Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Crohns Colitis ; 15(11): 1908-1919, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-33891011

ABSTRACT

BACKGROUND AND AIMS: Very early onset inflammatory bowel disease [VEOIBD] is characterized by intestinal inflammation affecting infants and children less than 6 years of age. To date, over 60 monogenic aetiologies of VEOIBD have been identified, many characterized by highly penetrant recessive or dominant variants in underlying immune and/or epithelial pathways. We sought to identify the genetic cause of VEOIBD in a subset of patients with a unique clinical presentation. METHODS: Whole exome sequencing was performed on five families with ten patients who presented with a similar constellation of symptoms including medically refractory infantile-onset IBD, bilateral sensorineural hearing loss and, in the majority, recurrent infections. Genetic aetiologies of VEOIBD were assessed and Sanger sequencing was performed to confirm novel genetic findings. Western analysis on peripheral blood mononuclear cells and functional studies with epithelial cell lines were employed. RESULTS: In each of the ten patients, we identified damaging heterozygous or biallelic variants in the Syntaxin-Binding Protein 3 gene [STXBP3], a protein known to regulate intracellular vesicular trafficking in the syntaxin-binding protein family of molecules, but not associated to date with either VEOIBD or sensorineural hearing loss. These mutations interfere with either intron splicing or protein stability and lead to reduced STXBP3 protein expression. Knock-down of STXBP3 in CaCo2 cells resulted in defects in cell polarity. CONCLUSION: Overall, we describe a novel genetic syndrome and identify a critical role for STXBP3 in VEOIBD, sensorineural hearing loss and immune dysregulation.


Subject(s)
Hearing Loss, Sensorineural/genetics , Immune System Diseases/genetics , Inflammatory Bowel Diseases/genetics , Qa-SNARE Proteins/analysis , Age of Onset , Female , Genetic Variation/genetics , Hearing Loss, Sensorineural/epidemiology , Humans , Immune System Diseases/epidemiology , Infant, Newborn , Inflammatory Bowel Diseases/epidemiology , Male , Qa-SNARE Proteins/genetics , Exome Sequencing
2.
Front Immunol ; 11: 545414, 2020.
Article in English | MEDLINE | ID: mdl-33162974

ABSTRACT

Neonatal hemophagocytic lymphohistiocytosis (HLH) is a medical emergency that can be associated with significant morbidity and mortality. Often these patients present with familial HLH (f-HLH), which is caused by gene mutations interfering with the cytolytic pathway of cytotoxic T-lymphocytes (CTLs) and natural killer cells. Here we describe a male newborn who met the HLH diagnostic criteria, presented with profound cholestasis, and carried a maternally inherited heterozygous mutation in syntaxin-binding protein-2 [STXBP2, c.568C>T (p.Arg190Cys)] in addition to a severe pathogenic variant in glucose 6-phosphate dehydrogenase [G6PD, hemizygous c.1153T>C (Cys385Arg)]. Although mutations in STXBP2 gene are associated with f-HLH type 5, the clinical and biological relevance of the p.Arg190Cys mutation identified in this patient was uncertain. To assess its role in disease pathogenesis, we performed functional assays and biochemical and microscopic studies. We found that p.Arg190Cys mutation did not alter the expression or subcellular localization of STXBP2 or STX11, neither impaired the STXBP2/STX11 interaction. In contrast, forced expression of the mutated protein into normal CTLs strongly inhibited degranulation and reduced the cytolytic activity outcompeting the effect of endogenous wild-type STXBP2. Interestingly, arginine 190 is located in a structurally conserved region of STXBP2 where other f-HLH-5 mutations have been identified. Collectively, data strongly suggest that STXBP2-R190C is a deleterious variant that may act in a dominant-negative manner by probably stabilizing non-productive interactions between STXBP2/STX11 complex and other still unknown factors such as the membrane surface or Munc13-4 protein and thus impairing the release of cytolytic granules. In addition to the contribution of STXBP2-R190C to f-HLH, the accompanied G6PD mutation may have compounded the clinical symptoms; however, the extent by which G6PD deficiency has contributed to HLH in our patient remains unclear.


Subject(s)
Exocytosis/genetics , Glucosephosphate Dehydrogenase Deficiency/diagnosis , Glucosephosphate Dehydrogenase Deficiency/genetics , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Munc18 Proteins/genetics , Mutation , Alleles , Amino Acid Sequence , Amino Acid Substitution , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Cytotoxicity, Immunologic , Disease Susceptibility , Gene Expression , Genetic Association Studies , Glucosephosphate Dehydrogenase Deficiency/complications , Humans , Infant, Newborn , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphohistiocytosis, Hemophagocytic/complications , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Munc18 Proteins/chemistry , Munc18 Proteins/metabolism , Protein Conformation , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Structure-Activity Relationship , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
3.
Traffic ; 18(7): 442-452, 2017 07.
Article in English | MEDLINE | ID: mdl-28471021

ABSTRACT

Adaptive and innate immunity utilize the perforin-killing pathway to eliminate virus-infected or cancer cells. Cytotoxic T-lymphocytes (CTLs) and natural killer cells mediate this process by releasing toxic proteins at the contact area with target cells known as immunological synapse (IS). Formation of a stable IS and exocytosis of toxic proteins requires persistent fusion of Rab11a recycling endosomes with the plasma membrane (PM) that may assure the delivery of key effector proteins. Despite the importance of the recycling endosomal compartment, the membrane fusion proteins that control this process at the IS remain elusive. Here, by performing knockdown experiments we found that syntaxin 4 (STX4) is necessary for cytotoxic activity and CD107a degranulation against target cells in a similar fashion to syntaxin 11, which is involved in lytic granule (LG) exocytosis and immunodeficiency when it is mutated. Using total internal reflection fluorescent microscopy we identified that STX4 mediates fusion of EGFP-Rab11a vesicles at the IS. Immunoprecipitation experiments in lysates of activated CTLs indicate that endogenous STX4 may drive this fusion step by interacting with cognate proteins: Munc18-3/SNAP23/VAMP7 and/or VAMP8. These results reveal the role of STX4 in mediating fusion of Rab11a endosomes upstream of lytic granules (LGs) exocytosis and further demonstrate the importance of this pathway in controlling CTL-mediated cytotoxicity.


Subject(s)
Cytoplasmic Granules/metabolism , Endosomes/metabolism , Exocytosis/immunology , Qa-SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Cell Degranulation , Cell Line , Cytoplasmic Granules/immunology , Cytotoxicity, Immunologic , Gene Knockdown Techniques , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Protein Transport , Qa-SNARE Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology
4.
Proc Natl Acad Sci U S A ; 114(11): E2176-E2185, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28265073

ABSTRACT

The atypical lipid-anchored Syntaxin 11 (STX11) and its binding partner, the Sec/Munc (SM) protein Munc18-2, facilitate cytolytic granule release by cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Patients carrying mutations in these genes develop familial hemophagocytic lymphohistiocytosis, a primary immunodeficiency characterized by impaired lytic granule exocytosis. However, whether a SNARE such as STX11, which lacks a transmembrane domain, can support membrane fusion in vivo is uncertain, as is the precise role of Munc18-2 during lytic granule exocytosis. Here, using a reconstituted "flipped" cell-cell fusion assay, we show that lipid-anchored STX11 and its cognate SNARE proteins mainly support exchange of lipids but not cytoplasmic content between cells, resembling hemifusion. Strikingly, complete fusion is stimulated by addition of wild-type Munc18-2 to the assay, but not of Munc18-2 mutants with abnormal STX11 binding. Our data reveal that Munc18-2 is not just a chaperone of STX11 but also directly contributes to complete membrane merging by promoting SNARE complex assembly. These results further support the concept that SM proteins in general are part of the core fusion machinery. This fusion mechanism likely contributes to other cell-type-specific exocytic processes such as platelet secretion.


Subject(s)
Cytotoxicity, Immunologic , Membrane Fusion , Membrane Lipids/metabolism , Munc18 Proteins/metabolism , Qa-SNARE Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , 3T3 Cells , Animals , CHO Cells , Carrier Proteins/metabolism , Cricetulus , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Multiprotein Complexes/metabolism , Munc18 Proteins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/genetics , SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
5.
Blood ; 125(10): 1566-77, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25564401

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (F-HLH) and Griscelli syndrome type 2 (GS) are life-threatening immunodeficiencies characterized by impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell lytic activity. In the majority of cases, these disorders are caused by biallelic inactivating germline mutations in genes such as RAB27A (GS) and PRF1, UNC13D, STX11, and STXBP2 (F-HLH). Although monoallelic (ie, heterozygous) mutations have been identified in certain patients, the clinical significance and molecular mechanisms by which these mutations influence CTL and NK cell function remain poorly understood. Here, we characterize 2 novel monoallelic hemophagocytic lymphohistiocytosis (HLH)-associated mutations affecting codon 65 of STXPB2, the gene encoding Munc18-2, a member of the SEC/MUNC18 family. Unlike previously described Munc18-2 mutants, Munc18-2(R65Q) and Munc18-2(R65W) retain the ability to interact with and stabilize syntaxin 11. However, presence of Munc18-2(R65Q/W) in patient-derived lymphocytes and forced expression in control CTLs and NK cells diminishes degranulation and cytotoxic activity. Mechanistic studies reveal that mutations affecting R65 hinder membrane fusion in vitro by arresting the late steps of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-complex assembly. Collectively, these results reveal a direct role for SEC/MUNC18 proteins in promoting SNARE-complex assembly in vivo and suggest that STXBP2 R65 mutations operate in a novel dominant-negative fashion to impair lytic granule fusion and contribute to HLH.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Munc18 Proteins/genetics , Mutant Proteins/genetics , Mutation, Missense , SNARE Proteins/immunology , Adult , Amino Acid Substitution , Child , Child, Preschool , Codon/genetics , Female , Genes, Dominant , HeLa Cells , Heterozygote , Humans , Infant , Killer Cells, Natural/immunology , Lymphohistiocytosis, Hemophagocytic/metabolism , Male , Membrane Fusion/genetics , Membrane Fusion/immunology , Middle Aged , Models, Biological , Models, Molecular , Munc18 Proteins/chemistry , Munc18 Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology
6.
Curr Biol ; 24(17): 1958-68, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25131674

ABSTRACT

BACKGROUND: Class II myosins generate contractile forces in cells by polymerizing into bipolar filaments and pulling on anchored actin filaments. Nonmuscle myosin II (NMII) plays central roles during cell adhesion, migration, cytokinesis, and tissue morphogenesis. NMII is present in virtually all mammalian cell types as tissue-specific combinations of NMIIA, NMIIB, and NMIIC isoforms. It remains poorly understood how the highly dynamic NMII-actin contractile system begins to assemble at new cellular locations during cell migration and how incorporation of different NMII isoforms into this system is coordinated. RESULTS: Using platinum replica electron microscopy in combination with immunogold labeling, we demonstrate that individual activated (phosphorylated on the regulatory light chain and unfolded) NMIIA and NMIIB molecules represent a functional form of NMII in motile cells and that NMIIA and NMIIB copolymerize into nascent bipolar filaments during contractile system assembly. Using subdiffraction stimulated emission depletion microscopy together with a pharmacological block-and-release approach, we report that NMIIA and NMIIB simultaneously incorporate into the cytoskeleton during initiation of contractile system assembly, whereas the characteristic rearward shift of NMIIB relative to NMIIA is established later in the course of NMII turnover. CONCLUSIONS: We show existence of activated NMII monomers in cells, copolymerization of endogenous NMIIA and NMIIB molecules, and contribution of both isoforms, rather than only NMIIA, to early stages of the contractile system assembly. These data change the current paradigms about dynamics and functions of NMII and provide new conceptual insights into the organization and dynamics of the ubiquitous cellular machinery for contraction that acts in multiple cellular contexts.


Subject(s)
Actin Cytoskeleton/metabolism , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Animals , Cell Adhesion , Cells, Cultured , Embryo, Mammalian , Phosphorylation , Protein Isoforms/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...