Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958468

ABSTRACT

Primary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening disorder associated with autosomal recessive variants in genes required for perforin-mediated lymphocyte cytotoxicity. A rapid diagnosis is crucial for successful treatment. Although defective cytotoxic T lymphocyte (CTL) function causes pathogenesis, quantification of natural killer (NK) cell exocytosis triggered by K562 target cells currently represents a standard diagnostic procedure for primary HLH. We have prospectively evaluated different lymphocyte exocytosis assays in 213 patients referred for evaluation for suspected HLH and related hyperinflammatory syndromes. A total of 138 patients received a molecular diagnosis consistent with primary HLH. Compared to routine K562 cell-based assays, assessment of Fc receptor-triggered NK-cell and T cell receptor-triggered CTL exocytosis displayed higher sensitivity and improved specificity for the diagnosis of primary HLH, with these assays combined providing a sensitivity of 100% and specificity of 98.3%. By comparison, NK-cell exocytosis following K562 target cell stimulation displayed a higher inter-individual variability, in part explained by differences in NK-cell differentiation or large functional reductions following shipment. We thus recommend combined analysis of T cell receptor-triggered CTL and Fc receptor-triggered NK-cell exocytosis for the diagnosis of patients with suspected familial HLH or atypical manifestations of congenital defects in lymphocyte exocytosis.

2.
Immunity ; 46(2): 287-300, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28214226

ABSTRACT

Tissue-resident memory T (Trm) cells form a heterogeneous population that provides localized protection against pathogens. Here, we identify CD49a as a marker that differentiates CD8+ Trm cells on a compartmental and functional basis. In human skin epithelia, CD8+CD49a+ Trm cells produced interferon-γ, whereas CD8+CD49a- Trm cells produced interleukin-17 (IL-17). In addition, CD8+CD49a+ Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response. In skin from patients with vitiligo, where melanocytes are eradicated locally, CD8+CD49a+ Trm cells that constitutively expressed perforin and granzyme B accumulated both in the epidermis and dermis. Conversely, CD8+CD49a- Trm cells from psoriasis lesions predominantly generated IL-17 responses that promote local inflammation in this skin disease. Overall, CD49a expression delineates CD8+ Trm cell specialization in human epithelial barriers and correlates with the effector cell balance found in distinct inflammatory skin diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Integrin alpha1/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology , Cell Separation , Flow Cytometry , Humans , Immunologic Memory/immunology , Integrin alpha1/biosynthesis , Lymphocyte Activation/immunology , Microscopy, Confocal , Psoriasis/immunology , Vitiligo/immunology
3.
Blood ; 140(7): 706-715, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35687753

ABSTRACT

XIAP (X-linked inhibitor of apoptosis) deficiency is a rare inborn error of immunity. XIAP deficiency causes hyperinflammatory disease manifestations due to dysregulated TNF (tumor necrosis factor)-receptor signaling and NLRP3 (NOD- [nucleotide-binding oligomerization domain], LRR- [leucine-rich repeat] and pyrin domain-containing protein 3) inflammasome function. Safe and effective long-term treatments are needed and are especially important to help prevent the need for high-risk allogeneic hematopoietic cell transplantation. Here we evaluated inflammasome inhibitors as potential therapeutics with a focus on the natural flavonoid antioxidant quercetin. Bone marrow (BM)-derived macrophages were derived from XIAP-deficient or wild-type (WT) mice. Human monocytes were obtained from control or XIAP-deficient patients. Cells were stimulated with TLR (Toll-like receptor) agonists or TNF-α ± inhibitors or quercetin. For in vivo lipopolysaccharide (LPS) challenge experiments, XIAP-deficient or WT mice were fed mouse chow ± supplemental quercetin (50 mg/kg per day exposure) for 7 days followed by a challenge with 10 ng/kg LPS. IL-1ß (interleukin-1ß) and IL-18 were measured by ELISA (enzyme-linked immunosorbent assay). In murine studies, quercetin prevented IL-1ß secretion from XIAP knockout cells following TLR agonists or TNF-α stimulation (P < .05) and strongly reduced constitutive production of IL-18 by both WT and XIAP-deficient cells (P < .05). At 4 hours after in vivo LPS challenge, blood levels of IL-1ß and IL-18 were significantly decreased in mice that had received quercetin-supplemented chow (P < .05). In experiments using human cells, quercetin greatly reduced IL-1ß secretion by monocytes following TNF-α stimulation (P < .05). Our data suggest that quercetin may be an effective natural therapeutic for the prevention of XIAP deficiency-associated hyperinflammation. Clinical trials, including careful pharmacokinetic and pharmacodynamic studies to ensure that effective levels of quercetin can be obtained, are warranted.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Genetic Diseases, X-Linked , Humans , Inhibitor of Apoptosis Proteins , Interleukin-18 , Interleukin-1beta , Lipopolysaccharides/pharmacology , Lymphoproliferative Disorders , Mice , Quercetin/pharmacology , Quercetin/therapeutic use , Tumor Necrosis Factor-alpha , X-Linked Inhibitor of Apoptosis Protein/genetics
4.
Immunity ; 42(3): 443-56, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25786176

ABSTRACT

The mechanisms underlying human natural killer (NK) cell phenotypic and functional heterogeneity are unknown. Here, we describe the emergence of diverse subsets of human NK cells selectively lacking expression of signaling proteins after human cytomegalovirus (HCMV) infection. The absence of B and myeloid cell-related signaling protein expression in these NK cell subsets correlated with promoter DNA hypermethylation. Genome-wide DNA methylation patterns were strikingly similar between HCMV-associated adaptive NK cells and cytotoxic effector T cells but differed from those of canonical NK cells. Functional interrogation demonstrated altered cytokine responsiveness in adaptive NK cells that was linked to reduced expression of the transcription factor PLZF. Furthermore, subsets of adaptive NK cells demonstrated significantly reduced functional responses to activated autologous T cells. The present results uncover a spectrum of epigenetically unique adaptive NK cell subsets that diversify in response to viral infection and have distinct functional capabilities compared to canonical NK cell subsets.


Subject(s)
Antibodies/immunology , Cytomegalovirus Infections/genetics , Epigenesis, Genetic/immunology , Killer Cells, Natural/immunology , Kruppel-Like Transcription Factors/immunology , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Cell Proliferation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , DNA Methylation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Profiling , Humans , Immunophenotyping , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/classification , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Microarray Analysis , NK Cell Lectin-Like Receptor Subfamily C/deficiency , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction , Syk Kinase , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/immunology
5.
Blood ; 137(15): 2033-2045, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33513601

ABSTRACT

Exocytosis of cytotoxic granules (CG) by lymphocytes is required for the elimination of infected and malignant cells. Impairments in this process underly a group of diseases with dramatic hyperferritinemic inflammation termed hemophagocytic lymphohistiocytosis (HLH). Although genetic and functional studies of HLH have identified proteins controlling distinct steps of CG exocytosis, the molecular mechanisms that spatiotemporally coordinate CG release remain partially elusive. We studied a patient exhibiting characteristic clinical features of HLH associated with markedly impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell exocytosis functions, who beared biallelic deleterious mutations in the gene encoding the small GTPase RhoG. Experimental ablation of RHOG in a model cell line and primary CTLs from healthy individuals uncovered a hitherto unappreciated role of RhoG in retaining CGs in the vicinity of the plasma membrane (PM), a fundamental prerequisite for CG exocytotic release. We discovered that RhoG engages in a protein-protein interaction with Munc13-4, an exocytosis protein essential for CG fusion with the PM. We show that this interaction is critical for docking of Munc13-4+ CGs to the PM and subsequent membrane fusion and release of CG content. Thus, our study illuminates RhoG as a novel essential regulator of human lymphocyte cytotoxicity and provides the molecular pathomechanism behind the identified here and previously unreported genetically determined form of HLH.


Subject(s)
Killer Cells, Natural/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , T-Lymphocytes, Cytotoxic/pathology , rho GTP-Binding Proteins/genetics , Cell Line , Cells, Cultured , Gene Deletion , Germ-Line Mutation , Humans , Infant , Killer Cells, Natural/metabolism , Lymphohistiocytosis, Hemophagocytic/pathology , Male , Models, Molecular , T-Lymphocytes, Cytotoxic/metabolism , rho GTP-Binding Proteins/chemistry
6.
Eur J Anaesthesiol ; 38(7): 692-701, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33186307

ABSTRACT

BACKGROUND: Severe pandemic influenza has been associated with the hyperinflammatory condition secondary haemophagocytic lymphohistiocytosis (HLH). OBJECTIVES: To determine the frequency, degree, character and possible cause of influenza-associated HLH in critically ill patients with severe acute respiratory distress syndrome due to influenza A (H1N1) infection requiring extracorporeal membrane oxygenation (ECMO) support at our hospital. DESIGN: A retrospective observational study. PATIENTS AND SETTING: Medical data were retrieved retrospectively from 11 consenting patients of thirteen adults infected with pandemic influenza A (H1N1) 2009 requiring ECMO between July 2009 and January 2010 at the ECMO Centre of Karolinska University Hospital, Stockholm, Sweden. All patients were evaluated for HLH using HLH-2004 criteria and HScore. RESULTS: Eleven patients (median age 31 years) were included in the study and all survived. All patients showed signs of multiple organ dysfunction and pronounced inflammation, more severe in the four patients with HLH who had significantly higher peak serum concentrations of ferritin (P = 0.024), alkaline phosphatase (P = 0.012) and gamma-glutamyl transferase (P = 0.024), lower concentration of albumin (P = 0.0086) and more frequently hepatomegaly (P = 0.048). Abnormal lymphocyte cytotoxicity (lytic units <10) and a low proportion of natural killer (NK) cells were observed in three of four patients with HLH. Notably, we found a significant inverse correlation between serum ferritin concentration and NK cell and cytotoxic T lymphocyte percentages (rs = -0.74, P = 0.0013 and rs = -0.79, P = 0.0025, respectively). One HLH patient received HLH-directed cytotoxic therapy, another intravenous immunoglobulin and the other two no specific HLH-directed therapy. CONCLUSION: Critically ill patients, including healthy young adults, with pandemic influenza may develop HLH and should be monitored for signs of hyperinflammation and increasing organ dysfunction, and evaluated promptly for HLH because HLH-directed therapy may then be beneficial. The association of low NK percentages with hyperferritinaemia may suggest a role for reduced NK cell numbers, possibly also cytotoxic T lymphocytes, and subsequently reduced lymphocyte cytotoxicity, in the pathogenesis of hyperinflammation and secondary HLH.


Subject(s)
Extracorporeal Membrane Oxygenation , Influenza A Virus, H1N1 Subtype , Influenza, Human , Lymphohistiocytosis, Hemophagocytic , Adult , Humans , Influenza, Human/diagnosis , Influenza, Human/epidemiology , Influenza, Human/therapy , Laboratories , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/epidemiology , Lymphohistiocytosis, Hemophagocytic/therapy , Retrospective Studies , Sweden , Young Adult
7.
Pediatr Blood Cancer ; 67(4): e28184, 2020 04.
Article in English | MEDLINE | ID: mdl-31994322

ABSTRACT

Mutations in SH2D1A, encoding the intracellular adaptor signaling lymphocyte activation molecule associated protein (SAP), are associated with X-linked lymphoproliferative disease type 1 (XLP1). We identified a novel hemizygous SH2D1A c.49G > A (p.E17K) variant in a 21-year-old patient with fatal Epstein-Barr virus infection-associated hemophagocytic lymphohistiocytosis. Cellular and biochemical assays revealed normal expression of the SAP variant protein, yet binding to phosphorylated CD244 receptor was reduced by >95%. Three healthy brothers carried the SH2D1A c.49G > A variant. Thus, data suggest that this variant represents a pathogenic mutation, but with variable expressivity. Importantly, our results highlight challenges in the clinical interpretation of SH2D1A variants and caution in using functional flow cytometry assays for the diagnosis of XLP1.


Subject(s)
Epstein-Barr Virus Infections , Hemizygote , Herpesvirus 4, Human , Lymphohistiocytosis, Hemophagocytic , Lymphoproliferative Disorders , Mutation, Missense , Neoplasm Proteins , Signaling Lymphocytic Activation Molecule Associated Protein , Adult , Amino Acid Substitution , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Fatal Outcome , Gene Expression Regulation, Leukemic , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/metabolism , Lymphohistiocytosis, Hemophagocytic/virology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Lymphoproliferative Disorders/virology , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein/biosynthesis , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Signaling Lymphocytic Activation Molecule Family/genetics
8.
J Pediatr Hematol Oncol ; 42(7): e627-e629, 2020 10.
Article in English | MEDLINE | ID: mdl-31651726

ABSTRACT

Although familial hemophagocytic lymphohistiocytosis (FHL) generally manifest with a combination of unremitting fever, hepatosplenomegaly, and pancytopenia; unusual presentations should also be taken into account. Herein, we present 3 FHL cases with 2 novel mutations with different initial presentations. The first patient bearing a homozygous truncation mutation in UNC13D (c.2650C>T.p.Gln884Ter) presented with central nervous system involvement and skin rash. The patient responded to the HLH-2004 protocol, and allogenic hematopoietic stem cell transplantation was performed from her healthy sister. The second and third patients with homozygous splice site mutation (c.430-1G>A) in STXBP2 were siblings who presented at birth with fevers, elevated aspartate aminotransferase, alanine aminotransferase, and hyperferritinemia but did not fulfill FHL criteria. The last 2 infants died despite intervention. Hematologists should be vigilant about the different presentation of FHL in children.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Proteins/genetics , Munc18 Proteins/genetics , Female , Humans , Infant , Infant, Newborn , Male , Mutation , Phenotype
9.
J Pediatr Hematol Oncol ; 42(2): 156-159, 2020 03.
Article in English | MEDLINE | ID: mdl-31033788

ABSTRACT

Gain of function mutations in the p110δ catalytic subunit of the phosphatidylinositol-3-OH kinase (PIK3CD) classified as activated phosphoinositide 3-kinase delta syndrome (APDS) are the cause of a primary immunodeficiency characterized by recurrent sinopulmonary infections, and lymphoproliferation. Previously, autoimmunity and Epstein-Barr virus-related B-cell lymphoma have been documented for patients with APDS; here, we present a case that extends the picture, as the patient shows the full diagnostic criteria of hemophagocytic lymphohistiocytosis at 6 months of age. He experienced Hodgkin lymphoma as a 2.5-year-old baby. Next-generation sequencing returned a de novo heterozygous missense variant in PIK3CD (LRG_191t1: c.3061G>A; p.Glu1021Lys), confirming the primary immunodeficiency. After 2 courses of ifosfamide, cisplatin, and etoposide combined with brentuximab, the patient successfully underwent allogeneic hematopoietic stem cell transplantation from his HLA full matched sister, and he has been well for 18 months after that. The hematologist treating Hodgkin lymphoma and/or hemophagocytic lymphohistiocytosis should be vigilant about the possible underlying immune deficiency, and they should consider APDS in their differential diagnosis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Hodgkin Disease/pathology , Lymphohistiocytosis, Hemophagocytic/pathology , Mutation , Primary Immunodeficiency Diseases/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/complications , Hodgkin Disease/genetics , Hodgkin Disease/therapy , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/therapy , Prognosis
10.
Blood ; 129(14): 1927-1939, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28209719

ABSTRACT

Heterozygous GATA2 mutation is associated with immunodeficiency, lymphedema, and myelodysplastic syndrome. Disease presentation is variable, often coinciding with loss of circulating dendritic cells, monocytes, B cells, and natural killer (NK) cells. Nonetheless, in a proportion of patients carrying GATA2 mutation, NK cells persist. We found that peripheral blood NK cells in symptomatic patients uniformly lacked expression of the transcription factor promyelocytic leukemia zinc finger (PLZF), as well as expression of intracellular signaling proteins FcεRγ, spleen tyrosine kinase (SYK), and EWS/FLI1-Activated Transcript 2 (EAT-2) in a variegated manner. Moreover, consistent with an adaptive identity, NK cells from patients with GATA2 mutation displayed altered expression of cytotoxic granule constituents and produced interferon-γ upon Fc-receptor engagement but not following combined interleukin-12 (IL-12) and IL-18 stimulation. Canonical, PLZF-expressing NK cells were retained in asymptomatic carriers of GATA2 mutation. Developmentally, GATA-binding protein-2 (GATA-2) was expressed in hematopoietic stem cells, but not in NK-cell progenitors, CD3-CD56bright, canonical, or adaptive CD3-CD56dim NK cells. Peripheral blood NK cells from individuals with GATA2 mutation proliferated normally in vitro, whereas lineage-negative progenitors displayed impaired NK-cell differentiation. In summary, adaptive NK cells can persist in patients with GATA2 mutation, even after NK-cell progenitors expire. Moreover, our data suggest that adaptive NK cells are more long-lived than canonical, immunoregulatory NK cells.


Subject(s)
Cell Proliferation , GATA2 Transcription Factor , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/immunology , Mutation , Adolescent , Adult , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/immunology , Child , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Male , Middle Aged , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, IgE/genetics , Receptors, IgE/immunology , Syk Kinase/genetics , Syk Kinase/immunology , Transcription Factors/genetics , Transcription Factors/immunology
11.
Blood ; 129(16): 2266-2279, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28202457

ABSTRACT

Several monogenic causes of familial myelodysplastic syndrome (MDS) have recently been identified. We studied 2 families with cytopenia, predisposition to MDS with chromosome 7 aberrations, immunodeficiency, and progressive cerebellar dysfunction. Genetic studies uncovered heterozygous missense mutations in SAMD9L, a tumor suppressor gene located on chromosome arm 7q. Consistent with a gain-of-function effect, ectopic expression of the 2 identified SAMD9L mutants decreased cell proliferation relative to wild-type protein. Of the 10 individuals identified who were heterozygous for either SAMD9L mutation, 3 developed MDS upon loss of the mutated SAMD9L allele following intracellular infections associated with myeloid, B-, and natural killer (NK)-cell deficiency. Five other individuals, 3 with spontaneously resolved cytopenic episodes in infancy, harbored hematopoietic revertant mosaicism by uniparental disomy of 7q, with loss of the mutated allele or additional in cisSAMD9L truncating mutations. Examination of 1 individual indicated that somatic reversions were postnatally selected. Somatic mutations were tracked to CD34+ hematopoietic progenitor cell populations, being further enriched in B and NK cells. Stimulation of these cell types with interferon (IFN)-α or IFN-γ induced SAMD9L expression. Clinically, revertant mosaicism was associated with milder disease, yet neurological manifestations persisted in 3 individuals. Two carriers also harbored a rare, in trans germ line SAMD9L missense loss-of-function variant, potentially counteracting the SAMD9L mutation. Our results demonstrate that gain-of-function mutations in the tumor suppressor SAMD9L cause cytopenia, immunodeficiency, variable neurological presentation, and predisposition to MDS with -7/del(7q), whereas hematopoietic revertant mosaicism commonly ameliorated clinical manifestations. The findings suggest a role for SAMD9L in regulating IFN-driven, demand-adapted hematopoiesis.


Subject(s)
Cognitive Dysfunction/diagnosis , Immunologic Deficiency Syndromes/diagnosis , Mutation , Myelodysplastic Syndromes/diagnosis , Pancytopenia/diagnosis , Tumor Suppressor Proteins/genetics , Adult , Alleles , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Proliferation , Child , Chromosomes, Human, Pair 7/chemistry , Cognitive Dysfunction/complications , Cognitive Dysfunction/genetics , Cognitive Dysfunction/immunology , Female , Gene Expression , Hematopoiesis/immunology , Heterozygote , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunophenotyping , Interferon Type I/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Middle Aged , Mosaicism , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Pancytopenia/complications , Pancytopenia/genetics , Pancytopenia/immunology , Pedigree , Tumor Suppressor Proteins/metabolism
12.
J Pediatr Hematol Oncol ; 39(6): e309-e311, 2017 08.
Article in English | MEDLINE | ID: mdl-28328614

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is characterized by activation of cytotoxic T and natural killer (NK) cells, and macrophages related to a spectrum of hyperinflammatory disorders. The clinical findings mainly include high fever, cytopenia, splenomegaly, phagocytosis, and proliferation of histiocytes in lymphoreticular tissue. To the best of our knowledge, transfusion-associated graft versus host disease (TA-GVHD) in a 13-day old male newborn with HLH is being reported first time in the literature. The aim of this report was to emphasize the importance of blood products irradiation in the prevention of the development of graft versus host disease especially among high-risk subjects such as newborns with HLH.


Subject(s)
Graft vs Host Disease/prevention & control , Lymphohistiocytosis, Hemophagocytic/complications , Transfusion Reaction , Blood Preservation , Blood Safety/methods , Blood Transfusion/standards , Graft vs Host Disease/etiology , Humans , Infant, Newborn , Lymphohistiocytosis, Hemophagocytic/congenital , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/pathology , Male
13.
J Pediatr Hematol Oncol ; 39(6): e321-e324, 2017 08.
Article in English | MEDLINE | ID: mdl-28692549

ABSTRACT

BACKGROUND: Familial hemophagocytic lymphohistiocytosis (HLH) is a fatal disease affecting infants and very young children. Central nervous system involvement of HLH can cause catastrophic results. METHOD: We present a case with cranial involvement of familial HLH type 4 who showed diffuse infiltration of white matter complicated with intracranial thrombosis. A 5-year-old girl from a consanguineous couple presented with fever and pancytopenia, and was referred to our hematology unit. Examination revealed fever, lymphadenopathy, and hepatosplenomegaly. Ultrasound examination revealed hepatosplenomegaly and free intra-abdominal fluid. HLH was revealed on bone marrow aspiration biopsy. Defective natural killer and T lymphocyte cytotoxicity using degranulation tests was determined. In the genetic analysis, syntaxin gene mutation was found. On T2-weighted and T2-fluid-attenuated inversion recovery magnetic resonance imaging (MRI), diffuse hyperintense signal changes of cerebral white matter, indicating white matter demyelination, were observed. A second brain MRI showed an acute infarct involving the left temporooccipital region. Immunosuppressive therapy according to the HLH 2004 protocol was started. The infarct resolved but white matter lesions were stable on the brain MRI that was performed 1 month later. Brain MRI taken 4 months after the first examination showed stable cerebral white matter lesions, but hyperintense signal changes appeared in the cerebellar white matter and were regarded as progression. The patient died because of infection despite immunosuppressive therapy. CONCLUSIONS: Physicians managing patients with HLH must be vigilant about the possibility of central nervous system involvement including stroke.


Subject(s)
Central Nervous System Diseases/complications , Infarction/etiology , Lymphohistiocytosis, Hemophagocytic/pathology , Child, Preschool , Disease Progression , Fatal Outcome , Female , Humans , Intracranial Thrombosis/etiology , Leukoencephalopathies/etiology , Leukoencephalopathies/pathology , Lymphohistiocytosis, Hemophagocytic/complications , Magnetic Resonance Imaging/methods
14.
J Allergy Clin Immunol ; 137(4): 1165-1177, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26478006

ABSTRACT

BACKGROUND: Mutations in lysosomal trafficking regulator (LYST) cause Chediak-Higashi syndrome (CHS), a rare immunodeficiency with impaired cytotoxic lymphocyte function, mainly that of natural killer (NK) cells. Our understanding of NK cell function deficiency in patients with CHS and how LYST regulates lytic granule exocytosis is very limited. OBJECTIVE: We sought to delineate cellular defects associated with LYST mutations responsible for the impaired NK cell function seen in patients with CHS. METHODS: We analyzed NK cells from patients with CHS with missense mutations in the LYST ARM/HEAT (armadillo/huntingtin, elongation factor 3, protein phosphatase 2A, and the yeast kinase TOR1) or BEACH (beige and Chediak-Higashi) domains. RESULTS: NK cells from patients with CHS displayed severely reduced cytotoxicity. Mutations in the ARM/HEAT domain led to a reduced number of perforin-containing granules, which were significantly increased in size but able to polarize to the immunologic synapse; however, they were unable to properly fuse with the plasma membrane. Mutations in the BEACH domain resulted in formation of normal or slightly enlarged granules that had markedly impaired polarization to the IS but could be exocytosed on reaching the immunologic synapse. Perforin-containing granules in NK cells from patients with CHS did not acquire certain lysosomal markers (lysosome-associated membrane protein 1/2) but were positive for markers of transport vesicles (cation-independent mannose 6-phosphate receptor), late endosomes (Ras-associated binding protein 27a), and, to some extent, early endosomes (early endosome antigen 1), indicating a lack of integrity in the endolysosomal compartments. NK cells from patients with CHS had normal cytokine compartments and cytokine secretion. CONCLUSION: LYST is involved in regulation of multiple aspects of NK cell lytic activity, ranging from governance of lytic granule size to control of their polarization and exocytosis, as well as regulation of endolysosomal compartment identity. LYST functions in the regulated exocytosis but not in the constitutive secretion pathway.


Subject(s)
Chediak-Higashi Syndrome/physiopathology , Cytokines/metabolism , Exocytosis/physiology , Killer Cells, Natural/metabolism , Lysosomes/physiology , Vesicular Transport Proteins/genetics , Adult , Chediak-Higashi Syndrome/genetics , Female , Genetic Markers , Humans , Male , Mutation, Missense , Vesicular Transport Proteins/physiology
16.
J Clin Immunol ; 36(5): 480-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27146671

ABSTRACT

PURPOSE: Autosomal recessive mutations in LRBA, encoding for LPS-responsive beige-like anchor protein, were described in patients with a common variable immunodeficiency (CVID)-like disease characterized by hypogammaglobulinemia, autoimmune cytopenias, and enteropathy. Here, we detail the clinical, immunological, and genetic features of a patient with severe autoimmune manifestations. METHODS: Whole exome sequencing was performed to establish a molecular diagnosis. Evaluation of lymphocyte subsets was performed for immunological characterization. Medical files were reviewed to collect clinical and immunological data. RESULTS: A 7-year-old boy, born to consanguineous parents, presented with autoimmune hemolytic anemia, hepatosplenomegaly, autoimmune thyroiditis, and severe autoimmune gastrointestinal manifestations. Immunological investigations revealed low immunoglobulin levels and low numbers of B and NK cells. Treatment included immunoglobulin replacement and immunosuppressive therapy. Seven years after disease onset, the patient developed severe neurological symptoms resembling acute disseminated encephalomyelitis, prompting allogeneic hematopoietic stem cell transplantation (HSCT) with the HLA-identical mother as donor. Whole exome sequencing of the patient uncovered a homozygous 1 bp deletion in LRBA (c.7162delA:p.T2388Pfs*7). Importantly, during 2 years of follow-up post-HSCT, marked clinical improvement and recovery of immune function was observed. CONCLUSIONS: Our data suggest a beneficial effect of HSCT in patients with LRBA deficiency.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Anemia, Hemolytic, Autoimmune/therapy , B-Lymphocytes/immunology , Common Variable Immunodeficiency/therapy , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Sequence Deletion/genetics , Anemia, Hemolytic, Autoimmune/diagnosis , Anemia, Hemolytic, Autoimmune/genetics , Autoimmunity , Child , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/genetics , Consanguinity , DNA Mutational Analysis , HLA Antigens/immunology , Histocompatibility , Humans , Immunoglobulins/blood , Male , Treatment Outcome
17.
Bioconjug Chem ; 27(9): 2095-102, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27491005

ABSTRACT

Antibodies are widely used reagents for recognition in both clinic and research laboratories all over the world. For many applications, antibodies are labeled through conjugation to different reporter molecules or therapeutic agents. Traditionally, antibodies are covalently conjugated to reporter molecules via primary amines on lysines or thiols on cysteines. While efficient, such labeling is variable and nonstoichiometric and may affect an antibody's binding to its target. Moreover, an emerging field for therapeutics is antibody-drug conjugates, where a toxin or drug is conjugated to an antibody in order to increase or incorporate a therapeutic effect. It has been shown that homogeneity and controlled conjugation are crucial in these therapeutic applications. Here we present two novel protein domains developed from an IgG-binding domain of Streptococcal Protein G. These domains show obligate Fab binding and can be used for site-specific and covalent attachment exclusively to the constant part of the Fab fragment of an antibody. The two different domains can covalently label IgG of mouse and human descent. The labeled antibodies were shown to be functional in both an ELISA and in an NK-cell antibody-dependent cellular cytotoxicity assay. These engineered protein domains provide novel tools for controlled labeling of Fab fragments and full-length IgG.


Subject(s)
Bacterial Proteins/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin G/chemistry , Amino Acid Sequence , Animals , Humans , Immunoglobulin Fab Fragments/metabolism , Mice , Models, Molecular , Photochemical Processes , Protein Conformation, beta-Strand , Protein Domains , Staining and Labeling
18.
Blood ; 122(8): 1411-8, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23847195

ABSTRACT

Cytotoxic lymphocytes are important for immune responses against viral infections and cancer. They are able to kill target cells through the release of cytotoxic granules (CGs) without being harmed in the process. Because the lysosomal-associated membrane proteins (LAMPs) appear on the cell surface after CG exocytosis, we hypothesized that some of these proteins might be involved in transiently protecting cytotoxic lymphocytes from self-destruction. Intracellular expression of CD107a/LAMP-1, and to a lesser extent that of CD107b/LAMP-2, correlated with lymphocyte CG content. Engineered surface expression of CD107a/LAMP-1, but not of CD107b/LAMP-2, reduced the granule-mediated killing of transfected target cells. This was dependent on glycosylation of the CD107a/LAMP-1 hinge. Moreover, surface expression of CD107a/LAMP-1 reduced binding of perforin to cells. Importantly, knockdown of CD107a/LAMP-1 in primary human natural killer (NK) cells and deficiency of CD107a/LAMP-1 in mice resulted in increased NK cell apoptosis upon target cell-induced degranulation. Thus, our data support a novel role of CD107a/LAMP-1 in the protection of NK cells from degranulation-associated suicide, which may represent a general mechanism to transiently limit self-destruction by cytotoxic lymphocytes upon target cell killing.


Subject(s)
Cell Degranulation , Cell Membrane/metabolism , Killer Cells, Natural/cytology , Lymphocytes/cytology , Lysosomal-Associated Membrane Protein 1/metabolism , Animals , Apoptosis , Cell Line , Cytotoxicity, Immunologic , Genetic Engineering , Glycosylation , HeLa Cells , Humans , K562 Cells , Mice , Perforin/metabolism , RNA, Small Interfering/metabolism
19.
Blood ; 121(8): 1345-56, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23287865

ABSTRACT

Cytotoxic lymphocytes, encompassing cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, kill pathogen-infected, neoplastic, or certain hematopoietic cells through the release of perforin-containing lytic granules. In the present study, we first performed probability-state modeling of differentiation and lytic granule markers on CD8(+) T cells to enable the comparison of bona fide CTLs with NK cells. Analysis identified CD57(bright) expression as a reliable phenotype of granule marker-containing CTLs. We then compared CD3(+)CD8(+)CD57(bright) CTLs with NK cells. Healthy adult peripheral blood CD3(+)CD8(+)CD57(bright) CTLs expressed more granzyme B but less perforin than CD3(-)CD56(dim) NK cells. On stimulation, such CTLs degranulated more readily than other T-cell subsets, but had a propensity to degranulate that was similar to NK cells. Remarkably, the CTLs produced cytokines more rapidly and with greater frequency than NK cells. In patients with biallelic mutations in UNC13D, STX11, or STXBP2 associated with familial hemophagocytic lymphohistiocytosis, CTL and NK cell degranulation were similarly impaired. Therefore, cytotoxic lymphocyte subsets have similar requirements for Munc13-4, syntaxin-11, and Munc18-2 in lytic granule exocytosis. The present results provide a detailed comparison of human CD3(+)CD8(+)CD57(bright) CTLs and NK cells and suggest that analysis of CD57(bright) CTL function may prove useful in the diagnosis of primary immunodeficiencies including familial hemophagocytic lymphohistiocytosis.


Subject(s)
CD57 Antigens/metabolism , Cytokines/metabolism , Cytoplasmic Granules/metabolism , Immunologic Deficiency Syndromes/metabolism , Killer Cells, Natural/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Adult , Biomarkers/metabolism , CD3 Complex/metabolism , CD8 Antigens/metabolism , Cell Degranulation/immunology , Cytokines/biosynthesis , Cytoplasmic Granules/immunology , Exocytosis/immunology , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/immunology , Immunophenotyping , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Munc18 Proteins/metabolism , Perforin , Pore Forming Cytotoxic Proteins/metabolism , Qa-SNARE Proteins/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
20.
Pediatr Blood Cancer ; 62(12): 2094-100, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26184781

ABSTRACT

BACKGROUND: Perforin, encoded by PRF1, is a pore-forming protein crucial for lymphocyte cytotoxicity. Biallelic PRF1 nonsense mutations invariably result in early-onset hemophagocytic lymphohistiocytosis (HLH), termed familial HLH type 2 (FHL2). In contrast, biallelic PRF1 missense mutations may give rise to later-onset disease and more variable manifestations. PROCEDURE: We retrospectively searched our database for patients from families with siblings carrying biallelic PRF1 missense mutations where at least one sibling did not develop HLH, and for patients with biallelic PRF1 missense mutations and an atypical presentation of disease. We reviewed their clinical, genetic, and immunological characteristics. RESULTS: In all, we identified 10 such patients, including three sibling pairs with discordant manifestations. Interestingly, in two families, siblings of late-onset HLH patients developed Hodgkin lymphoma but no HLH. In a third family, one sibling presented with recurrent HLH episodes, whereas the other remains healthy. Of note, the affected sibling also suffered from systemic lupus erythematosus. Additional unrelated patients with biallelic PRF1 missense mutations were affected by neurological disease without classical signs of HLH, gastrointestinal inflammation as initial presentation of disease, as well as a hematological malignancy. Compared to early-onset FHL2 patients, the patients with an atypical presentation displayed a partial recovery of NK cell cytotoxicity upon IL-2 stimulation in vitro. CONCLUSIONS: Our findings substantiate and expand the spectrum of clinical presentations of perforin deficiency, linking PRF1 missense mutations to lymphoma susceptibility and highlighting clinical variability within families. PRF1 mutations should, therefore, be considered as a cause of several diseases disparate to HLH.


Subject(s)
Hodgkin Disease/genetics , Lupus Erythematosus, Systemic/genetics , Lymphohistiocytosis, Hemophagocytic/genetics , Mutation, Missense , Nervous System Diseases/genetics , Perforin/genetics , Adolescent , Adult , Female , Humans , Male , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL