25.Casanova JL, Abel L, Quintana-Murci L: Immunology Taught by Human Genetics. Cold Spring Harbor symposia on quantitative biology 2013, 78:157–172. [DOI] [PubMed] [Google Scholar]
  • 26.Miller LH, Mason SJ, Clyde DF, McGinniss MH: The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl J Med 1976, 295:302–304. [DOI] [PubMed] [Google Scholar]
  • 27.Barnwell JW, Nichols ME, Rubinstein P: In vitro evaluation of the role of the Duffy blood group in erythrocyte invasion by Plasmodium vivax. J Exp Med 1989, 169:1795–1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Miller LH, Mason SJ, Dvorak JA, McGinniss MH, Rothman IK: Erythrocyte receptors for (Plasmodium knowlesi) malaria: Duffy blood group determinants. Science 1975, 189:561–563. [DOI] [PubMed] [Google Scholar]
  • 29.Tournamille C, Colin Y, Cartron JP, Le Van Kim C: Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individuals. Nat Genet 1995, 10:224–228. [DOI] [PubMed] [Google Scholar]
  • 30.Howes RE, Patil AP, Piel FB, Nyangiri OA, Kabaria CW, Gething PW, Zimmerman PA, Barnadas C, Beall CM, Gebremedhin A, et al. : The global distribution of the Duffy blood group. Nat Commun 2011, 2:266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.McManus KF, Taravella AM, Henn BM, Bustamante CD, Sikora M, Cornejo OE: Population genetic analysis of the DARC locus (Duffy) reveals adaptation from standing variation associated with malaria resistance in humans. PLoS Genet 2017, 13:e1006560. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Population genetic study of the DARC locus estimating the age of the most recent common ancestor carrying the most common FY*O null mutation to be ~42,000 years. This mutation swept to fixation in Africa from a very low frequency (0.1%), with one of the strongest selection coefficients in the human genome.
  • 32.Casanova JL, Abel L: Human genetics of infectious diseases: Unique insights into immunological redundancy. Semin Immunol 2018, 36:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]; * A classification of human genes into four groups, according to their redundancy in host defense. This is of potential interest to biologists testing immunological hypotheses experimentally and physicians managing patients with immunological or infectious conditions.
  • 33.Menard D, Barnadas C, Bouchier C, Henry-Halldin C, Gray LR, Ratsimbasoa A, Thonier V, Carod JF, Domarle O, Colin Y, et al. : Plasmodium vivax clinical malaria is commonly observed in Duffy-negative Malagasy people. Proc Natl Acad Sci U S A 2010, 107:5967–5971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Niangaly A, Karthigayan G, Amed O, Coulibaly D, Sa JM, Adams M, Travassos MA, Ferrero J, Laurens MB, Kone AK, et al. : Plasmodium vivax Infections over 3 Years in Duffy Blood Group Negative Malians in Bandiagara, Mali. Am J Trop MedHyg 2017, 97:744–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Le Pendu J, Ruvoen-Clouet N, Kindberg E, Svensson L: Mendelian resistance to human norovirus infections. Semin Immunol 2006, 18:375–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Marionneau S, Cailleau-Thomas A, Rocher J, Le Moullac-Vaidye B, Ruvoen N, Clement M, Le Pendu J: ABH and Lewis histo-blood group antigens, a model for the meaning of oligosaccharide diversity in the face of a changing world. Biochimie 2001, 83:565–573. [DOI] [PubMed] [Google Scholar]
  • 37.Huang P, Xia M, Tan M, Zhong W, Wei C, Wang L, Morrow A, Jiang X: Spike protein VP8* of human rotavirus recognizes histo-blood group antigens in a type-specific manner. J Virol 2012, 86:4833–4843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lindesmith L, Moe C, Marionneau S, Ruvoen N, Jiang X, Lindblad L, Stewart P, LePendu J, Baric R: Human susceptibility and resistance to Norwalk virus infection. Nat Med 2003, 9:548–553. [DOI] [PubMed] [Google Scholar]
  • 39.Thorven M, Grahn A, Hedlund KO, Johansson H, Wahlfrid C, Larson G, Svensson L: A homozygous nonsense mutation (428G-->A) in the human secretor (FUT2) gene provides resistance to symptomatic norovirus (GGII) infections. J Virol 2005, 79:15351–15355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nordgren J, Sharma S, Bucardo F, Nasir W, Gunaydin G, Ouermi D, Nitiema LW, Becker-Dreps S, Simpore J, Hammarstrom L, et al. : Both Lewis and secretor status mediate susceptibility to rotavirus infections in a rotavirus genotype-dependent manner. Clin Infect Dis 2014, 59:1567–1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Payne DC, Currier RL, Staat MA, Sahni LC, Selvarangan R, Halasa NB, Englund JA, Weinberg GA, Boom JA, Szilagyi PG, et al. : Epidemiologic Association Between FUT2 Secretor Status and Severe Rotavirus Gastroenteritis in Children in the United States. JAMA Pediatr 2015, 169:1040–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Marionneau S, Ruvoen N, Le Moullac-Vaidye B, Clement M, Cailleau-Thomas A, Ruiz-Palacois G, Huang P, Jiang X, Le Pendu J: Norwalk virus binds to histo-blood group antigens present on gastroduodenal epithelial cells of secretor individuals. Gastroenterology 2002, 122:1967–1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ma X, Li DD, Sun XM, Guo YQ, Xiang JY, Wang WH, Zhang LX, Gu QJ, Duan ZJ: Binding Patterns of Rotavirus Genotypes P[4], P[6], and P[8] in China with Histo-Blood Group Antigens. PLoS One 2015, 10:e0134584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Le Pendu J, Nystrom K, Ruvoen-Clouet N: Host-pathogen co-evolution and glycan interactions. Curr Opin Virol 2014, 7:88–94. [DOI] [PubMed] [Google Scholar]
  • 45.Kambhampati A, Payne DC, Costantini V, Lopman BA: Host Genetic Susceptibility to Enteric Viruses: A Systematic Review and Metaanalysis. Clin Infect Dis 2016, 62:11–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Barbe L, Le Moullac-Vaidye B, Echasserieau K, Bernardeau K, Carton T, Bovin N, Nordgren J, Svensson L, Ruvoen-Clouet N, Le Pendu J: Histo-blood group antigenbinding specificities of human rotaviruses are associated with gastroenteritis but not with in vitro infection. Sci Rep 2018, 8:12961. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This study shows that fucosylation is not a requirement for in vitro infection of transformed cells by P[4] and P[8] rotavirus strains, suggesting that HBGAs could enhance viral replication and development of disease but may not be essential for infection per se.
  • 47.Zhang D, Tan M, Zhong W, Xia M, Huang P, Jiang X: Human intestinal organoids express histo-blood group antigens, bind norovirus VLPs, and support limited norovirus replication. Sci Rep 2017, 7:12621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.McGovern DP, Jones MR, Taylor KD, Marciante K, Yan X, Dubinsky M, Ippoliti A, Vasiliauskas E, Berel D, Derkowski C, et al. : Fucosyltransferase 2 (FUT2) non-secretor status is associated with Crohn’s disease. Hum Mol Genet 2010, 19:3468–3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Takeuchi M, Mizuki N, Meguro A, Ombrello MJ, Kirino Y, Satorius C, Le J, Blake M, Erer B, Kawagoe T, et al. : Dense genotyping of immune-related loci implicates host responses to microbial exposure in Behcet’s disease susceptibility. Nat Genet 2017, 49:438–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Santos-Cortez RLP, Chiong CM, Frank DN, Ryan AF, Giese APJ, Bootpetch Roberts T, Daly KA, Steritz MJ, Szeremeta W, Pedro M, et al. : FUT2 Variants Confer Susceptibility to Familial Otitis Media. Am JHum Genet 2018, 103:679–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ferrer-Admetlla A, Sikora M, Laayouni H, Esteve A, Roubinet F, Blancher A, Calafell F, Bertranpetit J, Casals F: A natural history of FUT2 polymorphism in humans. Mol Biol Evol 2009, 26:1993–2003. [DOI] [PubMed] [Google Scholar]
  • 52.Rausch P, Rehman A, Kunzel S, Hasler R, Ott SJ, Schreiber S, Rosenstiel P, Franke A, Baines JF: Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (Secretor) genotype. Proc Natl Acad Sci US A 2011, 108:19030–19035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Walport MJ: Complement. First of two parts. N Engl JMed 2001, 344:1058–1066. [DOI] [PubMed] [Google Scholar]
  • 54.Alper CA, Abramson N, Johnston RB Jr., Jandl JH, Rosen FS: Increased susceptibility to infection associated with abnormalities of complement-mediated functions and of the third component of complement (C3). N Engl J Med 1970, 282:349–354. [DOI] [PubMed] [Google Scholar]
  • 55.Miller ME, Seals J, Kaye R, Levitsky LC: A familial, plasma associated defect of phagocytosis: a new cause of recurrent bacterial infections. Lancet 1968, ii:60–63. [Google Scholar]
  • 56.Figueroa JE, Densen P: Infectious diseases associated with complement deficiencies. Clin Microbiol Rev 1991, 4:359–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ram S, Lewis LA, Rice PA: Infections of people with complement deficiencies and patients who have undergone splenectomy. Clin Microbiol Rev 2010, 23:740–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Klemperer MR, Woodworth HC, Rosen FS, Austen KF: Hereditary deficiency of the second component of complement (C′2) in man. J Clin Invest 1966, 45:880–890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Klemperer MR, Austen KF, Rosen FS: Hereditary deficiency of the second component of complement (C′2) in man: further observations on a second kindred. J Immunol 1967, 98:72–78. [PubMed] [Google Scholar]
  • 60.Sarma JV, Ward PA: The complement system. Cell Tissue Res 2011, 343:227–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hajishengallis G, Reis ES, Mastellos DC, Ricklin D, Lambris JD: Novel mechanisms and functions of complement. Nat Immunol 2017, 18:1288–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** A comprehensive review of recent advances in the field of complement. It provides an updated view of new and previously unanticipated functions of complement in health and disease.
  • 62.Alper CA, Johnson AM, Birtch AG, Moore FD: Human C′3: evidence for the liver as the primary site of synthesis. Science 1969, 163:286–288. [DOI] [PubMed] [Google Scholar]
  • 63.Morris KM, Aden DP, Knowles BB, Colten HR: Complement biosynthesis by the human hepatoma-derived cell line HepG2. J Clin Invest 1982, 70:906–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Perlmutter DH, Colten HR: Molecular immunobiology of complement biosynthesis: a model of single-cell control of effector-inhibitor balance. Annu Rev Immunol 1986, 4:231–251. [DOI] [PubMed] [Google Scholar]
  • 65.Baumann H, Richards C, Gauldie J: Interaction among hepatocyte-stimulating factors, interleukin 1, and glucocorticoids for regulation of acute phase plasma proteins in human hepatoma (HepG2) cells. J Immunol 1987, 139:4122–4128. [PubMed] [Google Scholar]
  • 66.Mier JW, Dinarello CA, Atkins MB, Punsal PI, Perlmutter DH: Regulation of hepatic acute phase protein synthesis by products of interleukin 2 (IL 2)-stimulated human peripheral blood mononuclear cells. J Immunol 1987, 139:1268–1272. [PubMed] [Google Scholar]
  • 67.Morgan BP, Gasque P: Extrahepatic complement biosynthesis: where, when and why? Clin Exp Immunol 1997, 107:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tenner AJ, Volkin DB: Complement subcomponent C1q secreted by cultured human monocytes has subunit structure identical with that of serum C1q. Biochem J 1986, 233:451–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gulati P, Lemercier C, Guc D, Lappin D, Whaley K: Regulation of the synthesis of C1 subcomponents and C1-inhibitor. Behring Inst Mitt 1993:196–203. [PubMed] [Google Scholar]
  • 70.Choy LN, Rosen BS, Spiegelman BM: Adipsin and an endogenous pathway of complement from adipose cells. J Biol Chem 1992, 267:12736–12741. [PubMed] [Google Scholar]
  • 71.White RT, Damm D, Hancock N, Rosen BS, Lowell BB, Usher P, Flier JS, Spiegelman BM: Human adipsin is identical to complement factor D and is expressed at high levels in adipose tissue. J Biol Chem 1992, 267:9210–9213. [PubMed] [Google Scholar]
  • 72.Maves KK, Weiler JM: Properdin: approaching four decades of research. Immunol Res 1993, 12:233–243. [DOI] [PubMed] [Google Scholar]
  • 73.Wurzner R, Joysey VC, Lachmann PJ: Complement component C7. Assessment of in vivo synthesis after liver transplantation reveals that hepatocytes do not synthesize the majority of human C7. J Immunol 1994, 152:4624–4629. [PubMed] [Google Scholar]
  • 74.Alper CA, Raum D, Awdeh ZL, Petersen BH, Taylor PD, Starzl TE: Studies of hepatic synthesis in vivo of plasma proteins, including orosomucoid, transferrin, alpha 1-antitrypsin, C8, and factor B. Clin ImmunolImmunopathol 1980, 16:84–89. [DOI] [PubMed] [Google Scholar]
  • 75.Ross SC, Densen P: Complement deficiency states and infection: epidemiology, pathogenesis and consequences of neisserial and other infections in an immune deficiency. Medicine (Baltimore) 1984, 63:243–273. [PubMed] [Google Scholar]
  • 76.Schur PH: Inherited complement component abnormalities. Annu Rev Med 1986, 37:333–346. [DOI] [PubMed] [Google Scholar]
  • 77.Schifferli JA, Peters DK: Complement, the immune-complex lattice, and the pathophysiology of complement-deficiency syndromes. Lancet 1983, 2:957–959. [DOI] [PubMed] [Google Scholar]
  • 78.Hannema AJ, Kluin-Nelemans JC, Hack CE, Eerenberg-Belmer AJ, Mallee C, van Helden HP: SLE like syndrome and functional deficiency of C1q in members of a large family. Clin Exp Immunol 1984, 55:106–114. [PMC free article] [PubMed] [Google Scholar]
  • 79.Meyer O, Hauptmann G, Tappeiner G, Ochs HD, Mascart-Lemone F: Genetic deficiency of C4, C2 or C1q and lupus syndromes. Association with anti-Ro (SS-A) antibodies. Clin Exp Immunol 1985, 62:678–684. [PMC free article] [PubMed] [Google Scholar]
  • 80.Gelfand EW, Rao CP, Minta JO, Ham T, Purkall DB, Ruddy S: Inherited deficiency of properdin and C2 in a patient with recurrent bacteremia. Am J Med 1987, 82:671–675. [DOI] [PubMed] [Google Scholar]
  • 81.Sjoholm AG, Hallberg T, Oxelius VA, Hammarstrom L, Smith CI, Lindgren F: C2 deficiency, moderately low IgG2 concentrations and lack of the G2m(23) allotype marker in a child with repeated bacterial infections. Acta Paediatr Scand 1987, 76:533–538. [DOI] [PubMed] [Google Scholar]
  • 82.Hauptmann G, Goetz J, Uring-Lambert B, Grosshans E: Component deficiencies. 2. The fourth component. Prog Allergy 1986, 39:232–249. [PubMed] [Google Scholar]
  • 83.Uring-Lambert B, Mascart-Lemone F, Tongio MM, Goetz J, Hauptmann G: Molecular basis of complete C4 deficiency. A study of three patients. Hum Immunol 1989, 24:125–132. [DOI] [PubMed] [Google Scholar]
  • 84.Grumach AS, Vilela MM, Gonzalez CH, Starobinas N, Pereira AB, Dias-da-Silva W, Carneiro-Sampaio MM: Inherited C3 deficiency of the complement system. Braz J Med Biol Res 1988, 21:247–257. [PubMed] [Google Scholar]
  • 85.Fijen CA, Kuijper EJ, Hannema AJ, Sjoholm AG, van Putten JP: Complement deficiencies in patients over ten years old with meningococcal disease due to uncommon serogroups. Lancet 1989, 2:585–588. [DOI] [PubMed] [Google Scholar]
  • 86.Botto M, Fong KY, So AK, Rudge A, Walport MJ: Molecular basis of hereditary C3 deficiency. J Clin Invest 1990, 86:1158–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Borzy MS, Houghton D: Mixed-pattern immune deposit glomerulonephritis in a child with inherited deficiency of the third component of complement. Am J Kidney Dis 1985, 5:54–59. [DOI] [PubMed] [Google Scholar]
  • 88.Barrett DJ, Boyle MD: Restoration of complement function in vivo by plasma infusion in factor I (C3b inactivator) deficiency. J Pediatr 1984, 104:76–81. [DOI] [PubMed] [Google Scholar]
  • 89.Rasmussen JM, Teisner B, Brandslund I, Svehag SE: A family with complement factor I deficiency. Scand J Immunol 1986, 23:711–715. [DOI] [PubMed] [Google Scholar]
  • 90.Nielsen HE, Christensen KC, Koch C, Thomsen BS, Heegaard NH, Tranum-Jensen J: Hereditary, complete deficiency of complement factor H associated with recurrent meningococcal disease. Scand J Immunol 1989, 30:711–718. [DOI] [PubMed] [Google Scholar]
  • 91.Alper CA, Colten HR, Rosen FS, Rabson AR, Macnab GM, Gear JS: Homozygous deficiency of C3 in a patient with repeated infections. Lancet 1972, 2:1179–1181. [DOI] [PubMed] [Google Scholar]
  • 92.Botto M, Fong KY, So AK, Barlow R, Routier R, Morley BJ, Walport MJ: Homozygous hereditary C3 deficiency due to a partial gene deletion. Proc Natl Acad Sci U S A 1992, 89:4957–4961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wang X, Fleischer DT, Whitehead WT, Haviland DL, Rosenfeld SI, Leddy JP, Snyderman R, Wetsel RA: Inherited human complement C5 deficiency. Nonsense mutations in exons 1 (Gln1 to Stop) and 36 (Arg1458 to Stop) and compound heterozygosity in three African-American families. J Immunol 1995, 154:5464–5471. [PubMed] [Google Scholar]
  • 94.Wurzner R, Hobart MJ, Fernie BA, Mewar D, Potter PC, Orren A, Lachmann PJ: Molecular basis of subtotal complement C6 deficiency. A carboxy-terminally truncated but functionally active C6. J Clin Invest 1995, 95:1877–1883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kaufmann T, Hansch G, Rittner C, Spath P, Tedesco F, Schneider PM: Genetic basis of human complement C8 beta deficiency. J Immunol 1993, 150:4943–4947. [PubMed] [Google Scholar]
  • 96.Kojima T, Horiuchi T, Nishizaka H, Fukumori Y, Amano T, Nagasawa K, Niho Y, Hayashi K: Genetic basis of human complement C8 alpha-gamma deficiency. J Immunol 1998, 161:3762–3766. [PubMed] [Google Scholar]
  • 97.Westberg J, Fredrikson GN, Truedsson L, Sjoholm AG, Uhlen M: Sequence-based analysis of properdin deficiency: identification of point mutations in two phenotypic forms of an X-linked immunodeficiency. Genomics 1995, 29:1–8. [DOI] [PubMed] [Google Scholar]
  • 98.Biesma DH, Hannema AJ, van Velzen-Blad H, Mulder L, van Zwieten R, Kluijt I, Roos D: A family with complement factor D deficiency. J Clin Invest 2001, 108:233–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lim D, Gewurz A, Lint TF, Ghaze M, Sepheri B, Gewurz H: Absence of the sixth component of complement in a patient with repeated episodes of meningococcal meningitis. J Pediatr 1976, 89:42–47. [DOI] [PubMed] [Google Scholar]
  • 100.Petersen BH, Graham JA, Brooks GF: Human deficiency of the eighth component of complement. The requirement of C8 for serum Neisseria gonorrhoeae bactericidal activity. J Clin Invest 1976, 57:283–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hiemstra PS, Langeler E, Compier B, Keepers Y, Leijh PC, van den Barselaar MT, Overbosch D, Daha MR: Complete and partial deficiencies of complement factor D in a Dutch family. J Clin Invest 1989, 84:1957–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Rosenfeld SI, Kelly ME, Leddy JP: Hereditary deficiency of the fifth component of complement in man. I. Clinical, immunochemical, and family studies. J Clin Invest 1976, 57:1626–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Harriman GR, Esser AF, Podack ER, Wunderlich AC, Braude AI, Lint TF, Curd JG: The role of C9 in complement-mediated killing of Neisseria. J Immunol 1981, 127:2386–2390. [PubMed] [Google Scholar]
  • 104.Snyderman R, Durack DT, McCarty GA, Ward FE, Meadows L: Deficiency of the fifth component of complement in human subjects. Clinical, genetic and immunologic studies in a large kindred. Am J Med 1979, 67:638–645. [DOI] [PubMed] [Google Scholar]
  • 105.Lee TJ, Utsinger PD, Snyderman R, Yount WJ, Sparling PF: Familial deficiency of the seventh component of complement associated with recurrent bacteremic infections due to Neisseria. J Infect Dis 1978, 138:359–368. [DOI] [PubMed] [Google Scholar]
  • 106.Sjoholm AG, Braconier JH, Soderstrom C: Properdin deficiency in a family with fulminant meningococcal infections. Clin Exp Immunol 1982, 50:291–297. [PMC free article] [PubMed] [Google Scholar]
  • 107.Owen EP, Wurzner R, Leisegang F, Rizkallah P, Whitelaw A, Simpson J, Thomas AD, Harris CL, Giles JL, Hellerud BC, et al. : A complement C5 gene mutation, c.754G>A:p.A252T, is common in the Western Cape, South Africa and found to be homozygous in seven percent of Black African meningococcal disease cases. Mol Immunol 2015, 64:170–176. [DOI] [PubMed] [Google Scholar]
  • 108.Nishizaka H, Horiuchi T, Zhu ZB, Fukumori Y, Volanakis JE: Genetic bases of human complement C7 deficiency. J Immunol 1996, 157:4239–4243. [PubMed] [Google Scholar]
  • 109.Nagata M, Hara T, Aoki T, Mizuno Y, Akeda H, Inaba S, Tsumoto K, Ueda K: Inherited deficiency of ninth component of complement: an increased risk of meningococcal meningitis. J Pediatr 1989, 114:260–264. [DOI] [PubMed] [Google Scholar]
  • 110.Petersen BH, Lee TJ, Snyderman R, Brooks GF: Neisseria meningitidis and Neisseria gonorrhoeae bacteremia associated with C6, C7, or C8 deficiency. Ann Intern Med 1979, 90:917–920. [DOI] [PubMed] [Google Scholar]
  • 111.Page NM, Butlin DJ, Lomthaisong K, Lowry PJ: The human apolipoprotein L gene cluster: identification, classification, and sites of distribution. Genomics 2001, 74:71–78. [DOI] [PubMed] [Google Scholar]
  • 112.Duchateau PN, Pullinger CR, Orellana RE, Kunitake ST, Naya-Vigne J, O’Connor PM, Malloy MJ, Kane JP: Apolipoprotein L, a new human high density lipoprotein apolipoprotein expressed by the pancreas. Identification, cloning, characterization, and plasma distribution of apolipoprotein L. J Biol Chem 1997, 272:25576–25582. [DOI] [PubMed] [Google Scholar]
  • 113.Duchateau PN, Pullinger CR, Cho MH, Eng C, Kane JP: Apolipoprotein L gene family: tissue-specific expression, splicing, promoter regions; discovery of a new gene. J Lipid Res 2001, 42:620–630. [PubMed] [Google Scholar]
  • 114.Vanhollebeke B, Truc P, Poelvoorde P, Pays A, Joshi PP, Katti R, Jannin JG, Pays E: Human Trypanosoma evansi infection linked to a lack of apolipoprotein L-I. N Engl J Med 2006, 355:2752–2756. [DOI] [PubMed] [Google Scholar]
  • 115.Fontaine F, Lecordier L, Vanwalleghem G, Uzureau P, Van Reet N, Fontaine M, Tebabi P, Vanhollebeke B, Buscher P, Perez-Morga D, et al. : APOLs with low pH dependence can kill all African trypanosomes. Nat Microbiol 2017, 2:1500–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** This study shows that the difference in trypanolytic activity between recombinant APOL3 and APOL1 is accounted for by their different dependence on acidic pH.
  • 116.Pays E, Vanhollebeke B, Uzureau P, Lecordier L, Perez-Morga D: The molecular arms race between African trypanosomes and humans. Nat Rev Microbiol 2014, 12:575–584. [DOI] [PubMed] [Google Scholar]
  • 117.Vanhamme L, Paturiaux-Hanocq F, Poelvoorde P, Nolan DP, Lins L, Van Den Abbeele J, Pays A, Tebabi P, Van Xong H, Jacquet A, et al. : Apolipoprotein L-I is the trypanosome lytic factor of human serum. Nature 2003, 422:83–87. [DOI] [PubMed] [Google Scholar]
  • 118.Perez-Morga D, Vanhollebeke B, Paturiaux-Hanocq F, Nolan DP, Lins L, Homble F, Vanhamme L, Tebabi P, Pays A, Poelvoorde P, et al. : Apolipoprotein L-I promotes trypanosome lysis by forming pores in lysosomal membranes. Science 2005, 309:469–472. [DOI] [PubMed] [Google Scholar]
  • 119.Kennedy PG: Human African trypanosomiasis of the CNS: current issues and challenges. J Clin Invest 2004, 113:496–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Chappuis F, Loutan L, Simarro P, Lejon V, Buscher P: Options for field diagnosis of human african trypanosomiasis. Clin Microbiol Rev 2005, 18:133–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Checchi F, Filipe JA, Barrett MP, Chandramohan D: The natural progression of Gambiense sleeping sickness: what is the evidence? PLoS Negl Trop Dis 2008, 2:e303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Uzureau P, Uzureau S, Lecordier L, Fontaine F, Tebabi P, Homble F, Grelard A, Zhendre V, Nolan DP, Lins L, et al. : Mechanism of Trypanosoma brucei gambiense resistance to human serum. Nature 2013, 501:430–434. [DOI] [PubMed] [Google Scholar]
  • 123.Xong HV, Vanhamme L, Chamekh M, Chimfwembe CE, Van Den Abbeele J, Pays A, Van Meirvenne N, Hamers R, De Baetselier P, Pays E: A VSG expression site-associated gene confers resistance to human serum in Trypanosoma rhodesiense. Cell 1998, 95:839–846. [DOI] [PubMed] [Google Scholar]
  • 124.Genovese G, Friedman DJ, Ross MD, Lecordier L, Uzureau P, Freedman BI, Bowden DW, Langefeld CD, Oleksyk TK, Uscinski Knob AL, et al. : Association of trypanolytic ApoL1 variants with kidney disease in African Americans. Science 2010, 329:841–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Cooper A, Ilboudo H, Alibu VP, Ravel S, Enyaru J, Weir W, Noyes H, Capewell P, Camara M, Milet J, et al. : APOL1 renal risk variants have contrasting resistance and susceptibility associations with African trypanosomiasis. Elife 2017, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lowy DH, P M: Papillomaviruses Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, Roizman B, et al. , editors Fields Virology 4th ed Philadelphia: Lippincott Williams & Wilkins; 2001:2231–2246. [Google Scholar]
  • 127.Orth G: Papillomaviruses-Human (Papovaviridae). General Features . Granoff A, Webster RG, editors Encyclopedia of Virology 2nd ed London: Academic Press; 1999:1105–1114. [Google Scholar]
  • 128.Casanova JL: Severe infectious diseases of childhood as monogenic inborn errors of immunity. Proc Natl Acad Sci U S A 2015, 112:E7128–7137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Casanova JL: Human genetic basis of interindividual variability in the course of infection. Proc Natl Acad Sci US A 2015, 112:E7118–7127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Orth G: Genetics of epidermodysplasia verruciformis: Insights into host defense against papillomaviruses. Semin Immunol 2006, 18:362–374. [DOI] [PubMed] [Google Scholar]
  • 131.Lewandowsky F, Lutz W: Ein Fall einer bisher nicht beschriebenen Hauterkrankung (Epidermodysplasia verruciformis). Arch Dermatol Syphilol 1922, 141:193–203. [Google Scholar]
  • 132.Cockayne E: Inherited abnormalities of the skin and its appendages. Oxford University Press; 1933, London. [Google Scholar]
  • 133.Lutz W: A propos de l’epidermodysplasie verruciforme. Dermatologica 1946:30–43. [PubMed] [Google Scholar]
  • 134.Lutzner MA: Epidermodysplasia verruciformis. An autosomal recessive disease characterized by viral warts and skin cancer. A model for viral oncogenesis. Bull Cancer 1978:169–182. [PubMed] [Google Scholar]
  • 135.Burger B, Itin PH: Epidermodysplasia verruciformis. Curr Probl Dermatol 2014, 45:123–131. [DOI] [PubMed] [Google Scholar]
  • 136.Orth G: Host defenses against human papillomaviruses: lessons from epidermodysplasia verruciformis. Curr Top Microbiol Immunol 2008, 321:59–83. [DOI] [PubMed] [Google Scholar]
  • 137.de Jong SJ, Imahorn E, Itin P, Uitto J, Orth G, Jouanguy E, Casanova JL, Burger B: Epidermodysplasia Verruciformis: Inborn Errors of Immunity to Human Beta-Papillomaviruses. Front Microbiol 2018, 9:1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Huang S, Wu JH, Lewis DJ, Rady PL, Tyring SK: A novel approach to the classification of epidermodysplasia verruciformis. Int J Dermatol 2018, 57:1344–1350. [DOI] [PubMed] [Google Scholar]
  • 139.Orth G, Jablonska S, Breitburd F, Favre M, Croissant O: The human papillomaviruses. Bull Cancer 1978, 65:151–164. [PubMed] [Google Scholar]
  • 140.Antonsson A, Erfurt C, Hazard K, Holmgren V, Simon M, Kataoka A, Hossain S, Hakangard C, Hansson BG: Prevalence and type spectrum of human papillomaviruses in healthy skin samples collected in three continents. J Gen Virol 2003, 84:1881–1886. [DOI] [PubMed] [Google Scholar]
  • 141.Antonsson A, Karanfilovska S, Lindqvist PG, Hansson BG: General acquisition of human papillomavirus infections of skin occurs in early infancy. J Clin Microbiol 2003, 41:2509–2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Youssefian L, Vahidnezhad H, Mahmoudi H, Saeidian AH, Daneshpazhooh M, Kamyab Hesari K, Zeinali S, de Jong SJ, Orth G, Blanchet-Bardon C, et al. : Epidermodysplasia Verruciformis: Genetic Heterogeneity and EVER1 and EVER2 Mutations Revealed by Genome-Wide Analysis. J Invest Dermatol 2019, 139:241–244. [DOI] [PubMed] [Google Scholar]
  • 143.Ramoz N, Rueda LA, Bouadjar B, Montoya LS, Orth G, Favre M: Mutations in two adjacent novel genes are associated with epidermodysplasia verruciformis. Nat Genet 2002, 32:579–581. [DOI] [PubMed] [Google Scholar]
  • 144.Ramoz N, Taieb A, Rueda LA, Montoya LS, Bouadjar B, Favre M, Orth G: Evidence for a nonallelic heterogeneity of epidermodysplasia verruciformis with two susceptibility loci mapped to chromosome regions 2p21-p24 and 17q25. J Invest Dermatol 2000, 114:1148–1153. [DOI] [PubMed] [Google Scholar]
  • 145.Arnold AW, Burger B, Kump E, Rufle A, Tyring SK, Kempf W, Hausermann P, Itin PH: Homozygosity for the c.917A-->T (p.N306l) polymorphism in the EVER2/TMC8 gene of two sisters with epidermodysplasia verruciformis Lewandowsky-Lutz originally described by Wilhelm Lutz. Dermatology 2011, 222:81–86. [DOI] [PubMed] [Google Scholar]
  • 146.Imahorn E, Yuksel Z, Spoerri I, Gurel G, Imhof C, Saracoglu ZN, Koku Aksu AE, Rady PL, Tyring SK, Kempf W, et al. : Novel TMC8 splice site mutation in epidermodysplasia verruciformis and review of HPV infections in patients with the disease. J Eur Acad Dermatol Venereol 2017, 31:1722–1726. [DOI] [PubMed] [Google Scholar]
  • 147.Landini MM, Zavattaro E, Borgogna C, Azzimonti B, De Andrea M, Colombo E, Marenco F, Amantea A, Landolfo S, Gariglio M: Lack of EVER2 protein in two epidermodysplasia verruciformis patients with skin cancer presenting previously unreported homozygous genetic deletions in the EVER2 gene. J Invest Dermatol 2012, 132:1305–1308. [DOI] [PubMed] [Google Scholar]
  • 148.Crequer A, Picard C, Pedergnana V, Lim A, Zhang SY, Abel L, Majewski S, Casanova JL, Jablonska S, Orth G, et al. : EVER2 deficiency is associated with mild T-cell abnormalities. J Clin Immunol 2013, 33:14–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Lazarczyk M, Dalard C, Hayder M, Dupre L, Pignolet B, Majewski S, Vuillier F, Favre M, Liblau RS: EVER proteins, key elements of the natural anti-human papillomavirus barrier, are regulated upon T-cell activation. PLoS One 2012, 7:e39995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Laffort C, Le Deist F, Favre M, Caillat-Zucman S, Radford-Weiss I, Debre M, Fraitag S, Blanche S, Cavazzana-Calvo M, de Saint Basile G, et al. : Severe cutaneous papillomavirus disease after haemopoietic stem-cell transplantation in patients with severe combined immune deficiency caused by common gammac cytokine receptor subunit or JAK-3 deficiency. Lancet 2004, 363:2051–2054. [DOI] [PubMed] [Google Scholar]
  • 151.Gaspar HB, Harwood C, Leigh I, Thrasher AJ: Severe cutaneous papillomavirus disease after haematopoietic stem-cell transplantation in patients with severe combined immunodeficiency. Br J Haematol 2004, 127:232–233. [DOI] [PubMed] [Google Scholar]
  • 152.de Jong SJ, Crequer A, Matos I, Hum D, Gunasekharan V, Lorenzo L, Jabot-Hanin F, Imahorn E, Arias AA, Vahidnezhad H, et al. : The human CIB1-EVER1-EVER2 complex governs keratinocyte-intrinsic immunity to beta-papillomaviruses. J Exp Med 2018, 215:2289–2310. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This report of autosomal recessive CIB1 deficiency a novel genetic etiology of isolated epidermodysplasia verruciformis, clinically indistinguishable from EVER1 and EVER2 deficiency, suggests that the disruption of CIB1-EVER1-EVER2-dependent keratinocyte-intrinsic immunity underlies the selective susceptibility of these patients to β-human papillomaviruses.
  • 153.Vahidnezhad H, Youssefian L, Saeidian AH, Mansoori B, Jazayeri A, Azizpour A, Kamyab Hesari K, Yousefi M, Zeinali S, Jouanguy E, et al. : A CIB1 Splice-site Founder Mutation in Families with Typical Epidermodysplasia Verruciformis. J Invest Dermatol 2018. [DOI] [PubMed] [Google Scholar]
  • 154.Leisner TM, Freeman TC, Black JL, Parise LV: CIB1: a small protein with big ambitions. FASEB J 2016, 30:2640–2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Yuan W, Leisner TM, McFadden AW, Clark S, Hiller S, Maeda N, O’Brien DA, Parise LV: CIB1 is essential for mouse spermatogenesis. Mol Cell Biol 2006, 26:8507–8514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Naik MU, Nigam A, Manrai P, Millili P, Czymmek K, Sullivan M, Naik UP: CIB1 deficiency results in impaired thrombosis: the potential role of CIB1 in outside-in signaling through integrin alpha lib beta 3. J Thromb Haemost 2009, 7:1906–1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Heineke J, Auger-Messier M, Correll RN, Xu J, Benard MJ, Yuan W, Drexler H, Parise LV, Molkentin JD: CIB1 is a regulator of pathological cardiac hypertrophy. Nat Med 2010, 16:872–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zayed MA, Yuan W, Leisner TM, Chalothom D, McFadden AW, Schaller MD, Hartnett ME, Faber JE, Parise LV: CIB1 regulates endothelial cells and ischemia-induced pathological and adaptive angiogenesis. CircRes 2007, 101:1185–1193. [DOI] [PubMed] [Google Scholar]
  • 159.Lazarczyk M, Pons C, Mendoza JA, Cassonnet P, Jacob Y, Favre M: Regulation of cellular zinc balance as a potential mechanism of EVER-mediated protection against pathogenesis by cutaneous oncogenic human papillomaviruses. J Exp Med 2008, 205:35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Vuillier F, Gaud G, Guillemot D, Commere PH, Pons C, Favre M: Loss of the HPV-infection resistance EVER2 protein impairs NF-kappaB signaling pathways in keratinocytes. PLoS One 2014, 9:e89479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Gaud G, Guillemot D, Jacob Y, Favre M, Vuillier F: EVER2 protein binds TRADD to promote TNF-alpha-induced apoptosis. Cell death & disease 2013, 4:e499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Bieniasz PD: Intrinsic immunity: a front-line defense against viral attack. Nat Immunol 2004, 5:1109–1115. [DOI] [PubMed] [Google Scholar]
  • 163.Randow F, MacMicking JD, James LC: Cellular self-defense: how cell-autonomous immunity protects against pathogens. Science 2013, 340:701–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Yan N, Chen ZJ: Intrinsic antiviral immunity. Nat Immunol 2012, 13:214–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.MacMicking JD: Interferon-inducible effector mechanisms in cell-autonomous immunity. Nat Rev Immunol 2012, 12:367–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Taubenberger JK, Hultin JV, Morens DM: Discovery and characterization of the 1918 pandemic influenza virus in historical context. Antivir Ther 2007, 12:581–591. [PMC free article] [PubMed] [Google Scholar]
  • 167.Ciancanelli MJ, Abel L, Zhang SY, Casanova JL: Host genetics of severe influenza: from mouse Mxl to human IRF7. Curr Opin Immunol 2016, 38:109–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Nair H, Brooks WA, Katz M, Roca A, Berkley JA, Madhi SA, Simmerman JM, Gordon A, Sato M, Howie S, et al. : Global burden of respiratory infections due to seasonal influenza in young children: a systematic review and meta-analysis. Lancet 2011, 378:1917–1930. [DOI] [PubMed] [Google Scholar]
  • 169.Short KR, Kroeze E, Fouchier RAM, Kuiken T: Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect Dis 2014, 14:57–69. [DOI] [PubMed] [Google Scholar]
  • 170.Skalka AMR, G. F; Flint J; Enquist LW; Racaniello VR: Principles of Virology 4th Edition 2015. [Google Scholar]
  • 171.Picard C, Bobby Gaspar H, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, Crow YJ, Cunningham-Rundles C, Etzioni A, Franco JL, et al. : International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on Inborn Errors of Immunity. J Clin Immunol 2018, 38:96–128. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This is the report of the classification of human inborn errors of immunity by the international committee of the IUIS.
  • 172.Haller O, Arnheiter H, Pavlovic J, Staeheli P: The Discovery of the Antiviral Resistance Gene Mx: A Story of Great Ideas, Great Failures, and Some Success. Annu Rev Virol 2018, 5:33–51. [DOI] [PubMed] [Google Scholar]; ** A comprehensive review of the Mx-influenza saga, from the 1960s onward, by its main characters, who discovered the underlying gene and deciphered its mechanism of action.
  • 173.Bigley V, Haniffa M, Doulatov S, Wang XN, Dickinson R, McGovern N, Jardine L, Pagan S, Dimmick I, Chua I, et al. : The human syndrome of dendritic cell, monocyte, B and NK lymphoid deficiency. J Exp Med 2011, 208:227–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sologuren I, Martinez-Saavedra MT, Sole-Violan J, de Borges de Oliveira E Jr., Betancor E, Casas I, Oleaga-Quintas C, Martinez-Gallo M, Zhang SY, Pestano J, et al. : Lethal Influenza in Two Related Adults with Inherited GATA2 Deficiency. J Clin Immunol 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ciancanelli MJ, Huang SX, Luthra P, Garner H, Itan Y, Volpi S, Lafaille FG, Trouillet C, Schmolke M, Albrecht RA, et al. : Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science 2015, 348:448–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Hernandez N, Melki I, Jing H, Habib T, Huang SSY, Danielson J, Kula T, Drutman S, Belkaya S, Rattina V, et al. : Life-threatening influenza pneumonitis in a child with inherited IRF9 deficiency. J Exp Med 2018, 215:2567–2585. [DOI] [PMC free article] [PubMed] [Google Scholar]; * Autosomal recessive IRF9 deficiency is the second genetic etiology of severe influenza pneumonitis in children. Cells from the patient do not activate IRF9- and ISGF3-dependent genes in response to type I and III IFNs, thereby failing to control influenza virus in vitro and in vivo.
  • 177.Duncan CJ, Mohamad SM, Young DF, Skelton AJ, Leahy TR, Munday DC, Butler KM, Morfopoulou S, Brown JR, Hubank M, et al. : Human IFNAR2 deficiency: Lessons for antiviral immunity. Sci Transl Med 2015, 7:307ra154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Dupuis S, Jouanguy E, Al-Hajjar S, Fieschi C, Al-Mohsen IZ, Al-Jumaah S, Yang K, Chapgier A, Eidenschenk C, Eid P, et al. : Impaired response to interferon-alpha/beta and lethal viral disease in human STAT1 deficiency. Nat Genet 2003, 33:388–391. [DOI] [PubMed] [Google Scholar]
  • 179.Averbuch D, Chapgier A, Boisson-Dupuis S, Casanova JL, Engelhard D: The clinical spectrum of patients with deficiency of Signal Transducer and Activator of Transcription-1. Pediatr Infect Dis J 2011, 30:352–355. [DOI] [PubMed] [Google Scholar]
  • 180.Boisson-Dupuis S, Kong XF, Okada S, Cypowyj S, Puel A, Abel L, Casanova JL: Inborn errors of human STAT1: allelic heterogeneity governs the diversity of immunological and infectious phenotypes. Current opinion in immunology 2012, 24:364–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Burns C, Cheung A, Stark Z, Choo S, Downie L, White S, Conyers R, Cole T: A novel presentation of homozygous loss-of-function STAT-1 mutation in an infant with hyperinflammation-A case report and review of the literature. J Allergy Clin ImmunolPract 2016, 4:777–779. [DOI] [PubMed] [Google Scholar]
  • 182.Hambleton S, Goodbourn S, Young DF, Dickinson P, Mohamad SM, Valappil M, McGovern N, Cant AJ, Hackett SJ, Ghazal P, et al. : STAT2 deficiency and susceptibility to viral illness in humans. Proc Natl Acad Sci U S A 2013, 110:3053–3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Moens L, Van Eyck L, Jochmans D, Mitera T, Frans G, Bossuyt X, Matthys P, Neyts J, Ciancanelli M, Zhang SY, et al. : A novel kindred with inherited STAT2 deficiency and severe viral illness. J Allergy Clin Immunol 2017, 139:1995–1997 e1999. [DOI] [PubMed] [Google Scholar]
  • 184.Hernandez NBG; Moens L; Le Pen J; Goudouris E; Habib T; Shirkani A; Changi-Ashtiani M; Rokni-Zadeh H; Zijlmans D; Jurado A; Drutman S; Belkaya S; Boudewijns R; Jochmans D; Seeleuthner Y; Lorenzo L; Enemchukwu C; Tietjen I; Hoffmann HH.; Momenilandi M: Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J Exp Med 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Swanson PA 2nd, McGavern DB: Viral diseases of the central nervous system. Curr Opin Virol 2015, 11:44–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Casanova JL, Abel L: The genetic theory of infectious diseases: brief history and selected illustrations. Annu Rev Genome Hum Genetics 2013, 14:215–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Casrouge A, Zhang SY, Eidenschenk C, Jouanguy E, Puel A, Yang K, Alcais A, Picard C, Mahfoufi N, Nicolas N, et al. : Herpes simplex virus encephalitis in human UNC-93B deficiency. Science 2006, 314:308–312. [DOI] [PubMed] [Google Scholar]
  • 188.Guo Y, Audry M, Ciancanelli M, Alsina L, Azevedo J, Herman M, Anguiano E, Sancho-Shimizu V, Lorenzo L, Pauwels E, et al. : Herpes simplex virus encephalitis in a patient with complete TLR3 deficiency: TLR3 is otherwise redundant in protective immunity. J Exp Med 2011, 208:2083–2098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Herman M, Ciancanelli M, Ou YH, Lorenzo L, Klaudel-Dreszler M, Pauwels E, Sancho-Shimizu V, Perez de Diego R, Abhyankar A, Israelsson E, et al. : Heterozygous TBK1 mutations impair TLR3 immunity and underlie herpes simplex encephalitis of childhood. J Exp Med 2012, 209:1567–1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Perez de Diego R, Sancho-Shimizu V, Lorenzo L, Puel A, Plancoulaine S, Picard C, Herman M, Cardon A, Durandy A, Bustamante J, et al. : Human TRAF3 Adaptor Molecule Deficiency Leads to Impaired Toll-like Receptor 3 Response and Susceptibility to Herpes Simplex Encephalitis. Immunity 2010, 33:400–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Sancho-Shimizu V, Perez de Diego R, Lorenzo L, Halwani R, Alangari A, Israelsson E, Fabrega S, Cardon A, Maluenda J, Tatematsu M, et al. : Herpes simplex encephalitis in children with autosomal recessive and dominant TRIF deficiency. J Clin Invest 2012, 121:4889–4902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Zhang SY, Jouanguy E, Ugolini S, Smahi A, Elain G, Romero P, Segal D, Sancho-Shimizu V, Lorenzo L, Puel A, et al. : TLR3 deficiency in patients with herpes simplex encephalitis. Science 2007, 317:1522–1527. [DOI] [PubMed] [Google Scholar]
  • 193.Lim HK, Seppanen M, Hautala T, Ciancanelli MJ, Itan Y, Lafaille FG, Dell W, Lorenzo L, Byun M, Pauwels E, et al. : TLR3 deficiency in herpes simplex encephalitis: high allelic heterogeneity and recurrence risk. Neurology 2014, 83:1888–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lafaille FG, Pessach IM, Zhang SY, Ciancanelli MJ, Herman M, Abhyankar A, Ying SW, Keros S, Goldstein PA, Mostoslavsky G, et al. : Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012, 491:769–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Andersen LL, Mork N, Reinert LS, Kofod-Olsen E, Narita R, Jorgensen SE, Skipper KA, Honing K, Gad HH, Ostergaard L, et al. : Functional IRF3 deficiency in a patient with herpes simplex encephalitis. J Exp Med 2015, 212:1371–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Zhang SY, Casanova JL: Inborn errors underlying herpes simplex encephalitis: From TLR3 to IRF3. J Exp Med 2015, 212:1342–1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chapgier A, Wynn RF, Jouanguy E, Filipe-Santos O, Zhang S, Feinberg J, Hawkins K, Casanova JL, Arkwright PD: Human complete Stat-1 deficiency is associated with defective type I and II IFN responses in vitro but immunity to some low virulence viruses in vivo. J Immunol 2006, 176:5078–5083. [DOI] [PubMed] [Google Scholar]
  • 198.Chapgier A, Kong XF, Boisson-Dupuis S, Jouanguy E, Averbuch D, Feinberg J, Zhang SY, Bustamante J, Vogt G, Lejeune J, et al. : A partial form of recessive STAT1 deficiency in humans. J Clin Invest 2009, 119:1502–1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Niehues T, Reichenbach J, Neubert J, Gudowius S, Puel A, Horneff G, Lainka E, Dirksen U, Schroten H, Doffinger R, et al. : A NEMO-deficient child with immunodeficiency yet without anhidrotic ectodermal dysplasia. J Allergy Clin Immunol 2004, 114:1456–1462. [DOI] [PubMed] [Google Scholar]
  • 200.Puel A, Reichenbach J, Bustamante J, Ku CL, Feinberg J, Doffinger R, Bonnet M, Filipe-Santos O, Beaucoudrey L, Durandy A, et al. : The NEMO Mutation Creating the Most-Upstream Premature Stop Codon Is Hypomorphic Because of a Reinitiation of Translation. Am J Hum Genet 2006, 78:691–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Audry M, Ciancanelli M, Yang K, Cobat A, Chang HH, Sancho-Shimizu V, Lorenzo L, Niehues T, Reichenbach J, Li XX, et al. : NEMO is a key component of NF-kappaB- and IRF-3-dependent TLR3-mediated immunity to herpes simplex virus. J Allergy Clin Immunol 128:610–617 e611–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Zimmer B, Ewaleifoh O, Harschnitz O, Lee YS, Peneau C, McAlpine JL, Liu B, Tchieu J, Steinbeck JA, Lafaille F, et al. : Human iPSC-derived trigeminal neurons lack constitutive TLR3-dependent immunity that protects cortical neurons from HSV-1 infection. Proc Natl Acad Sci U S A 2018, 115:E8775–E8782. [DOI] [PMC free article] [PubMed] [Google Scholar]; * Following the 2012 report that TLR3-deficient iPSC-derived cortical neurons are more susceptible to HSV-1 infection than controls, this paper shows that control and TLR3-deficient iPSC-derived trigeminal neurons are equally susceptible to HSV-1. Collectively, these studies suggest that deficiency in the constitutive TLR3-dependent response of cortical neurons drives the pathogenesis of HSV-1 encephalitis.
  • 203.Zhang SY, Clark NE, Freije CA, Pauwels E, Taggart AJ, Okada S, Mandel H, Garcia P, Ciancanelli MJ, Biran A, et al. : Inborn Errors of RNA Lariat Metabolism in Humans with Brainstem Viral Infection. Cell 2018, 172:952–965 e918. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This paper reports the discovery of autosomal recessive partial DBR1 deficiency in children with viral infections of the brainstem. DBR1 is the only known RNA lariat debranching enzyme, the expression of which normally peaks in the brainstem. Inborn errors of RNA lariat debranching underlies viral infection of the brainstem by disrupting brainstem-specific and cell-intrinsic imminity to viruses.
  • 204.Chapman KB, Boeke JD: Isolation and characterization of the gene encoding yeast debranching enzyme. Cell 1991, 65:483–492. [DOI] [PubMed] [Google Scholar]
  • 205.Ooi SL, Samarsky DA, Fournier MJ, Boeke JD: Intronic snoRNA biosynthesis in Saccharomyces cerevisiae depends on the lariat-debranching enzyme: intron length effects and activity of a precursor snoRNA. RNA 1998, 4:1096–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Petfalski E, Dandekar T, Henry Y, Tollervey D: Processing of the precursors to small nucleolar RNAs and rRNAs requires common components. Mol Cell Biol 1998, 18:1181–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Murray JL, Sheng J, Rubin DH: A role for H/ACA and C/D small nucleolar RNAs in viral replication. Mol Biotechnol 2014, 56:429–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Sedger LM: microRNA control of interferons and interferon induced anti-viral activity. Mol Immunol 2013, 56:781–793. [DOI] [PubMed] [Google Scholar]
  • 209.Han B, Park HK, Ching T, Panneerselvam J, Wang H, Shen Y, Zhang J, Li L, Che R, Garmire L, et al. : Human DBR1 modulates the recycling of snRNPs to affect alternative RNA splicing and contributes to the suppression of cancer development. Oncogene 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Ulfendahl PJ, Kreivi JP, Akusjarvi G: Role of the branch site/3′-splice site region in adenovirus-2 E1A pre-mRNA alternative splicing: evidence for 5′- and 3′-splice site co-operation. Nucleic Acids Res 1989, 17:925–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Plotch SJ, Krug RM: In vitro splicing of influenza viral NS1 mRNA and NS1-beta-globin chimeras: possible mechanisms for the control of viral mRNA splicing. Proc Natl Acad Sci U S A 1986, 83:5444–5448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Perng GC, Jones C: Towards an understanding of the herpes simplex virus type 1 latency-reactivation cycle. Interdiscip Perspect InfectDis 2010:262415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Galvis AE, Fisher HE, Fan H, Camerini D: Conformational Changes in the 5’ End of the HIV-1 Genome Dependent on the Debranching Enzyme DBR1 During Early Stages of Infection. J Virol 2017, 91(23). pii:e01377-01317. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES