Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Blood Adv ; 7(10): 2171-2176, 2023 05 23.
Article in English | MEDLINE | ID: mdl-36112425

ABSTRACT

Most children with high-risk Langerhans cell histiocytosis (LCH) have BRAFV600E mutation. BRAFV600E alleles are detectable in myeloid mononuclear cells at diagnosis but it is not known if the cellular distribution of mutation evolves over time. Here, the profiles of 16 patients with high-risk disease were analyzed. Two received conventional salvage chemotherapy, 4 patients on inhibitors were tracked at intervals of 3 to 6 years, and 10 patients, also given inhibitors, were analyzed more than 2 years after diagnosis. In contrast to the patients responding to salvage chemotherapy who completely cleared BRAFV600E within 6 months, children who received inhibitors maintained high BRAFV600E alleles in their blood. At diagnosis, mutation was detected predominantly in monocytes and myeloid dendritic cells. With time, mutation switched to the T-cell compartment, which accounted for most of the mutational burden in peripheral blood mononuclear cells, more than 2 years from diagnosis (median, 85.4%; range, 44.5%-100%). The highest level of mutation occurred in naïve CD4+ T cells (median, 51.2%; range, 3.8%-93.5%). This study reveals an unexpected lineage switch of BRAFV600E mutation in high-risk LCH, which may influence monitoring strategies for the potential withdrawal of inhibitor treatment and has new implications for the pathogenesis of neurodegeneration, which occurred in 4 patients.


Subject(s)
Dendritic Cells , Histiocytosis, Langerhans-Cell , Monocytes , T-Lymphocytes , Humans , Dendritic Cells/pathology , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Leukocytes, Mononuclear , Monocytes/pathology , Mutation , Male , Female , Infant , Child, Preschool , T-Lymphocytes/pathology , Cell Lineage/genetics
2.
Immunity ; 53(2): 353-370.e8, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32735845

ABSTRACT

The formation of mammalian dendritic cells (DCs) is controlled by multiple hematopoietic transcription factors, including IRF8. Loss of IRF8 exerts a differential effect on DC subsets, including plasmacytoid DCs (pDCs) and the classical DC lineages cDC1 and cDC2. In humans, cDC2-related subsets have been described including AXL+SIGLEC6+ pre-DC, DC2 and DC3. The origin of this heterogeneity is unknown. Using high-dimensional analysis, in vitro differentiation, and an allelic series of human IRF8 deficiency, we demonstrated that cDC2 (CD1c+DC) heterogeneity originates from two distinct pathways of development. The lymphoid-primed IRF8hi pathway, marked by CD123 and BTLA, carried pDC, cDC1, and DC2 trajectories, while the common myeloid IRF8lo pathway, expressing SIRPA, formed DC3s and monocytes. We traced distinct trajectories through the granulocyte-macrophage progenitor (GMP) compartment showing that AXL+SIGLEC6+ pre-DCs mapped exclusively to the DC2 pathway. In keeping with their lower requirement for IRF8, DC3s expand to replace DC2s in human partial IRF8 deficiency.


Subject(s)
Antigens, CD34/metabolism , Dendritic Cells/cytology , Hematopoiesis/physiology , Interferon Regulatory Factors/metabolism , Animals , Antigens, CD1/metabolism , Cell Line , Cell Lineage/immunology , Dendritic Cells/immunology , Glycoproteins/metabolism , Hematopoietic Stem Cells/cytology , Humans , Interleukin-3 Receptor alpha Subunit/metabolism , Lipopolysaccharide Receptors/metabolism , Mice , Receptors, Immunologic/metabolism
3.
J Clin Invest ; 130(9): 4574-4586, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32453711

ABSTRACT

Myelopoiesis is invariably present and contributes to pathology in animal models of graft-versus-host disease (GVHD). In humans, a rich inflammatory infiltrate bearing macrophage markers has also been described in histological studies. In order to determine the origin, functional properties, and role in pathogenesis of these cells, we isolated single-cell suspensions from acute cutaneous GVHD and subjected them to genotype, transcriptome, and in vitro functional analysis. A donor-derived population of CD11c+CD14+ cells was the dominant population of all leukocytes in GVHD. Surface phenotype and NanoString gene expression profiling indicated the closest steady-state counterpart of these cells to be monocyte-derived macrophages. In GVHD, however, there was upregulation of monocyte antigens SIRPα and S100A8/9 transcripts associated with leukocyte trafficking, pattern recognition, antigen presentation, and costimulation. Isolated GVHD macrophages stimulated greater proliferation and activation of allogeneic T cells and secreted higher levels of inflammatory cytokines than their steady-state counterparts. In HLA-matched mixed leukocyte reactions, we also observed differentiation of activated macrophages with a similar phenotype. These exhibited cytopathicity to a keratinocyte cell line and mediated pathological damage to skin explants independently of T cells. Together, these results define the origin, functional properties, and potential pathogenic roles of human GVHD macrophages.


Subject(s)
Gene Expression Regulation/immunology , Graft vs Host Disease/immunology , Macrophages/immunology , Monocytes/immunology , Skin Diseases/immunology , Tissue Donors , Graft vs Host Disease/pathology , Humans , Macrophages/pathology , Monocytes/pathology , Skin Diseases/pathology
4.
Blood Adv ; 3(20): 3052-3061, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31648336

ABSTRACT

Fms-like tyrosine kinase 3 (Flt3) is expressed on progenitor cells and acute myeloid leukemia (AML) blasts. Fms-like tyrosine kinase 3 ligand (Flt3L) is detectable during homeostasis and increases in hypoplasia due to genetic defects or treatment with cytoreductive agents. Conversely, Flt3+ AML is associated with depletion of Flt3L to undetectable levels. After induction chemotherapy, Flt3L is restored in patients entering complete remission (CR) but remains depressed in those with refractory disease. Weekly sampling reveals marked differences in the kinetics of Flt3L response during the first 6 weeks of treatment, proportionate to the clearance of blasts and cellularity of the bone marrow. In the UK NCRI AML17 trial, Flt3L was measured at day 26 in a subgroup of 140 patients with Flt3 mutation randomized to the tyrosine kinase inhibitor lestaurtinib or placebo. In these patients, attainment of CR was associated with higher Flt3L at day 26 (Mann-Whitney UP < .0001). Day 26 Flt3L was also associated with survival; Flt3L ≤291 pg/mL was associated with inferior event-free survival (EFS), and Flt3L >1185 pg/mL was associated with higher overall survival (OS; P = .0119). The separation of EFS and OS curves increased when minimal residual disease (MRD) status was combined with Flt3L measurement, and Flt3L retained a near-significant association with survival after adjusting for MRD in a proportional hazards model. Serial measurement of Flt3L in patients who had received a hematopoietic stem cell transplant for AML illustrates the potential value of monitoring Flt3L to identify relapse. Measurement of Flt3L is a noninvasive test with the potential to inform clinical decisions in patients with AML.


Subject(s)
Biomarkers , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/diagnosis , Membrane Proteins/blood , Neoplastic Stem Cells/metabolism , Gene Expression , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Induction Chemotherapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Prognosis , Proportional Hazards Models , Treatment Outcome
5.
EBioMedicine ; 14: 65-73, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27913155

ABSTRACT

High-risk primary biliary cholangitis (PBC), defined by inadequate response at one year to Ursodeoxycholic acid (UDCA), is associated with disease progression and liver transplantation. Stratifying high-risk patients early would facilitate improved approaches to care. Using long-term follow-up data to define risk at presentation, 6 high-risk PBC patients and 8 low-risk patients were identified from biopsy, transplant and biochemical archival records. Formalin-fixed paraffin-embedded (FFPE) liver biopsies taken at presentation were graded (Scheuer and Nakanuma scoring) and gene expression analysed using the NanoString® nCounter PanCancer Immunity 770-gene panel. Principle component analysis (PCA) demonstrated discrete gene expression clustering between controls and high- and low-risk PBC. High-risk PBC was characterised by up-regulation of genes linked to T-cell activation and apoptosis, INF-γ signalling and leukocyte migration and down-regulation of those linked to the complement pathway. CDKN1a, up-regulated in high-risk PBC, correlated with significantly increased expression of its gene product, the senescence marker p21WAF1/Cip, by biliary epithelial cells. Our findings suggest high- and low-risk PBC are biologically different from disease outset and senescence an early feature in high-risk disease. Identification of a high-risk 'signal' early from standard FFPE tissue sections has clear clinical utility allowing for patient stratification and second-line therapeutic intervention.


Subject(s)
Cholangitis/genetics , Cholangitis/pathology , Gene Expression Profiling , Transcriptome , Adult , Aged , Biomarkers , Biopsy , Cholangitis/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
J Leukoc Biol ; 97(4): 627-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25516751

ABSTRACT

Langerin is a C-type lectin expressed at high level by LCs of the epidermis. Langerin is also expressed by CD8(+)/CD103(+) XCR1(+) cross-presenting DCs of mice but is not found on the homologous human CD141(high) XCR1(+) myeloid DC. Here, we show that langerin is expressed at a low level on DCs isolated from dermis, lung, liver, and lymphoid tissue and that langerin(+) DCs are closely related to CD1c(+) myeloid DCs. They are distinguishable from LCs by the level of expression of CD1a, EpCAM, CD11b, CD11c, CD13, and CD33 and are found in tissues and tissue-draining LNs devoid of LCs. They are unrelated to CD141(high) XCR1(+) myeloid DCs, lacking the characteristic expression profile of cross-presenting DCs, conserved between mammalian species. Stem cell transplantation and DC deficiency models confirm that dermal langerin(+) DCs have an independent homeostasis to LCs. Langerin is not expressed by freshly isolated CD1c(+) blood DCs but is rapidly induced on CD1c(+) DCs by serum or TGF-ß via an ALK-3-dependent pathway. These results show that langerin is expressed outside of the LC compartment of humans and highlight a species difference: langerin is expressed by the XCR1(+) "DC1" population of mice but is restricted to the CD1c(+) "DC2" population of humans (homologous to CD11b(+) DCs in the mouse).


Subject(s)
Antigens, CD1/analysis , Antigens, CD/analysis , Dendritic Cells/classification , Glycoproteins/analysis , Lectins, C-Type/analysis , Mannose-Binding Lectins/analysis , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation/analysis , Antigens, Surface/analysis , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/physiology , Dendritic Cells/chemistry , Dendritic Cells/drug effects , Gene Expression Profiling , Homeostasis , Humans , Langerhans Cells/classification , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Liver/cytology , Lung/cytology , Lymphoid Tissue/cytology , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/genetics , Mice , Organ Specificity , Receptors, G-Protein-Coupled/analysis , Serum , Skin/cytology , Thrombomodulin , Transforming Growth Factor beta/pharmacology
8.
Immunity ; 41(3): 465-477, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25200712

ABSTRACT

Dendritic cells (DCs), monocytes, and macrophages are leukocytes with critical roles in immunity and tolerance. The DC network is evolutionarily conserved; the homologs of human tissue CD141(hi)XCR1⁺ CLEC9A⁺ DCs and CD1c⁺ DCs are murine CD103⁺ DCs and CD64⁻ CD11b⁺ DCs. In addition, human tissues also contain CD14⁺ cells, currently designated as DCs, with an as-yet unknown murine counterpart. Here we have demonstrated that human dermal CD14⁺ cells are a tissue-resident population of monocyte-derived macrophages with a short half-life of <6 days. The decline and reconstitution kinetics of human blood CD14⁺ monocytes and dermal CD14⁺ cells in vivo supported their precursor-progeny relationship. The murine homologs of human dermal CD14⁺ cells are CD11b⁺ CD64⁺ monocyte-derived macrophages. Human and mouse monocytes and macrophages were defined by highly conserved gene transcripts, which were distinct from DCs. The demonstration of monocyte-derived macrophages in the steady state in human tissue supports a conserved organization of human and mouse mononuclear phagocyte system.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Skin/immunology , Animals , CD11b Antigen/biosynthesis , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Humans , Immunologic Memory/immunology , Mice , Mice, Transgenic , Receptors, IgG/biosynthesis , Skin/cytology , T-Lymphocytes/immunology
9.
Blood ; 123(6): 863-74, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24345756

ABSTRACT

Constitutive heterozygous GATA2 mutation is associated with deafness, lymphedema, mononuclear cytopenias, infection, myelodysplasia (MDS), and acute myeloid leukemia. In this study, we describe a cross-sectional analysis of 24 patients and 6 relatives with 14 different frameshift or substitution mutations of GATA2. A pattern of dendritic cell, monocyte, B, and natural killer (NK) lymphoid deficiency (DCML deficiency) with elevated Fms-like tyrosine kinase 3 ligand (Flt3L) was observed in all 20 patients phenotyped, including patients with Emberger syndrome, monocytopenia with Mycobacterium avium complex (MonoMAC), and MDS. Four unaffected relatives had a normal phenotype indicating that cellular deficiency may evolve over time or is incompletely penetrant, while 2 developed subclinical cytopenias or elevated Flt3L. Patients with GATA2 mutation maintained higher hemoglobin, neutrophils, and platelets and were younger than controls with acquired MDS and wild-type GATA2. Frameshift mutations were associated with earlier age of clinical presentation than substitution mutations. Elevated Flt3L, loss of bone marrow progenitors, and clonal myelopoiesis were early signs of disease evolution. Clinical progression was associated with increasingly elevated Flt3L, depletion of transitional B cells, CD56(bright) NK cells, naïve T cells, and accumulation of terminally differentiated NK and CD8(+) memory T cells. These studies provide a framework for clinical and laboratory monitoring of patients with GATA2 mutation and may inform therapeutic decision-making.


Subject(s)
B-Lymphocytes/pathology , Dendritic Cells/pathology , GATA2 Transcription Factor/genetics , Killer Cells, Natural/pathology , Monocytes/pathology , Mutation/genetics , Myelodysplastic Syndromes/pathology , Adolescent , Adult , Aged , Aged, 80 and over , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Case-Control Studies , Child , Child, Preschool , Clonal Evolution , Cross-Sectional Studies , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Genetic Association Studies , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Pedigree , Prognosis , Young Adult , fms-Like Tyrosine Kinase 3/metabolism
10.
Leuk Lymphoma ; 54(1): 167-73, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22742576

ABSTRACT

Natural killer (NK) cell immunosurveillance may be impaired by malignant disease, resulting in tumor escape and disease progression. Therapies that enhance NK cytotoxicity may therefore prove valuable in remission-induction and maintenance treatment regimens. Acute lymphoblastic leukemia (ALL) has previously been considered resistant to NK cell lysis and not tractable to this approach. Our study demonstrates that bortezomib, valproate and troglitazone can up-regulate NK activating ligands on a B-ALL cell line and on a proportion but not all adult primary B-ALL samples. Drug-treated ALL cells trigger higher levels of NK degranulation, as measured by CD107a expression, and this effect is dependent on signaling through the NK activating receptor NKG2D. These results suggest that bortezomib, valproate and troglitazone may have clinical utility in sensitizing ALL to NK mediated lysis in vivo.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Boronic Acids/pharmacology , Bortezomib , Cell Degranulation/drug effects , Cell Line, Tumor , Chromans/pharmacology , Humans , Ligands , Pyrazines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Thiazolidinediones/pharmacology , Troglitazone , Up-Regulation/drug effects , Valproic Acid/pharmacology
11.
Front Immunol ; 4: 495, 2013.
Article in English | MEDLINE | ID: mdl-24416034

ABSTRACT

Dendritic cells (DCs) and monocytes are critical regulators and effectors of innate and adaptive immune responses. Monocyte expansion has been described in many pathological states while monocyte and DC deficiency syndromes are relatively recent additions to the catalog of human primary immunodeficiency disorders. Clinically applicable screening tests to diagnose and monitor these conditions are lacking. Conventional strategies for identifying human DCs and monocytes have been based on the use of a lineage gate to exclude lymphocytes, thus preventing simultaneous detection of DCs, monocytes, and lymphocyte subsets. Here we demonstrate that CD4 is a reliable lineage marker for the human peripheral blood antigen-presenting cell compartment that can be used to identify DCs and monocytes in parallel with lymphocytes. Based on this principle, simple modification of a standard lymphocyte phenotyping assay permits simultaneous enumeration of four lymphocyte and five DC/monocyte populations from a single sample. This approach is applicable to clinical samples and facilitates the diagnosis of DC and monocyte disorders in a wide range of clinical settings, including genetic deficiency, neoplasia, and inflammation.

12.
Immunity ; 37(1): 60-73, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22795876

ABSTRACT

Dendritic cell (DC)-mediated cross-presentation of exogenous antigens acquired in the periphery is critical for the initiation of CD8(+) T cell responses. Several DC subsets are described in human tissues but migratory cross-presenting DCs have not been isolated, despite their potential importance in immunity to pathogens, vaccines, and tumors and tolerance to self. Here, we identified a CD141(hi) DC present in human interstitial dermis, liver, and lung that was distinct from the majority of CD1c(+) and CD14(+) tissue DCs and superior at cross-presenting soluble antigens. Cutaneous CD141(hi) DCs were closely related to blood CD141(+) DCs, and migratory counterparts were found among skin-draining lymph node DCs. Comparative transcriptomic analysis with mouse showed tissue DC subsets to be conserved between species and permitted close alignment of human and mouse DC subsets. These studies inform the rational design of targeted immunotherapies and facilitate translation of mouse functional DC biology to the human setting.


Subject(s)
Antigens, CD/metabolism , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Animals , Antigens/immunology , Cell Movement/immunology , Chemokine CXCL10/biosynthesis , Gene Expression Profiling , Humans , Immunophenotyping , Langerhans Cells/immunology , Langerhans Cells/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Skin/immunology , Transcriptome , Tumor Necrosis Factor-alpha/biosynthesis
13.
Blood ; 118(10): 2656-8, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21765025

ABSTRACT

The human syndrome of dendritic cell, monocyte, B and natural killer lymphoid deficiency presents as a sporadic or autosomal dominant trait causing susceptibility to mycobacterial and other infections, predisposition to myelodysplasia and leukemia, and, in some cases, pulmonary alveolar proteinosis. Seeking a genetic cause, we sequenced the exomes of 4 unrelated persons, 3 with sporadic disease, looking for novel, heterozygous, and probably deleterious variants. A number of genes harbored novel variants in person, but only one gene, GATA2, was mutated in all 4 persons. Each person harbored a different mutation, but all were predicted to be highly deleterious and to cause loss or mutation of the C-terminal zinc finger domain. Because GATA2 is the only common mutated gene in 4 unrelated persons, it is highly probable to be the cause of dendritic cell, monocyte, B, and natural killer lymphoid deficiency. This disorder therefore constitutes a new genetic form of heritable immunodeficiency and leukemic transformation.


Subject(s)
B-Lymphocytes/pathology , Dendritic Cells/pathology , Disease Susceptibility/etiology , Exons/genetics , GATA2 Transcription Factor/genetics , Killer Cells, Natural/pathology , Lymphoid Tissue/pathology , Monocytes/pathology , Mutation/genetics , GATA2 Transcription Factor/chemistry , Humans , Protein Conformation
14.
J Exp Med ; 208(2): 227-34, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21242295

ABSTRACT

Congenital or acquired cellular deficiencies in humans have the potential to reveal much about normal hematopoiesis and immune function. We show that a recently described syndrome of monocytopenia, B and NK lymphoid deficiency additionally includes the near absence of dendritic cells. Four subjects showed severe depletion of the peripheral blood HLA-DR(+) lineage(-) compartment, with virtually no CD123(+) or CD11c(+) dendritic cells (DCs) and very few CD14(+) or CD16(+) monocytes. The only remaining HLA-DR(+) lineage(-) cells were circulating CD34(+) progenitor cells. Dermal CD14(+) and CD1a(+) DC were also absent, consistent with their dependence on blood-derived precursors. In contrast, epidermal Langerhans cells and tissue macrophages were largely preserved. Combined loss of peripheral DCs, monocytes, and B and NK lymphocytes was mirrored in the bone marrow by complete absence of multilymphoid progenitors and depletion of granulocyte-macrophage progenitors. Depletion of the HLA-DR(+) peripheral blood compartment was associated with elevated serum fms-like tyrosine kinase ligand and reduced circulating CD4(+)CD25(hi)FoxP3(+) T cells, supporting a role for DC in T reg cell homeostasis.


Subject(s)
Dendritic Cells/cytology , Disease Susceptibility/etiology , HLA-DR Antigens/blood , Leukopenia/genetics , Monocytes/cytology , Adult , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Child , Dendritic Cells/pathology , Disease Susceptibility/microbiology , Disease Susceptibility/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-gamma/blood , Leukopenia/blood , Leukopenia/complications , Microscopy, Fluorescence , Monocytes/pathology , Mycobacterium Infections/immunology , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...