Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Comput Struct Biotechnol J ; 21: 3522-3531, 2023.
Article in English | MEDLINE | ID: mdl-37484491

ABSTRACT

Interleukin 18 (IL18) is a pro-inflammatory cytokine that modulates innate and adaptive immune responses. IL18 activity is tightly controlled by the constitutively secreted IL18 binding protein (IL18BP). PDB structures of human IL18 showed that a short stretch of amino acids between 68 and 81 adopted a disordered conformation in all IL18-IL18BP complexes while adopting a 310 helical structure in other IL18 structures, including the receptor complexes. The C74 of human IL18, which was reported to form a novel intermolecular disulfide bond in the human tetrameric assembly, is also located in this short epitope. These observations reflected the importance of this short surface epitope for the structure and dynamics of the IL18-IL18BP heterodimers. We have analyzed all known IL18-IL18BP complexes in the PDB by all-atom MD simulations. The analysis also included two computed complex models adopting a helical structure for the surface epitope. Heterodimer simulations showed a stabilizing impact of the small surface region at the helical form by reducing flexibility of the complex backbone. Analysis of the symmetry-related human IL18-IL18BP tetramer showed that the unfolding of this small surface region also contributed to the IL18-IL18BP stability through a completely exposed C74 sidechain to form an intermolecular disulfide bond in the self-assembled human IL18-IL18BP dimer. Our findings showed how the conformation of the short IL18 epitope between amino acids 68 and 81 would affect IL18 activity by mediating the intermolecular interactions of IL18.

2.
Allergol Immunopathol (Madr) ; 51(4): 87-93, 2023.
Article in English | MEDLINE | ID: mdl-37422784

ABSTRACT

INTRODUCTION: Pentraxin-3 (PTX3) is a soluble long pentraxin molecule that regulates inflammatory responses. This study aimed to determine the plasma levels of plasma PTX-3 as an inflammation marker in chronic spontaneous urticaria (CSU) and whether the PTX3 levels correlate with disease activity and other clinical parameters, including acute phase reactants and biomarkers. METHODS: The study included 70 CSU patients and 30 healthy controls. Plasma PTX3 levels were measured by ELISA. CSU disease activity was evaluated with the urticaria activity score summed over 7 days. Complete blood count, C-reactive protein (CRP), transaminases, total IgE, antinuclear antibody, anti-thyroid peroxidase, anti-thyroglobulin, and D-dimer levels were recorded. RESULTS: Of the 70 patients, 52 (74.3%) were female, with a mean age of 37.51 ± 11.80 years. Disease activity was severe in 43, moderate in 15, and mild in 12 patients. Mean PTX3 levels were elevated in CSU patients compared to healthy controls (0.81 vs. 0.55 ng/mL, p = 0.031). The mean CRP levels were higher in patients than in the controls (4.26 vs. 1.57 mg/L, p = 0.023). Patients also had higher D-dimer levels than the controls (5.96 vs. 0.59 mg/L, p < 0.001). A significant positive correlation was found between PTX3 and CRP levels (r = 0.508, p < 0.001) and between D-dimer levels and UAS7 (r = 0.338, p = 0.004) and CRP (r = 0.213, p = 0.034) levels. A multivariable stepwise regression analysis showed that the one-unit increase in the CRP level increased to 38.19 units in the PTX3 level (95% confidence interval [17.40-58.98], p < 0.001). CONCLUSION: Circulating levels of CRP and PTX3, two members of the pentraxin family, are significantly correlated and elevated in CSU patients with increasing disease activity, indicating their utility as inflammatory markers in CSU.


Subject(s)
Chronic Urticaria , Urticaria , Humans , Female , Adult , Middle Aged , Male , C-Reactive Protein/analysis , Chronic Disease , Biomarkers
3.
Mikrobiyol Bul ; 57(3): 473-480, 2023 Jul.
Article in Turkish | MEDLINE | ID: mdl-37462310

ABSTRACT

Bovine tuberculosis might be seen in low-income countries, especially in children fed with raw milk. The most common transmission route is fecal-oral way, and it is most likely through unpasteurized dairy products. Although clinical and radiological findings are like non-zoonotic tuberculosis, treatment approaches may differ in individuals with zoonotic tuberculosis. Prevention of zoonotic diseases requires multidisciplinary approaches. These approaches include the development of veterinary and surveillance studies for the detection of communicable diseases in farm animals, as well as informing the public about raw milk consumption. In this case report, a patient with zoonotic pulmonary tuberculosis related to Mycobacterium bovis because of consumption of raw milk was presented. A five-month-old male was admitted to the hospital due to a persistent, feverless, non-productive cough since birth. Empirical antibiotic treatment was started with a preliminary diagnosis of pneumonia because of left upper lobe and right pericardial infiltration on chest X-ray. However, after two weeks of antimicrobial therapy, the patient's clinical and laboratory findings did not improve. This led to the referral for a computed tomography imaging, which revealed tracheomalacia, consolidation on the right upper lobe, an indistinguishable mass or consolidation on the left middle lobe of the lung, peribronchial thickening on the basal segment of the lower lobe, and mediastinal lymphadenopathy. Three consecutive days of fasting gastric lavage fluid was sent to the reference laboratory for acid-resistant bacillus examination, polymerase chain reaction (PCR) and culture studies. As the clinical findings were compatible and PCR was positive, the patient was started on quadruple antituberculous therapy. After initiation of anti-tuberculosis drugs, the patient's findings radiologically and clinically were improved. Mycobacterium bovis was grown in the culture. In the meantime, it was discovered that the patient was fed with raw milk. Due to the patient's clinical symptoms and the growth of Mycobacterium bovis in the gastric lavage fluid culture, the diagnosis of bovine tuberculosis was made. The culprit was that the milk of the cow belonging to the patient's family, which was later found to be infected with M.bovis, was milked and given to the patient without boiling. Today, unpasteurized dairy products continue to be consumed, especially in rural areas. One of the most important steps to prevent zoonotic diseases is to raise awareness about not consuming raw milk and undercooked meat. To elucidate the epidemiological link in childhood, taking a good anamnesis, including questioning raw milk consumption, is essential in the diagnosis of tuberculosis.


Subject(s)
Mycobacterium bovis , Tuberculosis, Bovine , Tuberculosis, Pulmonary , Tuberculosis , Animals , Female , Cattle , Male , Tuberculosis, Bovine/diagnosis , Tuberculosis, Bovine/drug therapy , Tuberculosis, Bovine/epidemiology , Tuberculosis/microbiology , Tuberculosis, Pulmonary/diagnostic imaging , Tuberculosis, Pulmonary/drug therapy , Zoonoses , Antitubercular Agents
5.
Genes Immun ; 24(1): 12-20, 2023 02.
Article in English | MEDLINE | ID: mdl-36517554

ABSTRACT

Insufficient dietary folate intake, hereditary malabsorption, or defects in folate transport may lead to combined immunodeficiency (CID). Although loss of function mutations in the major intestinal folate transporter PCFT/SLC46A1 was shown to be associated with CID, the evidence for pathogenic variants of RFC/SLC19A1 resulting in immunodeficiency was lacking. We report two cousins carrying a homozygous pathogenic variant c.1042 G > A, resulting in p.G348R substitution who showed symptoms of immunodeficiency associated with defects of folate transport. SLC19A1 expression by peripheral blood mononuclear cells (PBMC) was quantified by real-time qPCR and immunostaining. T cell proliferation, methotrexate resistance, NK cell cytotoxicity, Treg cells and cytokine production by T cells were examined by flow cytometric assays. Patients were treated with and benefited from folinic acid. Studies revealed normal NK cell cytotoxicity, Treg cell counts, and naive-memory T cell percentages. Although SLC19A1 mRNA and protein expression were unaltered, remarkably, mitogen induced-T cell proliferation was significantly reduced at suboptimal folic acid and supraoptimal folinic acid concentrations. In addition, patients' PBMCs were resistant to methotrexate-induced apoptosis supporting a functionally defective SLC19A1. This study presents the second pathogenic SLC19A1 variant in the literature, providing the first experimental evidence that functionally defective variants of SLC19A1 may present with symptoms of immunodeficiency.


Subject(s)
Immunologic Deficiency Syndromes , Leucovorin , Reduced Folate Carrier Protein , Humans , Folic Acid/genetics , Folic Acid/metabolism , Leucovorin/therapeutic use , Leucovorin/metabolism , Leukocytes, Mononuclear/metabolism , Methotrexate/pharmacology , Methotrexate/therapeutic use , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism , Reduced Folate Carrier Protein/genetics , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism
6.
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
7.
J Clin Immunol ; 43(1): 192-205, 2023 01.
Article in English | MEDLINE | ID: mdl-36205835

ABSTRACT

Human herpesvirus-6 (HHV-6) infection can rarely cause life-threatening conditions, such as encephalitis, in otherwise healthy children, with unclear pathogenesis. We studied a child who presented with acute HHV-6 encephalitis at the age of 10 months and who was homozygous for a novel missense mutation in IRAK4, encoding interleukin-1 receptor-associated kinase 4, identified by whole-exome sequencing. We tested the damaging impact of this mutation in silico by molecular dynamics simulations and in vitro by biochemical and functional experiments utilizing cell lines and patient's cells. We found that the mutation is severely hypomorphic, impairing both the expression and function of IRAK-4. Patient's leukocytes had barely detectable levels of IRAK-4 and diminished anti-viral immune responses to various stimuli inducing different Toll-like receptors and cytosolic nucleic acid sensors. Overall, these findings suggest that acute HHV-6 encephalitis can result from inborn errors of immunity to virus. This study represents the first report of isolated acute HHV-6 infection causing encephalitis in an inherited primary immunodeficiency, notably autosomal recessive (AR) partial IRAK-4 deficiency, and the first report of AR IRAK-4 deficiency presenting with a severe viral disease, notably HHV-6 encephalitis upon an acute infection, thereby expanding the clinical spectrum of IRAK-4 deficiency.


Subject(s)
Encephalitis, Viral , Herpesvirus 6, Human , Primary Immunodeficiency Diseases , Roseolovirus Infections , Child , Humans , Infant , Interleukin-1 Receptor-Associated Kinases/genetics , Toll-Like Receptors/metabolism , Primary Immunodeficiency Diseases/genetics , Encephalitis, Viral/diagnosis , Encephalitis, Viral/genetics , Herpesvirus 6, Human/genetics , Roseolovirus Infections/diagnosis , Roseolovirus Infections/genetics
8.
Allergol. immunopatol ; 51(4): 87-93, 2023. tab, graf
Article in English | IBECS | ID: ibc-222638

ABSTRACT

Introduction: Pentraxin-3 (PTX3) is a soluble long pentraxin molecule that regulates inflammatory responses. This study aimed to determine the plasma levels of plasma PTX-3 as an inflammation marker in chronic spontaneous urticaria (CSU) and whether the PTX3 levels correlate with disease activity and other clinical parameters, including acute phase reactants and biomarkers. Methods: The study included 70 CSU patients and 30 healthy controls. Plasma PTX3 levels were measured by ELISA. CSU disease activity was evaluated with the urticaria activity score summed over 7 days. Complete blood count, C-reactive protein (CRP), transaminases, total IgE, antinuclear antibody, anti-thyroid peroxidase, anti-thyroglobulin, and D-dimer levels were recorded. Results: Of the 70 patients, 52 (74.3%) were female, with a mean age of 37.51 ± 11.80 years. Disease activity was severe in 43, moderate in 15, and mild in 12 patients. Mean PTX3 levels were elevated in CSU patients compared to healthy controls (0.81 vs. 0.55 ng/mL, p = 0.031). The mean CRP levels were higher in patients than in the controls (4.26 vs. 1.57 mg/L, p = 0.023). Patients also had higher D-dimer levels than the controls (5.96 vs. 0.59 mg/L, p < 0.001). A significant positive correlation was found between PTX3 and CRP levels (r = 0.508, p < 0.001) and between D-dimer levels and UAS7 (r = 0.338, p = 0.004) and CRP (r = 0.213, p = 0.034) levels. A multivariable stepwise regression analysis showed that the one-unit increase in the CRP level increased to 38.19 units in the PTX3 level (95% confidence interval [17.40–58.98], p < 0.001). Conclusion: Circulating levels of CRP and PTX3, two members of the pentraxin family, are significantly correlated and elevated in CSU patients with increasing disease activity, indicating their utility as inflammatory markers in CSU (AU)


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Serum Amyloid P-Component/analysis , C-Reactive Protein/blood , Urticaria/blood , Urticaria/metabolism , Case-Control Studies , Prospective Studies , Biomarkers/blood , Chronic Disease
9.
Cell Rep ; 40(11): 111321, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103835

ABSTRACT

Advanced non-alcoholic fatty liver disease (NAFLD) is a rapidly emerging global health problem associated with pre-disposing genetic polymorphisms, most strikingly an isoleucine to methionine substitution in patatin-like phospholipase domain-containing protein 3 (PNPLA3-I148M). Here, we study how human hepatocytes with PNPLA3 148I and 148M variants engrafted in the livers of broadly immunodeficient chimeric mice respond to hypercaloric diets. As early as four weeks, mice developed dyslipidemia, impaired glucose tolerance, and steatosis with ballooning degeneration selectively in the human graft, followed by pericellular fibrosis after eight weeks of hypercaloric feeding. Hepatocytes with the PNPLA3-148M variant, either from a homozygous 148M donor or overexpressed in a 148I donor background, developed microvesicular and severe steatosis with frequent ballooning degeneration, resulting in more active steatohepatitis than 148I hepatocytes. We conclude that PNPLA3-148M in human hepatocytes exacerbates NAFLD. These models will facilitate mechanistic studies into human genetic variant contributions to advanced fatty liver diseases.


Subject(s)
Non-alcoholic Fatty Liver Disease , Acyltransferases , Animals , Hepatocytes/metabolism , Humans , Lipase/genetics , Lipase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Non-alcoholic Fatty Liver Disease/genetics , Phospholipases A2, Calcium-Independent
10.
J Clin Invest ; 131(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34623332

ABSTRACT

We studied a child with severe viral, bacterial, fungal, and parasitic diseases, who was homozygous for a loss-of-function mutation of REL, encoding c-Rel, which is selectively expressed in lymphoid and myeloid cells. The patient had low frequencies of NK, effector memory cells reexpressing CD45RA (Temra) CD8+ T cells, memory CD4+ T cells, including Th1 and Th1*, Tregs, and memory B cells, whereas the counts and proportions of other leukocyte subsets were normal. Functional deficits of myeloid cells included the abolition of IL-12 and IL-23 production by conventional DC1s (cDC1s) and monocytes, but not cDC2s. c-Rel was also required for induction of CD86 expression on, and thus antigen-presenting cell function of, cDCs. Functional deficits of lymphoid cells included reduced IL-2 production by naive T cells, correlating with low proliferation and survival rates and poor production of Th1, Th2, and Th17 cytokines by memory CD4+ T cells. In naive CD4+ T cells, c-Rel is dispensable for early IL2 induction but contributes to later phases of IL2 expression. The patient's naive B cells displayed impaired MYC and BCL2L1 induction, compromising B cell survival and proliferation and preventing their differentiation into Ig-secreting plasmablasts. Inherited c-Rel deficiency disrupts the development and function of multiple myeloid and lymphoid cells, compromising innate and adaptive immunity to multiple infectious agents.


Subject(s)
Genes, rel , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Child , Consanguinity , Female , Hematopoietic Stem Cell Transplantation , Homozygote , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Lymphocyte Activation , Lymphocytes/classification , Lymphocytes/immunology , Mutation , Myeloid Cells/immunology , Primary Immunodeficiency Diseases/therapy , Protein Isoforms
11.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34214472

ABSTRACT

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Subject(s)
CD28 Antigens/deficiency , Inheritance Patterns/genetics , Papillomaviridae/physiology , Skin/virology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Animals , Base Sequence , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Child , Endopeptidases/metabolism , Female , Genes, Recessive , HEK293 Cells , Homozygote , Humans , Immunity, Humoral , Immunologic Memory , Jurkat Cells , Keratinocytes/pathology , Male , Mice, Inbred C57BL , Oncogenes , Papilloma/pathology , Papilloma/virology , Pedigree , Protein Sorting Signals , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Front Microbiol ; 11: 1631, 2020.
Article in English | MEDLINE | ID: mdl-32849337

ABSTRACT

Nearly one third of the world's population is infected with Mycobacterium tuberculosis (Mtb). While much work has focused on the role of different Mtb encoded proteins in pathogenesis, recent studies have revealed that Mtb also transcribes many noncoding RNAs whose functions remain poorly characterized. We performed RNA sequencing and identified a subset of Mtb H37Rv-encoded small RNAs (<30 nts in length) that were produced in infected macrophages. Designated as smaller noncoding RNAs (sncRNAs), three of these predominated the read counts. Each of the three, sncRNA-1, sncRNA-6, and sncRNA-8 had surrounding sequences with predicted stable secondary RNA stem loops. Site-directed mutagenesis of the precursor sequences suggest the existence of a hairpin loop dependent RNA processing mechanism. A functional assessment of sncRNA-1 suggested that it positively regulated two mycobacterial transcripts involved in oleic acid biosynthesis. Complementary loss- and gain- of-function approaches revealed that sncRNA-1 positively supports Mtb growth and survival in nutrient-depleted cultures as well as in infected macrophages. Overall, the findings reveal that Mtb produces sncRNAs in infected cells, with sncRNA-1 modulating mycobacterial gene expression including genes coupled to oleic acid biogenesis.

13.
N Engl J Med ; 382(5): 437-445, 2020 01 30.
Article in English | MEDLINE | ID: mdl-31995689

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) can cause severe disease in children and adults with a variety of inherited or acquired T-cell immunodeficiencies, who are prone to multiple infections. It can also rarely cause disease in otherwise healthy persons. The pathogenesis of idiopathic CMV disease is unknown. Inbred mice that lack the gene encoding nitric oxide synthase 2 (Nos2) are susceptible to the related murine CMV infection. METHODS: We studied a previously healthy 51-year-old man from Iran who after acute CMV infection had an onset of progressive CMV disease that led to his death 29 months later. We hypothesized that the patient may have had a novel type of inborn error of immunity. Thus, we performed whole-exome sequencing and tested candidate mutant alleles experimentally. RESULTS: We found a homozygous frameshift mutation in NOS2 encoding a truncated NOS2 protein that did not produce nitric oxide, which determined that the patient had autosomal recessive NOS2 deficiency. Moreover, all NOS2 variants that we found in homozygosity in public databases encoded functional proteins, as did all other variants with an allele frequency greater than 0.001. CONCLUSIONS: These findings suggest that inherited NOS2 deficiency was clinically silent in this patient until lethal infection with CMV. Moreover, NOS2 appeared to be redundant for control of other pathogens in this patient. (Funded by the National Center for Advancing Translational Sciences and others.).


Subject(s)
Cytomegalovirus Infections , Frameshift Mutation , Nitric Oxide Synthase Type II/deficiency , Fatal Outcome , Female , Genotype , Homozygote , Humans , Loss of Function Mutation , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pedigree , Exome Sequencing
14.
Proc Natl Acad Sci U S A ; 116(38): 19055-19063, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484767

ABSTRACT

Juvenile-onset recurrent respiratory papillomatosis (JRRP) is a rare and debilitating childhood disease that presents with recurrent growth of papillomas in the upper airway. Two common human papillomaviruses (HPVs), HPV-6 and -11, are implicated in most cases, but it is still not understood why only a small proportion of children develop JRRP following exposure to these common viruses. We report 2 siblings with a syndromic form of JRRP associated with mild dermatologic abnormalities. Whole-exome sequencing of the patients revealed a private homozygous mutation in NLRP1, encoding Nucleotide-Binding Domain Leucine-Rich Repeat Family Pyrin Domain-Containing 1. We find the NLRP1 mutant allele to be gain of function (GOF) for inflammasome activation, as demonstrated by the induction of inflammasome complex oligomerization and IL-1ß secretion in an overexpression system. Moreover, patient-derived keratinocytes secrete elevated levels of IL-1ß at baseline. Finally, both patients displayed elevated levels of inflammasome-induced cytokines in the serum. Six NLRP1 GOF mutations have previously been described to underlie 3 allelic Mendelian diseases with differing phenotypes and modes of inheritance. Our results demonstrate that an autosomal recessive, syndromic form of JRRP can be associated with an NLRP1 GOF mutation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Gain of Function Mutation , Homozygote , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/pathology , Child, Preschool , Cytokines/metabolism , Female , Humans , Infant , Inflammasomes , Keratinocytes/cytology , Keratinocytes/immunology , Keratinocytes/metabolism , Male , NLR Proteins , Pedigree , Siblings , Syndrome
15.
J Exp Med ; 216(9): 2057-2070, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31270247

ABSTRACT

Vaccination against measles, mumps, and rubella (MMR) and yellow fever (YF) with live attenuated viruses can rarely cause life-threatening disease. Severe illness by MMR vaccines can be caused by inborn errors of type I and/or III interferon (IFN) immunity (mutations in IFNAR2, STAT1, or STAT2). Adverse reactions to the YF vaccine have remained unexplained. We report two otherwise healthy patients, a 9-yr-old boy in Iran with severe measles vaccine disease at 1 yr and a 14-yr-old girl in Brazil with viscerotropic disease caused by the YF vaccine at 12 yr. The Iranian patient is homozygous and the Brazilian patient compound heterozygous for loss-of-function IFNAR1 variations. Patient-derived fibroblasts are susceptible to viruses, including the YF and measles virus vaccine strains, in the absence or presence of exogenous type I IFN. The patients' fibroblast phenotypes are rescued with WT IFNAR1 Autosomal recessive, complete IFNAR1 deficiency can result in life-threatening complications of vaccination with live attenuated measles and YF viruses in previously healthy individuals.


Subject(s)
Inheritance Patterns/genetics , Measles Vaccine/adverse effects , Receptor, Interferon alpha-beta/deficiency , Yellow Fever Vaccine/adverse effects , Adolescent , Alleles , Child , Female , Humans , Immunity , Infant , Interferon Type I/metabolism , Male , Measles Vaccine/immunology , Mutant Proteins/metabolism , Mutation/genetics , Pedigree , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Yellow Fever Vaccine/immunology
16.
J Exp Med ; 216(8): 1777-1790, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31213488

ABSTRACT

Fulminant viral hepatitis (FVH) is a devastating and unexplained condition that strikes otherwise healthy individuals during primary infection with common liver-tropic viruses. We report a child who died of FVH upon infection with hepatitis A virus (HAV) at age 11 yr and who was homozygous for a private 40-nucleotide deletion in IL18BP, which encodes the IL-18 binding protein (IL-18BP). This mutation is loss-of-function, unlike the variants found in a homozygous state in public databases. We show that human IL-18 and IL-18BP are both secreted mostly by hepatocytes and macrophages in the liver. Moreover, in the absence of IL-18BP, excessive NK cell activation by IL-18 results in uncontrolled killing of human hepatocytes in vitro. Inherited human IL-18BP deficiency thus underlies fulminant HAV hepatitis by unleashing IL-18. These findings provide proof-of-principle that FVH can be caused by single-gene inborn errors that selectively disrupt liver-specific immunity. They also show that human IL-18 is toxic to the liver and that IL-18BP is its antidote.


Subject(s)
Genetic Diseases, Inborn/complications , Hepatitis A/genetics , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Massive Hepatic Necrosis/genetics , Child , Cohort Studies , Female , Gene Frequency , Hep G2 Cells , Hepatitis A/virology , Hepatitis A Virus, Human , Hepatocytes/metabolism , Homozygote , Humans , Interleukin-18/metabolism , Killer Cells, Natural/immunology , Liver/metabolism , Loss of Function Mutation , Lymphocyte Activation/genetics , Macrophages/metabolism , Male , Massive Hepatic Necrosis/virology , Pedigree , Exome Sequencing
17.
J Exp Med ; 215(10): 2567-2585, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30143481

ABSTRACT

Life-threatening pulmonary influenza can be caused by inborn errors of type I and III IFN immunity. We report a 5-yr-old child with severe pulmonary influenza at 2 yr. She is homozygous for a loss-of-function IRF9 allele. Her cells activate gamma-activated factor (GAF) STAT1 homodimers but not IFN-stimulated gene factor 3 (ISGF3) trimers (STAT1/STAT2/IRF9) in response to IFN-α2b. The transcriptome induced by IFN-α2b in the patient's cells is much narrower than that of control cells; however, induction of a subset of IFN-stimulated gene transcripts remains detectable. In vitro, the patient's cells do not control three respiratory viruses, influenza A virus (IAV), parainfluenza virus (PIV), and respiratory syncytial virus (RSV). These phenotypes are rescued by wild-type IRF9, whereas silencing IRF9 expression in control cells increases viral replication. However, the child has controlled various common viruses in vivo, including respiratory viruses other than IAV. Our findings show that human IRF9- and ISGF3-dependent type I and III IFN responsive pathways are essential for controlling IAV.


Subject(s)
Alleles , Homozygote , Influenza, Human , Interferon-Stimulated Gene Factor 3, gamma Subunit/deficiency , Orthomyxoviridae/immunology , Pneumonia, Viral , Female , Humans , Infant , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/pathology , Interferon alpha-2/genetics , Interferon alpha-2/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology
18.
Eur J Med Genet ; 60(8): 426-432, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28576691

ABSTRACT

BACKGROUND: Acute liver failure (ALF) in children can be life-threatening. Although many causes are known, ALF remains unexplained in about half of the cases. Recently, bi-allelic mutations in NBAS were reported to underlie recurrent episodes of elevated liver transaminases (ELT) and ALF in the context of diverse extrahepatic phenotypes. METHODS AND RESULTS: We here describe two sisters, born to non-consanguineous Portuguese parents, who had short stature and presented with recurrent episodes of severe ELT triggered by febrile respiratory viral infections since early childhood. Patient 1 had mild facial dysmorphism and died during the second ELT crisis at 3-11/12 years of age. Patient 2, currently 9 years old, had multiple episodes of ELT (>30), twice with ALF, often accompanied by extensive urticaria and facial angioedema. Whole-exome and Sanger sequencing revealed that both patients carried previously undescribed compound heterozygous mutations of NBAS (NM_015909.3): c.680A > C (p.His227Pro), affecting an evolutionarily conserved residue, and c.1749G > A (p.Trp583*), causing a premature stop codon. Both mutations are predicted to be highly damaging. The parents and two younger siblings are healthy and heterozygous for one or another mutant allele. CONCLUSION: The multiplex kindred reported herein expands the genotypic and phenotypic spectrum of this recently described clinical syndrome due to autosomal recessive NBAS deficiency.


Subject(s)
Liver Failure, Acute/genetics , Mutation, Missense , Neoplasm Proteins/genetics , Transaminases/metabolism , Adult , Alleles , Child , Child, Preschool , Codon, Terminator , Female , Humans , Liver/enzymology , Liver/pathology , Liver Failure, Acute/diagnosis , Male , Pedigree , Siblings
19.
J Am Coll Cardiol ; 69(13): 1653-1665, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28359509

ABSTRACT

BACKGROUND: Myocarditis is inflammation of the heart muscle that can follow various viral infections. Why children only rarely develop life-threatening acute viral myocarditis (AVM), given that the causal viral infections are common, is unknown. Genetic lesions might underlie such susceptibilities. Mouse genetic studies demonstrated that interferon (IFN)-α/ß immunity defects increased susceptibility to virus-induced myocarditis. Moreover, variations in human TLR3, a potent inducer of IFNs, were proposed to underlie AVM. OBJECTIVES: This study sought to evaluate the hypothesis that human genetic factors may underlie AVM in previously healthy children. METHODS: We tested the role of TLR3-IFN immunity using human induced pluripotent stem cell-derived cardiomyocytes. We then performed whole-exome sequencing of 42 unrelated children with acute myocarditis (AM), some with proven viral causes. RESULTS: We found that TLR3- and STAT1-deficient cardiomyocytes were not more susceptible to Coxsackie virus B3 (CVB3) infection than control cells. Moreover, CVB3 did not induce IFN-α/ß and IFN-α/ß-stimulated genes in control cardiomyocytes. Finally, exogenous IFN-α did not substantially protect cardiomyocytes against CVB3. We did not observe a significant enrichment of rare variations in TLR3- or IFN-α/ß-related genes. Surprisingly, we found that homozygous but not heterozygous rare variants in genes associated with inherited cardiomyopathies were significantly enriched in AM-AVM patients compared with healthy individuals (p = 2.22E-03) or patients with other diseases (p = 1.08E-04). Seven of 42 patients (16.7%) carried rare biallelic (homozygous or compound heterozygous) nonsynonymous or splice-site variations in 6 cardiomyopathy-associated genes (BAG3, DSP, PKP2, RYR2, SCN5A, or TNNI3). CONCLUSIONS: Previously silent recessive defects of the myocardium may predispose to acute heart failure presenting as AM, notably after common viral infections in children.


Subject(s)
Cardiomyopathies/genetics , Enterovirus B, Human/physiology , Myocarditis/genetics , STAT1 Transcription Factor/genetics , Toll-Like Receptor 3/genetics , Cardiomyopathies/complications , Case-Control Studies , Female , Host-Pathogen Interactions/genetics , Humans , Induced Pluripotent Stem Cells , Male , Myocarditis/virology , Myocytes, Cardiac/virology
20.
PLoS One ; 9(1): e85274, 2014.
Article in English | MEDLINE | ID: mdl-24416377

ABSTRACT

Physiological stress resulting from infections, trauma, surgery, alcoholism, malnutrition, and/or pregnancy results in a substantial depletion of immature CD4(+)CD8(+) thymocytes. We previously identified 18 distinct stress-responsive microRNAs (miRs) in the thymus upon systemic stress induced by lipopolysaccharide (LPS) or the synthetic glucocorticoid, dexamethasone (Dex). MiRs are short, non-coding RNAs that play critical roles in the immune system by targeting diverse mRNAs, suggesting that their modulation in the thymus in response to stress could impact thymopoiesis. MiR-181d is one such stress-responsive miR, exhibiting a 15-fold down-regulation in expression. We utilized both transgenic and gene-targeting approaches to study the impact of miR-181d on thymopoiesis under normal and stress conditions. The over-expression of miR-181d in developing thymocytes reduced the total number of immature CD4(+)CD8(+) thymocytes. LPS or Dex injections caused a 4-fold greater loss of these cells when compared with the wild type controls. A knockout mouse was developed to selectively eliminate miR-181d, leaving the closely spaced and contiguous family member miR-181c intact. The targeted elimination of just miR-181d resulted in a thymus stress-responsiveness similar to wild-type mice. These experiments suggest that one or more of three other miR-181 family members have overlapping or compensatory functions. Gene expression comparisons of thymocytes from the wild type versus transgenic mice indicated that miR-181d targets a number of stress, metabolic, and signaling pathways. These findings demonstrate that selected miRs enhance stress-mediated thymic involution in vivo.


Subject(s)
MicroRNAs/genetics , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , Base Sequence , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Dexamethasone/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Transgenic , MicroRNAs/immunology , Molecular Sequence Data , Primary Cell Culture , Stress, Physiological , Thymocytes/drug effects , Thymocytes/immunology , Thymus Gland/drug effects , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...