Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 865773, 2022.
Article in English | MEDLINE | ID: mdl-35769478

ABSTRACT

GATA2 deficiency is a disease with a broad spectrum of clinical presentation, ranging from lymphedema, deafness, pulmonary dysfunction to miscarriage and urogenital anomalies, but it is mainly recognized as an immune system and bone marrow disorder. It is caused by various heterozygous mutations in the GATA2 gene, encoding for a zinc finger transcription factor with a key role for the development and maintenance of a pool of hematopoietic stem cells; notably, most of these mutations arise de novo. Patients carrying a mutated allele usually develop a loss of some cell populations, such as B-cell, dendritic cell, natural killer cell, and monocytes, and are predisposed to disseminated human papilloma virus and mycobacterial infections. Also, these patients have a predisposition to myeloid neoplasms, including myelodysplastic syndromes, myeloproliferative neoplasms, chronic myelomonocytic leukaemia. The age of symptoms onset can vary greatly even also within the same family, ranging from early childhood to late adulthood; incidence increases by age and most frequently clinical presentation is between the second and third decade of life. Currently, haematopoietic stem cell transplantation represents the only curative treatment, restoring both the hematopoietic and immune system function.


Subject(s)
GATA2 Deficiency , GATA2 Transcription Factor , Disease Susceptibility , GATA2 Deficiency/genetics , GATA2 Deficiency/immunology , GATA2 Transcription Factor/immunology , Humans , Immune System
3.
J Immunol ; 205(12): 3311-3318, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33188077

ABSTRACT

IL-13 plays a critical role in mediating many biological processes responsible for allergic inflammation. Mast cells express Il13 mRNA and produce IL-13 protein in response to antigenic stimulation. Enhancers are essential in promoting gene transcription and are thought to activate transcription by delivering essential accessory cofactors to the promoter to potentiate gene transcription. However, enhancers mediating Il13 have not been identified. Furthermore, which Il13 enhancers detect signals triggered by antigenic stimulation have not yet been defined. In this study, we identified potential mouse Il13 enhancers using histone modification monomethylation at lysine residue 4 on histone 3 (H3K4me1) chromatin immunoprecipitation sequencing and acetylation at lysine residue 27 on histone 3 (H3K27ac) chromatin immunoprecipitation sequencing. We used Omni-assay for transposase-accessible chromatin sequencing to determine which accessible regions within the potential Il13 enhancers that responded to IgE receptor crosslinking. We also demonstrated that the transcription factor cluster consisting of the NFATC2, STAT5, GATA2, AP1, and RUNX1 binding sites at the proximal Il13 enhancer and the transcription factor cluster consisting of the EGR2 binding site at the distal Il13 E+6.5 enhancer are critical in sensing the signals triggered by antigenic stimulation. Those enhancers, which are responsive to antigenic stimulation and are constitutively active, cooperate to generate greater transcriptional outputs. Our study reveals a novel mechanism underlying how antigenic stimulation induces robust Il13 mRNA expression in mouse mast cells.


Subject(s)
Antigens/immunology , Core Binding Factor Alpha 2 Subunit/immunology , Early Growth Response Protein 2/immunology , GATA2 Transcription Factor/immunology , Interleukin-13/immunology , Mast Cells/immunology , NFATC Transcription Factors/immunology , Response Elements/immunology , STAT5 Transcription Factor/immunology , Transcription Factor AP-1/immunology , Transcription, Genetic/immunology , Animals , Cell Line , Mast Cells/cytology , Mice
4.
Immunol Cell Biol ; 97(2): 142-151, 2019 02.
Article in English | MEDLINE | ID: mdl-30221399

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) represent a group of immature myeloid cells composed of myeloid progenitor cells and immature myeloid cells that can negatively regulate immune responses by inhibiting T-cell function. In mice, MDSCs are broadly defined by the expression of CD11b and Gr1. We and others have shown that injection of a lethal or sublethal dose of lipopolysaccharide (LPS) into mice could result in the expansion of MDSCs in the bone marrow (BM), spleen and blood. Until now, the molecular mechanisms responsible for this expansion are poorly studied; specifically, the roles of the individual microRNAs (miRNAs) which may be involved remain largely unknown. We performed microarray analysis to compare the miRNA expression profiles of CD11b+ Gr1+ cells sorted from the BM of LPS-injected and phosphate-buffered saline-injected mice. We identified let-7e, which was highly upregulated in the LPS-treated group, as a potent regulator of LPS-induced MDSC expansion. Furthermore, let-7e overexpression in BM chimeric mice led to a noticeable increase in the population of CD11b+ Gr1+ cells, which resulted from reduced cellular apoptosis. Further studies showed that let-7e could directly target caspase-3 to inhibit cell apoptosis, and upregulation of let-7e in LPS-stimulated MDSCs could be due to the relieved repression of let-7e transcription exerted by downregulated GATA2. Our findings suggest that LPS expands MDSCs by inhibiting apoptosis through the regulation of the GATA2/let-7e axis.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , GATA2 Transcription Factor/immunology , GATA2 Transcription Factor/metabolism , MicroRNAs/immunology , MicroRNAs/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Animals , Apoptosis/drug effects , Bone Marrow/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Mice , Myeloid-Derived Suppressor Cells/immunology
5.
Front Immunol ; 9: 2766, 2018.
Article in English | MEDLINE | ID: mdl-30564229

ABSTRACT

Two unrelated patients with GATA2-haploinsufficiency developed a hemophagocytic lymphohistiocytosis (HLH)-like disease during a varicella zoster virus (VZV) infection. High copy numbers of VZV were detected in the blood, and the patients were successfully treated with acyclovir and intravenous immunoglobulins. After treatment with corticosteroids for the HLH, both patients made a full recovery. Although the mechanisms leading to this disease constellation have yet to be characterized, we hypothesize that impairment of the immunoregulatory role of NK cells in GATA2-haploinsufficiency may have accentuated the patients' susceptibility to HLH. Expansion of a double negative T-lymphocytic population identified with CyTOF could be a further factor contributing to HLH in these patients. This is the first report of VZV-triggered HLH-like disease in a primary immunodeficiency and the third report of HLH in GATA2-haploinsufficiency. Since HLH was part of the presentation in one of our patients, GATA2-haploinsufficiency represents a potential differential diagnosis in patients presenting with the clinical features of HLH-especially in cases of persisting cytopenia after recovery from HLH.


Subject(s)
GATA2 Deficiency/immunology , GATA2 Transcription Factor/immunology , Herpesvirus 3, Human/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , T-Lymphocytes/immunology , Varicella Zoster Virus Infection/immunology , Child , Female , GATA2 Deficiency/virology , Humans , Lymphohistiocytosis, Hemophagocytic/virology , Male , Pilot Projects , T-Lymphocytes/virology , Varicella Zoster Virus Infection/virology
6.
J Cell Mol Med ; 22(3): 1464-1474, 2018 03.
Article in English | MEDLINE | ID: mdl-28994199

ABSTRACT

The MEK/ERK pathway is found to be important in regulating different biological processes such as proliferation, differentiation and survival in a wide variety of cells. However, its role in self-renewal of haematopoietic stem cells is controversial and remains to be clarified. The aim of this study was to understand the role of MEK/ERK pathway in ex vivo expansion of mononuclear cells (MNCs) and purified CD34+ cells, both derived from human umbilical cord blood (hUCB). Based on our results, culturing the cells in the presence of an inhibitor of MEK/ERK pathway-PD0325901 (PD)-significantly reduces the expansion of CD34+ and CD34+  CD38- cells, while there is no change in the expression of stemness-related genes (HOXB4, BMI1). Moreover, in vivo analysis demonstrates that PD reduces engraftment capacity of ex vivo expanded CD34+ cells. Notably, when ERK pathway is blocked in UCB-MNCs, spontaneous erythroid differentiation is promoted, found in concomitant with increasing number of burst-forming unit-erythroid colony (BFU-E) as well as enhancement of erythroid glycophorin-A marker. These results are in total conformity with up-regulation of some erythroid enhancer genes (TAL1, GATA2, LMO2) and down-regulation of some erythroid repressor genes (JUN, PU1) as well. Taken together, our results support the idea that MEK/ERK pathway has a critical role in achieving the correct balance between self-renewal and differentiation of UCB cells. Also, we suggest that inhibition of ERK signalling could likely be a new key for erythroid induction of UCB-haematopoietic progenitor cells.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Erythroid Cells/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , MAP Kinase Signaling System/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Animals, Newborn , Antigens, CD/genetics , Antigens, CD/immunology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Diphenylamine/pharmacology , Erythroid Cells/cytology , Erythroid Cells/immunology , Female , Fetal Blood/cytology , Fetal Blood/immunology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , Gene Expression Regulation , Glycophorins/genetics , Glycophorins/immunology , Graft Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , LIM Domain Proteins/genetics , LIM Domain Proteins/immunology , Mice , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/immunology , Transplantation, Heterologous
7.
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
8.
Int J Infect Dis ; 54: 89-91, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27894982

ABSTRACT

GATA2 deficiency is a genetic disorder caused by inherited or sporadic haploinsufficient mutations in the GATA2 gene. Patients have abnormalities in hematopoiesis, lymphangiogenesis and immunity; encompassing a broad range of clinical syndromes, mainly characterized by monocytopenia, B and NK cell cytopenia, severe or recurrent infections, and a high risk of developing myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). We report a case of an Argentinean woman who presented with multiple opportunistic infections as her first manifestation of GATA2 deficiency.


Subject(s)
GATA2 Transcription Factor/genetics , Mutation , Opportunistic Infections/genetics , Female , GATA2 Transcription Factor/immunology , Humans , Killer Cells, Natural/immunology , Opportunistic Infections/complications , Opportunistic Infections/immunology , Virus Diseases/etiology , Virus Diseases/virology , Young Adult
9.
Hematology Am Soc Hematol Educ Program ; 2016(1): 598-604, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27913534

ABSTRACT

Myelodysplastic syndrome (MDS) and myeloproliferative disorders are rare in children; they are divided into low-grade MDS (refractory cytopenia of childhood [RCC]), advanced MDS (refractory anemia with excess blasts in transformation), and juvenile myelomonocytic leukemia (JMML), each with different characteristics and management strategies. Underlying genetic predisposition is recognized in an increasing number of patients. Germ line GATA2 mutation is found in 70% of adolescents with MDS and monosomy 7. It is challenging to distinguish RCC from aplastic anemia, inherited bone marrow failure, and reactive conditions. RCC is often hypoplastic and may respond to immunosuppressive therapy. In case of immunosuppressive therapy failure, hypercellular RCC, or RCC with monosomy 7, hematopoietic stem cell transplantation (HSCT) using reduced-intensity conditioning regimens is indicated. Almost all patients with refractory anemia with excess blasts are candidates for HSCT; children age 12 years or older have a higher risk of treatment-related death, and the conditioning regimens should be adjusted accordingly. Unraveling the genetics of JMML has demonstrated that JMML in patients with germ line PTPN11 and CBL mutations often regresses spontaneously, and therapy is seldom indicated. Conversely, patients with JMML and neurofibromatosis type 1, somatic PTPN11, KRAS, and most of those with NRAS mutations have a rapidly progressive disease, and early HSCT is indicated. The risk of relapse after HSCT is high, and prophylaxis for graft-versus-host disease and monitoring should be adapted to this risk.


Subject(s)
Anemia, Aplastic , Anemia, Refractory, with Excess of Blasts , Leukemia, Myelomonocytic, Juvenile , Adolescent , Anemia, Aplastic/diagnosis , Anemia, Aplastic/genetics , Anemia, Aplastic/immunology , Anemia, Aplastic/therapy , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/immunology , Anemia, Refractory, with Excess of Blasts/therapy , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/immunology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Humans , Immunosuppression Therapy/methods , Infant , Leukemia, Myelomonocytic, Juvenile/diagnosis , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/immunology , Leukemia, Myelomonocytic, Juvenile/therapy , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology
10.
Blood ; 128(4): 508-18, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27259979

ABSTRACT

Dendritic cells (DCs) are critical immune response regulators; however, the mechanism of DC differentiation is not fully understood. Heterozygous germ line GATA2 mutations induce GATA2-deficiency syndrome, characterized by monocytopenia, a predisposition to myelodysplasia/acute myeloid leukemia, and a profoundly reduced DC population, which is associated with increased susceptibility to viral infections, impaired phagocytosis, and decreased cytokine production. To define the role of GATA2 in DC differentiation and function, we studied Gata2 conditional knockout and haploinsufficient mice. Gata2 conditional deficiency significantly reduced the DC count, whereas Gata2 haploinsufficiency did not affect this population. GATA2 was required for the in vitro generation of DCs from Lin(-)Sca-1(+)Kit(+) cells, common myeloid-restricted progenitors, and common dendritic cell precursors, but not common lymphoid-restricted progenitors or granulocyte-macrophage progenitors, suggesting that GATA2 functions in the myeloid pathway of DC differentiation. Moreover, expression profiling demonstrated reduced expression of myeloid-related genes, including mafb, and increased expression of T-lymphocyte-related genes, including Gata3 and Tcf7, in Gata2-deficient DC progenitors. In addition, GATA2 was found to bind an enhancer element 190-kb downstream region of Gata3, and a reporter assay exhibited significantly reduced luciferase activity after adding this enhancer region to the Gata3 promoter, which was recovered by GATA sequence deletion within Gata3 +190. These results suggest that GATA2 plays an important role in cell-fate specification toward the myeloid vs T-lymphocyte lineage by regulating lineage-specific transcription factors in DC progenitors, thereby contributing to DC differentiation.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , GATA2 Transcription Factor/immunology , Animals , Cell Differentiation/genetics , Dendritic Cells/cytology , GATA2 Transcription Factor/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
11.
Blood ; 125(21): 3306-15, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25855601

ABSTRACT

GATA2 plays a crucial role for the mast cell fate decision. We herein demonstrate that GATA2 is also required for the maintenance of the cellular identity in committed mast cells derived from mouse bone marrow (BMMCs). The deletion of the GATA2 DNA binding domain (GATA2ΔCF) in BMMCs resulted in a loss of the mast cell phenotype and an increase in the number of CD11b- and/or Ly6G/C-positive cells. These cells showed the ability to differentiate into macrophage- and neutrophil-like cells but not into eosinophils. Although the mRNA levels of basophil-specific genes were elevated, CD49b, a representative basophil marker, never appeared on these cells. GATA2 ablation led to a significant upregulation of C/EBPα, and forced expression of C/EBPα in wild-type BMMCs phenocopied the GATA2ΔCF cells. Interestingly, simultaneous deletion of the Gata2 and Cebpa genes in BMMCs restored the aberrant increases of CD11b and Ly6G/C while retaining the reduced c-Kit expression. Chromatin immunoprecipitation assays indicated that GATA2 directly binds to the +37-kb region of the Cebpa gene and thereby inhibits the RUNX1 and PU.1 binding to the neighboring region. Upregulation of C/EBPα following the loss of GATA2 was not observed in cultured mast cells derived from peritoneal fluid, whereas the repression of c-Kit and other mast cell-specific genes were observed in these cells. Collectively, these results indicate that GATA2 maintains cellular identity by preventing Cebpa gene activation in a subpopulation of mast cells, whereas it plays a fundamental role as a positive regulator of mast cell-specific genes throughout development of this cell lineage.


Subject(s)
Bone Marrow Cells/cytology , Cell Dedifferentiation/immunology , GATA2 Transcription Factor/metabolism , Mast Cells/cytology , Stem Cells/cytology , Animals , Blotting, Western , Cell Differentiation/immunology , Chromatin Immunoprecipitation , Flow Cytometry , GATA2 Transcription Factor/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
12.
BMC Pulm Med ; 15: 8, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25879889

ABSTRACT

BACKGROUND: GATA-2 transcription factor deficiency has recently been described in patients with a propensity towards myeloid malignancy associated with other highly variable phenotypic features: chronic leukocytopenias (dendritic cell-, monocyto-, granulocyto-, lymphocytopenia), increased susceptibility to infections, lymphatic vasculature abnormalities, and sensorineural deafness. Patients often suffer from opportunistic respiratory infections; chronic pulmonary changes have been found in advanced disease. CASE PRESENTATION: We present a case of a 17-year-old previously healthy Caucasian male who was admitted to the hospital with fever, malaise, headache, cough and dyspnea. A chest X-ray revealed bilateral interstitial infiltrates and pneumonia was diagnosed. Despite prompt clinical improvement under antibiotic therapy, interstitial changes remained stable. A high resolution computer tomography showed severe diffuse parenchymal lung disease, while the patient's pulmonary function tests were normal and he was asymptomatic. Lung tissue biopsy revealed chronic reparative and resorptive reaction with organizing vasculitis. At the time of the initial presentation to the hospital, serological signs of acute infection with Epstein-Barr virus (EBV) were present; EBV viremia with atypical serological response persisted during two-year follow up. No other infectious agents were found. Marked monocytopenia combined with B-cell lymphopenia led to a suspicion of GATA-2 deficiency. Diagnosis was confirmed by detection of the previously published heterozygous mutation in GATA2 (c.1081 C > T, p.R361C). The patient's brother and father were both carriers of the same genetic defect. The brother had no clinically relevant ailments despite leukocyte changes similar to the index patient. The father suffered from spondylarthritis, and apart from B-cell lymphopenia, no other changes within the leukocyte pool were seen. CONCLUSION: We conclude that a diagnosis of GATA-2 deficiency should be considered in all patients with diffuse parenchymal lung disease presenting together with leukocytopenia, namely monocyto-, dendritic cell- and B-lymphopenia, irrespective of severity of the clinical phenotype. Genetic counseling and screening for GATA2 mutations within the patient's family should be provided as the phenotype is highly variable and carriers without apparent immunodeficiency are still in danger of developing myeloid malignancy. A prompt recognition of this rare condition helps to direct clinical treatment strategies and follow-up procedures.


Subject(s)
Epstein-Barr Virus Infections/genetics , GATA2 Transcription Factor/deficiency , Lung Diseases, Interstitial/genetics , Lung/pathology , Lymphopenia/genetics , Adolescent , B-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , Humans , Leukopenia/genetics , Leukopenia/immunology , Lung/diagnostic imaging , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/immunology , Lymphopenia/immunology , Male , Monocytes/immunology , Mutation , Radiography , Syndrome , Vasculitis/diagnosis , Vasculitis/genetics , Vasculitis/immunology
13.
Blood ; 125(2): 214-5, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25573968

ABSTRACT

In this issue of Blood, Sasaki and colleagues demonstrate a strict requirement for the transcription factor interferon regulator factor 8 (IRF8) in the development of mouse basophils.


Subject(s)
Basophils/cytology , Basophils/immunology , GATA2 Transcription Factor/immunology , Interferon Regulatory Factors/immunology , Mast Cells/cytology , Mast Cells/immunology , Animals
14.
J Immunol ; 194(5): 2190-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25624456

ABSTRACT

Germline GATA2 mutations have been identified as the cause of familial syndromes with immunodeficiency and predisposition to myeloid malignancies. GATA2 mutations appear to cause loss of function of the mutated allele leading to haploinsufficiency; however, this postulate has not been experimentally validated as the basis of these syndromes. We hypothesized that mutations that are translated into abnormal proteins could affect the transcription of GATA2, triggering GATA2 deficiency. Chromatin immunoprecipitation and luciferase assays showed that the human GATA2 protein activates its own transcription through a specific region located at -2.4 kb, whereas the p.Thr354Met, p.Thr355del, and p.Arg396Gln germline mutations impair GATA2 promoter activation. Accordingly, GATA2 expression was decreased to ∼58% in a patient with p.Arg396Gln, compared with controls. p.Arg396Gln is the second most common mutation in these syndromes, and no previous functional analyses have been performed. We therefore analyzed p.Arg396Gln. Our data show that p.Arg396Gln is a loss-of-function mutation affecting DNA-binding ability and, as a consequence, it fails to maintain the immature characteristics of hematopoietic stem and progenitor cells, which could result in defects in this cell compartment. In conclusion, we show that human GATA2 binds to its own promoter, activating its transcription, and that the aforementioned mutations impair the transcription of GATA2. Our results indicate that they can affect other GATA2 target genes, which could partially explain the variability of symptoms in these diseases. Moreover, we show that p.Arg396Gln is a loss-of-function mutation, which is unable to retain the progenitor phenotype in cells where it is expressed.


Subject(s)
GATA2 Transcription Factor/genetics , Germ-Line Mutation , Immunologic Deficiency Syndromes/immunology , Mycobacterium avium-intracellulare Infection/immunology , Transcription, Genetic , Alleles , Binding Sites , Cell Line, Tumor , Female , GATA2 Transcription Factor/immunology , Gene Expression Regulation , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Middle Aged , Models, Molecular , Mycobacterium avium Complex/immunology , Mycobacterium avium-intracellulare Infection/complications , Mycobacterium avium-intracellulare Infection/genetics , Mycobacterium avium-intracellulare Infection/pathology , Phenotype , Promoter Regions, Genetic , Protein Binding , Signal Transduction
15.
Dev Comp Immunol ; 49(1): 38-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25445909

ABSTRACT

Dendritic cells (DCs) are among the most important professional antigen-presenting cells (APCs) that participate in various biological activities in mammals. However, evidence of the existence of DCs in teleost fish and other lower vertebrates remains limited. In this study, phenotypic and functional characteristics of teleost DCs were described in a zebrafish model. An improved method to efficiently enrich DCs was established. Immunofluorescence staining revealed that the surface phenotypic hallmarks of mammalian DCs, including MHC-II, CD80/86, CD83, and CD209, were distributed on the surfaces of zebrafish DCs (DrDCs). Functional analysis results showed that DrDCs could initiate antigen-specific CD4(+) T cell activation, in which MHC-II, CD80/86, CD83, and CD209 are implicated. Hence, teleost DCs exhibit conserved immunophenotypes and functions similar to those of their mammalian counterparts. Our findings contributed to the current understanding of the evolutionary history of DCs and the DC-regulatory mechanisms of adaptive immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Cell Membrane/immunology , Dendritic Cells/immunology , Zebrafish/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Dendritic Cells/metabolism , Female , Flow Cytometry , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , GATA2 Transcription Factor/metabolism , Gene Expression/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunoglobulins/genetics , Immunoglobulins/immunology , Immunoglobulins/metabolism , Immunophenotyping , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish/genetics , Zebrafish/metabolism
16.
Blood ; 125(2): 358-69, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25398936

ABSTRACT

Basophils and mast cells play critical roles in host defense against pathogens and allergic disorders. However, the molecular mechanism by which these cells are generated is not completely understood. Here we demonstrate that interferon regulatory factor-8 (IRF8), a transcription factor essential for the development of several myeloid lineages, also regulates basophil and mast cell development. Irf8(-/-) mice displayed a severe reduction in basophil counts, which was accounted for by the absence of pre-basophil and mast cell progenitors (pre-BMPs). Although Irf8(-/-) mice retained peripheral tissue mast cells, remaining progenitors from Irf8(-/-) mice including granulocyte progenitors (GPs) were unable to efficiently generate either basophils or mast cells, indicating that IRF8 also contributes to the development of mast cells. IRF8 appeared to function at the GP stage, because IRF8 was expressed in GPs, but not in basophils, mast cells, and basophil/mast cell-restricted progenitor cells. Furthermore, we demonstrate that GATA2, a transcription factor known to promote basophil and mast cell differentiation, acts downstream of IRF8. These results shed light on the pathways and mechanism underlying the development of basophils and mast cells.


Subject(s)
Basophils/cytology , Basophils/immunology , GATA2 Transcription Factor/immunology , Interferon Regulatory Factors/immunology , Mast Cells/cytology , Mast Cells/immunology , Animals , Basophils/metabolism , Cell Differentiation/immunology , GATA2 Transcription Factor/metabolism , Interferon Regulatory Factors/metabolism , Mast Cells/metabolism , Mice , Mice, Knockout , Stem Cells/immunology , Stem Cells/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
18.
Curr Opin Allergy Clin Immunol ; 13(6): 589-95, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24135998

ABSTRACT

PURPOSE OF REVIEW: Inborn errors of human natural killer (NK) cells may affect the development of these cells, their function, or both. There are two broad categories of genetic defects of NK cell development, depending on whether the deficiency is apparently specific to NK cells or clearly affects multiple hematopoietic lineages. We review here recent progress in the genetic dissection of these NK deficiencies (NKDs). RECENT FINDINGS: Patients with severe combined immunodeficiencies bearing mutations of adenosine deaminase, adenylate kinase 2, interleukin-2 receptor gamma chain, and Janus kinase 3 genes present NKDs and are prone to a broad range of infections. Patients with GATA binding protein 2 deficiency are susceptible to both mycobacterial and viral infections, and display NKDs and a lack of monocytes. Rare patients with mini chromosomal maintenance 4 deficiency display an apparently selective NKD associated with viral infections, but they also display various nonhematopoietic phenotypes, including adrenal insufficiency and growth retardation. SUMMARY: These studies have initiated a genetic dissection of the development of human NK cells. Further studies are warranted, including the search for genetic causes of NKD in particular. This research may lead to the discovery of molecules specifically controlling the development of NK cells and to improvements in our understanding of the hitherto elusive function of these cells in humans.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Adenylate Kinase/genetics , Adenylate Kinase/immunology , Agammaglobulinemia/pathology , Agammaglobulinemia/physiopathology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , Humans , Janus Kinase 2 , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Killer Cells, Natural , Minichromosome Maintenance Complex Component 4/genetics , Minichromosome Maintenance Complex Component 4/immunology , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Mycobacterium Infections/pathology , Mycobacterium Infections/physiopathology , Severe Combined Immunodeficiency/pathology , Severe Combined Immunodeficiency/physiopathology , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/pathology , Virus Diseases/physiopathology
19.
Blood ; 121(14): 2669-77, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23365458

ABSTRACT

Mutations in the transcription factor GATA2 underlie the syndrome of monocytopenia and B- and natural killer (NK)-cell lymphopenia associated with opportunistic infections and cancers. In addition, patients have recurrent and severe viral infections. NK cells play a critical role in mediating antiviral immunity. Human NK cells are thought to mature in a linear fashion, with the CD56(bright) stage preceding terminal maturation to the CD56(dim) stage, considered the most enabled for cytotoxicity. Here we report an NK cell functional defect in GATA2-deficient patients and extend this genetic lesion to what is considered to be the original NK cell-deficient patient. In most cases, GATA2 deficiency is accompanied by a severe reduction in peripheral blood NK cells and marked functional impairment. The NK cells detected in peripheral blood of some GATA2-deficient patients are exclusively of the CD56(dim) subset, which is recapitulated on in vitro NK cell differentiation. In vivo, interferon α treatment increased NK cell number and partially restored function but did not correct the paucity of CD56(bright) cells. Thus, GATA2 is required for the maturation of human NK cells and the maintenance of the CD56(bright) pool in the periphery. Defects in GATA2 are a novel cause of profound NK cell dysfunction.


Subject(s)
CD56 Antigen/immunology , Cell Differentiation/immunology , GATA2 Transcription Factor/genetics , Killer Cells, Natural/immunology , Lymphopenia/genetics , Antigens, CD34/metabolism , CD56 Antigen/metabolism , Cytotoxicity, Immunologic/immunology , GATA2 Transcription Factor/immunology , GATA2 Transcription Factor/metabolism , Humans , Immunophenotyping , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphopenia/immunology , Lymphopenia/metabolism , Stromal Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...