Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Autoimmun ; 116: 102571, 2021 01.
Article in English | MEDLINE | ID: mdl-33223341

ABSTRACT

Guillain-Barré syndrome (GBS), including its variant Miller Fisher syndrome (MFS), is an acute peripheral neuropathy that involves autoimmune mechanisms leading to the production of autoantibodies to gangliosides; sialic acid-containing glycosphingolipids. Although association with various genetic polymorphisms in the major histocompatibility complex (MHC) is shown in other autoimmune diseases, GBS is an exception, showing no such link. No significant association was found by genome wide association studies, suggesting that GBS is not associated with common variants. To address the involvement of rare variants in GBS, we analyzed Siglec-10, a sialic acid-recognizing inhibitory receptor expressed on B cells. Here we demonstrate that two rare variants encoding R47Q and A108V substitutions in the ligand-binding domain are significantly accumulated in patients with GBS. Because of strong linkage disequilibrium, there was no patient carrying only one of them. Recombinant Siglec-10 protein containing R47Q but not A108V shows impaired binding to gangliosides. Homology modeling revealed that the R47Q substitution causes marked alteration in the ligand-binding site. Thus, GBS is associated with a rare variant of the SIGLEC10 gene that impairs ligand binding of Siglec-10. Because Siglec-10 regulates antibody production to sialylated antigens, our finding suggests that Siglec-10 regulates development of GBS by suppressing antibody production to gangliosides, with defects in its function predisposing to disease.


Subject(s)
Gangliosides/immunology , Genetic Predisposition to Disease , Guillain-Barre Syndrome/immunology , Lectins/immunology , Mutation, Missense/immunology , Polymorphism, Single Nucleotide/immunology , Receptors, Cell Surface/immunology , Alleles , Amino Acid Sequence , Autoantibodies/immunology , Binding Sites/genetics , Female , Gangliosides/metabolism , Gene Frequency , Genotype , Guillain-Barre Syndrome/genetics , Guillain-Barre Syndrome/metabolism , Humans , Lectins/genetics , Lectins/metabolism , Male , Middle Aged , Miller Fisher Syndrome/genetics , Miller Fisher Syndrome/immunology , Miller Fisher Syndrome/metabolism , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid
2.
J Biol Chem ; 290(9): 5462-9, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25568323

ABSTRACT

Human immunoglobulin G isotype 4 (IgG4) antibodies (Abs) are potential candidates for immunotherapy when reduced effector functions are desirable. IgG4 Abs are dynamic molecules able to undergo a process known as Fab arm exchange (FAE). This results in functionally monovalent, bispecific antibodies (bsAbs) with unknown specificity and hence, potentially, reduced therapeutic efficacy. IgG4 FAE is suggested to be an important biological mechanism that provides the basis for the anti-inflammatory activity attributed to IgG4 Abs. To date, the mechanism of FAE is not entirely understood and studies measuring FAE in ex vivo matrices have been hampered by the presence and abundance of endogenous IgG4 wild-type (WT) Abs. Using representative humanized WT IgG4 monoclonal Abs, namely, anti-IL-6 and anti-TNF, and a core-hinge stabilized serine 228 to proline (S228P) anti-IL-6 IgG4 mutant, it is demonstrated for the first time how anti-IgG4 affinity chromatography can be used to prepare physiologically relevant matrices for assessing and quantifying FAE. A novel method for quantifying FAE using a single MSD immunoassay is also reported and confirms previous findings that, dependent on the redox conditions, the S228P mutation can prevent IgG4 FAE to undetectable levels both in vitro and in vivo. Together, the findings and novel methodologies will allow researchers to monitor and quantify FAE of their own IgG4 molecules in physiologically relevant matrices.


Subject(s)
Immunoglobulin Fab Fragments/genetics , Immunoglobulin G/genetics , Mutation, Missense/genetics , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Blotting, Western , Humans , Immunoglobulin Fab Fragments/blood , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mutation, Missense/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
PLoS Pathog ; 9(5): e1003354, 2013.
Article in English | MEDLINE | ID: mdl-23671418

ABSTRACT

Surveillance data indicate that most circulating A(H1N1)pdm09 influenza viruses have remained antigenically similar since they emerged in humans in 2009. However, antigenic drift is likely to occur in the future in response to increasing population immunity induced by infection or vaccination. In this study, sequential passaging of A(H1N1)pdm09 virus by contact transmission through two independent series of suboptimally vaccinated ferrets resulted in selection of variant viruses with an amino acid substitution (N156K, H1 numbering without signal peptide; N159K, H3 numbering without signal peptide; N173K, H1 numbering from first methionine) in a known antigenic site of the viral HA. The N156K HA variant replicated and transmitted efficiently between naïve ferrets and outgrew wildtype virus in vivo in ferrets in the presence and absence of immune pressure. In vitro, in a range of cell culture systems, the N156K variant rapidly adapted, acquiring additional mutations in the viral HA that also potentially affected antigenic properties. The N156K escape mutant was antigenically distinct from wildtype virus as shown by binding of HA-specific antibodies. Glycan binding assays demonstrated the N156K escape mutant had altered receptor binding preferences compared to wildtype virus, which was supported by computational modeling predictions. The N156K substitution, and culture adaptations, have been detected in human A(H1N1)pdm09 viruses with N156K preferentially reported in sequences from original clinical samples rather than cultured isolates. This study demonstrates the ability of the A(H1N1)pdm09 virus to undergo rapid antigenic change to evade a low level vaccine response, while remaining fit in a ferret transmission model of immunization and infection. Furthermore, the potential changes in receptor binding properties that accompany antigenic changes highlight the importance of routine characterization of clinical samples in human A(H1N1)pdm09 influenza surveillance.


Subject(s)
Antigens, Viral , Genetic Drift , Influenza A Virus, H1N1 Subtype , Influenza, Human , Mutation, Missense/immunology , Pandemics , Amino Acid Substitution , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Chick Embryo , Disease Models, Animal , Dogs , Female , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/epidemiology , Influenza, Human/genetics , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Male
4.
Mol Biol (Mosk) ; 49(2): 342-50, 2015.
Article in Russian | MEDLINE | ID: mdl-26065262

ABSTRACT

Changes associated with the resistance to physical and chemical factors in the hemagglutinin (HA) of influenza A viruses may play an important role in the selection of different influenza variants during circulation in nature. Here, we studied the escape mutants of influenza virus A/mallard/Pennsylvania/10218/84 (H5N2) that were selected by the monoclonal antibody. The escape mutant m4F11(4) carried a single amino acid substitution in large subunit (HA1) of the HA, S145P1, and two ones, m4G10(10) and m4G10(6), had additional amino acid changes in the small subunit (HA2), namely: L124F2 and L124F2 + N79D2, respectively. As it has been found the substitutions appeared in the HA2 of m4G(10) and m4G(6) viruses compensated negative effect of the S145P1 mutation and provided a significant increase in the viral replication ability at the early stage of infection in embryonated chicken eggs as well as in HA thermostability in comparison with m4F11(4) mutant. Phenotypic properties that provide advantages in the process of virus replication can play a role of the positive selection factor in viral population.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H5N2 Subtype , Mutation, Missense/immunology , Amino Acid Substitution , Animals , Chick Embryo , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N2 Subtype/immunology , Influenza in Birds/genetics , Influenza in Birds/immunology
5.
J Clin Immunol ; 33(1): 127-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22996269

ABSTRACT

PURPOSE: Immunological and molecular evaluation of a patient presenting with recurrent infections caused by Streptococcus pneumoniae and low complement component 3 (C3) levels. METHODS: Immunological evaluation included complement components and immunoglobulin level quantification as well as number and function of T cells, B cells and neutrophils. Serotype-specific immunoglobulin G antibodies against S. pneumoniae capsular polysaccharides were quantified by ELISA in serum samples before and after vaccination with unconjugated polysaccharide vaccine. For the molecular analysis, genomic DNA from the patient and parents were isolated and all exons as well as exon-intron boundaries of the C3 gene were sequenced by Sanger sequencing. RESULTS: A 16-year-old male, born to consanguineous parents, presented with recurrent episodes of pneumonia caused by S. pneumoniae and bronchiectasis. The patient showed severely reduced C3 and immunoglobulin A levels, while the parents showed moderately reduced levels of C3. Mutational analysis revealed a novel, homozygous missense mutation in the C3 gene (c. C4554G, p. Cys1518Trp), substituting a highly conserved amino acid in the C345C domain of C3 and interrupting one of its disulfide bonds. Both parents were found to be carriers of the affected allele. Vaccination against S. pneumoniae resulted in considerable clinical improvement. CONCLUSIONS: We report a novel homozygous mutation in the C3 gene in a patient with concomitant selective IgA deficiency who presented with a marked clinical improvement after vaccination against S. pneumoniae. This observation underlines the notion that vaccination against this microorganism is an important strategy for treatment of PID patients, particularly those presenting with increased susceptibility to infections caused by this agent.


Subject(s)
Complement C3/genetics , IgA Deficiency/genetics , IgA Deficiency/immunology , Mutation, Missense , Adolescent , Bronchiectasis/complications , Bronchiectasis/genetics , Bronchiectasis/immunology , Child , Child, Preschool , Comorbidity , Complement C3/antagonists & inhibitors , Complement C3/biosynthesis , Female , Humans , IgA Deficiency/complications , Male , Mutation, Missense/genetics , Mutation, Missense/immunology , Pedigree , Pneumococcal Vaccines/therapeutic use , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/prevention & control , Point Mutation/genetics , Point Mutation/immunology , Sequence Homology, Amino Acid
6.
J Clin Immunol ; 33(1): 162-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22926405

ABSTRACT

PURPOSE: Acute Hemorrhagic Leukoencephalitis (AHLE) is a rare demyelinating disorder of acute onset, rapid deterioration and significant morbidity and mortality. Most often described as a post-infectious complication of an upper respiratory illness, its precise pathophysiology remains unclear. We describe two pediatric patients with AHLE with partial complement factor I (FI) deficiency whose successful treatment included the interleukin-1 (IL-1) receptor antagonist, anakinra, implicating a role for FI and IL-1 in this disorder. METHODS: Extensive clinical workup of two patients presenting with AHLE revealed complement abnormalities, specifically related to the alternative pathway and its regulator, FI. Aggressive management with steroids, immunoglobulin, and anakinra ultimately led to improvement of clinical status and near return to neurologic baseline in both patients. Genetic sequencing of the FI coding regions of the patients and their families was performed. In vitro protein expression studies and immunohistochemistry of fixed brain tissue was used to investigate pathogenic mechanisms. RESULTS: Two novel mutations in FI were identified in our patients, which result in failure to secrete FI. Immunohistochemical evaluation of brain tissue demonstrated positive staining for C3, membrane attack complex (MAC) and IL-1. CONCLUSIONS: We propose AHLE is an unreported, rare phenotype for partial FI deficiency. The upregulation of C3, MAC and IL-1 with subsequent demyelination support a pathologic role for complement activation in AHLE, and suggest anakinra as an important adjunctive therapy in this disease.


Subject(s)
Complement Factor I/genetics , Leukoencephalitis, Acute Hemorrhagic/genetics , Leukoencephalitis, Acute Hemorrhagic/immunology , Mutation, Missense/immunology , Neurons/immunology , Neurons/pathology , Adolescent , Adult , Child , Complement Activation/genetics , Complement Activation/immunology , Complement C3/physiology , Complement Factor I/deficiency , Complement Factor I/metabolism , Complement Membrane Attack Complex/physiology , Female , HEK293 Cells , Humans , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1/physiology , Leukoencephalitis, Acute Hemorrhagic/pathology , Male , Neurons/metabolism , Pedigree
7.
Proc Natl Acad Sci U S A ; 107(7): 3024-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133615

ABSTRACT

DNA ligase IV (LIG4) is an essential component of the nonhomologous end-joining (NHEJ) repair pathway and plays a key role in V(D)J recombination. Hypomorphic LIG4 mutations in humans are associated with increased cellular radiosensitivity, microcephaly, facial dysmorphisms, growth retardation, developmental delay, and a variable degree of immunodeficiency. We have generated a knock-in mouse model with a homozygous Lig4 R278H mutation that corresponds to the first LIG4 mutation reported in humans. The phenotype of homozygous mutant mice Lig4(R278H/R278H) (Lig4(R/R)) includes growth retardation, a decreased life span, a severe cellular sensitivity to ionizing radiation, and a very severe, but incomplete block in T and B cell development. Peripheral T lymphocytes show an activated and anergic phenotype, reduced viability, and a restricted repertoire, reminiscent of human leaky SCID. Genomic instability is associated with a high rate of thymic tumor development. Finally, Lig4(R/R) mice spontaneously produce low-affinity antibodies that include autoreactive specificities, but are unable to mount high-affinity antibody responses. These findings highlight the importance of LIG4 in lymphocyte development and function, and in genomic stability maintenance, and provide a model for the complex phenotype of LIG4 syndrome in humans.


Subject(s)
Abnormalities, Multiple/genetics , Antibody Formation/genetics , DNA Ligases/genetics , Developmental Disabilities/genetics , Disease Models, Animal , Mutation, Missense/genetics , Severe Combined Immunodeficiency/genetics , Animals , Apoptosis/immunology , Blotting, Southern , Child , DNA Ligase ATP , DNA Ligases/immunology , Flow Cytometry , Humans , Immunoglobulins/blood , Immunophenotyping , Mice , Mutation, Missense/immunology , Syndrome
8.
J Biol Chem ; 286(39): 34457-67, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21841002

ABSTRACT

The microtubule-associated protein Tau plays a critical role in the pathogenesis of Alzheimer disease and several related disorders (tauopathies). In the disease Tau aggregates and becomes hyperphosphorylated forming paired helical and straight filaments, which can further condense into higher order neurofibrillary tangles in neurons. The development of this pathology is consistently associated with progressive neuronal loss and cognitive decline. The identification of tractable therapeutic targets in this pathway has been challenging, and consequently very few clinical studies addressing Tau pathology are underway. Recent active immunization studies have raised the possibility of modulating Tau pathology by activating the immune system. Here we report for the first time on passive immunotherapy for Tau in two well established transgenic models of Tau pathogenesis. We show that peripheral administration of two antibodies against pathological Tau forms significantly reduces biochemical Tau pathology in the JNPL3 mouse model. We further demonstrate that peripheral administration of the same antibodies in the more rapidly progressive P301S tauopathy model not only reduces Tau pathology quantitated by biochemical assays and immunohistochemistry, but also significantly delays the onset of motor function decline and weight loss. This is accompanied by a reduction in neurospheroids, providing direct evidence of reduced neurodegeneration. Thus, passive immunotherapy is effective at preventing the buildup of intracellular Tau pathology, neurospheroids, and associated symptoms, although the exact mechanism remains uncertain. Tau immunotherapy should therefore be considered as a therapeutic approach for the treatment of Alzheimer disease and other tauopathies.


Subject(s)
Alzheimer Disease/therapy , Antibodies/immunology , Antibodies/pharmacology , Immunization, Passive/methods , tau Proteins/immunology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amino Acid Substitution/immunology , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Motor Activity/immunology , Mutation, Missense/immunology , tau Proteins/genetics
9.
J Immunol ; 184(8): 4362-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20212095

ABSTRACT

Two missense variants (D299G and T399I) of TLR4 are cosegregated in individuals of European descent and, in a number of test systems, result in reduced responsiveness to endotoxin. How these changes within the ectodomain (ecd) of TLR4 affect TLR4 function is unclear. For both wild-type and D299G.T399I TLR4, we used endotoxinCD14 and endotoxinMD-2 complexes of high specific radioactivity to measure: 1) interaction of recombinant MD-2TLR4 with endotoxinCD14 and TLR4 with endotoxinMD-2; 2) expression of functional MD-2TLR4 and TLR4; and 3) MD-2TLR4 and TLR4-dependent cellular endotoxin responsiveness. Both wild-type and D299G.T399I TLR4(ecd) demonstrated high affinity (K(d) approximately 200 pM) interaction of endotoxinCD14 with MD-2TLR4(ecd) and endotoxinMD-2 with TLR4(ecd). However, levels of functional TLR4 were reduced up to 2-fold when D299G.T399I TLR4 was coexpressed with MD-2 and >10-fold when expressed without MD-2, paralleling differences in cellular endotoxin responsiveness. The dramatic effect of the D299G.T399I haplotype on expression of functional TLR4 without MD-2 suggests that cells expressing TLR4 without MD-2 are most affected by these polymorphisms.


Subject(s)
Genetic Variation , Lymphocyte Antigen 96/genetics , Mutation, Missense , Polymorphism, Genetic , Toll-Like Receptor 4/genetics , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Cell Line , Dose-Response Relationship, Immunologic , Endotoxins/metabolism , Endotoxins/pharmacology , Genetic Variation/immunology , Haplotypes , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/biosynthesis , Lymphocyte Antigen 96/metabolism , Mutation, Missense/immunology , Polymorphism, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/metabolism
10.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34889940

ABSTRACT

B cell self-tolerance is maintained through multiple checkpoints, including restraints on intracellular signaling and cell trafficking. P2RY8 is a receptor with established roles in germinal center (GC) B cell migration inhibition and growth regulation. Somatic P2RY8 variants are common in GC-derived B cell lymphomas. Here, we identify germline novel or rare P2RY8 missense variants in lupus kindreds or the related antiphospholipid syndrome, including a "de novo" variant in a child with severe nephritis. All variants decreased protein expression, F-actin abundance, and GPCR-RhoA signaling, and those with stronger effects increased AKT and ERK activity and cell migration. Remarkably, P2RY8 was reduced in B cell subsets from some SLE patients lacking P2RY8 gene variants. Low P2RY8 correlated with lupus nephritis and increased age-associated B cells and plasma cells. By contrast, P2RY8 overexpression in cells and mice restrained plasma cell development and reinforced negative selection of DNA-reactive developing B cells. These findings uncover a role of P2RY8 in immunological tolerance and lupus pathogenesis.


Subject(s)
Antiphospholipid Syndrome/immunology , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Mutation, Missense/immunology , Receptors, Purinergic P2Y/immunology , Animals , Antiphospholipid Syndrome/genetics , Antiphospholipid Syndrome/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immune Tolerance/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Male , Mice, Inbred C57BL , Mutation, Missense/genetics , Pedigree , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Purinergic P2Y/genetics , Receptors, Purinergic P2Y/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
11.
J Mol Biol ; 434(2): 167332, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34717971

ABSTRACT

Extensive glycosylation of viral glycoproteins is a key feature of the antigenic surface of viruses and yet glycan processing can also be influenced by the manner of their recombinant production. The low yields of the soluble form of the trimeric spike (S) glycoprotein from SARS-CoV-2 has prompted advances in protein engineering that have greatly enhanced the stability and yields of the glycoprotein. The latest expression-enhanced version of the spike incorporates six proline substitutions to stabilize the prefusion conformation (termed SARS-CoV-2 S HexaPro). Although the substitutions greatly enhanced expression whilst not compromising protein structure, the influence of these substitutions on glycan processing has not been explored. Here, we show that the site-specific N-linked glycosylation of the expression-enhanced HexaPro resembles that of an earlier version containing two proline substitutions (2P), and that both capture features of native viral glycosylation. However, there are site-specific differences in glycosylation of HexaPro when compared to 2P. Despite these discrepancies, analysis of the serological reactivity of clinical samples from infected individuals confirmed that both HexaPro and 2P protein are equally able to detect IgG, IgA, and IgM responses in all sera analysed. Moreover, we extend this observation to include an analysis of glycan engineered S protein, whereby all N-linked glycans were converted to oligomannose-type and conclude that serological activity is not impacted by large scale changes in glycosylation. These observations suggest that variations in glycan processing will not impact the serological assessments currently being performed across the globe.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Mutation, Missense/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/blood , Binding Sites/genetics , COVID-19/virology , Glycosylation , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mannose/metabolism , Mutation, Missense/genetics , Oligosaccharides/metabolism , Polysaccharides/metabolism , Proline/genetics , Proline/immunology , Proline/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism
12.
Blood ; 113(2): 338-46, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18927437

ABSTRACT

Perforin-mediated lymphocyte cytotoxicity is critical for pathogen elimination and immune homeostasis. Perforin disruption of target cell membranes is hypothesized to require binding of a calcium-dependent, lipid-inserting, C2 domain. In a family affected by hemophagocytic lymphohistiocytosis, a severe inflammatory disorder caused by perforin deficiency, we identified 2 amino acid substitutions in the perforin C2 domain: T435M, a previously identified mutant with disputed pathogenicity, and Y438C, a novel substitution. Using biophysical modeling, we predicted that the T435M substitution, but not Y438C, would interfere with calcium binding and thus cytotoxic function. The capacity for cytotoxic function was tested after expression of the variant perforins in rat basophilic leukemia cells and murine cytotoxic T lymphocytes. As predicted, cells transduced with perforin-T435M lacked cytotoxicity, but those expressing perforin-Y438C displayed intact cytotoxic function. Using novel antibody-capture and liposome-binding assays, we found that both mutant perforins were secreted; however, only nonmutated and Y438C-substituted perforins were capable of calcium-dependent lipid binding. In addition, we found that perforin-Y438C was capable of mediating cytotoxicity without apparent proteolytic maturation. This study clearly demonstrates the pathogenicity of the T435M mutation and illustrates, for the first time, the critical role of the human perforin C2 domain for calcium-dependent, cytotoxic function.


Subject(s)
Calcium/immunology , Cell Membrane/immunology , Membrane Lipids/immunology , Mutation, Missense/immunology , Perforin/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Substitution/immunology , Animals , Cell Line, Tumor , Cell Membrane/genetics , Homeostasis/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Mice , Perforin/genetics , Protein Structure, Tertiary/genetics , Rats
13.
Proc Natl Acad Sci U S A ; 105(32): 11311-6, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18678888

ABSTRACT

Recombinant immunotoxins are hybrid proteins composed of an Fv that binds to a tumor antigen fused to a bacterial or plant toxin. Immunotoxin BL22 targets CD22 positive malignancies and is composed of an anti-CD22 Fv fused to a 38-kDa fragment of Pseudomonas exotoxin A (PE38). BL22 has produced many complete remissions in drug-resistant Hairy cell leukemia, where many treatment cycles can be given, because neutralizing antibodies do not form. In marked contrast, only minor responses have been observed in trials with immunotoxins targeting solid tumors, because only a single treatment cycle can be given before antibodies develop. To allow more treatment cycles and increase efficacy, we have produced a less immunogenic immunotoxin by identifying and eliminating most of the B cell epitopes on PE38. This was accomplished by mutation of specific large hydrophilic amino acids (Arg, Gln, Glu, Lys) to Ala, Ser, or Gly. The new immunotoxin (HA22-8X) is significantly less immunogenic in three strains of mice, yet retains full cytotoxic and anti-tumor activities. Elimination of B-cell epitopes is a promising approach to the production of less immunogenic proteins for therapeutic purposes.


Subject(s)
ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Antibodies/genetics , Antibodies/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Enterotoxins/genetics , Enterotoxins/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Exotoxins/genetics , Exotoxins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Virulence Factors/genetics , Virulence Factors/immunology , ADP Ribose Transferases/therapeutic use , Amino Acid Substitution/immunology , Animals , Antibodies/therapeutic use , Bacterial Toxins/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Enterotoxins/therapeutic use , Exotoxins/therapeutic use , Humans , Leukemia, Hairy Cell/drug therapy , Leukemia, Hairy Cell/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation, Missense/immunology , Rabbits , Recombinant Fusion Proteins/therapeutic use , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
14.
Sci Immunol ; 6(63): eabf6723, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34533979

ABSTRACT

Inhibitor of nuclear factor kappa B kinase alpha (IKKα) is critical for p100/NF-κB2 phosphorylation and processing into p52 and activation of the noncanonical NF-κB pathway. A patient with recurrent infections, skeletal abnormalities, absent secondary lymphoid structures, reduced B cell numbers, hypogammaglobulinemia, and lymphocytic infiltration of intestine and liver was found to have a homozygous p.Y580C mutation in the helix-loop-helix domain of IKKα. The mutation preserves IKKα kinase activity but abolishes the interaction of IKKα with its activator NF-κB­inducing kinase and impairs lymphotoxin-ß­driven p100/NF-κB2 processing and VCAM1 expression. Homozygous IKKαY580C/Y580C mutant mice phenocopy the patient findings; lack marginal zone B cells, germinal centers, and antigen-specific T cell response to cutaneous immunization; have impaired Il17a expression; and are susceptible to cutaneous Staphylococcus aureus infection. In addition, these mice demonstrate a severe reduction in medullary thymic epithelial cells, impaired thymocyte negative selection, a restricted TCRVß repertoire, a selective expansion of potentially autoreactive T cell clones, a decreased frequency of regulatory T cells, and infiltration of liver, pancreas, and lung by activated T cells coinciding with organ damage. Hence, this study identifies IKKα deficiency as a previously undescribed cause of primary immunodeficiency with associated autoimmunity.


Subject(s)
Autoimmunity/immunology , I-kappa B Kinase/immunology , Mutation, Missense/genetics , Animals , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense/immunology
15.
Front Immunol ; 12: 759308, 2021.
Article in English | MEDLINE | ID: mdl-34975848

ABSTRACT

Rare autosomal-recessive variants in tetratricopeptide repeat domain 7A (TTC7A) gene have been shown to cause intestinal and immune disorders of variable severity. Missense mutations in TTC7A gene, usually retaining most of the functional motifs, is associated with relative milder clinical presentations. In this study, we reported a patient who was suffering from severe multiple intestinal atresia (MIA) with combined immunodeficiency (CID) that led to the pyloric diaphragm, ileum atresia, colon stenosis, and multiple episodes of sepsis. In spite of several surgeries and supportive treatment, the patient died of severe sepsis and multiple organ failure at age of 3 months. The whole exome sequencing (WES) of peripheral blood samples identified a novel homozygous TTC7A missense mutation (c. 206T>C, p. L69P), inherited from his parents with consanguineous marriage. In silico analysis revealed that a hydrogen bond present between Gly65 and Leu69 in the wild-type TTC7A was disrupted by the Leu69Pro mutation. Moreover, this homozygous missense mutation led to a reduced TTC7A expression in lymphocytes and intestinal tissues, accompanied by impeded lymphocyte development. Further studies demonstrated that the PI4K-FAM126A-EFR3A pathway was impaired in colon tissues. Our data strongly support the linkage of severe MIA-CID with the missense mutation in TTC7A gene. More knowledge of the TTC7A protein functions will have important therapeutic implications for patients with MIA-CID.


Subject(s)
Intestinal Atresia/genetics , Mutation, Missense/genetics , Proteins/genetics , Severe Combined Immunodeficiency/genetics , Child , Humans , Intestinal Atresia/immunology , Male , Mutation, Missense/immunology , Proteins/immunology , Severe Combined Immunodeficiency/immunology
16.
Front Immunol ; 12: 779502, 2021.
Article in English | MEDLINE | ID: mdl-35095854

ABSTRACT

Ataxia-telangiectasia (AT) is a rare autosomal recessive neurodegenerative multisystem disorder. A minority of AT patients can present late-onset atypical presentations due to unknown mechanisms. The demographic, clinical, immunological and genetic data were collected by direct interview and examining the Iranian AT patients with late-onset manifestations. We also conducted a systematic literature review for reported atypical AT patients. We identified three Iranian AT patients (3/249, 1.2% of total registry) with later age at ataxia onset and slower neurologic progression despite elevated alpha-fetoprotein levels, history of respiratory infections, and immunological features of the syndrome. Of note, all patients developed autoimmunity in which a decrease of naïve T cells and regulatory T cells were observed. The literature searches also summarized data from 73 variant AT patients with atypical presentation indicating biallelic mild mutations mainly lead to an atypical phenotype with an increased risk of cancer. Variant AT patients present with milder phenotype or atypical form of classical symptoms causing under- or mis- diagnosis. Although missense mutations are more frequent, an atypical presentation can be associated with deleterious mutations due to unknown modifying factors.


Subject(s)
Ataxia Telangiectasia/genetics , Ataxia/genetics , Mutation, Missense/genetics , Adolescent , Adult , Ataxia/immunology , Ataxia Telangiectasia/immunology , Child , Child, Preschool , Female , Humans , Iran , Male , Mutation, Missense/immunology , Phenotype , T-Lymphocytes, Regulatory/immunology , Young Adult , alpha-Fetoproteins/genetics , alpha-Fetoproteins/immunology
17.
Retrovirology ; 7: 38, 2010 Apr 26.
Article in English | MEDLINE | ID: mdl-20420700

ABSTRACT

BACKGROUND: In the acute phase of infection with feline immunodeficiency virus (FIV), the virus targets activated CD4+ T cells by utilising CD134 (OX40) as a primary attachment receptor and CXCR4 as a co-receptor. The nature of the virus-receptor interaction varies between isolates; strains such as GL8 and CPGammer recognise a "complex" determinant on CD134 formed by cysteine-rich domains (CRDs) 1 and 2 of the molecule while strains such as PPR and B2542 require a more "simple" determinant comprising CRD1 only for infection. These differences in receptor recognition manifest as variations in sensitivity to receptor antagonists. In this study, we ask whether the nature of the virus-receptor interaction evolves in vivo. RESULTS: Following infection with a homogeneous viral population derived from a pathogenic molecular clone, a quasispecies emerged comprising variants with distinct sensitivities to neutralising antibody and displaying evidence of conversion from a "complex" to a "simple" interaction with CD134. Escape from neutralising antibody was mediated primarily by length and sequence polymorphisms in the V5 region of Env, and these alterations in V5 modulated the virus-receptor interaction as indicated by altered sensitivities to antagonism by both anti-CD134 antibody and soluble CD134. CONCLUSIONS: The FIV-receptor interaction evolves under the selective pressure of the host humoral immune response, and the V5 loop contributes to the virus-receptor interaction. Our data are consistent with a model whereby viruses with distinct biological properties are present in early versus late infection and with a shift from a "complex" to a "simple" interaction with CD134 with time post-infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Feline Acquired Immunodeficiency Syndrome/virology , Immunodeficiency Virus, Feline/immunology , Mutation, Missense/immunology , Viral Envelope Proteins/immunology , Virus Attachment , Animals , Cats , Feline Acquired Immunodeficiency Syndrome/immunology , Immunodeficiency Virus, Feline/genetics , Immunodeficiency Virus, Feline/isolation & purification , Immunodeficiency Virus, Feline/physiology , Models, Molecular , Protein Conformation , Receptors, Virus/metabolism , Selection, Genetic , Viral Envelope Proteins/genetics
18.
J Virol ; 83(8): 3993-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19091871

ABSTRACT

Human immunodeficiency virus effectively evades CD8(+) T-cell responses through the development of CD8 escape mutations. Recent reports documenting reversion of transmitted mutations and the impact of specific escape mutations upon viral replication suggest that complex forces limit the accumulation of CD8 escape mutations at the population level. However, the presence of compensatory mutations capable of alleviating the impact of CD8 escape mutations on replication capacity may enable their persistence in an HLA-mismatched host. Herein, we illustrate the long-term stability of stereotypic escape mutations in the immunodominant HLA-B27-restricted epitope KK10 in p24/Gag following transmission when accompanied by a specific compensatory mutation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HIV/growth & development , HIV/immunology , Mutation, Missense/immunology , Amino Acid Sequence , Animals , HIV/genetics , HIV Core Protein p24/genetics , Humans , Molecular Sequence Data , Virus Replication
19.
J Virol ; 83(4): 1845-55, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036810

ABSTRACT

The control of human immunodeficiency virus type 1 (HIV-1) associated with particular HLA class I alleles suggests that some CD8(+) T-cell responses may be more effective than others at containing HIV-1. Unfortunately, substantial diversities in the breadth, magnitude, and function of these responses have impaired our ability to identify responses most critical to this control. It has been proposed that CD8 responses targeting conserved regions of the virus may be particularly effective, since the development of cytotoxic T-lymphocyte (CTL) escape mutations in these regions may significantly impair viral replication. To address this hypothesis at the population level, we derived near-full-length viral genomes from 98 chronically infected individuals and identified a total of 76 HLA class I-associated mutations across the genome, reflective of CD8 responses capable of selecting for sequence evolution. The majority of HLA-associated mutations were found in p24 Gag, Pol, and Nef. Reversion of HLA-associated mutations in the absence of the selecting HLA allele was also commonly observed, suggesting an impact of most CTL escape mutations on viral replication. Although no correlations were observed between the number or location of HLA-associated mutations and protective HLA alleles, limiting the analysis to mutations selected by acute-phase immunodominant responses revealed a strong positive correlation between mutations at conserved residues and protective HLA alleles. These data suggest that control of HIV-1 may be associated with acute-phase CD8 responses capable of selecting for viral escape mutations in highly conserved regions of the virus, supporting the inclusion of these regions in the design of an effective vaccine.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HIV-1/genetics , HIV-1/immunology , Mutation, Missense/immunology , HIV Core Protein p24/genetics , HIV Core Protein p24/immunology , Humans , Molecular Sequence Data , Sequence Analysis, DNA , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology , pol Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/immunology
20.
J Virol ; 83(7): 3407-12, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19153230

ABSTRACT

Elite controllers (EC) of human immunodeficiency virus type 1 (HIV-1) maintain viremia below the limit of detection without antiretroviral treatment. Virus-specific cytotoxic CD8(+) T lymphocytes are believed to play a crucial role in viral containment, but the degree of immune imprinting and compensatory mutations in EC is unclear. We obtained plasma gag, pol, and nef sequences from HLA-diverse subjects and found that 30 to 40% of the predefined HLA-associated polymorphic sites show evidence of immune selection pressure in EC, compared to approximately 50% of the sites in chronic progressors. These data indicate ongoing viral replication and escape from cytotoxic T lymphocytes are present even in strictly controlled HIV-1 infection.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV Long-Term Survivors , HIV-1/genetics , HIV-1/immunology , Mutation, Missense/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Plasma/virology , Polymorphism, Genetic , Selection, Genetic , Viral Load , Viremia/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL