Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Commun ; 15(1): 6790, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117654

ABSTRACT

Immunochemotherapy has been the mainstay of treatment for newly diagnosed diffuse large B-cell lymphoma (ndDLBCL) yet is inadequate for many patients. In this work, we perform unsupervised clustering on transcriptomic features from a large cohort of ndDLBCL patients and identify seven clusters, one called A7 with poor prognosis, and develop a classifier to identify these clusters in independent ndDLBCL cohorts. This high-risk cluster is enriched for activated B-cell cell-of-origin, low immune infiltration, high MYC expression, and copy number aberrations. We compare and contrast our methodology with recent DLBCL classifiers to contextualize our clusters and show improved prognostic utility. Finally, using pre-clinical models, we demonstrate a mechanistic rationale for IKZF1/3 degraders such as lenalidomide to overcome the low immune infiltration phenotype of A7 by inducing T-cell trafficking into tumors and upregulating MHC I and II on tumor cells, and demonstrate that TCF4 is an important regulator of MYC-related biology in A7.


Subject(s)
Gene Expression Regulation, Neoplastic , Ikaros Transcription Factor , Lenalidomide , Lymphoma, Large B-Cell, Diffuse , Proto-Oncogene Proteins c-myc , Transcription Factor 4 , Transcriptome , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Lenalidomide/therapeutic use , Lenalidomide/pharmacology , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Prognosis , Animals , Cell Line, Tumor , Gene Expression Profiling/methods , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , DNA Copy Number Variations
2.
Haematologica ; 109(4): 1082-1094, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37941406

ABSTRACT

Oral azacitidine (oral-Aza) treatment results in longer median overall survival (OS) (24.7 vs. 14.8 months in placebo) in patients with acute myeloid leukemia (AML) in remission after intensive chemotherapy. The dosing schedule of oral-Aza (14 days/28-day cycle) allows for low exposure of Aza for an extended duration thereby facilitating a sustained therapeutic effect. However, the underlying mechanisms supporting the clinical impact of oral-Aza in maintenance therapy remain to be fully understood. In this preclinical work, we explore the mechanistic basis of oral-Aza/extended exposure to Aza through in vitro and in vivo modeling. In cell lines, extended exposure to Aza results in sustained DNMT1 loss, leading to durable hypomethylation, and gene expression changes. In mouse models, extended exposure to Aza, preferentially targets immature leukemic cells. In leukemic stem cell (LSC) models, the extended dose of Aza induces differentiation and depletes CD34+CD38- LSC. Mechanistically, LSC differentiation is driven in part by increased myeloperoxidase (MPO) expression. Inhibition of MPO activity either by using an MPO-specific inhibitor or blocking oxidative stress, a known mechanism of MPO, partly reverses the differentiation of LSC. Overall, our preclinical work reveals novel mechanistic insights into oral-Aza and its ability to target LSC.


Subject(s)
Azacitidine , Leukemia, Myeloid, Acute , Animals , Mice , Humans , Azacitidine/pharmacology , Azacitidine/therapeutic use , Antigens, CD34/metabolism , Leukemia, Myeloid, Acute/genetics , Peroxidase , Stem Cells/metabolism
3.
Clin Cancer Res ; 28(15): 3367-3377, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35583604

ABSTRACT

PURPOSE: Cereblon (CRBN), a substrate receptor of the E3 ubiquitin ligase complex CRL4CRBN, is the target of the small molecules lenalidomide and avadomide. Upon binding of the drugs, Aiolos and Ikaros are recruited to the E3 ligase, ubiquitylated, and subsequently degraded. In diffuse large B-cell lymphoma (DLBCL) cells, Aiolos and Ikaros are direct transcriptional repressors of interferon-stimulated genes (ISG) and degradation of these substrates results in increased ISG protein levels resulting in decreased proliferation and apoptosis. Herein, we aimed to uncover the mechanism(s) Aiolos and Ikaros use to repress ISG transcription and provide a mechanistic rationale for a combination strategy to enhance cell autonomous activities of CRBN modulators (CELMoD). EXPERIMENTAL DESIGN: We conducted paired RNA sequencing with histone modification and Aiolos/Ikaros chromatin immunoprecipitation sequencing to identify genes regulated by these transcription factors and to elucidate correlations to drug sensitivity. We confirmed Aiolos/Ikaros mediated transcriptional complex formation in DLBCL patient samples including those treated with avadomide. RESULTS: In DLBCL, the repression of ISG transcription is accomplished in part through recruitment of large transcriptional complexes such as the nucleosome remodeling and deacetylase, which modify the chromatin landscape of these promoters. A rational combination approach of avadomide with a specific histone deacetylase inhibitor leads to a significant increase in ISG transcription compared with either single agent, and synergistic antiproliferative activity in DLBCL cell lines. CONCLUSIONS: Our results provide a novel role for lineage factors Aiolos and Ikaros in DLBCL as well as further insight into the mechanism(s) of Aiolos and Ikaros-mediated transcriptional repression and unique therapeutic combination strategies.


Subject(s)
Histone Deacetylase Inhibitors , Lymphoma, Large B-Cell, Diffuse , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Immunologic Factors/therapeutic use , Lenalidomide/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Ubiquitin-Protein Ligases/genetics
4.
Blood ; 137(2): 216-231, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33024998

ABSTRACT

Cancer treatment has been transformed by checkpoint blockade therapies, with the highest anti-tumor activity of anti-programmed death 1 (PD-1) antibody therapy seen in Hodgkin lymphoma. Disappointingly, response rates have been low in the non-Hodgkin lymphomas, with no activity seen in relapsed/refractory chronic lymphocytic leukemia (CLL) with PD-1 blockade. Thus, identifying more powerful combination therapy is required for these patients. Here, we preclinically demonstrate enhanced anti-CLL activity following combinational therapy with anti-PD-1 or anti-PD-1 ligand (PD-L1) and avadomide, a cereblon E3 ligase modulator (CELMoD). Avadomide induced type I and II interferon (IFN) signaling in patient T cells, triggering a feedforward cascade of reinvigorated T-cell responses. Immune modeling assays demonstrated that avadomide stimulated T-cell activation, chemokine expression, motility and lytic synapses with CLL cells, as well as IFN-inducible feedback inhibition through upregulation of PD-L1. Patient-derived xenograft tumors treated with avadomide were converted to CD8+ T cell-inflamed tumor microenvironments that responded to anti-PD-L1/PD-1-based combination therapy. Notably, clinical analyses showed increased PD-L1 expression on T cells, as well as intratumoral expression of chemokine signaling genes in B-cell malignancy patients receiving avadomide-based therapy. These data illustrate the importance of overcoming a low inflammatory T-cell state to successfully sensitize CLL to checkpoint blockade-based combination therapy.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation/drug effects , Piperidones/pharmacology , Quinazolinones/pharmacology , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents/pharmacology , Humans , Immunotherapy/methods , Interferons/immunology , Mice , Signal Transduction/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
6.
Br J Haematol ; 185(2): 240-253, 2019 04.
Article in English | MEDLINE | ID: mdl-30767211

ABSTRACT

Chemotherapy plus rituximab has been the mainstay of treatment for follicular lymphoma (FL) for two decades but is associated with immunosuppression and relapse. In phase 2 studies, lenalidomide combined with rituximab (R2 ) has shown clinical synergy in front-line and relapsed/refractory FL. Here, we show that lenalidomide reactivated dysfunctional T and Natural Killer (NK) cells ex vivo from FL patients by enhancing proliferative capacity and T-helper cell type 1 (Th1) cytokine release. In combination with rituximab, lenalidomide improved antibody-dependent cellular cytotoxicity in sensitive and chemo-resistant FL cells, via a cereblon-dependent mechanism. While single-agent lenalidomide and rituximab increased formation of lytic NK cell immunological synapses with primary FL tumour cells, the combination was superior and correlated with enhanced cytotoxicity. Immunophenotyping of FL patient samples from a phase 3 trial revealed that R2 treatment increased circulating T- and NK-cell counts, while R-chemotherapy was associated with reduced cell numbers. Finally, using an in vitro model of myeloid differentiation, we demonstrated that lenalidomide caused a reversible arrest in neutrophil maturation that was distinct from a cytotoxic chemotherapeutic agent, which may help explain the lower rates of neutropenia observed with R2 versus R-chemotherapy. Taken together, we believe these data support a paradigm shift in the treatment of FL - moving from combination immunochemotherapy to chemotherapy-free immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lenalidomide/administration & dosage , Lymphoma, Follicular/drug therapy , Rituximab/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cyclophosphamide/therapeutic use , Cytokines/biosynthesis , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/immunology , Humans , Immunological Synapses/drug effects , Immunological Synapses/immunology , Immunotherapy/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lenalidomide/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphoma, Follicular/immunology , Neutrophils/drug effects , Prednisone/therapeutic use , Rituximab/immunology , Rituximab/therapeutic use , Tumor Cells, Cultured , Vincristine/therapeutic use
7.
Oncotarget ; 9(74): 34009-34021, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30338042

ABSTRACT

We recently showed that the outcome of multiple myeloma (MM) patients treated in the REPEAT study (evaluation of lenalidomide combined with low-dose cyclophosphamide and prednisone (REP) in lenalidomide-refractory MM) was markedly better than what has been described with cyclophosphamide-prednisone alone. The outcome with REP was not associated with plasma cell Cereblon expression levels, suggesting that the effect of REP treatment may involve mechanisms independent of plasma cell Cereblon-mediated direct anti-tumor activity. We therefore hypothesized that immunomodulatory effects contribute to the anti-MM activity of REP treatment, rather than plasma cell Cereblon-mediated effects. Consequently, we now characterized the effect of REP treatment on immune cell subsets in peripheral blood samples collected on day 1 and 14 of cycle 1, as well as on day 1 of cycle 2. We observed a significant mid-cycle decrease in the Cereblon substrate proteins Ikaros and Aiolos in diverse lymphocyte subsets, which was paralleled by an increase in T-cell activation. These effects were restored to baseline at day one of the second cycle, one week after lenalidomide interruption. In vitro, lenalidomide enhanced peripheral blood mononuclear cell-mediated killing of both lenalidomide-sensitive and lenalidomide-resistant MM cells in a co-culture system. These results indicate that the Cereblon-mediated immunomodulatory properties of lenalidomide are maintained in lenalidomide-refractory MM patients and may contribute to immune-mediated killing of MM cells. Therefore, combining lenalidomide with other drugs can have potent effects through immunomodulation, even in patients considered to be lenalidomide-refractory.

8.
Br J Haematol ; 179(3): 399-409, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28771673

ABSTRACT

Lenalidomide is an immunomodulatory agent that has demonstrated clinical benefit for patients with relapsed or refractory mantle cell lymphoma (MCL); however, despite this observed clinical activity, the mechanism of action (MOA) of lenalidomide has not been characterized in this setting. We investigated the MOA of lenalidomide in clinical samples from patients enrolled in the CC-5013-MCL-002 trial (NCT00875667) comparing single-agent lenalidomide versus investigator's choice single-agent therapy and validated our findings in pre-clinical models of MCL. Our results revealed a significant increase in natural killer (NK) cells relative to total lymphocytes in lenalidomide responders compared to non-responders that was associated with a trend towards prolonged progression-free survival and overall survival. Clinical response to lenalidomide was independent of baseline tumour microenvironment expression of its molecular target, cereblon, as well as genetic mutations reported to impact clinical response to the Bruton tyrosine kinase inhibitor ibrutinib. Preclinical experiments revealed lenalidomide enhanced NK cell-mediated cytotoxicity against MCL cells via increased lytic immunological synapse formation and secretion of granzyme B. In contrast, lenalidomide exhibited minimal direct cytotoxic effects against MCL cells. Taken together, these data provide the first insight into the clinical activity of lenalidomide against MCL, revealing a predominately immune-mediated MOA.


Subject(s)
Immunologic Factors/pharmacology , Killer Cells, Natural/drug effects , Lymphoma, Mantle-Cell/drug therapy , Thalidomide/analogs & derivatives , Adaptor Proteins, Signal Transducing , Adenine/analogs & derivatives , Coculture Techniques , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Killer Cells, Natural/immunology , Lenalidomide , Lymphocyte Count , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/metabolism , Mutation , Peptide Hydrolases/metabolism , Piperidines , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Thalidomide/administration & dosage , Thalidomide/pharmacology , Thalidomide/therapeutic use , Treatment Outcome , Tumor Cells, Cultured , Tumor Microenvironment , Ubiquitin-Protein Ligases
9.
Blood ; 126(6): 779-89, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26002965

ABSTRACT

Cereblon (CRBN), a substrate receptor of the Cullin 4 RING E3 ubiquitin ligase complex, is the target of the immunomodulatory drugs lenalidomide and pomalidomide. Recently, it was demonstrated that binding of these drugs to CRBN promotes the ubiquitination and subsequent degradation of 2 common substrates, transcription factors Aiolos and Ikaros. Here we report that CC-122, a new chemical entity termed pleiotropic pathway modifier, binds CRBN and promotes degradation of Aiolos and Ikaros in diffuse large B-cell lymphoma (DLBCL) and T cells in vitro, in vivo, and in patients, resulting in both cell autonomous as well as immunostimulatory effects. In DLBCL cell lines, CC-122-induced degradation or short hairpin RNA-mediated knockdown of Aiolos and Ikaros correlates with increased transcription of interferon (IFN)-stimulated genes independent of IFN-α, -ß, and -γ production and/or secretion and results in apoptosis in both activated B-cell (ABC) and germinal center B-cell DLBCL cell lines. Our results provide mechanistic insight into the cell-of-origin independent antilymphoma activity of CC-122, in contrast to the ABC subtype selective activity of lenalidomide.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Ikaros Transcription Factor/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/genetics , Piperidones/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/chemistry , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ikaros Transcription Factor/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferons/genetics , Interferons/metabolism , Lenalidomide , Lentivirus/genetics , Lentivirus/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, SCID , Molecular Mimicry , Peptide Hydrolases/metabolism , Piperidones/chemistry , Proteolysis/drug effects , Quinazolinones/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
10.
Prostate ; 68(2): 152-60, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18058812

ABSTRACT

BACKGROUND: Although a field effect in which transformed cells extend beyond morphologically evident tumor has been proposed in cancer, little direct evidence exists as to its magnitude and spatial resolution. We tested this hypothesis using molecular techniques to detect epigenetic changes in the primary tumor and surrounding tissues. METHODS: Ex vivo core biopsies, each spaced approximately 1 mm apart, were generated from 37 unique prostatectomy samples. The first core biopsy was confirmed to be histologically positive for cancer, and the subsequent biopsies were confirmed to be histologically negative. The methylation ratio of GSTP1, APC, RARbeta2, and RASSF1A were measured for all of the 159 cores. RESULTS: No field effect, defined as absence of epigenetically transformed cells, for GSTP1 was observed whereas APC, RARbeta2, and RASSF1A showed a field effect up to 3 mm from the malignant core in three prostatectomy samples. Furthermore, for each case, different patterns of the field effect were observed. The field effect appeared most pronounced with RARbeta2. In 11 prostatectomy samples in which a second focus of cancer was identified, cells harboring RARbeta2 methylation extended a large distance away from the primary tumor in one sample. Bisulfite sequencing of RARbeta2 confirmed the presence of epigenetic aberrations. CONCLUSIONS: This study quantifies previous observations of methylation in histologically negative samples and provides important assessment of field effects based on epigenetic events in cancer. These molecular approaches set the stage for consideration of such data in prospective trials for assessment of surgical margins and prediction of recurrence.


Subject(s)
DNA Methylation , DNA, Neoplasm/genetics , Epigenesis, Genetic/genetics , Prostatic Neoplasms/genetics , Adenomatous Polyposis Coli Protein/genetics , Base Sequence , Biomarkers, Tumor/genetics , Biopsy , Glutathione S-Transferase pi/genetics , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/pathology , Receptors, Retinoic Acid/genetics , Retrospective Studies , Tumor Suppressor Proteins/genetics
11.
J Gen Virol ; 87(Pt 2): 461-470, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16432035

ABSTRACT

Lamellocytes are specialized larval blood cells of Drosophila that carry out encapsulation of metazoan pathogens such as parasitoid wasps. Large virus-like particles (VLPs) from two closely related virulent parasitoid wasp species, Leptopilina heterotoma and Leptopilina victoriae, suppress the host encapsulation response by promoting lysis of lamellocytes. The molecular basis of VLP-lamellocyte interaction and lamellocyte lysis is not understood. Here, it was shown that mature VLPs are composed of at least four major proteins. Polyclonal antisera against the most abundant L. heterotoma VLP protein, p40, cross-reacted with the most abundant L. victoriae VLP protein, p47.5. Immuno-electron microscopy (EM) of the long gland-reservoir complex revealed that p40 was expressed early in VLP biogenesis and was detected along with VLP precursors within the long gland cells and lumen. In the reservoir, VLPs had an angular core, resembled mature particles and p40 was detected outside the VLP cores. Immuno-EM staining of mature VLPs from both species localized the p40 and p47.5 proteins largely to the periphery of the VLPs and along the VLP spike-like projections. p40 staining was observed in VLP-treated host haemocytes. In vitro, anti-p40 antibody almost completely blocked the ability of L. heterotoma VLPs to promote lamellocyte lysis. Anti-p40 antibody blocked lysis by L. victoriae VLPs by >50%. It is proposed that the VLP surface proteins p40 and p47.5 share antigenic determinants and significantly contribute to the strong virulence of their Hymenopteran hosts.


Subject(s)
Drosophila/parasitology , Hymenoptera/physiology , Insect Proteins/isolation & purification , Wasps/chemistry , Animals , Antibodies/analysis , Drosophila/immunology , Insect Proteins/ultrastructure , Larva/immunology , Larva/parasitology , Microscopy, Immunoelectron , Wasps/immunology , Wasps/pathogenicity , Wasps/ultrastructure
12.
Dev Biol ; 288(1): 60-72, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16248995

ABSTRACT

Highly conserved during evolution, the enzyme Ubc9 activates the small ubiquitin-like modifier (SUMO) prior to its covalent ligation to target proteins. We have used mutations in the Drosophila Ubc9 (dUbc9) gene to understand Ubc9 functions in vivo. Loss-of-function mutations in dUbc9 cause strong mitotic defects in larval hematopoietic tissues, an increase in the number of hematopoietic precursors in the lymph gland and of mature blood cells in circulation, and an increase in the proportion of cyclin-B-positive cells. Some blood cells are polyploid and multinucleate, exhibiting signs of genomic instability. We also observe an overabundance of highly differentiated blood cells (lamellocytes), normally not found in healthy larvae. Lamellocytes in mutants are either free in circulation or recruited to form tumorous masses. Hematopoietic defects of dUbc9 mutants are strongly suppressed in the absence of the Rel/NF-kappaB-family transcription factors Dorsal and Dif or in the presence of a non-signaling allele of Cactus, the IkappaB protein in Drosophila. In the larval fat body, dUbc9 negatively regulates the expression of the antifungal peptide gene drosomycin, which is constitutively expressed in dUbc9 mutants in the absence of immune challenge. dUbc9-mediated drosomycin expression requires Dorsal and Dif. Together, our results support a role for dUbc9 in the negative regulation of the Drosophila NF-kappaB signaling pathways in larval hematopoiesis and humoral immunity.


Subject(s)
Down-Regulation/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Hemocytes/cytology , NF-kappa B/antagonists & inhibitors , Toll-Like Receptors/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Hemocytes/physiology , Larva/cytology , Larva/physiology , Mutation , NF-kappa B/physiology , Phosphoproteins/physiology , Toll-Like Receptors/physiology , Ubiquitin-Conjugating Enzymes/genetics
13.
J Insect Physiol ; 51(2): 181-95, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15749103

ABSTRACT

Drosophila melanogaster larvae are attacked by virulent strains of parasitoid wasps. Females of Leptopilina heterotoma produce virus-like particles (VLPs) that efficiently destroy lamellocytes, a major larval immune effector cell type. We report here that L. victoriae, a closely related wasp species, also produces VLPs that trigger immune suppression responses in fly hosts. We compare the ability of immune suppression of the two parasitoids using a mutant host strain hopscotch(Tumorous-lethal) (hop(Tum-l)). hop(Tum-l) larvae have two defects of hematopoietic origin: overproliferation of hemocytes and constitutive encapsulation of self-tissue by lamellocytes. The encapsulation phenotype is suppressed weakly by L. victoriae and strongly by L. heterotoma. In vitro studies on hop(Tum-l) lamellocytes show that VLP-containing fluid from either wasp species induces lamellocyte lysis, but with different kinetics. Previously undocumented precursors of L. victoriae VLPs are synthesized in the long gland and are first visible within canals connecting secretory cells to the long gland lumen. VLP assembly occurs in the lumen. VLPs show multiple electron-dense projections surrounding a central core. Maturing particles appear segmented, singly or in arrays, embedded in the reservoir matrix. In sections, mature particles are pentagonal or hexagonal; the polygon vertices extending into spikes. Our results suggest that L. victoriae is likely to promote immune suppression by an active mechanism that is mediated by VLPs, similar to that used by L. heterotoma.


Subject(s)
Drosophila/parasitology , Hemocytes/drug effects , Immune Tolerance/immunology , Insect Proteins/biosynthesis , Wasps/metabolism , Animals , Drosophila/immunology , Exocrine Glands/anatomy & histology , Exocrine Glands/metabolism , Female , Histological Techniques , Immune Tolerance/drug effects , Insect Proteins/immunology , Insect Proteins/toxicity , Larva/immunology , Larva/parasitology , Species Specificity , Wasps/immunology
SELECTION OF CITATIONS
SEARCH DETAIL