Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
J Leukoc Biol ; 107(2): 323-339, 2020 02.
Article in English | MEDLINE | ID: mdl-31749181

ABSTRACT

Myeloid lineage cells present in human peripheral blood include dendritic cells (DC) and monocytes. The DC are identified phenotypically as HLA-DR+ cells that lack major cell surface lineage markers for T cells (CD3), B cells (CD19, CD20), NK cells (CD56), red blood cells (CD235a), hematopoietic stem cells (CD34), and Mo that express CD14. Both DC and Mo can be phenotypically divided into subsets. DC are divided into plasmacytoid DC, which are CD11c- , CD304+ , CD85g+ , and myeloid DC that are CD11c+ . The CD11c+ DC are readily classified as CD1c+ DC and CD141+ DC. Monocytes are broadly divided into the CD14+ CD16- (classical) and CD14dim CD16+ subsets (nonclassical). A population of myeloid-derived cells that have DC characteristics, that is, HLA-DR+ and lacking lineage markers including CD14, but express CD16 are generally clustered with CD14dim CD16+ monocytes. We used high-dimensional clustering analyses of fluorescence and mass cytometry data, to delineate CD14+ monocytes, CD14dim CD16+ monocytes (CD16+ Mo), and CD14- CD16+ DC (CD16+ DC). We sought to identify the functional and kinetic relationship of CD16+ DC to CD16+ Mo. We demonstrate that differentiation of CD16+ DC and CD16+ Mo during activation with IFNγ in vitro and as a result of an allo-hematopoietic cell transplant (HCT) in vivo resulted in distinct populations. Recovery of blood CD16+ DC in both auto- and allo-(HCT) patients after myeloablative conditioning showed similar reconstitution and activation kinetics to CD16+ Mo. Finally, we show that expression of the cell surface markers CD300c, CCR5, and CLEC5a can distinguish the cell populations phenotypically paving the way for functional differentiation as new reagents become available.


Subject(s)
Antigen-Presenting Cells/immunology , Biomarkers/analysis , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Monocytes/immunology , Myeloid Cells/immunology , Receptors, IgG/metabolism , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , Cell Differentiation , Cell Lineage , Dendritic Cells/metabolism , GPI-Linked Proteins/metabolism , Graft vs Host Disease/diagnosis , Graft vs Host Disease/metabolism , HLA-DR Antigens/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Lectins, C-Type/metabolism , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Myeloid Cells/metabolism , Receptors, CCR5/metabolism , Receptors, Cell Surface/metabolism , Transplantation, Homologous
2.
Mol Oncol ; 13(10): 2107-2120, 2019 10.
Article in English | MEDLINE | ID: mdl-31338922

ABSTRACT

Antibody-based therapy in acute myeloid leukemia (AML) has been marred by significant hematologic toxicity due to targeting of both hematopoietic stem and progenitor cells (HSPCs). Achieving greater success with therapeutic antibodies requires careful characterization of the potential target molecules on AML. One potential target is CD300f, which is an immunoregulatory molecule expressed predominantly on myeloid lineage cells. To confirm the value of CD300f as a leukemic target, we showed that CD300f antibodies bind to AML from 85% of patient samples. While one CD300f monoclonal antibody (mAb) reportedly did not bind healthy hematopoietic stem cells, transcriptomic analysis found that CD300f transcripts are expressed by healthy HSPC. Several CD300f protein isoforms exist as a result of alternative splicing. Importantly for antibody targeting, the extracellular region of CD300f can be present with or without the exon 4-encoded sequence. This results in CD300f isoforms that are differentially bound by CD300f-specific antibodies. Furthermore, binding of one mAb, DCR-2, to CD300f exposes a structural epitope recognized by a second CD300f mAb, UP-D2. Detailed analysis of publicly available transcriptomic data indicated that CD34+ HSPC expressed fewer CD300f transcripts that lacked exon 4 compared to AML with monocytic differentiation. Analysis of a small cohort of AML cells revealed that the UP-D2 conformational binding site could be induced in cells from AML patients with monocytic differentiation but not those from other AML or HSPC. This provides the opportunity to develop an antibody-based strategy to target AMLs with monocytic differentiation but not healthy CD34+ HSPCs. This would be a major step forward in developing effective anti-AML therapeutic antibodies with reduced hematologic toxicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Epitopes/immunology , Leukemia, Myeloid, Acute/drug therapy , Receptors, Immunologic/immunology , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/immunology , Molecular Targeted Therapy , Monocytes/drug effects , Monocytes/immunology , Receptors, Immunologic/antagonists & inhibitors
3.
Front Immunol ; 10: 1312, 2019.
Article in English | MEDLINE | ID: mdl-31231400

ABSTRACT

CD83 is a member of the immunoglobulin (Ig) superfamily and is expressed in membrane bound or soluble forms. Membrane CD83 (mCD83) can be detected on a variety of activated immune cells, although it is most highly and stably expressed by mature dendritic cells (DC). mCD83 regulates maturation, activation and homeostasis. Soluble CD83 (sCD83), which is elevated in the serum of patients with autoimmune disease and some hematological malignancies is reported to have an immune suppressive function. While CD83 is emerging as a promising immune modulator with therapeutic potential, some important aspects such as its ligand/s, intracellular signaling pathways and modulators of its expression are unclear. In this review we discuss the recent biological findings and the potential clinical value of CD83 based therapeutics in various conditions including autoimmune disease, graft-vs.-host disease, transplantation and hematological malignancies.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, CD/immunology , Immunoglobulins/immunology , Membrane Glycoproteins/immunology , Animals , Humans , CD83 Antigen
4.
PLoS One ; 14(5): e0216368, 2019.
Article in English | MEDLINE | ID: mdl-31075107

ABSTRACT

Acute myeloid leukemia (AML) is the most common form of adult acute leukemia with ~20,000 new cases yearly. The disease develops in people of all ages, but is more prominent in the elderly, who due to limited treatment options, have poor overall survival rates. Monoclonal antibodies (mAb) targeting specific cell surface molecules have proven to be safe and effective in different haematological malignancies. However, AML target molecules are currently limited so discovery of new targets would be highly beneficial to patients. We examined the C-type lectin receptor CD302 as a potential therapeutic target for AML due to its selective expression in myeloid immune populations. In a cohort of 33 AML patients with varied morphological and karyotypic classifications, 88% were found to express CD302 on the surface of blasts and 80% on the surface of CD34+ CD38- population enriched with leukemic stem cells. A mAb targeting human CD302 was effective in mediating antibody dependent cell cytotoxicity and was internalised, making it amenable to toxin conjugation. Targeting CD302 with antibody limited in vivo engraftment of the leukemic cell line HL-60 in NOD/SCID mice. While CD302 was expressed in a hepatic cell line, HepG2, this molecule was not detected on the surface of HepG2, nor could HepG2 be killed using a CD302 antibody-drug conjugate. Expression was however found on the surface of haematopoietic stem cells suggesting that targeting CD302 would be most effective prior to haematopoietic transplantation. These studies provide the foundation for examining CD302 as a potential therapeutic target for AML.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/pharmacology , Blast Crisis , Drug Delivery Systems , Lectins, C-Type/metabolism , Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Receptors, Cell Surface/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Blast Crisis/drug therapy , Blast Crisis/metabolism , Blast Crisis/pathology , Female , HL-60 Cells , Hematopoietic Stem Cell Transplantation , Hep G2 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
5.
Semin Cell Dev Biol ; 86: 3-14, 2019 02.
Article in English | MEDLINE | ID: mdl-29499385

ABSTRACT

Dendritic cells (DC) are bone marrow derived leucocytes that are part of the mononuclear phagocytic system. These are surveillance cells found in all tissues and, as specialised antigen presenting cells, direct immune responses. Membrane molecules on the DC surface form a landscape that defines them as leucocytes and part of the mononuclear phagocytic system, interacts with their environment and directs interactions with other cells. This review describes the DC surface landscape, reflects on the different molecules confirmed to be on their surface and how they provide the basis for manipulation and translation of the potent functions of these cells into new diagnostics and immune therapies for the clinic.


Subject(s)
Dendritic Cells/cytology , Phenotype , Dendritic Cells/immunology , Humans
6.
Semin Cell Dev Biol ; 86: 77-88, 2019 02.
Article in English | MEDLINE | ID: mdl-29454038

ABSTRACT

The ability of immune therapies to control cancer has recently generated intense interest. This therapeutic outcome is reliant on T cell recognition of tumour cells. The natural function of dendritic cells (DC) is to generate adaptive responses, by presenting antigen to T cells, hence they are a logical target to generate specific anti-tumour immunity. Our understanding of the biology of DC is expanding, and they are now known to be a family of related subsets with variable features and function. Most clinical experience to date with DC vaccination has been using monocyte-derived DC vaccines. There is now growing experience with alternative blood-derived DC derived vaccines, as well as with multiple forms of tumour antigen and its loading, a wide range of adjuvants and different modes of vaccine delivery. Key insights from pre-clinical studies, and lessons learned from early clinical testing drive progress towards improved vaccines. The potential to fortify responses with other modalities of immunotherapy makes clinically effective "second generation" DC vaccination strategies a priority for cancer immune therapists.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Humans , T-Lymphocytes/immunology
7.
Haematologica ; 103(4): 655-665, 2018 04.
Article in English | MEDLINE | ID: mdl-29351987

ABSTRACT

Chemotherapy and hematopoietic stem cell transplantation are effective treatments for most Hodgkin lymphoma patients, however there remains a need for better tumor-specific target therapy in Hodgkin lymphoma patients with refractory or relapsed disease. Herein, we demonstrate that membrane CD83 is a diagnostic and therapeutic target, highly expressed in Hodgkin lymphoma cell lines and Hodgkin and Reed-Sternberg cells in 29/35 (82.9%) Hodgkin lymphoma patient lymph node biopsies. CD83 from Hodgkin lymphoma tumor cells was able to trogocytose to surrounding T cells and, interestingly, the trogocytosing CD83+T cells expressed significantly more programmed death-1 compared to CD83-T cells. Hodgkin lymphoma tumor cells secreted soluble CD83 that inhibited T-cell proliferation, and anti-CD83 antibody partially reversed the inhibitory effect. High levels of soluble CD83 were detected in Hodgkin lymphoma patient sera, which returned to normal in patients who had good clinical responses to chemotherapy confirmed by positron emission tomography scans. We generated a human anti-human CD83 antibody, 3C12C, and its toxin monomethyl auristatin E conjugate, that killed CD83 positive Hodgkin lymphoma cells but not CD83 negative cells. The 3C12C antibody was tested in dose escalation studies in non-human primates. No toxicity was observed, but there was evidence of CD83 positive target cell depletion. These data establish CD83 as a potential biomarker and therapeutic target in Hodgkin lymphoma.


Subject(s)
Antigens, CD/blood , Biomarkers, Tumor/blood , Hodgkin Disease/drug therapy , Immunoglobulins/blood , Membrane Glycoproteins/blood , Molecular Targeted Therapy/methods , Adolescent , Adult , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Female , Hodgkin Disease/diagnosis , Humans , Immunoglobulins/immunology , Male , Membrane Glycoproteins/immunology , Middle Aged , Salvage Therapy/methods , T-Lymphocytes/cytology , Young Adult , CD83 Antigen
8.
J Immunol ; 197(12): 4613-4625, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27837105

ABSTRACT

CD83 is a member of the Ig gene superfamily, first identified in activated lymphocytes. Since then, CD83 has become an important marker for defining activated human dendritic cells (DC). Several potential CD83 mRNA isoforms have been described, including a soluble form detected in human serum, which may have an immunosuppressive function. To further understand the biology of CD83, we examined its expression in different human immune cell types before and after activation using a panel of mouse and human anti-human CD83 mAb. The mouse anti-human CD83 mAbs, HB15a and HB15e, and the human anti-human CD83 mAb, 3C12C, were selected to examine cytoplasmic and surface CD83 expression, based on their different binding characteristics. Glycosylation of CD83, the CD83 mRNA isoforms, and soluble CD83 released differed among blood DC, monocytes, and monocyte-derived DC, and other immune cell types. A small T cell population expressing surface CD83 was identified upon T cell stimulation and during allogeneic MLR. This subpopulation appeared specifically during viral Ag challenge. We did not observe human CD83 on unstimulated human natural regulatory T cells (Treg), in contrast to reports describing expression of CD83 on mouse Treg. CD83 expression was increased on CD4+, CD8+ T, and Treg cells in association with clinical acute graft-versus-host disease in allogeneic hematopoietic cell transplant recipients. The differential expression and function of CD83 on human immune cells reveal potential new roles for this molecule as a target of therapeutic manipulation in transplantation, inflammation, and autoimmune diseases.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Monocytes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Antigens, CD/genetics , Antigens, Viral/immunology , Cells, Cultured , Glycosylation , Humans , Immunoglobulins/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , RNA Isoforms/genetics , RNA, Messenger/genetics , Transplantation, Homologous , CD83 Antigen
9.
Oncoimmunology ; 5(6): e1168555, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471645

ABSTRACT

There are numerous transcriptional, proteomic and functional differences between monocyte-derived dendritic cells (Mo-DC) and primary blood dendritic cells (BDC). The CMRF-56 monoclonal antibody (mAb) recognizes a cell surface marker, which is upregulated on BDC following overnight culture. Given its unique ability to select a heterogeneous population of BDC, we engineered a human chimeric (h)CMRF-56 IgG4 mAb to isolate primary BDC for potential therapeutic vaccination. The ability to select multiple primary BDC subsets from patients and load them with in vitro transcribed (IVT) mRNA encoding tumor antigen might circumvent the issues limiting the efficacy of Mo-DC. After optimizing and validating the purification of hCMRF-56(+) BDC, we showed that transfection of hCMRF-56(+) BDC with mRNA resulted in efficient mRNA translation and antigen presentation by myeloid BDC subsets, while preserving superior DC functions compared to Mo-DC. Immune selected and transfected hCMRF-56(+) BDC migrated very efficiently in vitro and as effectively as cytokine matured Mo-DC in vivo. Compared to Mo-DC, hCMRF-56(+) BDC transfected with influenza matrix protein M1 displayed superior MHC peptide presentation and generated potent antigen specific CD8(+) T-cell recall responses, while Wilms tumor 1 (WT1) transfected CMRF-56(+) BDC generated effective primary autologous cytotoxic T-cell responses. The ability of the combined DC subsets within hCMRF-56(+) BDC to present mRNA delivered tumor antigens merits phase I evaluation as a reproducible generic platform for the next generation of active DC immune therapies.

10.
J Immunol ; 197(3): 885-98, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27316686

ABSTRACT

C-type lectin receptors play important roles in immune cell interactions with the environment. We described CD302 as the simplest, single domain, type I C-type lectin receptor and showed it was expressed mainly on the myeloid phagocytes in human blood. CD302 colocalized with podosomes and lamellopodia structures, so we hypothesized that it played a role in cell adhesion or migration. In this study, we used mouse models to obtain further insights into CD302 expression and its potential immunological function. Mouse CD302 transcripts were, as in humans, highest in the liver, followed by lungs, lymph nodes (LN), spleen, and bone marrow. In liver, CD302 was expressed by hepatocytes, liver sinusoidal endothelial cells, and Kupffer cells. A detailed analysis of CD302 transcription in mouse immune cells revealed highest expression by myeloid cells, particularly macrophages, granulocytes, and myeloid dendritic cells (mDC). Interestingly, 2.5-fold more CD302 was found in migratory compared with resident mDC populations and higher CD302 expression in mouse M1 versus M2 macrophages was also noteworthy. CD302 knockout (CD302KO) mice were generated. Studies on the relevant immune cell populations revealed a decrease in the frequency and numbers of migratory mDC within CD302KO LN compared with wild-type LN. In vitro studies showed CD302KO and wild-type DC had an equivalent capacity to undergo maturation, prime T cells, uptake Ags, and migrate toward the CCL19/CCL21 chemokines. Nevertheless, CD302KO migratory DC exhibited reduced in vivo migration into LN, confirming a functional role for CD302 in mDC migration.


Subject(s)
Chemotaxis, Leukocyte/physiology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Line , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Knockout , Polymerase Chain Reaction
11.
J Immunother ; 38(2): 71-6, 2015.
Article in English | MEDLINE | ID: mdl-25658616

ABSTRACT

Preclinical studies have suggested that purified populations of CD1c (BDCA-1) blood-derived dendritic cells (BDC) loaded with tumor-specific peptides may be a feasible option for prostate cancer immunotherapy. We performed an open-label dose-finding Phase I study to evaluate the safe use of CD1c BDC in patients with advanced metastatic hormone refractory prostate cancer. HLA-A*0201-positive patients with advanced metastatic prostate cancer were recruited and consented. The vaccine was manufactured by pulsing autologous CD1c BDC, prepared by magnetic bead immunoselection from apheresed peripheral blood mononuclear cells, with a cocktail of HLA-A*0201-restricted peptides (prostate-specific antigen, prostate acid phosphatase, prostate specific membrane antigen, and control influenza peptide) and keyhole limpet hemocyanin. The vaccine was administered intradermally or intravenously and peripheral blood was taken at predetermined intervals for clinical and immunologic monitoring. The vaccine was manufactured with a median purity of 82% CD1c BDC and administered successfully to 12 patients. Each patient received between 1 and 5 × 10 fresh CD1c BDC on day 0, followed by cryopreserved product in the same dose on days 28 and 56. The vaccine was well tolerated in all patients, with the most frequent adverse events being grade 1-2 fever, pain, or injection-site reactions. Vaccination with CD1c BDC is therefore feasible, safe, and well tolerated in patients with advanced-stage metastatic prostate cancer.


Subject(s)
Cancer Vaccines , Dendritic Cells/immunology , HLA-A2 Antigen/metabolism , Immunotherapy, Adoptive , Peptide Fragments/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , Acid Phosphatase/immunology , Acid Phosphatase/metabolism , Administration, Intravenous , Aged , Antigens, CD1/metabolism , Dendritic Cells/transplantation , Feasibility Studies , Glycoproteins/metabolism , Humans , Injections, Intradermal , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Organ Specificity , Prostate-Specific Antigen/immunology , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms, Castration-Resistant/immunology
12.
Br J Haematol ; 164(4): 481-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24321020

ABSTRACT

Novel therapies with increased efficacy and decreased toxicity are desperately needed for the treatment of acute myeloid leukaemia (AML). The anti CD33 immunoconjugate, gemtuzumab ozogamicin (GO), was withdrawn with concerns over induction mortality and lack of efficacy. However a number of recent trials suggest that, particularly in AML with favourable cytogenetics, GO may improve overall survival. This data and the development of alternative novel monoclonal antibodies (mAb) have renewed interest in the area. Leukaemic stem cells (LSC) are identified as the subset of AML blasts that reproduces the leukaemic phenotype upon transplantation into immunosuppressed mice. AML relapse may be caused by chemoresistant LSC and this has refocused interest on identifying and targeting antigens specific for LSC. Several mAb have been developed that target LSC effectively in xenogeneic models but only a few have begun clinical evaluation. Antibody engineering may improve the activity of potential new therapeutics for AML. The encouraging results seen with bispecific T cell-engaging mAb-based molecules against CD19 in the treatment of B-cell acute lymphobalstic leukaemia, highlight the potential efficacy of engineered antibodies in the treatment of acute leukaemia. Potent engineered mAb, possibly targeting novel LSC antigens, offer hope for improving the current poor prognosis for AML.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/radiotherapy
13.
Transplantation ; 96(8): 753-62, 2013 Oct 27.
Article in English | MEDLINE | ID: mdl-23903010

ABSTRACT

BACKGROUND: Dendritic cells (DC) are important in the development of acute graft-versus-host disease (GVHD) after allogeneic hemopoietic cell transplantation (alloHCT). The trafficking of immature DC from blood to GVHD target organs is likely to be regulated by chemokine receptors. METHODS: We performed flow cytometry to document the expression of chemokine receptors on circulating DC and correlated the findings after alloHCT with occurrence of acute GVHD. RESULTS: In normal individuals, plasmacytoid DC (pDC) expressed high levels of CCR5, whereas the major CD16 myeloid DC subpopulation lacked CCR5. However, its expression on CD16 cells was induced by culture in allogeneic mixed lymphocyte reaction supernatant, an effect largely mediated by interferon-γ. CCR5 was expressed on a significant proportion of CD16 DC in 42 alloHCT patients, whereas it was down-regulated on pDC. The maximum percentage of CCR5CD16 DC, at any time after transplantation, correlated with acute GVHD, whereas the minimum CCR5 on pDC showed a similar correlation. Before developing signs of GVHD, the maximum percentage CCR5CD16 DC was higher in patients with GVHD grades II to IV than in GVHD grades 0 and I, whereas the minimum percentage CCR5 on pDC was lower in GVHD grades II to IV than in GVHD grades 0 and I. CCR5 levels more than 20.5% on CD16 myeloid DC and less than 22.6% on CD123 pDC correlated with subsequent GVHD grades II to IV with high sensitivities and specificities. CONCLUSIONS: These observations may reflect DC activation and altered homing during the alloimmune response and could allow early diagnosis and therapeutic intervention before the clinical diagnosis of GVHD.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Postoperative Complications/immunology , Receptors, CCR5/immunology , Acute Disease , Adolescent , Adult , Aged , Dendritic Cells/metabolism , Female , Flow Cytometry , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Graft vs Host Disease/metabolism , Humans , Male , Middle Aged , Postoperative Complications/metabolism , Receptors, CCR5/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Transplantation, Homologous , Young Adult
14.
Immunol Lett ; 149(1-2): 93-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23072861

ABSTRACT

Dendritic cells (DC) are a heterogeneous population of leucocytes which play a key role in initiating and modulating immune responses. The human CD300 family consists of six immunoregulatory leucocyte membrane molecules that regulate cellular activity including differentiation, viability, cytokine and chemokine secretion, phagocytosis and chemotaxis. Recent work has identified polar lipids as probable ligands for these molecules in keeping with the known evolutionary conservation of this family. CD300 molecules are all expressed by DC; CD300b, d, e and f are restricted to different subpopulations of the myeloid DC lineage. They have been shown to regulate DC function both in vitro and in vivo. In addition DC are able to regulate their CD300 expression in an autocrine manner. The potential to form different CD300 heterodimers adds further complexity to their role in fine tuning DC function. Expression of CD300 molecules is altered in a number of diseases including many where DC are implicated in the pathogenesis. CD300 antibodies have been demonstrated to have significant therapeutic effect in animal models. The mechanisms underlying the immunoregulatory effects of the CD300 family are complex. Deciphering their physiology will allow effective targeting of these molecules as novel therapies in a wide variety of inflammatory diseases.


Subject(s)
Dendritic Cells/immunology , Protein Multimerization/immunology , Receptors, Immunologic/immunology , Autocrine Communication/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Receptors, Immunologic/biosynthesis
15.
Eur J Immunol ; 42(6): 1512-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22678905

ABSTRACT

Human blood myeloid DCs can be subdivided into CD1c (BDCA-1)(+) and CD141 (BDCA-3)(+) subsets that display unique gene expression profiles, suggesting specialized functions. CD1c(+) DCs express TLR4 while CD141(+) DCs do not, thus predicting that these two subsets have differential capacities to respond to Escherichia coli. We isolated highly purified CD1c(+) and CD141(+) DCs and compared them to in vitro generated monocyte-derived DCs (MoDCs) following stimulation with whole E. coli. As expected, MoDCs produced high levels of the proinflammatory cytokines TNF, IL-6, and IL-12, were potent inducers of Th1 responses, and processed E. coli-derived Ag. In contrast, CD1c(+) DCs produced only low levels of TNF, IL-6, and IL-12 and instead produced high levels of the anti-inflammatory cytokine IL-10 and regulatory molecules IDO and soluble CD25. Moreover, E. coli-activated CD1c(+) DCs suppressed T-cell proliferation in an IL-10-dependent manner. Contrary to their mouse CD8(+) DC counterparts, human CD141(+) DCs did not phagocytose or process E. coli-derived Ag and failed to secrete cytokines in response to E. coli. These data demonstrate substantial differences in the nature of the response of human blood DC subsets to E. coli.


Subject(s)
Antigens, Surface/analysis , Dendritic Cells/immunology , Escherichia coli/immunology , Interleukin-10/biosynthesis , Myeloid Cells/immunology , Antigens, CD1 , Dendritic Cells/metabolism , Glycoproteins , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Phenotype , T-Lymphocytes/immunology , Thrombomodulin
16.
Cancer Immunol Immunother ; 61(2): 169-179, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21874303

ABSTRACT

Immunotherapy is a promising new treatment for patients with advanced prostate and ovarian cancer, but its application is limited by the lack of suitable target antigens that are recognized by CD8+ cytotoxic T lymphocytes (CTL). Human kallikrein 4 (KLK4) is a member of the kallikrein family of serine proteases that is significantly overexpressed in malignant versus healthy prostate and ovarian tissue, making it an attractive target for immunotherapy. We identified a naturally processed, HLA-A*0201-restricted peptide epitope within the signal sequence region of KLK4 that induced CTL responses in vitro in most healthy donors and prostate cancer patients tested. These CTL lysed HLA-A*0201+ KLK4 + cell lines and KLK4 mRNA-transfected monocyte-derived dendritic cells. CTL specific for the HLA-A*0201-restricted KLK4 peptide were more readily expanded to a higher frequency in vitro compared to the known HLA-A*0201-restricted epitopes from prostate cancer antigens; prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA) and prostatic acid phosphatase (PAP). These data demonstrate that KLK4 is an immunogenic molecule capable of inducing CTL responses and identify it as an attractive target for prostate and ovarian cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Kallikreins/metabolism , Peptide Fragments/metabolism , Prostatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adult , Aged , Antigens, Neoplasm/immunology , Cell Proliferation , Computational Biology , Dendritic Cells/immunology , Female , HLA-A2 Antigen/metabolism , Humans , Immunodominant Epitopes/genetics , Kallikreins/immunology , Lymphocyte Activation , Male , Middle Aged , Peptide Fragments/immunology , Prostatic Neoplasms/pathology , Protein Sorting Signals/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
17.
Pathology ; 43(6): 605-15, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21897329

ABSTRACT

The recent Food and Drug Administration (FDA) approval of a cellular therapy to treat castration resistant prostate cancer has reinforced the potential of cellular therapy to consolidate current pharmacological approaches to treating cancer. The emergence of the cell manufacturing facility to facilitate clinical translation of these new methodologies allows greater access to these novel therapies. Here we review different strategies currently being explored to treat haematological malignancies with a focus on adoptive allogeneic or autologous transfer of antigen specific T cells, NK cells or dendritic cells. These approaches all aim to generate immunological responses against overexpressed tissue antigens, mismatched minor histocompatability antigens or tumour associated antigens. Current successes and limitations of these different approaches will be discussed with an emphasis on challenges encountered in generating long term engraftment, antigen selection and implementation as well as therapeutic immune monitoring of clinical responses, with examples from recent clinical trials.


Subject(s)
Cell Transplantation/methods , Hematologic Neoplasms/therapy , Cell Transplantation/trends , Clinical Trials as Topic , Dendritic Cells/immunology , Dendritic Cells/transplantation , Hematologic Neoplasms/immunology , Humans , Immunotherapy , Natural Killer T-Cells/immunology , Natural Killer T-Cells/transplantation , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation
18.
J Immunol ; 187(8): 3987-96, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21908738

ABSTRACT

The graft-versus-myeloma (GVM) effect represents a powerful form of immune attack exerted by alloreactive T cells against multiple myeloma cells, which leads to clinical responses in multiple myeloma transplant recipients. Whether myeloma cells are themselves able to induce alloreactive T cells capable of the GVM effect is not defined. Using adoptive transfer of T naive cells into myeloma-bearing mice (established by transplantation of human RPMI8226-TGL myeloma cells into CD122(+) cell-depleted NOD/SCID hosts), we found that myeloma cells induced alloreactive T cells that suppressed myeloma growth and prolonged survival of T cell recipients. Myeloma-induced alloreactive T cells arising in the myeloma-infiltrated bones exerted cytotoxic activity against resident myeloma cells, but limited activity against control myeloma cells obtained from myeloma-bearing mice that did not receive T naive cells. These myeloma-induced alloreactive T cells were derived through multiple CD8(+) T cell divisions and enriched in double-positive (DP) T cells coexpressing the CD8αα and CD4 coreceptors. MHC class I expression on myeloma cells and contact with T cells were required for CD8(+) T cell divisions and DP-T cell development. DP-T cells present in myeloma-infiltrated bones contained a higher proportion of cells expressing cytotoxic mediators IFN-γ and/or perforin compared with single-positive CD8(+) T cells, acquired the capacity to degranulate as measured by CD107 expression, and contributed to an elevated perforin level seen in the myeloma-infiltrated bones. These observations suggest that myeloma-induced alloreactive T cells arising in myeloma-infiltrated bones are enriched with DP-T cells equipped with cytotoxic effector functions that are likely to be involved in the GVM effect.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Tumor Effect/immunology , Multiple Myeloma/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Cell Separation , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Homologous
19.
Immunol Lett ; 134(2): 161-6, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-20970455

ABSTRACT

Dendritic cells (DC) are a heterogeneous population of bone marrow derived leucocytes that are essential in the initiation of primary T lymphocyte responses. DC are identified as Lineage negative, HLA-DR(+) blood cells that can be further subdivided by CD11c to distinguish CD11c(+) DC and the CD11c(-) plasmacytoid DC. Plasmacytoid DC are the primary IFNα producing cells and express CD303, CD304 and CD123. The CD11c(+) myeloid DC can be divided into populations by CD1c, CD16 and CD141 expression. Despite DC being a functionally unique population, they share many cell surface antigens with myeloid lineage cells and B lymphocytes. We used flow cytometry to screen fresh human blood DC populations with the HLDA9 panel of 63 directly labelled mAb which included mAb specific for a number of B lymphocyte antigens. Of this panel, 23 mAb did not bind Lin(-)HLA-DR(+) DC and 10 bound all four populations. Eight mAb bound to the three CD11c(+) DC populations whilst no mAb tested bound to only pDC. Some of the mAb expected to bind to DC populations failed in this analysis. Overall, this screening highlighted similarities between the CD11c(+) DC subsets and the relatively immature state of peripheral blood DC.


Subject(s)
Dendritic Cells/immunology , Antibodies, Monoclonal/immunology , CD11c Antigen/immunology , Cells, Cultured , Humans , Immunophenotyping , Protein Binding , Proteome
20.
Immunology ; 132(2): 296-305, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21091907

ABSTRACT

Mannose-binding lectin (MBL) is a serum lectin that plays a significant role in innate host defence. Individuals with mutations in exon 1 of the MBL2 gene have reduced MBL ligand binding and complement activation function and increased incidence of infection. We proposed that, during infection, MBL deficiency may impact on dendritic cell (DC) function. We analysed the blood myeloid DC (MDC) surface phenotype, inflammatory cytokine production and antigen-presenting capacity in MBL-deficient (MBL-D) individuals and MBL-sufficient (MBL-S) individuals using whole blood culture supplemented with zymosan (Zy) or MBL-opsonized zymosan (MBL-Zy) as a model of infection. Zy-stimulated MDCs from MBL-D individuals had significantly increased production of interleukin (IL)-6 and tumour necrosis factor (TNF)-α. Stimulation with MBL-Zy significantly decreased IL-6 production by MDCs from MBL-D, but had no effect on TNF-α production. MDCs from both MBL-S and MBL-D individuals up-regulated expression of the activation molecule CD83, and down-regulated expression of homing (CXCR4), adhesion (CD62L, CD49d) and costimulatory (CD40, CD86) molecules in response to Zy and MBL-Zy. MDC from both MBL-D and MBL-S individuals induced proliferation of allogeneic (allo) T cells following Zy or MBL-Zy stimulation; however, MBL-D individuals demonstrated a reduced capacity to induce effector allo-T cells. These data indicate that MBL deficiency is associated with unique functional characteristics of pathogen-stimulated blood MDCs manifested by increased production of IL-6, combined with a poor capacity to induce effector allo-T-cell responses. In MBL-D individuals, these functional features of blood MDCs may influence their ability to mount an immune response.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Immunity, Innate , Interleukin-6/metabolism , Mannose-Binding Lectin/deficiency , Myeloid Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Zymosan/immunology , Zymosan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...