Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Front Immunol ; 12: 677994, 2021.
Article in English | MEDLINE | ID: mdl-34557186

ABSTRACT

Neutrophils are key players in innate immunity and originate from the bone marrow of the adult mammalian organism. In mammals, mature neutrophils are released from the bone marrow into the peripheral blood where they circulate until their recruitment to sites of inflammation in a multistep adhesion cascade. Here, adhesion molecules of the ß2 integrin family (CD11/CD18) are critically required for the initial neutrophil adhesion to the inflamed endothelium and several post-adhesion steps allowing their extravasation into the inflamed tissue. Within the mammalian tissue, interstitial neutrophil migration can occur widely independent of ß2 integrins. This is in sharp contrast to neutrophil recruitment in zebrafish larvae (Danio rerio) where neutrophils originate from the caudal hematopoietic tissue and mainly migrate interstitially to sites of lesion upon the early onset of inflammation. However, neutrophils extravasate from the circulation to the inflamed tissue in zebrafish larvae at later-time points. Although zebrafish larvae are a widely accepted model system to analyze neutrophil trafficking in vivo, the functional impact of ß2 integrins for neutrophil trafficking during acute inflammation is completely unknown in this model. In this study, we generated zebrafish with a genetic deletion of CD18, the ß subunit of ß2 integrins, using CRISPR/Cas9 technology. Sequence alignments demonstrated a high similarity of the amino acid sequences between zebrafish and human CD18 especially in the functionally relevant I-like domain. In addition, the cytoplasmic domain of CD18 harbors two highly conserved NXXF motifs suggesting that zebrafish CD18 may share functional properties of human CD18. Accordingly, CD18 knock-out (KO) zebrafish larvae displayed the key symptoms of patients suffering from leukocyte adhesion deficiency (LAD) type I due to defects in ITGB2, the gene for CD18. Importantly, CD18 KO zebrafish larvae showed reduced neutrophil trafficking to sites of sterile inflammation despite the fact that an increased number of neutrophils was detectable in the circulation. By demonstrating the functional importance of CD18 for neutrophil trafficking in zebrafish larvae, our findings shed new light on neutrophil biology in vertebrates and introduce a new model organism for studying LAD type I.


Subject(s)
CD18 Antigens/metabolism , Cell Adhesion/genetics , Cell Movement/genetics , Neutrophil Infiltration/genetics , Neutrophils/immunology , Zebrafish/genetics , Zebrafish/immunology , Amino Acid Sequence , Animals , Animals, Genetically Modified , CD11 Antigens/chemistry , CD11 Antigens/genetics , CD11 Antigens/metabolism , CD18 Antigens/chemistry , CD18 Antigens/genetics , Cell Adhesion/immunology , Cell Movement/immunology , Disease Models, Animal , Gene Deletion , Gene Knockout Techniques , Inflammation/genetics , Inflammation/immunology , Integrins/metabolism , Larva/genetics , Larva/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Neutrophil Infiltration/immunology
2.
Front Immunol ; 11: 1065, 2020.
Article in English | MEDLINE | ID: mdl-32625202

ABSTRACT

The field of primary immunodeficiencies (PIDs) is rapidly evolving. Indeed, the number of described diseases is constantly increasing thanks to the rapid identification of novel genetic defects by next-generation sequencing. PIDs are now rather referred to as "inborn errors of immunity" due to the association between a wide range of immune dysregulation-related clinical features and the "prototypic" increased infection susceptibility. The phenotypic spectrum of PIDs is therefore very large and includes several orofacial features. However, the latter are often overshadowed by severe systemic manifestations and remain underdiagnosed. Patients with impaired innate immunity are predisposed to a variety of oral manifestations including oral infections (e.g., candidiasis, herpes gingivostomatitis), aphthous ulcers, and severe periodontal diseases. Although less frequently, they can also show orofacial developmental abnormalities. Oral lesions can even represent the main clinical manifestation of some PIDs or be inaugural, being therefore one of the first features indicating the existence of an underlying immune defect. The aim of this review is to describe the orofacial features associated with the different PIDs of innate immunity based on the new 2019 classification from the International Union of Immunological Societies (IUIS) expert committee. This review highlights the important role played by the dentist, in close collaboration with the multidisciplinary medical team, in the management and the diagnostic of these conditions.


Subject(s)
Immunity, Innate , Mouth Diseases/etiology , Primary Immunodeficiency Diseases/complications , Disease Susceptibility , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Humans , Immunity, Innate/genetics , Leukocyte-Adhesion Deficiency Syndrome/complications , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Mouth Diseases/genetics , Mouth Diseases/immunology , Mutation , Neutropenia/complications , Neutropenia/genetics , Neutropenia/immunology , Papillon-Lefevre Disease/complications , Papillon-Lefevre Disease/genetics , Papillon-Lefevre Disease/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology
3.
J Leukoc Biol ; 108(5): 1501-1514, 2020 11.
Article in English | MEDLINE | ID: mdl-32421906

ABSTRACT

ß2 Integrins mediate neutrophil-endothelial adhesion and recruitment of neutrophils to sites of inflammation. The diminished expression of ß2 integrins in patients with mutations in the ITGB2 (CD18) gene (leukocyte adhesion deficiency-Type 1; LAD1) results in few or no neutrophils in peripheral tissues. In the periodontium, neutrophil paucity is associated with up-regulation of IL-23 and IL-17, which drive inflammatory bone loss. Using a relevant mouse model, we investigated whether diminished efferocytosis (owing to neutrophil scarcity) is associated with LAD1 periodontitis pathogenesis and aimed to develop approaches to restore the missing efferocytosis signals. We first showed that CD18-/- mice phenocopied human LAD1 in terms of IL-23/IL-17-driven inflammatory bone loss. Ab-mediated blockade of c-Mer tyrosine kinase (Mer), a major efferocytic receptor, mimicked LAD1-associated up-regulation of gingival IL-23 and IL-17 mRNA expression in wild-type (WT) mice. Consistently, soluble Mer-Fc reversed the inhibitory effect of efferocytosis on IL-23 expression in LPS-activated Mϕs. Adoptive transfer of WT neutrophils to CD18-/- mice down-regulated IL-23 and IL-17 expression to normal levels, but not when CD18-/- mice were treated with blocking anti-Mer Ab. Synthetic agonist-induced activation of liver X receptors (LXR) and peroxisome proliferator-activated receptors (PPAR), which link efferocytosis to generation of homeostatic signals, inhibited the expression of IL-23 and IL-17 and favorably affected the bone levels of CD18-/- mice. Therefore, our data link diminished efferocytosis-associated signaling due to impaired neutrophil recruitment to dysregulation of the IL-23-IL-17 axis and, moreover, suggest LXR and PPAR as potential therapeutic targets for treating LAD1 periodontitis.


Subject(s)
Homeostasis/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Liver X Receptors/immunology , Periodontitis/immunology , Periodontium/immunology , Peroxisome Proliferator-Activated Receptors/immunology , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , Homeostasis/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Liver X Receptors/genetics , Mice , Mice, Knockout , Periodontitis/genetics , Periodontitis/pathology , Periodontium/pathology , Peroxisome Proliferator-Activated Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Up-Regulation/immunology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/immunology
4.
Int J Mol Sci ; 21(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32092981

ABSTRACT

ß2 integrins are heterodimeric surface receptors composed of a variable α (CD11a-CD11d) and a constant ß (CD18) subunit and are specifically expressed by leukocytes. The α subunit defines the individual functional properties of the corresponding ß2 integrin, but all ß2 integrins show functional overlap. They mediate adhesion to other cells and to components of the extracellular matrix (ECM), orchestrate uptake of extracellular material like complement-opsonized pathogens, control cytoskeletal organization, and modulate cell signaling. This review aims to delineate the tremendous role of ß2 integrins for immune functions as exemplified by the phenotype of LAD-I (leukocyte adhesion deficiency 1) patients that suffer from strong recurrent infections. These immune defects have been largely attributed to impaired migratory and phagocytic properties of polymorphonuclear granulocytes. The molecular base for this inherited disease is a functional impairment of ß2 integrins due to mutations within the CD18 gene. LAD-I patients are also predisposed for autoimmune diseases. In agreement, polymorphisms within the CD11b gene have been associated with autoimmunity. Consequently, ß2 integrins have received growing interest as targets in the treatment of autoimmune diseases. Moreover, ß2 integrin activity on leukocytes has been implicated in tumor development.


Subject(s)
Autoimmune Diseases , CD18 Antigens/metabolism , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocytes/immunology , Neoplasms/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/metabolism , CD18 Antigens/genetics , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Humans , Infections/immunology , Infections/metabolism , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocytes/metabolism , Lymphocyte Function-Associated Antigen-1/immunology , Macrophage-1 Antigen/immunology , Neoplasms/genetics , Neoplasms/metabolism , Non-Fibrillar Collagens/immunology , Phagocytosis/genetics , Phagocytosis/immunology , Collagen Type XVII
5.
Front Immunol ; 10: 254, 2019.
Article in English | MEDLINE | ID: mdl-30837997

ABSTRACT

Beta2-integrins are complex leukocyte-specific adhesion molecules that are essential for leukocyte (e.g., neutrophil, lymphocyte) trafficking, as well as for other immunological processes such as neutrophil phagocytosis and ROS production, and T cell activation. Intriguingly, however, they have also been found to negatively regulate cytokine responses, maturation, and migratory responses in myeloid cells such as macrophages and dendritic cells, revealing new, and unexpected roles of these molecules in immunity. Because of their essential role in leukocyte function, a lack of expression or function of beta2-integrins causes rare immunodeficiency syndromes, Leukocyte adhesion deficiency type I, and type III (LAD-I and LAD-III). LAD-I is caused by reduced or lost expression of beta2-integrins, whilst in LAD-III, beta2-integrins are expressed but dysfunctional because a major integrin cytoplasmic regulator, kindlin-3, is mutated. Interestingly, some LAD-related phenotypes such as periodontitis have recently been shown to be due to an uncontrolled inflammatory response rather than to an uncontrolled infection, as was previously thought. This review will focus on the recent advances concerning the regulation and functions of beta2-integrins in leukocyte trafficking, immune suppression, and immune deficiency disease.


Subject(s)
CD18 Antigens/immunology , Cell Movement/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocytes/immunology , Animals , Humans , Immunologic Deficiency Syndromes/immunology , Immunosuppression Therapy/methods , Lymphocyte Activation/immunology
7.
J Coll Physicians Surg Pak ; 28(6): S87-S88, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29866230

ABSTRACT

Leukocyte adhesion deficiency type 1 (LAD-1) is a rare autosomal recessive disorder caused by mutations in the gene that codes for CD18, the beta chain of beta-2 integrins, located on the long arm of chromosome 21. This defect results in failure of leukocyte migration to the site of infection due to the absence of surface integrins. Leukocyte adhesion deficiency should be suspected in any patient with recurrent infections, impaired wound healing, history of delayed umbilical cord separation, periodontitis, leukocytosis, recurrent soft tissue and oral infections. Diagnosis is based on the analysis of neutrophils for the surface expression of CD18, CD11a, CD11b and CD11c by flow cytometry. Here, we present a 55-day male infant with umbilical cord separation on the 10th day of life and no history of infection, who was identified with LAD-1 with low expression of CD11b. The purpose of performing LAD flow cytometric analysis in this patient was to screen him for LAD-1 as his elder brother had LAD-1 and one elder sister died undiagnosed with recurrent skin and chest infections at 8 months of age.


Subject(s)
CD11b Antigen/metabolism , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , CD11c Antigen/genetics , CD11c Antigen/metabolism , CD18 Antigens/genetics , CD18 Antigens/metabolism , Humans , Infant , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Male
8.
Immunol Lett ; 187: 7-13, 2017 07.
Article in English | MEDLINE | ID: mdl-28445705

ABSTRACT

BACKGROUND AND AIM: Leukocyte adhesion deficiency type 1 (LAD-I) is a rare, autosomal recessive disorder of neutrophil migration, characterized by severe, recurrent bacterial infections, inadequate pus formation and impaired wound healing. The ITGB2 gene encodes the ß2 integrin subunit (CD18) of the leukocyte adhesion cell molecules, and mutations in this gene cause LAD-I. The aim of the current study was to investigate the mutations in patients diagnosed with LAD-I and functional studies of the impact of two previously reported and a novel mutation on the expression of the CD18/CD11a heterodimer. MATERIALS AND METHODS: Blood samples were taken from three patients who had signed the consent form. Genomic DNA was extracted and ITGB2 exons and flanking intronic regions were amplified by polymerase chain reaction. Mutation screening was performed after Sanger sequencing of PCR products. For functional studies, COS-7 cells were co-transfected with an expression vector containing cDNA encoding mutant CD18 proteins and normal CD11a. Flow cytometry analysis of CD18/CD11a expression was assessed by dimer-specific IB4 monoclonal antibody. RESULTS: Two previously reported mutations and one novel mutation,p. Cys562Tyr, were found. All mutations reduced CD18/CD11 heterodimer expression. CONCLUSION: Our strategy recognized the p.Cys562Tyr mutation as a pathogenic alteration that does not support CD18 heterodimer formation. Therefore, it can be put into a panel of carrier and prenatal diagnosis programs.


Subject(s)
CD11a Antigen , CD18 Antigens , Leukocyte-Adhesion Deficiency Syndrome , Mutation, Missense , Amino Acid Substitution , Animals , CD11a Antigen/genetics , CD11a Antigen/immunology , CD18 Antigens/genetics , CD18 Antigens/immunology , COS Cells , Chlorocebus aethiops , Humans , Infant, Newborn , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/pathology , Male
11.
Indian J Pediatr ; 83(8): 799-804, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26924654

ABSTRACT

OBJECTIVE: To describe clinical and flow cytometric immunophenotyping details of 26 patients of Leukocyte adhesion deficiency-I (LAD-I) along with molecular characterization of 7 patients. METHODS: Diagnosis of LAD-I was suspected on the basis of clinical features, white blood cell count and absolute neutrophil counts and flow cytometric assessment of expression of CD18 and CD11(a, b, c) on leukocytes. Mutation analysis was performed using DNA PCR and conformation sensitive gel electrophoresis (CSGE) technique followed by sequencing. RESULTS: All the patients were symptomatic by the age of 6 mo, with history of recurrent bacterial infections involving skin, mucosa or umbilical cord (omphalitis) being the most frequent presenting symptoms. White blood cells (WBC) and absolute neutrophil counts (ANC) were markedly elevated, without any specific morphological findings. On flow cytometry, CD11a and CD11c showed moderate correlation with CD18 expression. Mutation analysis was performed in 7 patients and six different mutations (4 missense, 2 nonsense and 1 splice site) were identified, all of which were homozygous in nature. CONCLUSIONS: A presentation of repeated bacterial infections during infancy, especially omphalitis, with markedly elevated absolute neutrophil counts should trigger investigations for LAD-I including flow cytometric analysis of CD11/CD18 expression.


Subject(s)
CD18 Antigens , DNA Mutational Analysis , Leukocyte-Adhesion Deficiency Syndrome , Bacterial Infections , Child, Preschool , Homozygote , Humans , India , Infant , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocytes
12.
Hum Immunol ; 77(2): 191-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26639818

ABSTRACT

Leukocyte adhesion deficiency type 1 (LAD1) is an autosomal recessive disorder clinically characterized by severe, recurrent bacterial infections, impaired pus formation and wound healing. It is caused by mutation in the ITGB2 gene, encoding the ß2 integrin subunit of the leukocyte adhesion cell molecule. This study aimed to identify disease causing mutations in 19 consanguineous families diagnosed with LAD1. Blood samples were collected after informed and written consent was obtained. Genomic DNA was extracted from peripheral blood of patients and their parents. PCR amplification of the ITGB2 gene was done using specific primers followed by sequencing for mutation detection. A total number of 14 alterations scattered throughout the ITGB2 gene were ascertained in which 10 mutations were previously reported, including c.329-6C>A, c.382G>T, c.715G>A, c.843delC, c.897+1G>A, c.1062A>T, c.1143delC, c.1877+2T>C, c.1907delA and c.2147G>C. Four novel likely pathogenic mutations consisting of c.576dupC (Asn193GlnfsX72), c.706G>A (Gly236Arg), c.897+1G>T and c.1030G>T (Glu344(∗)), were identified. The majority of these mutations were located in exon six, suggesting this exon as a hotspot region probably. This study emphasis on allelic heterogeneity of the ITGB2 gene in Iranian patients diagnosed with LAD1. Our results suggest that every population should develop a mutation database for rare genetic disorders to take advantage in genetic counseling clinic as well as genetic testing for rapid diagnostic purposes.


Subject(s)
CD18 Antigens/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Mutation/genetics , Consanguinity , DNA Mutational Analysis , Exons/genetics , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Iran , Leukocyte-Adhesion Deficiency Syndrome/immunology , Pedigree , Polymorphism, Genetic
13.
Microb Pathog ; 94: 21-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26375893

ABSTRACT

Leukocyte adhesion deficiency Type I (LAD-I)-associated periodontitis is an aggressive form of inflammatory bone loss that has been historically attributed to lack of neutrophil surveillance of the periodontal infection. However, this form of periodontitis has proven unresponsive to antibiotics and/or mechanical removal of the tooth-associated biofilm. Recent studies in LAD-I patients and relevant animal models have shown that the fundamental cause of LAD-I periodontitis involves dysregulation of a granulopoietic cytokine cascade. This cascade includes interleukin IL-23 (IL-23) and IL-17 that drive inflammatory bone loss in LAD-I patients and animal models and, moreover, foster a nutritionally favorable environment for bacterial growth and development of a compositionally unique microbiome. Although the lack of neutrophil surveillance in the periodontal pockets might be expected to lead to uncontrolled bacterial invasion of the underlying connective tissue, microbiological analyses of gingival biopsies from LAD-I patients did not reveal tissue-invasive infection. However, bacterial lipopolysaccharide was shown to translocate into the lesions of LAD-I periodontitis. It is concluded that the bacteria serve as initial triggers for local immunopathology through translocation of bacterial products into the underlying tissues where they unleash the dysregulated IL-23-IL-17 axis. Subsequently, the IL-23/IL-17 inflammatory response sustains and shapes a unique local microbiome which, in turn, can further exacerbate inflammation and bone loss in the susceptible host.


Subject(s)
Bacterial Physiological Phenomena , Leukocyte-Adhesion Deficiency Syndrome/microbiology , Periodontitis/microbiology , Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Animals , Gingiva/immunology , Gingiva/microbiology , Gingiva/pathology , Humans , Interleukin-17/immunology , Interleukin-23/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/pathology , Lipopolysaccharides , Neutrophils/immunology , Periodontitis/diagnostic imaging , Periodontitis/immunology , Periodontitis/pathology , Radiography, Panoramic
14.
Article in English | MEDLINE | ID: mdl-26076788

ABSTRACT

Neutrophil functional disorders thought to be uncommon, yet important as a cause of morbidity and mortality in infants and children. During the first years of life, when the immune system is still not completely mature, when the viral infections are frequent and antibiotic overuse can damage and alter the immune response, the inadequate nutrition followed with iron deficient anemia and malnutrition can lead the child`s organism in state of immunodeficiency. Sometimes is difficult to distinguish at the beginning weather the cause of patient suffering from frequent infections is existing of primary immunodeficiency disorder or the cause of the immunodeficiency state is just from exogenous factors. Fortunately, primary immune deficiencies are rare diseases and only 6-7% of all of them, due to the neutrophilic functional disorders. Unfortunately, many exogenous and environmental factors have influence to the immune system, and the percentage of secondary caused neutrophilic functional disorders is much higher and should be considered when children are investigated for immunodeficiency. So, when to suspect neutrophil functional disorder? The hallmarks for diseases related to the neutrophilic functional disorders are discussed in this article.


Subject(s)
Immunity, Innate/immunology , Immunologic Deficiency Syndromes/immunology , Neutrophils/immunology , Phagocytosis/immunology , Protein-Energy Malnutrition/immunology , Amino Acid Metabolism, Inborn Errors/immunology , Chediak-Higashi Syndrome/immunology , Child Development , Child, Preschool , Glucosephosphate Dehydrogenase Deficiency/immunology , Glutathione Synthase/deficiency , Glutathione Synthase/immunology , Granulomatous Disease, Chronic/immunology , Humans , Infant , Infant, Newborn , Job Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Metabolism, Inborn Errors/immunology , Phagocytes/immunology
15.
J Immunol ; 195(1): 105-15, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25987740

ABSTRACT

Kindlin-3 is an important integrin regulator that is mutated in the rare genetic disorder, leukocyte adhesion deficiency type III, a disorder characterized by defective neutrophil trafficking and platelet function, leading to recurrent bacterial infections and bleeding. Kindlin-3 is also known to regulate T cell adhesion in vitro and trafficking in vivo, but whether the integrin/kindlin interaction regulates T or B cell activation in vivo is unclear. In this study, we used TTT/AAA ß2-integrin knock-in (KI) mice and TCR-transgenic (OT-II) KI mice, in which the integrin/kindlin connection is disrupted, to investigate the role of the integrin/kindlin interaction in T cell activation. We show that basal T cell activation status in these animals in vivo is normal, but they display reduced T cell activation by wild-type Ag-loaded dendritic cells in vitro. In addition, T cell activation in vivo is reduced. We also show that basal Ab levels are normal in TTT/AAA ß2-integrin KI mice, but B cell numbers in lymph nodes and IgG and IgM production after immunization are reduced. In conclusion, we show that the integrin/kindlin interaction is required for trafficking of immune cells, as well as for T cell activation and B cell Ab responses in vivo. These results imply that the immunodeficiency found in leukocyte adhesion deficiency type III patients, in addition to being caused by defects in neutrophil function, may be due, in part, to defects in lymphocyte trafficking and activation.


Subject(s)
B-Lymphocytes/immunology , CD18 Antigens/immunology , Cytoskeletal Proteins/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , B-Lymphocytes/pathology , CD18 Antigens/genetics , Cell Movement , Cytoskeletal Proteins/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/pathology
16.
PLoS Pathog ; 11(3): e1004698, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25741691

ABSTRACT

Leukocyte Adhesion Deficiency I (LAD-I) is a primary immunodeficiency caused by single gene mutations in the CD18 subunit of ß2 integrins which result in defective transmigration of neutrophils into the tissues. Affected patients suffer from recurrent life threatening infections and severe oral disease (periodontitis). Microbial communities in the local environment (subgingival plaque) are thought to be the triggers for inflammatory periodontitis, yet little is known regarding the microbial communities associated with LAD-I periodontitis. Here we present the first comprehensive characterization of the subgingival communities in LAD-I, using a 16S rRNA gene-based microarray, and investigate the relationship of this tooth adherent microbiome to the local immunopathology of periodontitis. We show that the LAD subgingival microbiome is distinct from that of health and Localized Aggressive Periodontitits. Select periodontitis-associated species in the LAD microbiome included Parvimonas micra, Porphyromonas endodontalis, Eubacterium brachy and Treponema species. Pseudomonas aeruginosa, a bacterium not typically found in subgingival plaque is detected in LAD-I. We suggest that microbial products from LAD-associated communities may have a role in stimulating the local inflammatory response. We demonstrate that bacterial LPS translocates into the lesions of LAD-periodontitis potentially triggering immunopathology. We also show in in vitro assays with human macrophages and in vivo in animal models that microbial products from LAD-associated subgingival plaque trigger IL-23-related immune responses, which have been shown to dominate in patient lesions. In conclusion, our current study characterizes the subgingival microbial communities in LAD-periodontitis and supports their role as triggers of disease pathogenesis.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocytes/immunology , Periodontitis/microbiology , Porphyromonas gingivalis , Animals , DNA, Bacterial/genetics , DNA, Bacterial/immunology , Dental Plaque/genetics , Humans , Interleukin-23/metabolism , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/therapy , Mice , Microbiota/immunology , RNA, Ribosomal, 16S/genetics
18.
J Clin Immunol ; 34(7): 788-91, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25135596

ABSTRACT

Leukocyte adhesion deficiency type 1 (LAD-1) is an autosomal recessive disorder, caused by the absence or reduced expression of the beta-2 integrins on granulocytes, and characterized by the inability of these cells to emigrate from the bloodstream towards the sites of tissue inflammation. A twelve-year-old girl with a diagnosis of LAD-1 syndrome and recurrent skin and mucosal infections since birth, presented with a two week history of fever, abdominal pain, vomiting, weight loss and polyarthralgia. She underwent an exploratory laparotomy with the finding of inflamed terminal ileum and colon and a normal appendix. Colonoscopy and videocapsule endoscopy showed multiple ileal and colonic mucosal ulcerations, which were compatible with inflammatory bowel disease, confirmed on histological examination. Given the lack of response to conventional therapy (prednisone and mesalamine), a monoclonal anti-TNF-α antibody was started at a dosage of 5 mg/kg at weeks 0,2,4,6 and then every 8 weeks. We observed a significant improvement of all clinical and laboratory parameters after the first weeks of therapy. Five months later, we anticipated the drug's administration every 5 weeks because of a precocious recurrence of symptoms. After 30 months of treatment no relapse nor any relevant side effects have been observed, and corticosteroids were withdrawn. Interestingly, our patient presented a small subset of CD18+ T cells, similarly to previously reported LAD-1 patients with mild phenotype, inflammatory bowel disease and CD18+ somatic revertant T cells. To the best of our knowledge, this is the first LAD-1 pediatric patient with inflammatory autoimmune complications who experienced a positive response to anti-TNF-α treatment.


Subject(s)
Granulocytes/physiology , Immunotherapy/methods , Inflammatory Bowel Diseases/therapy , Integrin beta Chains/metabolism , Leukocyte-Adhesion Deficiency Syndrome/therapy , Antibodies, Monoclonal/administration & dosage , Cell Movement/genetics , Child , Disease-Free Survival , Female , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Integrin beta Chains/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Tumor Necrosis Factor-alpha/immunology
19.
Aust Vet J ; 92(8): 299-302, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24954630

ABSTRACT

BACKGROUND: Leucocyte adhesion deficiencies are inherited disorders characterised by immunodeficiency leading to recurrent infections and a marked leucocytosis. We describe the clinical characteristics, diagnosis and management of an Australian mixed- breed dog with leucocyte adhesion deficiency III. CASE REPORT: A 16-month-old male, neutered, German Shepherd × Rottweiler dog was investigated for pyrexia, persistent leucocytosis, marked periodontal disease, lameness, increased mucosal haemorrhages and poor wound healing. Numerous diagnostics were performed including a leucocyte adhesion deficiency III PCR test, which was positive. The patient was managed with topical pressure at bleeding sites, antibiotics, analgesics and dental prophylaxis when required. DISCUSSION: Leucocyte adhesion deficiency III is a rare disorder that manifests because of impaired activation of beta integrins. This results in an absence of neutrophil chemotaxis and adhesion, and platelet dysfunction. Mutations within the KINDLIN3 gene resulting in the absence of the kindlin-3 protein have been identified as the cause of this disease. Leucocyte adhesion deficiency III has previously been reported in humans and a German Shepherd dog. This report describes the first reported case of leucocyte adhesion deficiency III in Australia and the first reported case in a mixed-breed dog worldwide.


Subject(s)
Dog Diseases/immunology , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Animals , Dog Diseases/blood , Dog Diseases/diagnosis , Dogs/immunology , Leukocyte-Adhesion Deficiency Syndrome/blood , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , Leukocyte-Adhesion Deficiency Syndrome/immunology , Male , Polymerase Chain Reaction/veterinary
20.
Expert Rev Clin Immunol ; 10(8): 973-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24931458

ABSTRACT

In leukocyte adhesion deficiency type I, neutrophils fail to adhere to blood vessel walls and thus cannot transmigrate to peripheral tissues. Leukocyte adhesion deficiency type I patients invariably experience an aggressive form of generalized periodontitis, which has been historically attributed to defective neutrophil surveillance of the periodontal infection. This time-honored notion has now been challenged by a recent study, which showed that the underlying etiology involves a dysregulated host response that leads to overexpression of the proinflammatory and bone-resorptive cytokine IL-17.


Subject(s)
Bacterial Infections/immunology , Interleukin-17/metabolism , Leukocyte-Adhesion Deficiency Syndrome/immunology , Neutrophils/immunology , Periodontitis/immunology , Animals , Bacterial Infections/complications , Bone Resorption , Chemotaxis , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Interleukin-17/genetics , Leukocyte-Adhesion Deficiency Syndrome/complications , Mice , Molecular Targeted Therapy , Neutrophils/microbiology , Periodontitis/etiology , Transendothelial and Transepithelial Migration , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...