Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
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
2.
Gene ; 715: 144027, 2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31374327

ABSTRACT

OBJECTIVES: To explore the clinical and molecular characteristics of a Chinese Zhuang minority patient with leukocyte adhesion deficiency type-1 (LAD-1) and glucose-6-phosphate dehydrogenase deficiency (G6PDD). METHODS: Routine clinical and physical examinations were performed, and patient data was collected and analyzed. Protein expression levels of Itgb2 and glucose-6-phosphate dehydrogenase (G6pd) proteins were assessed by flow cytometry and the glucose-6-phosphate (G6P) substrate method, respectively. Whole exome sequencing was performed to investigate genetic variations of the patient and his parents. RESULTS: The patient had fester disease and delayed separation of the umbilical cord at birth. Staphylococcus was detected in the fluid secretion of the auditory meatus of the patient. He exhibited a recurrent cheek scab, swollen hand, and swollen gum. Hematological examination indicated dramatic elevation of leukocytes including lymphocytes, monocytes, neutrophils and eosinophils. A novel homozygous mutation was detected in the ITGB2 gene of the patient, which was determined to be a two nucleotide deletion at the site of c.1537-1538 (c.1537-1538delGT), causing a frameshift of 24 amino acids from p.513 and inducing a stop codon (p.V513Lfs*24). A base substitution mutation was identified at c.1466 (c.1466G>T) of G6PD on chromosome X of the patient, which resulted in an amino acid change from arginine to leucine at p.489 (p.R489L). The patient also showed deficient lymphocyte expression of CD18 (2.99%) and significant downregulation of the G6pd protein. CONCLUSIONS: The patient was diagnosed with G6PDD and moderate LAD-1. The combination of LAD-1 and G6PDD in this case may have been due to the high incidence of genetic disease in this minority ethnic population. Analyzing existing LAD-1 and G6PDD cases from different populations can facilitate disease diagnosis and treatment. Particularly, reporting pathogenic mutations of LAD-1 and G6PDD will be crucial for genetic testing and prenatal diagnosis in an effort to decrease the incidence of these diseases.


Subject(s)
CD18 Antigens/genetics , Glucosephosphate Dehydrogenase Deficiency/genetics , Homozygote , Leukocyte-Adhesion Deficiency Syndrome/genetics , Mutation, Missense , Amino Acid Substitution , Asian People , CD18 Antigens/metabolism , Glucosephosphate Dehydrogenase Deficiency/pathology , Humans , Infant , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/pathology , Leukocytes/metabolism , Leukocytes/pathology , Male
3.
Integr Biol (Camb) ; 11(6): 293-300, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31329860

ABSTRACT

Leukocyte adhesion is important for the proper functioning of the immune system. While leukocyte homing is mediated by adhesion receptors, the activation of these receptors is modulated by intracellular signaling molecules. In Leukocyte Adhesion Deficiency Type 3, the loss of the kindlin-3 prevents the activation of Leukocyte Function-associated Antigen-1 (LFA-1), which leads to a defect in adhesion, causing recurrent infections and bleeding disorders. Here, we use Integrated Signaling Adhesive Dynamics, a computer model of leukocyte rolling and adhesion combined with a simulated intracellular signaling cascade, to predict the response of T cells to depletion of kindlin-3. Our model predicts that cell adhesion is hypersensitive to the amount of kindlin-3 in the cell, while the rolling velocity is independent of kindlin-3 concentration. In addition, our simulation predicted that the time to stop, an important metric of adhesion, would increase with decreasing kindlin-3 expression. These predictions were confirmed experimentally in experiments using Jurkat cells with reduced expression of kindlin-3. These results suggest that Adhesive Dynamics is a versatile tool for quantifying adhesion in the immune response and predicting the effects of engineering cellular components.


Subject(s)
Cell Adhesion , Leukocyte Rolling , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , T-Lymphocytes/cytology , Adsorption , Algorithms , Chemokine CXCL12/metabolism , Computer Simulation , Gene Knockdown Techniques , Humans , Immune System , Jurkat Cells , Kinetics , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocytes/cytology , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Signal Transduction , Surface Properties
4.
Curr Opin Hematol ; 24(1): 38-45, 2017 01.
Article in English | MEDLINE | ID: mdl-27749372

ABSTRACT

PURPOSE OF REVIEW: Since the discovery of the lack of kindlin-3 expression as the reason for the immunopathology leukocyte adhesion deficiency III syndrome, the role of kindlin-3 in inflammatory processes was investigated in a numerous studies. This review gives an overview about recent findings regarding the role of kindlin-3 in neutrophil activation and recruitment. RECENT FINDINGS: Kindlin-3, together with talin-1, contributes essentially to the activation of ß2-integrins in neutrophils. During inside-out signaling, kindlin-3 binds to the ß-cytoplasmic integrin tail and is indispensable for the integrin conformational shift into the high-affinity ligand binding conformation, but not for the intermediate (extended) conformation. During outside-in signaling (as a consequence of integrin ligand binding) kindlin-3 interacts with distinct signaling molecules and is required for cell-autonomous functions like migration and spreading. SUMMARY: Leukocyte adhesion deficiency III syndrome, which is caused by absence of kindlin-3, is a rarely occurring disease. However, the investigation of the clinical symptoms as well as the underlying molecular mechanisms gave rise to a huge amount of new insights into the processes of integrin activation in neutrophils and the consequences of defects in these processes.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Neutrophil Infiltration , Neutrophils/physiology , Animals , Carrier Proteins , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Gene Expression Regulation , Humans , Inflammation/pathology , Integrins/genetics , Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/etiology , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocytes/physiology , Protein Binding , Signal Transduction
5.
Immunol Res ; 64(2): 476-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26434744

ABSTRACT

Leukocyte adhesion deficiency type 1 (LAD-1) is an autosomal recessive primary immunodeficiency, hallmarked by defective polymorphonuclear transmigration. It is caused by mutations in the gene encoding CD18, which interfere with the CD18/CD11 heterodimerization and expression on leukocyte cell surface. LAD-1 diagnosis rests primarily on the measurement of CD18 expression. However, CD18 measurement entails its pitfalls. Here we present a cohort of ten LAD patients and a review of the relevant literature illustrating the difficulties in sole reliance on CD18 measurement for initial diagnosis. These include normal range expression in some mutations, great variability between patients with the same mutation and subjective interpretation of results. We think there is a need for additional markers as part of the initial LAD diagnostic algorithm. We suggest CD11a expression, which was near absent in all patients in our cohort. The dual use of CD18 and CD11a can increase testing sensitivity and prevent delayed diagnosis of LAD-1.


Subject(s)
CD18 Antigens/metabolism , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Biomarkers , CD18 Antigens/genetics , Case-Control Studies , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Infant , Infant, Newborn , Leukocyte Count , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocytes/immunology , Leukocytes/metabolism , Male , Mutation , Phenotype , Reproducibility of Results , Transplantation, Homologous
6.
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
9.
Blood ; 120(19): 3915-24, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22983444

ABSTRACT

Recent evidence suggests that kindlin-3 is a major coactivator, required for most, if not all, integrin activities. Here we studied the function of kindlin-3 in regulating NK cell activation by studying a patient with kindlin-3 deficiency (leukocyte adhesion deficiency-III). We found that kindlin-3 is required for NK cell migration and adhesion under shear force. Surprisingly, we also found that kindlin-3 lowers the threshold for NK cell activation. Loss of kindlin-3 has a pronounced effect on NK cell-mediated cytotoxicity triggered by single activating receptors. In contrast, for activation through multiple receptors, kindlin-3 deficiency is overcome and target cells killed. The realization that NK cell activity is impaired, but not absent in leukocyte adhesion deficiency, may lead to the development of more efficient therapy for this rare disease.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Membrane Proteins/deficiency , Neoplasm Proteins/deficiency , Actins/chemistry , Actins/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Codon, Terminator , Cytotoxicity, Immunologic , Genotype , Humans , Leukocyte-Adhesion Deficiency Syndrome/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pedigree , Protein Multimerization , Protein Transport , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, Natural Killer Cell/immunology , Receptors, Natural Killer Cell/metabolism , Shear Strength
10.
Methods Mol Biol ; 757: 369-97, 2012.
Article in English | MEDLINE | ID: mdl-21909923

ABSTRACT

Integrins are adhesive proteins that have evolved to mediate cell-cell and cell-matrix communication that is indispensable for development and postnatal physiology. Despite their widespread expression, the genetic deletion of specific integrin family members in lower organisms as well as mammals leads to relatively distinct abnormalities. Many of the processes in which integrins participate have a requirement for strong adhesion coincident with times of mechanical stress. In Drosophila, the absence of specific integrins leads to detachment of muscle from the gut and body wall and separation of the two epithelial layers in the wing. In mice and humans, a deletion of either subunit of the laminin-binding integrin, α6ß4 leads to severe skin blistering and defects in other epithelial layers. In addition, integrins have also evolved to serve more subspecialized roles ranging from the establishment of a stem cell niche in Drosophila and mammals, to the regulation of pathogenic tumor vascularization, platelet adhesion, and leukocyte transmigration in mammalian systems. However, some cells seem to function normally in the absence of all integrins, as revealed by the very surprising finding that deletion of all the major integrin types on dendritic cells of mice has no effect on the ability of these cells to migrate within the interstitium of the skin and enter into lymphatics. In addition to serving as transmembrane mechanical links, integrins in vertebrates synergize with a number of receptors including growth factor receptors, to enhance responses. This leads to the activation of a large signaling network that affects cell proliferation and differentiation, as well as cell shape and migration. In vivo studies, in lower organisms, knockout mouse models as well as in inherited human diseases together have provided important insights into how this major, primordial family of adhesion receptors have remained true to their name "integrins" as their diverse functions have in common the ability to integrate extracellular stimuli into intracellular signals that affect cell behavior.


Subject(s)
Integrins/genetics , Integrins/metabolism , Animals , Humans , Integrins/deficiency , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Signal Transduction , Thrombasthenia/genetics , Thrombasthenia/metabolism
11.
Blood ; 119(2): 583-6, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22065596

ABSTRACT

Patients with Glanzmann thrombasthenia or Leukocyte Adhesion Deficiency-III syndrome (LAD-III or LAD-1/variant) present with increased bleeding tendency because of the lack or dysfunction of the fibrinogen receptor GPIIb/IIIa (integrin αIIbß3), respectively. Although the bleeding disorder is more severe in LAD-III patients, classic aggregometry or perfusion of Glanzmann or LAD-III platelets over collagen-coated slides under physiologic shear rate does not discriminate between these 2 conditions. However, in a novel flow cytometry-based aggregation assay, Glanzmann platelets were still capable of forming small aggregates upon collagen stimulation, whereas LAD-III platelets were not. These aggregates required functional GPIa/IIa (integrin α2ß1) instead of integrin αIIbß3, thus explaining the clinically more severe bleeding manifestations in LAD-III patients, in which all platelet integrins are functionally defective. These findings provide genetic evidence for the differential requirements of platelet integrins in thrombus formation and demonstrate that correct integrin function assessment can be achieved with a combination of diagnostic methods.


Subject(s)
Hemorrhage/diagnosis , Integrin alpha2beta1/metabolism , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombasthenia/metabolism , Collagen/metabolism , Flow Cytometry , Hemorrhage/etiology , Hemorrhage/metabolism , Humans , Leukocyte-Adhesion Deficiency Syndrome/complications , Leukocyte-Adhesion Deficiency Syndrome/pathology , Phenotype , Thrombasthenia/complications , Thrombasthenia/pathology
12.
Blood ; 117(26): 7042-52, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21536861

ABSTRACT

Kindlin-3 is a key lymphocyte function-associated antigen-1 (LFA-1) coactivator deleted in leukocyte adhesion deficiency-III (LAD-III). In the present study, we investigated the involvement of this adaptor in lymphocyte motility and TCR-triggered arrest on ICAM-1 or on dendritic cells (DCs). Kindlin-3-null primary T cells from a LAD-III patient migrated normally on the major lymph node chemokine CCL21 and engaged in normal TCR signaling. However, TCR activation of Kindlin-3-null T lymphocytes failed to trigger the robust LFA-1-mediated T-cell spreading on ICAM-1 and ICAM-1-expressing DCs that is observed in normal lymphocytes. Kindlin-3 was also essential for cytoskeletal anchorage of the LFA-1 heterodimer and for microclustering of LFA-1 within ventral focal dots of TCR-stimulated lymphocytes spread on ICAM-1. Surprisingly, LFA-1 on Kindlin-3-null lymphocytes migrating over CCL21 acquired normal expression of an epitope associated with the conformational activation of the key headpiece domain, ß I. This activated LFA-1 was highly responsive to TCR-triggered ICAM-1-driven stop signals in normal T cells locomoting on CCL21, but not in their Kindlin-3-null T-cell counterparts. We suggest that Kindlin-3 selectively contributes to a final TCR-triggered outside-in stabilization of bonds generated between chemokine-primed LFA-1 molecules and cell-surface ICAM-1.


Subject(s)
Cell Communication , Dendritic Cells/immunology , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Cell Adhesion , Cell Movement , Cell Shape , Cells, Cultured , Chemokine CCL21/metabolism , Cytoskeleton/metabolism , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Humans , Immunological Synapses/immunology , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/pathology , Lymphocyte Activation , Membrane Microdomains/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Microvilli/metabolism , Microvilli/ultrastructure , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Protein Multimerization , Protein Transport , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/ultrastructure
13.
J Immunol ; 186(9): 5273-83, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441448

ABSTRACT

Leukocyte adhesion deficiency type III is a recently described condition involving a Glanzmann-type bleeding syndrome and leukocyte adhesion deficiency. This was ascribed to a defect of the FERMT3 gene resulting in abnormal expression of kindlin-3, a protein expressed in hematopoietic cells with a major role in the regulation of integrin activation. In this article, we describe a patient with a new mutation of FERMT3 and lack of kindlin-3 expression in platelets and leukocytes. We assayed quantitatively the first steps of kindlin-3-defective leukocyte adhesion, namely, initial bond formation, bond strengthening, and early spreading. Initial bond formation was readily stimulated with neutrophils stimulated by fMLF, and neutrophils and lymphocytes stimulated by a phorbol ester or Mn(2+). In contrast, attachment strengthening was defective in the patient's lymphocytes treated with PMA or Mn(2+), or fMLF-stimulated neutrophils. However, attachment strengthening was normal in patient's neutrophils treated with phorbol ester or Mn(2+). In addition, the patient's T lymphocytes displayed defective integrin-mediated spreading and a moderate but significant decrease of spreading on anti-CD3-coated surfaces. Patient's neutrophils displayed a drastic alteration of integrin-mediated spreading after fMLF or PMA stimulation, whereas signaling-independent Mn(2+) allowed significant spreading. In conclusion, the consequences of kindlin-3 deficiency on ß(2) integrin function depend on both cell type and the stimulus used for integrin activation. Our results suggest looking for a possible kindlin-3 involvement in membrane dynamical event independent of integrin-mediated adhesion.


Subject(s)
Blood Platelets/metabolism , Leukocyte-Adhesion Deficiency Syndrome/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neutrophils/metabolism , Amino Acid Sequence , Base Sequence , Cell Adhesion/genetics , Cell Separation , Child, Preschool , Female , Flow Cytometry , Humans , Immunoblotting , Integrin alpha2/genetics , Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/physiopathology , Male , Molecular Sequence Data , Mutation , Pedigree , Reverse Transcriptase Polymerase Chain Reaction
14.
Dis Markers ; 29(3-4): 177-87, 2010.
Article in English | MEDLINE | ID: mdl-21178276

ABSTRACT

Rho GTPases are members of the Ras superfamily of GTPases that regulate a wide variety of cellular functions. While Rho GTPase pathways have been implicated in various pathological conditions in humans, to date coding mutations in only the hematopoietic specific GTPase, RAC2, have been found to cause a human disease, a severe phagocytic immunodeficiency characterized by life-threatening infections in infancy. Interestingly, the phenotype was predicted by a mouse knock-out of Rac2 and resembles leukocyte adhesion deficiency (LAD). Here we review Rho GTPases with a specific focus on Rac GTPases. In particular, we discuss a new understanding of the unique and overlapping roles of Rac2 in blood cells that has developed since the generation of mice deficient in Rac1, Rac2 and Rac3 proteins. We propose that Rac2 mutations leading to disease be termed LAD type IV.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , rac GTP-Binding Proteins , Animals , Genetic Association Studies , Hematopoietic System/physiopathology , Humans , Infant , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/physiopathology , Mice , Mice, Knockout , Mutation , Opportunistic Infections/blood , Phagocytes , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/immunology , rac GTP-Binding Proteins/metabolism
15.
J Intern Med ; 268(1): 25-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20497300

ABSTRACT

Neutrophil granules store proteins that are critically important for the neutrophil to move from the vascular bed to tissues and to kill microorganisms. This is illustrated in nature when individual proteins are deleted due to inherited mutations of their cognate genes, and such deficiencies result in the conditions leucocyte adhesion deficiency and chronic granulomatous disease. The granules of the neutrophil have traditionally been divided into two or three major types but are instead a continuum where several subtypes can be identified with differences in protein content and propensity for mobilization. This is explained by the 'targeting by timing hypothesis' which states that granules are filled with granule proteins that are synthesized at the time the granule is formed. The heterogeneity of granules arises because the synthesis of granule proteins is individually controlled and major differences exist in the timings of biosynthesis during granulocytopoiesis. This is largely controlled by gene transcription.


Subject(s)
Leukocyte Disorders/pathology , Neutrophils/physiology , Animals , Cell Differentiation/genetics , Cytoplasmic Granules/metabolism , Gene Expression Regulation , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Humans , Leukocyte Disorders/metabolism , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/pathology , Mice , Neutropenia/metabolism , Neutropenia/pathology , Neutrophils/pathology
16.
Blood ; 115(23): 4834-42, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20357244

ABSTRACT

In the disorder leukocyte adhesion deficiency III (LAD-III), integrins on platelets and leukocytes are expressed but fail to function and this leads to severe bleeding and infections at an early age. Mutation in the KINDLIN3 (FERMT3) gene is the cause of LAD-III in patients from the Middle East, Malta, and Turkey. We describe 2 novel homozygous mutations in the KINDLIN3 gene of a new African-American patient that destabilize KINDLIN3 mRNA leading to loss of kindlin-3 protein. Transfection of wild-type (WT) KINDLIN3 cDNA restored integrin-related adhesion and migration in the LAD-III patient's T and B lymphocytes. We analyzed the individual mutations separately in vitro to learn more about the function of the kindlin-3 protein. The first G>A mutation gives rise to a Gly308Arg change at the end of FERM (protein 4.1, ezrin, radixin, moesin) subdomain 2, and the second mutation is a base deletion causing early termination within the pleckstrin homology (PH) domain. This second mutation prevented membrane association of kindlin-3 and did not restore either adhesion or migration, whereas the FERM subdomain 2 mutation affected only migration. Thus, these LAD-III patient mutations have highlighted functionally important regions of kindlin-3 that alter leukocyte integrin-dependent function in 2 distinct ways.


Subject(s)
B-Lymphocytes/metabolism , Genetic Diseases, Inborn/metabolism , Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Membrane Proteins/metabolism , Mutation, Missense , Neoplasm Proteins/metabolism , T-Lymphocytes/metabolism , Black or African American , Amino Acid Substitution , Cell Adhesion/genetics , Cell Movement/genetics , Female , Genetic Diseases, Inborn/genetics , Homozygote , Humans , Infant , Integrins/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Protein Structure, Tertiary , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Infect Dev Ctries ; 4(3): 175-8, 2010 Mar 29.
Article in English | MEDLINE | ID: mdl-20351460

ABSTRACT

Leukocyte adhesion deficiency type 1 (LAD-1) is a rare, inherited immunodeficiency that affects one per million people yearly and usually presents with recurrent, indolent bacterial infections of the skin, mouth, and respiratory tract and impaired pus formation and wound healing. A 13-year-old girl diagnosed LAD-I at the age of 7 years was brought to the Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, because of a draining plaque on the left leg for 2.5 years. She had recurrent skin infections and had been treated with repeated courses of different antibiotic combinations, with temporary responses, since 5 years of age. Examination revealed a 7 x 8 cm minimally erythematous hyperpigmented plaque with multiple draining sinuses on the left leg. Tissue culture yielded Pseudomonas aeruginosa. Flow cytometry showed CD18 (18.79%), CD11a (51.59%), CD11b (18.61%) and CD11c(10.60%). A plain radiography of the left leg revealed osteomyelitis. It is highly suggested that patients diagnosed mild to moderate LAD-1 with recurrent skin infection and simultaneous weak response to conventional therapy undergo (BMT) marrow transplant to prohibit subsequent life-threatening complications.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome/complications , Osteomyelitis/etiology , Adolescent , Anti-Bacterial Agents/therapeutic use , Bone Marrow Transplantation , CD18 Antigens/biosynthesis , Female , Humans , Leg/diagnostic imaging , Leg/microbiology , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/surgery , Leukocytes/metabolism , Osteomyelitis/diagnostic imaging , Osteomyelitis/metabolism , Pseudomonas Infections/drug therapy , Pseudomonas Infections/etiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/drug effects , Radiography , Treatment Failure
18.
Thromb Haemost ; 103(5): 1053-64, 2010 May.
Article in English | MEDLINE | ID: mdl-20216991

ABSTRACT

Leukocyte adhesion deficiency-III (LAD-III) also called leukocyte adhesion deficiency-1/variant (LAD1v) is a rare congenital disease caused by defective integrin activation of leukocytes and platelets. Patients with LAD-III present with non-purulent infections and increased bleeding symptoms. We report on a novel integrin-dependent platelet dysfunction in two brothers with LAD-III syndrome caused by a homozygous mutation 1717C>T in the FERMT3 gene leading to a premature stop codon R573X in the focal adhesion protein kindlin-3. Stimulation of patients platelets with all used agonists resulted in a severely decreased binding of soluble fibrinogen indicating a defect in inside-out activation of the integrin alpha(IIb) beta(3) (GPIIb/IIIa). Patients platelets did not respond to the alpha(2)beta(1)-integrin agonist aggretin-A at all. Our data on granula secretion indicate for the first time that the thrombin receptor PAR-4 but not PAR-1 may be important in integrin-triggered granule secretion in response to thrombin. In contrast, collagen mediated platelet granule secretion was not affected in LAD-III-patients. Thus, integrin-signalling may be not essential in collagen-induced granule secretion. The patients' peripheral blood mononuclear cells showed a severe loss of adhesion capacity to VCAM-1 and to endothelial cells compared to cells from healthy donors. Rap-1 activation after PMA stimulation could be observed in controls but not in patients cells. After haematogenesis stem cell transplantation (HSCT) the brothers showed no symptoms of bleeding or immunodeficiency and the integrin-dependent platelet and leukocyte functions normalised.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome/blood , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocytes, Mononuclear/metabolism , Membrane Proteins/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Cell Adhesion/genetics , Cell Degranulation/genetics , Cells, Cultured , Child , Child, Preschool , Chimerism , Collagen/metabolism , GTPase-Activating Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Hemorrhage , Humans , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocytes, Mononuclear/pathology , Male , Platelet Activation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
19.
Semin Immunopathol ; 32(2): 173-82, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20217414

ABSTRACT

Integrin binding to ligand plays essential roles in the differentiation and function of mammalian cells. beta2 integrins in leukocytes are needed for migration to sites of inflammation and in lymph nodes as well as for cellular events such as phagocytosis and the formation of the conjugates between T cells and antigen-presenting cells. In T cells, integrin adhesion is activated primarily by the antigen-receptor (TCR complex) and chemokines in a process known as 'inside-out' signalling. Great progress has been made in identifying mutations that are responsible for leukocyte adhesion deficiency (LAD) syndromes, a disorder that presents with an impaired ability to clear pathogens and recurrent life-threatening infections. LAD mutations have been identified with defects in integrins, fucosylation and in the new intracellular mediator kindlin-3. Here, we review the key players in the 'inside-out' and 'outside-in' signalling pathways that will serve as new potential targets in the design of novel therapeutics to treat various immunodeficiencies.


Subject(s)
Integrins/immunology , Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/immunology , Signal Transduction/immunology , Animals , Cell Adhesion/immunology , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/metabolism , Integrins/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Lymphocyte Activation/immunology , Signal Transduction/genetics
20.
Gastroenterology ; 138(3): 1079-90.e1-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19900444

ABSTRACT

BACKGROUND & AIMS: Leukocyte adhesion deficiency II (LAD II) is a rare condition caused by defective protein fucosylation, causing decreased leukocyte rolling, psychomotor retardation, and poor growth. The ligand-binding activity of Notch, a gastrointestinal signaling protein, depends on O-fucosylation. We investigated Notch signaling and intestinal epithelial architecture in a mouse model of LAD II. METHODS: Mice lacking 3,5-epimerase/4-reductase (FX) or FX(-/-) bone marrow chimeras (with either wild-type or FX(-/-) bone marrow) were maintained on a fucose-free diet. Intestinal secretory epithelial cells were quantified by histology and immunohistochemistry. Reverse transcription-polymerase chain reaction and immunoblot analyses were used to detect Notch-regulated genes in isolated crypt epithelium. Intestinal leukocyte-endothelial interaction was quantified by intravital microscopy. The intestinal epithelium of 2-week-old FX(-/-) mice was transfected with an adenoviral vector expressing a constitutively active form of Notch. RESULTS: FX(-/-) mice rapidly exhibited secretory epithelial cell hyperplasia, reduced cell proliferation, and altered epithelial gene expression patterns consistent with reduced Notch signaling. These effects were reversed when mice were given dietary fucose or by adenoviral transfection of the intestinal epithelium with the Notch intracellular domain. CONCLUSIONS: In a mouse model of LAD II, secretory cell hyperplasia occurs in the small intestine and colon; these effects depend on Notch signaling. Defects in Notch signaling might therefore be involved in the pathogenesis of this rare pediatric condition.


Subject(s)
Carbohydrate Epimerases/metabolism , Cell Proliferation , Colon/metabolism , Goblet Cells/metabolism , Hydro-Lyases/metabolism , Ileum/metabolism , Leukocyte Rolling , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Paneth Cells/metabolism , Receptors, Notch/metabolism , Adenoviridae/genetics , Animals , Carbohydrate Epimerases/deficiency , Carbohydrate Epimerases/genetics , Cell Lineage , Colon/pathology , Dietary Carbohydrates/administration & dosage , Disease Models, Animal , Fucose/administration & dosage , Fucose/deficiency , Gene Expression Regulation , Genetic Vectors , Genotype , Goblet Cells/pathology , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Hyperplasia , Ileum/pathology , Immunoblotting , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Video , Paneth Cells/pathology , Phenotype , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...