Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(3): 621-645.e33, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736301

ABSTRACT

Inborn errors of human IFN-γ-dependent macrophagic immunity underlie mycobacterial diseases, whereas inborn errors of IFN-α/ß-dependent intrinsic immunity underlie viral diseases. Both types of IFNs induce the transcription factor IRF1. We describe unrelated children with inherited complete IRF1 deficiency and early-onset, multiple, life-threatening diseases caused by weakly virulent mycobacteria and related intramacrophagic pathogens. These children have no history of severe viral disease, despite exposure to many viruses, including SARS-CoV-2, which is life-threatening in individuals with impaired IFN-α/ß immunity. In leukocytes or fibroblasts stimulated in vitro, IRF1-dependent responses to IFN-γ are, both quantitatively and qualitatively, much stronger than those to IFN-α/ß. Moreover, IRF1-deficient mononuclear phagocytes do not control mycobacteria and related pathogens normally when stimulated with IFN-γ. By contrast, IFN-α/ß-dependent intrinsic immunity to nine viruses, including SARS-CoV-2, is almost normal in IRF1-deficient fibroblasts. Human IRF1 is essential for IFN-γ-dependent macrophagic immunity to mycobacteria, but largely redundant for IFN-α/ß-dependent antiviral immunity.


Subject(s)
COVID-19 , Mycobacterium , Child , Humans , Interferon-gamma , SARS-CoV-2 , Interferon-alpha , Interferon Regulatory Factor-1
2.
Cell ; 172(5): 952-965.e18, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474921

ABSTRACT

Viruses that are typically benign sometimes invade the brainstem in otherwise healthy children. We report bi-allelic DBR1 mutations in unrelated patients from different ethnicities, each of whom had brainstem infection due to herpes simplex virus 1 (HSV1), influenza virus, or norovirus. DBR1 encodes the only known RNA lariat debranching enzyme. We show that DBR1 expression is ubiquitous, but strongest in the spinal cord and brainstem. We also show that all DBR1 mutant alleles are severely hypomorphic, in terms of expression and function. The fibroblasts of DBR1-mutated patients contain higher RNA lariat levels than control cells, this difference becoming even more marked during HSV1 infection. Finally, we show that the patients' fibroblasts are highly susceptible to HSV1. RNA lariat accumulation and viral susceptibility are rescued by wild-type DBR1. Autosomal recessive, partial DBR1 deficiency underlies viral infection of the brainstem in humans through the disruption of tissue-specific and cell-intrinsic immunity to viruses.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Brain Stem/metabolism , Brain Stem/virology , RNA/chemistry , RNA/metabolism , Alleles , Amino Acid Sequence , Animals , Brain Diseases, Metabolic, Inborn/pathology , Brain Stem/pathology , Encephalitis, Viral/genetics , Escherichia coli/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human , Humans , Interferons/metabolism , Introns/genetics , Male , Mice , Mutant Proteins/metabolism , Mutation/genetics , Open Reading Frames/genetics , Pedigree , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/deficiency , RNA Nucleotidyltransferases/genetics , Toll-Like Receptor 3/metabolism , Virus Replication
3.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235196

ABSTRACT

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lipopolysaccharides/metabolism , Membrane Glycoproteins/deficiency , Receptors, Interleukin-1/deficiency , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Teichoic Acids/metabolism , Adaptive Immunity , Child , Female , Fibroblasts/metabolism , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , Pedigree , Phagocytes/metabolism , Point Mutation , Protein Isoforms/analysis , Protein Isoforms/genetics , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/genetics , Staphylococcal Infections/drug therapy , Teichoic Acids/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
4.
Am J Hum Genet ; 108(6): 1012-1025, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34015270

ABSTRACT

The human genetic dissection of clinical phenotypes is complicated by genetic heterogeneity. Gene burden approaches that detect genetic signals in case-control studies are underpowered in genetically heterogeneous cohorts. We therefore developed a genome-wide computational method, network-based heterogeneity clustering (NHC), to detect physiological homogeneity in the midst of genetic heterogeneity. Simulation studies showed our method to be capable of systematically converging genes in biological proximity on the background biological interaction network, and capturing gene clusters harboring presumably deleterious variants, in an efficient and unbiased manner. We applied NHC to whole-exome sequencing data from a cohort of 122 individuals with herpes simplex encephalitis (HSE), including 13 individuals with previously published monogenic inborn errors of TLR3-dependent IFN-α/ß immunity. The top gene cluster identified by our approach successfully detected and prioritized all causal variants of five TLR3 pathway genes in the 13 previously reported individuals. This approach also suggested candidate variants of three reported genes and four candidate genes from the same pathway in another ten previously unstudied individuals. TLR3 responsiveness was impaired in dermal fibroblasts from four of the five individuals tested, suggesting that the variants detected were causal for HSE. NHC is, therefore, an effective and unbiased approach for unraveling genetic heterogeneity by detecting physiological homogeneity.


Subject(s)
Computational Biology/methods , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/pathology , Fibroblasts/immunology , Gene Regulatory Networks , Genetic Heterogeneity , Genetic Predisposition to Disease , Case-Control Studies , Encephalitis, Herpes Simplex/immunology , Fibroblasts/metabolism , Humans , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism , Exome Sequencing
5.
J Clin Immunol ; 43(2): 406-420, 2023 02.
Article in English | MEDLINE | ID: mdl-36308662

ABSTRACT

Fulminant viral hepatitis (FVH) caused by hepatitis A virus (HAV) is a life-threatening disease that typically strikes otherwise healthy individuals. The only known genetic etiology of FVH is inherited IL-18BP deficiency, which unleashes IL-18-dependent lymphocyte cytotoxicity and IFN-γ production. We studied two siblings who died from a combination of early-onset inflammatory bowel disease (EOIBD) and FVH due to HAV. The sibling tested was homozygous for the W100G variant of IL10RB previously described in an unrelated patient with EOIBD. We show here that the out-of-frame IL10RB variants seen in other EOIBD patients disrupt cellular responses to IL-10, IL-22, IL-26, and IFN-λs in overexpression conditions and in homozygous cells. By contrast, the impact of in-frame disease-causing variants varies between cases. When overexpressed, the W100G variant impairs cellular responses to IL-10, but not to IL-22, IL-26, or IFN-λ1, whereas cells homozygous for W100G do not respond to IL-10, IL-22, IL-26, or IFN-λ1. As IL-10 is a potent antagonist of IFN-γ in phagocytes, these findings suggest that the molecular basis of FVH in patients with IL-18BP or IL-10RB deficiency may involve excessive IFN-γ activity during HAV infections of the liver. Inherited IL-10RB deficiency, and possibly inherited IL-10 and IL-10RA deficiencies, confer a predisposition to FVH, and patients with these deficiencies should be vaccinated against HAV and other liver-tropic viruses.


Subject(s)
Hepatitis, Viral, Human , Interleukin-10 , Humans , Interleukin-10/genetics , Siblings , Interferon-gamma/genetics
6.
J Clin Immunol ; 40(8): 1156-1162, 2020 11.
Article in English | MEDLINE | ID: mdl-32936395

ABSTRACT

Puumala hantavirus (PUUV) hemorrhagic fever with renal syndrome (HFRS) is common in Northern Europe; this infection is usually self-limited and severe complications are uncommon. PUUV and other hantaviruses, however, can rarely cause encephalitis. The pathogenesis of these rare and severe events is unknown. In this study, we explored the possibility that genetic defects in innate anti-viral immunity, as analogous to Toll-like receptor 3 (TLR3) mutations seen in HSV-1 encephalitis, may explain PUUV encephalitis. We completed exome sequencing of seven adult patients with encephalitis or encephalomyelitis during acute PUUV infection. We found heterozygosity for the TLR3 p.L742F novel variant in two of the seven unrelated patients (29%, p = 0.0195). TLR3-deficient P2.1 fibrosarcoma cell line and SV40-immortalized fibroblasts (SV40-fibroblasts) from patient skin expressing mutant or wild-type TLR3 were tested functionally. The TLR3 p.L742F allele displayed low poly(I:C)-stimulated cytokine induction when expressed in P2.1 cells. SV40-fibroblasts from three healthy controls produced increasing levels of IFN-λ and IL-6 after 24 h of stimulation with increasing concentrations of poly(I:C), whereas the production of the cytokines was impaired in TLR3 L742F/WT patient SV40-fibroblasts. Heterozygous TLR3 mutation may underlie not only HSV-1 encephalitis but also PUUV hantavirus encephalitis. Such possibility should be further explored in encephalitis caused by these and other hantaviruses.


Subject(s)
Encephalitis, Viral/etiology , Hantavirus Infections/etiology , Heterozygote , Mutation , Orthohantavirus , Toll-Like Receptor 3/genetics , Alleles , Cell Line , Cells, Cultured , Disease Susceptibility , Encephalitis, Viral/diagnosis , Fibroblasts/immunology , Fibroblasts/metabolism , Genetic Predisposition to Disease , Orthohantavirus/immunology , Hantavirus Infections/diagnosis , Humans
7.
J Clin Immunol ; 40(2): 388-398, 2020 02.
Article in English | MEDLINE | ID: mdl-32008135

ABSTRACT

In 2014, a child with broad combined immunodeficiency (CID) who was homozygous for a private BCL10 allele was reported to have complete inherited human BCL10 deficiency. In the present study, we report a new BCL10 mutation in another child with CID who was homozygous for a BCL10 variant (R88X), previously reported as a rare allele in heterozygosis (minor allele frequency, 0.000003986). The mutant allele was a loss-of-expression and loss-of-function allele. As with the previously reported patient, this patient had complete BCL10 deficiency. The clinical phenotype shared features, such as respiratory infections, but differed from that of the previous patient that he did not develop significant gastroenteritis episodes or chronic colitis. Cellular and immunological phenotypes were similar to those of the previous patient. TLR4, TLR2/6, and Dectin-1 responses were found to depend on BCL10 in fibroblasts, and final maturation of T cell and B cell maturation into memory cells was affected. Autosomal-recessive BCL10 deficiency should therefore be considered in children with CID.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/genetics , B-Lymphocytes/immunology , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , T-Lymphocytes/immunology , Cells, Cultured , Chromosome Disorders , Homozygote , Humans , Immunologic Memory , Infant , Lectins, C-Type/metabolism , Male , Respiratory Tract Infections , Toll-Like Receptors/metabolism
8.
Immunity ; 33(3): 400-11, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20832341

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3) functions downstream of multiple TNF receptors and receptors that induce interferon-α (IFN-α), IFN-ß, and IFN-λ production, including Toll-like receptor 3 (TLR3), which is deficient in some patients with herpes simplex virus-1 encephalitis (HSE). Mice lacking TRAF3 die in the neonatal period, preventing direct investigation of the role of TRAF3 in immune responses and host defenses in vivo. Here, we report autosomal dominant, human TRAF3 deficiency in a young adult with a history of HSE in childhood. The TRAF3 mutant allele is loss-of-expression, loss-of-function, dominant-negative and associated with impaired, but not abolished, TRAF3-dependent responses upon stimulation of both TNF receptors and receptors that induce IFN production. TRAF3 deficiency is associated with a clinical phenotype limited to HSE resulting from the impairment of TLR3-dependent induction of IFN. Thus, TLR3-mediated immunity against primary infection by HSV-1 in the central nervous system is critically dependent on TRAF3.


Subject(s)
Encephalitis, Herpes Simplex/immunology , TNF Receptor-Associated Factor 3/physiology , Toll-Like Receptor 3/physiology , Cells, Cultured , Disease Susceptibility , Humans , Interferons/physiology , Mutation , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Factor 3/genetics
9.
J Clin Immunol ; 36(1): 19-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26686525

ABSTRACT

Collagen and calcium-binding EGF domain-containing protein 1 (CCBE1) bi-allelic mutations have been associated with syndromes of widespread congenital lymphatic dysplasia, including Hennekam Syndrome (HS). HS is characterized by lymphedema, lymphangiectasia, and intellectual disability. CCBE1 encodes a putative extracellular matrix protein but the HS-causing mutations have not been studied biochemically. We report two HS siblings, born to consanguineous parents of Turkish ancestry, whose clinical phenotype also includes protein losing enteropathy, painful relapsing chylous ascites, and hypogammaglobulinemia. We identified by whole exome and Sanger sequencing the homozygous CCBE1 C174Y mutation in both siblings. This mutation had been previously reported in another HS kindred from the Netherlands. In over-expression studies, we found increased intracellular expression of all forms (monomers, dimers, trimers) of the CCBE1 C174Y mutant protein, by Western blot, despite mutant mRNA levels similar to wild-type (WT). In addition, we detected increased secretion of the mutant CCBE1 protein by ELISA. We further found the mutant and WT proteins to be evenly distributed in the cytoplasm, by immunofluorescence and confocal microscopy. Finally, we found a strong decrease of lymphatic vessels, with a corresponding diminished expression of CCBE1, by immunohistochemistry of the patients' intestinal biopsies. In contrast, mucosal blood vessels and muscularis mucosae showed normal CCBE1 staining. Our findings show that the mutant CCBE1 C174Y protein is not loss-of-function by loss-of-expression.


Subject(s)
Calcium-Binding Proteins/metabolism , Craniofacial Abnormalities/diagnosis , Lymphangiectasis, Intestinal/diagnosis , Lymphedema/diagnosis , Tumor Suppressor Proteins/metabolism , Adult , Calcium-Binding Proteins/genetics , Consanguinity , Craniofacial Abnormalities/genetics , DNA Mutational Analysis , Female , Gene Expression Regulation/genetics , Homozygote , Humans , Lymphangiectasis, Intestinal/genetics , Lymphedema/genetics , Male , Mutation/genetics , Pedigree , Siblings , Tumor Suppressor Proteins/genetics , Turkey , Young Adult
12.
J Allergy Clin Immunol ; 131(4): 1157-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23434283

ABSTRACT

BACKGROUND: Inborn errors in Toll-like receptor 3 (TLR3)-IFN type I and III pathways have been implicated in susceptibility to herpes simplex virus encephalitis (HSE) in children, but most patients studied do not carry mutations in any of the genes presently associated with HSE susceptibility. Moreover, many patients do not display any TLR3-IFN-related fibroblastic phenotype. OBJECTIVE: To study other signaling pathways downstream of TLR3 and/or other independent pathways that may contribute to HSE susceptibility. METHODS: We used the stable isotope labeling of amino acids in cell culture proteomics methodology to measure changes in the human immortalized fibroblast proteome after TLR3 activation. RESULTS: Cells from healthy controls were compared with cells from a patient with a known genetic etiology of HSE (UNC-93B-/-) and also to cells from an HSE patient with an unknown gene defect. Consistent with known variation in susceptibility of individuals to viral infections, substantial variation in the response level of different healthy controls was observed, but common functional networks could be identified, including upregulation of superoxide dismutase 2. The 2 patients with HSE studied show clear differences in functional response networks when compared with healthy controls and also when compared with each other. CONCLUSIONS: The present study delineates a number of novel proteins, TLR3-related pathways, and cellular phenotypes that may help elucidate the genetic basis of childhood HSE. Furthermore, our results reveal superoxide dismutase 2 as a potential therapeutic target for amelioration of the neurologic sequelae caused by HSE.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Fibroblasts/immunology , Gene Expression Regulation , Proteome/genetics , Superoxide Dismutase/genetics , Toll-Like Receptor 3/genetics , Child , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/pathology , Fibroblasts/pathology , Genetic Predisposition to Disease , Genetic Variation , Humans , Male , Proteome/immunology , Signal Transduction , Superoxide Dismutase/immunology , Toll-Like Receptor 3/immunology
13.
Sci Immunol ; 8(82): eade2860, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37083451

ABSTRACT

Inborn errors of TLR3-dependent type I IFN immunity in cortical neurons underlie forebrain herpes simplex virus-1 (HSV-1) encephalitis (HSE) due to uncontrolled viral growth and subsequent cell death. We report an otherwise healthy patient with HSE who was compound heterozygous for nonsense (R422*) and frameshift (P493fs9*) RIPK3 variants. Receptor-interacting protein kinase 3 (RIPK3) is a ubiquitous cytoplasmic kinase regulating cell death outcomes, including apoptosis and necroptosis. In vitro, the R422* and P493fs9* RIPK3 proteins impaired cellular apoptosis and necroptosis upon TLR3, TLR4, or TNFR1 stimulation and ZBP1/DAI-mediated necroptotic cell death after HSV-1 infection. The patient's fibroblasts displayed no detectable RIPK3 expression. After TNFR1 or TLR3 stimulation, the patient's cells did not undergo apoptosis or necroptosis. After HSV-1 infection, the cells supported excessive viral growth despite normal induction of antiviral IFN-ß and IFN-stimulated genes (ISGs). This phenotype was, nevertheless, rescued by application of exogenous type I IFN. The patient's human pluripotent stem cell (hPSC)-derived cortical neurons displayed impaired cell death and enhanced viral growth after HSV-1 infection, as did isogenic RIPK3-knockout hPSC-derived cortical neurons. Inherited RIPK3 deficiency therefore confers a predisposition to HSE by impairing the cell death-dependent control of HSV-1 in cortical neurons but not their production of or response to type I IFNs.


Subject(s)
Encephalitis, Herpes Simplex , Herpes Simplex , Herpesvirus 1, Human , Humans , Cell Death , Encephalitis, Herpes Simplex/genetics , Herpesvirus 1, Human/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
14.
Nat Cell Biol ; 7(3): 286-94, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15723049

ABSTRACT

The mammalian target of rapamycin (mTOR) and Akt proteins regulate various steps of muscle development and growth, but the physiological relevance and the downstream effectors are under investigation. Here we show that S6 kinase 1 (S6K1), a protein kinase activated by nutrients and insulin-like growth factors (IGFs), is essential for the control of muscle cytoplasmic volume by Akt and mTOR. Deletion of S6K1 does not affect myoblast cell proliferation but reduces myoblast size to the same extent as that observed with mTOR inhibition by rapamycin. In the differentiated state, S6K1(-/-) myotubes have a normal number of nuclei but are smaller, and their hypertrophic response to IGF1, nutrients and membrane-targeted Akt is blunted. These growth defects reveal that mTOR requires distinct effectors for the control of muscle cell cycle and size, potentially opening new avenues of therapeutic intervention against neoplasia or muscle atrophy.


Subject(s)
Muscle, Skeletal/metabolism , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Animals , Atrophy , Body Weight , Cell Differentiation , Cell Line , Cells, Cultured , Collagen/pharmacology , Drug Combinations , Gene Deletion , Genetic Vectors , Genotype , Green Fluorescent Proteins/metabolism , Homozygote , Humans , Immunoblotting , Laminin/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Muscles/pathology , Plasmids/metabolism , Protein Binding , Proteoglycans/pharmacology , Retroviridae/genetics , Signal Transduction , Somatomedins/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transfection
15.
J Allergy Clin Immunol ; 128(3): 610-7.e1-4, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21722947

ABSTRACT

BACKGROUND: Children with germline mutations in Toll-like receptor 3 (TLR3), UNC93B1, TNF receptor-associated factor 3, and signal transducer and activator of transcription 1 are prone to herpes simplex virus-1 encephalitis, owing to impaired TLR3-triggered, UNC-93B-dependent, IFN-α/ß, and/or IFN-λ-mediated signal transducer and activator of transcription 1-dependent immunity. OBJECTIVE: We explore here the molecular basis of the pathogenesis of herpes simplex encephalitis in a child with a hypomorphic mutation in nuclear factor-κB (NF-κB) essential modulator, which encodes the regulatory subunit of the inhibitor of the Iκß kinase complex. METHODS: The TLR3 signaling pathway was investigated in the patient's fibroblasts by analyses of IFN-ß, IFN-λ, and IL-6 mRNA and protein levels, by quantitative PCR and ELISA, respectively, upon TLR3 stimulation (TLR3 agonists or TLR3-dependent viruses). NF-κB activation was assessed by electrophoretic mobility shift assay and interferon regulatory factor 3 dimerization on native gels after stimulation with a TLR3 agonist. RESULTS: The patient's fibroblasts displayed impaired responses to TLR3 stimulation in terms of IFN-ß, IFN-λ, and IL-6 production, owing to impaired activation of both NF-κB and IRF-3. Moreover, vesicular stomatitis virus, a potent IFN-inducer in human fibroblasts, and herpes simplex virus-1, induced only low levels of IFN-ß and IFN-λ in the patient's fibroblasts, resulting in enhanced viral replication and cell death, as reported for UNC-93B-deficient fibroblasts. CONCLUSION: Herpes simplex encephalitis may occur in patients carrying NF-κB essential modulator mutations, due to the impairment of NF-κB- and interferon regulatory factor 3-dependent-TLR3-mediated antiviral IFN production.


Subject(s)
Encephalitis, Herpes Simplex/immunology , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Mutation , NF-kappa B/metabolism , Toll-Like Receptor 3/immunology , Child, Preschool , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/virology , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , I-kappa B Kinase/genetics , Signal Transduction , Simplexvirus/immunology , Toll-Like Receptor 3/metabolism
16.
Adv Mater ; 34(18): e2107739, 2022 May.
Article in English | MEDLINE | ID: mdl-35077604

ABSTRACT

Polarizers are ubiquitous components in current optoelectronic devices as displays or photographic cameras. Yet, control over light polarization is an unsolved challenge, since the main drawback of the existing display technologies is the significant optical losses. In such a context, organometal halide perovskites (OMHP) can play a decisive role given their flexible synthesis with tunable optical properties such as bandgap and photoluminescence, and excellent light emission with a low non-radiative recombination rate. Therefore, along with their outstanding electrical properties have elevated hybrid perovskites as the material of choice in photovoltaics and optoelectronics. Among the different OMHP nanostructures, nanowires and nanorods have lately arisen as key players in the control of light polarization for lighting or detector applications. Herein, the fabrication of highly aligned and anisotropic methylammonium lead iodide perovskite nanowalls by glancing-angle deposition, which is compatible with most substrates, is presented. Their high alignment degree provides the samples with anisotropic optical properties such as light absorption and photoluminescence. Furthermore, their implementation in photovoltaic devices provides them with a polarization-sensitive response. This facile vacuum-based approach embodies a milestone in the development of last-generation polarization-sensitive perovskite-based optoelectronic devices such as lighting appliances or self-powered photodetectors.

17.
J Exp Med ; 219(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35442418

ABSTRACT

Globally, autosomal recessive IFNAR1 deficiency is a rare inborn error of immunity underlying susceptibility to live attenuated vaccine and wild-type viruses. We report seven children from five unrelated kindreds of western Polynesian ancestry who suffered from severe viral diseases. All the patients are homozygous for the same nonsense IFNAR1 variant (p.Glu386*). This allele encodes a truncated protein that is absent from the cell surface and is loss-of-function. The fibroblasts of the patients do not respond to type I IFNs (IFN-α2, IFN-ω, or IFN-ß). Remarkably, this IFNAR1 variant has a minor allele frequency >1% in Samoa and is also observed in the Cook, Society, Marquesas, and Austral islands, as well as Fiji, whereas it is extremely rare or absent in the other populations tested, including those of the Pacific region. Inherited IFNAR1 deficiency should be considered in individuals of Polynesian ancestry with severe viral illnesses.


Subject(s)
Receptor, Interferon alpha-beta , Virus Diseases , Alleles , Child , Homozygote , Humans , Polynesia
18.
Science ; 376(6599): eabm6380, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35587511

ABSTRACT

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Subject(s)
Bacterial Toxins , Cri-du-Chat Syndrome , Endopeptidases , Haploinsufficiency , Hemolysin Proteins , Staphylococcal Infections , Staphylococcus aureus , Bacterial Toxins/immunology , Cri-du-Chat Syndrome/genetics , Cri-du-Chat Syndrome/immunology , Endopeptidases/genetics , Haploinsufficiency/genetics , Haploinsufficiency/immunology , Hemolysin Proteins/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular/genetics , Necrosis , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology
19.
Front Immunol ; 12: 786572, 2021.
Article in English | MEDLINE | ID: mdl-34868072

ABSTRACT

The CARD-BCL10-MALT1 (CBM) complex is critical for the proper assembly of human immune responses. The clinical and immunological consequences of deficiencies in some of its components such as CARD9, CARD11, and MALT1 have been elucidated in detail. However, the scarcity of BCL10 deficient patients has prevented gaining detailed knowledge about this genetic disease. Only two patients with BCL10 deficiency have been reported to date. Here we provide an in-depth description of an additional patient with autosomal recessive complete BCL10 deficiency caused by a nonsense mutation that leads to a loss of expression (K63X). Using mass cytometry coupled with unsupervised clustering and machine learning computational methods, we obtained a thorough characterization of the consequences of BCL10 deficiency in different populations of leukocytes. We showed that in addition to the near absence of memory B and T cells previously reported, this patient displays a reduction in NK, γδT, Tregs, and TFH cells. The patient had recurrent respiratory infections since early childhood, and showed a family history of lethal severe infectious diseases. Fortunately, hematopoietic stem-cell transplantation (HSCT) cured her. Overall, this report highlights the importance of early genetic diagnosis for the management of BCL10 deficient patients and HSCT as the recommended treatment to cure this disease.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/deficiency , Lymphocytes/immunology , Primary Immunodeficiency Diseases/diagnosis , B-Cell CLL-Lymphoma 10 Protein/genetics , Child , Codon, Nonsense , DNA Mutational Analysis , Female , Hematopoietic Stem Cell Transplantation , Humans , Lymphocytes/metabolism , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/therapy
20.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34726731

ABSTRACT

Enterovirus (EV) infection rarely results in life-threatening infection of the central nervous system. We report two unrelated children with EV30 and EV71 rhombencephalitis. One patient carries compound heterozygous TLR3 variants (loss-of-function F322fs2* and hypomorphic D280N), and the other is homozygous for an IFIH1 variant (loss-of-function c.1641+1G>C). Their fibroblasts respond poorly to extracellular (TLR3) or intracellular (MDA5) poly(I:C) stimulation. The baseline (TLR3) and EV-responsive (MDA5) levels of IFN-ß in the patients' fibroblasts are low. EV growth is enhanced at early and late time points of infection in TLR3- and MDA5-deficient fibroblasts, respectively. Treatment with exogenous IFN-α2b before infection renders both cell lines resistant to EV30 and EV71, whereas post-infection treatment with IFN-α2b rescues viral susceptibility fully only in MDA5-deficient fibroblasts. Finally, the poly(I:C) and viral phenotypes of fibroblasts are rescued by the expression of WT TLR3 or MDA5. Human TLR3 and MDA5 are critical for cell-intrinsic immunity to EV, via the control of baseline and virus-induced type I IFN production, respectively.


Subject(s)
Encephalitis, Viral/immunology , Enterovirus Infections/immunology , Interferon-Induced Helicase, IFIH1/genetics , Toll-Like Receptor 3/genetics , Cells, Cultured , Child, Preschool , Encephalitis, Viral/genetics , Enterovirus/drug effects , Enterovirus/physiology , Enterovirus Infections/genetics , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Humans , Infant , Interferon alpha-2/pharmacology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-beta/immunology , Interferon-beta/metabolism , Loss of Function Mutation , Male , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/immunology , Poly I-C/pharmacology , Rhombencephalon/virology , Toll-Like Receptor 3/immunology , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL