Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Blood ; 134(14): 1176-1189, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31383640

ABSTRACT

Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an "onco"-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing-based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2's gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.


Subject(s)
Carcinogenesis/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Multiple Myeloma/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Humans , Methylation , Mice , Multiple Myeloma/pathology
2.
Blood ; 128(25): 2949-2959, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27733356

ABSTRACT

To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Stem Cell Niche , Tissue Scaffolds/chemistry , Xenograft Model Antitumor Assays , Animals , Cell Self Renewal , Cell Separation , Clone Cells , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/cytology , Mice , Phenotype , Stromal Cells/pathology
3.
Mol Ther ; 25(8): 1946-1958, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28506593

ABSTRACT

Chimeric antigen receptors (CARs) can effectively redirect cytotoxic T cells toward highly expressed surface antigens on tumor cells. The low expression of several tumor-associated antigens (TAAs) on normal tissues, however, hinders their safe targeting by CAR T cells due to on-target/off-tumor effects. Using the multiple myeloma (MM)-associated CD38 antigen as a model system, here, we present a rational approach for effective and tumor-selective targeting of such TAAs. Using "light-chain exchange" technology, we combined the heavy chains of two high-affinity CD38 antibodies with 176 germline light chains and generated ∼124 new antibodies with 10- to >1,000-fold lower affinities to CD38. After categorizing them into three distinct affinity classes, we incorporated the single-chain variable fragments of eight antibodies from each class into new CARs. T cells carrying these CD38-CARs were extensively evaluated for their on-tumor/off-tumor cytotoxicity as well as CD38-dependent proliferation and cytokine production. We identified CD38-CAR T cells of ∼1,000- fold reduced affinity, which optimally proliferated, produced Th1-like cytokines, and effectively lysed CD382+ MM cells, but spared CD38+ healthy hematopoietic cells in vitro and in vivo. Thus, this systematic approach is highly suitable for the generation of optimal CARs for effective and selective targeting of TAAs.


Subject(s)
ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase 1/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , ADP-ribosyl Cyclase 1/metabolism , Animals , Antibody Affinity/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Mice , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Protein Binding/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Xenograft Model Antitumor Assays
4.
J Immunol ; 194(3): 911-20, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25520398

ABSTRACT

The effectiveness of chimeric Ag receptor (CAR)-transduced T (CAR-T) cells has been attributed to supraphysiological signaling through CARs. Second- and later-generation CARs simultaneously transmit costimulatory signals with CD3ζ signals upon ligation, but may lead to severe adverse effects owing to the recognition of minimal Ag expression outside the target tumor. Currently, the threshold target Ag density for CAR-T cell lysis and further activation, including cytokine production, has not yet been investigated in detail. Therefore, we determined the threshold target Ag density required to induce CAR-T cell responses using novel anti-CD20 CAR-T cells with a CD28 intracellular domain and a CD20-transduced CEM cell model. The newly developed CD20CAR-T cells demonstrated Ag-specific lysis and cytokine secretion, which was a reasonable level as a second-generation CAR. For lytic activity, the threshold Ag density was determined to be ∼200 molecules per target cell, whereas the Ag density required for cytokine production of CAR-T cells was ∼10-fold higher, at a few thousand per target cell. CD20CAR-T cells responded efficiently to CD20-downregulated lymphoma and leukemia targets, including rituximab- or ofatumumab-refractory primary chronic lymphocytic leukemia cells. Despite the potential influence of the structure, localization, and binding affinity of the CAR/Ag, the threshold determined may be used for target Ag selection. An Ag density below the threshold may not result in adverse effects, whereas that above the threshold may be sufficient for practical effectiveness. CD20CAR-T cells also demonstrated significant lytic activity against CD20-downregulated tumor cells and may exhibit effectiveness for CD20-positive lymphoid malignancies.


Subject(s)
Antigens, Surface/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, Surface/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Gene Expression , Gene Order , Genes, Reporter , Humans , Immunophenotyping , Intracellular Space , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphocyte Activation , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rituximab , Signal Transduction , T-Cell Antigen Receptor Specificity/genetics , T-Cell Antigen Receptor Specificity/immunology , Transduction, Genetic
5.
Haematologica ; 101(5): 616-25, 2016 05.
Article in English | MEDLINE | ID: mdl-26858358

ABSTRACT

Adoptive transfer of chimeric antigen receptor-transduced T cells is a promising strategy for cancer immunotherapy. The CD38 molecule, with its high expression on multiple myeloma cells, appears a suitable target for antibody therapy. Prompted by this, we used three different CD38 antibody sequences to generate second-generation retroviral CD38-chimeric antigen receptor constructs with which we transduced T cells from healthy donors and multiple myeloma patients. We then evaluated the preclinical efficacy and safety of the transduced T cells. Irrespective of the donor and antibody sequence, CD38-chimeric antigen receptor-transduced T cells proliferated, produced inflammatory cytokines and effectively lysed malignant cell lines and primary malignant cells from patients with acute myeloid leukemia and multi-drug resistant multiple myeloma in a cell-dose, and CD38-dependent manner, despite becoming CD38-negative during culture. CD38-chimeric antigen receptor-transduced T cells also displayed significant anti-tumor effects in a xenotransplant model, in which multiple myeloma tumors were grown in a human bone marrow-like microenvironment. CD38-chimeric antigen receptor-transduced T cells also appeared to lyse the CD38(+) fractions of CD34(+) hematopoietic progenitor cells, monocytes, natural killer cells, and to a lesser extent T and B cells but did not inhibit the outgrowth of progenitor cells into various myeloid lineages and, furthermore, were effectively controllable with a caspase-9-based suicide gene. These results signify the potential importance of CD38-chimeric antigen receptor-transduced T cells as therapeutic tools for CD38(+) malignancies and warrant further efforts to diminish the undesired effects of this immunotherapy using appropriate strategies.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Immunotherapy , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Animals , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Flow Cytometry , Gene Expression , Gene Transfer Techniques , Genes, Transgenic, Suicide , Hematopoietic Stem Cells/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Burden/genetics , Tumor Burden/immunology
6.
Cancer Immunol Immunother ; 64(8): 951-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25920521

ABSTRACT

Immunotherapy with allogeneic natural killer (NK) cells offers therapeutic perspectives for multiple myeloma patients. Here, we aimed to refine NK cell therapy by evaluation of the relevance of HLA-class I and HLA-E for NK anti-myeloma reactivity. We show that HLA-class I was strongly expressed on the surface of patient-derived myeloma cells and on myeloma cell lines. HLA-E was highly expressed by primary myeloma cells but only marginally by cell lines. HLA-E(low) expression on U266 cells observed in vitro was strongly upregulated after in vivo (bone marrow) growth in RAG-2(-/-) γc(-/-) mice, suggesting that in vitro HLA-E levels poorly predict the in vivo situation. Concurrent analysis of inhibitory receptors (KIR2DL1, KIR2DL2/3, KIR3DL1 and NKG2A) and NK cell degranulation upon co-culture with myeloma cells revealed that KIR-ligand-mismatched NK cells degranulate more than matched subsets and that HLA-E abrogates degranulation of NKG2A+ subsets. Inhibition by HLA-class I and HLA-E was also observed with IL-2-activated NK cells and at low oxygen levels (0.6 %) mimicking hypoxic bone marrow niches where myeloma cells preferentially reside. Our study demonstrates that NKG2A-negative, KIR-ligand-mismatched NK cells are the most potent subset for clinical application. We envision that infusion of high numbers of this subclass will enhance clinical efficacy.


Subject(s)
Cell Separation/methods , Histocompatibility Antigens Class I/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , NK Cell Lectin-Like Receptor Subfamily C/immunology , Animals , Cell Degranulation , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , DNA-Binding Proteins/genetics , Flow Cytometry , Humans , Interleukin-2/immunology , Mice , Mice, Knockout , Multiple Myeloma/immunology , Neoplasm Transplantation , Oxygen/metabolism , HLA-E Antigens
7.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653974

ABSTRACT

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Subject(s)
Hematopoietic Stem Cells/cytology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Stem Cell Niche/immunology , Transplantation Chimera/immunology , Tumor Microenvironment/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear Ossicles/cytology , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Mutant Strains , Neoplasm Transplantation , Osteolysis/immunology , Tissue Scaffolds , Transplantation, Heterologous
9.
Biol Blood Marrow Transplant ; 18(7): 1023-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22579931

ABSTRACT

Graft-versus-host disease (GVHD) remains a frequently occurring and difficult-to-treat complication in human allogeneic stem cell transplantation. Murine transplantation models are often used to study and understand the complex pathogenesis of GVHD and to explore new treatment strategies. Although GVHD kinetics may differ in murine and human models, adequate models are essential for identification of the crucial factors responsible for the major pathology in GVHD. We present a detailed description of the specific histological features of a graft-versus-host-induced fibrotic response in xenogeneic RAG2(-/-)γc(-/-) mice after total body irradiation and injection with human peripheral blood mononuclear cells. We describe the full morphological features of this reaction, including a detailed analysis of the specific tissue infiltration patterns of the human peripheral blood mononuclear cells. Our data show the development of fibrosis, predominantly near blood vessels, and reveal different cell populations and specific cell migration patterns in the affected organs. The combination of immunohistochemical cell characterization and mRNA expression analysis of both human (donor)- and murine (host)-derived cytokines reveals an interaction between host tissues and donor-derived cells in an entangled cytokine profile, in which both donor- and host-derived cytokines contribute to the formation of fibrosis.


Subject(s)
Blood Vessels/pathology , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation , Leukocytes, Mononuclear/transplantation , Animals , Blood Vessels/immunology , Cell Movement/immunology , Cytokines/biosynthesis , Cytokines/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Female , Graft vs Host Disease/immunology , Humans , Immunohistochemistry , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Sclerosis , Transplantation, Heterologous , Whole-Body Irradiation
10.
Blood ; 115(3): 601-4, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19965677

ABSTRACT

Expression of the heparan sulfate proteoglycan syndecan-1 is a hallmark of both normal and multiple myeloma (MM) plasma cells. Syndecan-1 could affect plasma cell fate by strengthening integrin-mediated adhesion via its core protein and/or by accommodating and presenting soluble factors via its HS side chains. Here, we show that inducible RNAi-mediated knockdown of syndecan-1 in human MM cells leads to reduced growth rates and a strong increase of apoptosis. Importantly, knockdown of EXT1, a copolymerase critical for HS chain biosynthesis, had similar effects. Using an innovative myeloma xenotransplantation model in Rag-2(-/-)gamma(c)(-/-) mice, we demonstrate that induction of EXT1 knockdown in vivo dramatically suppresses the growth of bone marrow localized myeloma. Our findings provide direct evidence that the HS chains of syndecan-1 are crucial for the growth and survival of MM cells within the bone marrow environment, and indicate the HS biosynthesis machinery as a potential treatment target in MM.


Subject(s)
Cell Proliferation/drug effects , Heparitin Sulfate/physiology , Multiple Myeloma/pathology , N-Acetylglucosaminyltransferases/genetics , RNA, Small Interfering/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Doxycycline/administration & dosage , Drug Delivery Systems , Gene Targeting , Heparitin Sulfate/metabolism , Humans , Immunoglobulin gamma-Chains/genetics , Mice , Mice, Knockout , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/physiology , Syndecan-1/genetics , Syndecan-1/metabolism , Xenograft Model Antitumor Assays
11.
Haematologica ; 96(11): 1653-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21828122

ABSTRACT

BACKGROUND: Multiple myeloma is a hematologic malignancy characterized by a clonal expansion of malignant plasma cells in the bone marrow, which is accompanied by the development of osteolytic lesions and/or diffuse osteopenia. The intricate bi-directional interaction with the bone marrow microenvironment plays a critical role in sustaining the growth and survival of myeloma cells during tumor progression. Identification and functional analysis of the (adhesion) molecules involved in this interaction will provide important insights into the pathogenesis of multiple myeloma. DESIGN AND METHODS: Multiple myeloma cell lines and patients' samples were analyzed for expression of the adhesion molecule N-cadherin by immunoblotting, flow cytometry, immunofluorescence microscopy, immunohistochemistry and expression microarray. In addition, by means of blocking antibodies and inducible RNA interference we studied the functional consequence of N-cadherin expression for the myeloma cells, by analysis of adhesion, migration and growth, and for the bone marrow microenvironment, by analysis of osteogenic differentiation. RESULTS: The malignant plasma cells in approximately half of the multiple myeloma patients, belonging to specific genetic subgroups, aberrantly expressed the homophilic adhesion molecule N-cad-herin. N-cadherin-mediated cell-substrate or homotypic cell-cell adhesion did not contribute to myeloma cell growth in vitro. However, N-cadherin directly mediated the bone marrow localization/retention of myeloma cells in vivo, and facilitated a close interaction between myeloma cells and N-cadherin-positive osteoblasts. Furthermore, this N-cadherin-mediated interaction contributed to the ability of myeloma cells to inhibit osteoblastogenesis. CONCLUSIONS: Taken together, our data show that myeloma cells frequently display aberrant expression of N-cadherin and that N-cadherin mediates the interaction of myeloma cells with the bone marrow microenvironment, in particular the osteoblasts. This N-cadherin-mediated interaction inhibits osteoblast differentiation and may play an important role in the pathogenesis of myeloma bone disease.


Subject(s)
Cadherins/metabolism , Cell Communication , Cell Differentiation , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Osteoblasts/metabolism , Tumor Microenvironment , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cadherins/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Osteoblasts/pathology
13.
Haematologica ; 95(12): 2063-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20851867

ABSTRACT

BACKGROUND: Incorporation of the chimeric CD20 monoclonal antibody rituximab in the treatment schedule of patients with non-Hodgkin's lymphoma has significantly improved outcome. Despite this success, about half of the patients do not respond to treatment or suffer from a relapse and additional therapy is required. A low CD20-expression level may in part be responsible for resistance against rituximab. We therefore investigated whether the CD20-expression level related resistance to rituximab could be overcome by a new group of CD20 mAbs (HuMab-7D8 and ofatumumab) targeting a unique membrane-proximal epitope on the CD20 molecule. DESIGN AND METHODS: By retroviral transduction of the CD20 gene into CD20-negative cells and clonal selection of transduced cells a system was developed in which the CD20-expression level is the only variable. These CD20 transduced cells were used to study the impact of rituximab and HuMab-7D8 mediated complement-dependent cytotoxicity. To study the in vivo efficacy of these mAbs an in vivo imaging system was generated by retroviral expression of the luciferase gene in the CD20-positive cells. RESULTS: We show that HuMab-7D8 efficiently killed CD20(low) cells that are not susceptible to rituximab-induced killing in vitro. In a mouse xenograft model, we observed a comparable increase in survival time between HuMab-7D8 and rituximab-treated mice. Most significantly, however, HuMab-7D8 eradicated all CD20-expressing cells both in the periphery as well as in the bone marrow whereas after rituximab treatment CD20(low) cells survived. CONCLUSIONS: Cells that are insensitive to in vitro and in vivo killing by rituximab as the result of their low CD20-expression profile may be efficiently killed by an antibody against the membrane-proximal epitope on CD20. Such antibodies should, therefore, be explored to overcome rituximab resistance in the clinic.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD20/metabolism , Cytotoxicity, Immunologic/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, CD20/genetics , Antigens, CD20/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Epitopes/immunology , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Membrane Microdomains/metabolism , Mice , Protein Transport , Rituximab , Xenograft Model Antitumor Assays
14.
Cell Microbiol ; 11(5): 825-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19215224

ABSTRACT

Bioluminescence imaging (BLI) is a powerful new method to study virus dissemination in the live animal. Here we used this method to monitor the spatial and temporal progression of mouse hepatitis coronavirus (MHV) infection in mice using luciferase-expressing viruses. Upon intranasal inoculation, virus replication could initially be observed in the nasal cavity and the cervical lymph nodes, after which the infection spread to the brain and frequently to the eyes. The kinetics of virus spread to and clearance from the brain appeared to depend on the inoculation dose. After intraperitoneal inoculation, virus replication was predominantly observed in the liver and occasionally in the intestines, but interestingly also in the tail and paws. BLI thus elucidated new anatomic locations of virus replication. Furthermore, MHV dissemination was shown to be critically depended on the viral spike protein, but also on the mouse strain used. Widespread dissemination was observed in mice lacking a functional type I interferon response. The importance of the type I interferon system in limiting viral spread was also demonstrated by the administration of type I interferons to mice. Our results provide new insights in coronavirus pathogenesis and demonstrate the potential of BLI to study coronavirus-host interactions in vivo.


Subject(s)
Coronavirus Infections/virology , Murine hepatitis virus/physiology , Virus Replication , Animals , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Diagnostic Imaging/methods , Heparitin Sulfate/pharmacology , Interferon Type I/pharmacology , Luminescent Proteins/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Murine hepatitis virus/drug effects , Murine hepatitis virus/pathogenicity , Recombinant Proteins
15.
Calcif Tissue Int ; 85(5): 434-43, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19816649

ABSTRACT

Multiple myeloma (MM) is an incurable B-cell neoplasia in which progressive skeletal lesions are a characteristic feature. Earlier we established an animal model for human MM in the immune-deficient RAG2(-/-)gammac(-/-) mouse, in which the growth of luciferase-transduced MM cells was visualized using noninvasive bioluminescence imaging (BLI). This model appeared well suited to study disease progression and response to therapy by identifying the location of various foci of MM tumor growth scattered throughout the skeleton and at subsequent time points the quantitative assessment of the tumor load by using BLI. We report here on the corresponding high-resolution X-ray micro-computed tomographic (micro-CT) analysis to study skeletal defects in the mice with full-blown MM. Several anatomical derangements were observed, including abnormalities in geometry and morphology, asymmetrical bone structures, decreased overall density in the remaining bone, loss of trabecular bone mass, destruction of the inner microarchitecture, as well as cortical perforations. Using the combination of BLI, micro-CT imaging, and immune-histopathological techniques, we found a high correlation between the micro-CT-identified lesions, exact tumor location, and infiltration leading to structural lesions and local bone deformation. This confirms that this animal model strongly resembles human MM and has the potential for studying the biology of MM growth and for preclinical testing of novel therapies for MM and for repair of MM-induced bone lesions.


Subject(s)
Disease Models, Animal , Multiple Myeloma/diagnostic imaging , X-Ray Microtomography , Animals , Cell Line, Tumor , Humans , Luciferases , Luminescent Measurements , Mice , Multiple Myeloma/pathology , Neoplasm Transplantation , Sensitivity and Specificity , Tumor Burden
16.
J Immunol Res ; 2019: 3538963, 2019.
Article in English | MEDLINE | ID: mdl-31205954

ABSTRACT

Humanized mouse models can well be modified to study specific aspects of Graft-versus-Host Disease (GvHD). This paper shows the results of both macrophage depletion and (early) B-cell depletion in a humanized mouse model using RAG2-/- γc-/- mice injected with HuPBMCs. Macrophage depletion showed a significant decrease in survival and also lead to a change in the histomorphology of the xenogeneic reaction. Higher levels of infiltrating B-cells were observed in various organs of mice depleted for macrophages. With (early) B-cell depletion using Rituximab, a clear improvement on clinical symptoms was observed, even when probably only inactivated B-cells were deleted. However, the histological examinations only showed a significant morphological effect on liver fibrosis. This may be related to a difference in the mRNA levels of TGF-ß. Also, lower mRNA levels of Tregs in some organs were observed after Rituximab treatment, which contradicts that a higher number of Tregs would always be related to less severe GvHD. Our data show that both macrophage depletion and (early) B-cell depletion in a xenogeneic mouse model can influence the clinical, histological, and cytokine production of a GvHD response.


Subject(s)
B-Lymphocytes/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Leukocytes, Mononuclear/immunology , Liver Cirrhosis/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Female , Humans , Lymphocyte Depletion , Mice , Mice, SCID
17.
J Control Release ; 296: 232-240, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30682443

ABSTRACT

Glucocorticoids are the cornerstone in the clinic for treatment of hematological malignancies, including multiple myeloma. Nevertheless, poor pharmacokinetic properties of glucocorticoids require high and frequent dosing with the off-target adverse effects defining the maximum dose. Recently, nanomedicine formulations of glucocorticoids have been developed that improve the pharmacokinetic profile, limit adverse effects and improve solid tumor accumulation. Multiple myeloma is a hematological malignancy characterized by uncontrolled growth of plasma cells. These tumors initiate increased angiogenesis and microvessel density in the bone marrow, which might be exploited using nanomedicines, such as liposomes. Nano-sized particles can accumulate as a result of the increased vascular leakiness at the bone marrow tumor lesions. Pre-clinical screening of novel anti-myeloma therapeutics in vivo requires a suitable animal model that represents key features of the disease. In this study, we show that fluorescently labeled long circulating liposomes were found in plasma up to 24 h after injection in an advanced human-mouse hybrid model of multiple myeloma. Besides the organs involved in clearance, liposomes were also found to accumulate in tumor bearing human-bone scaffolds. The therapeutic efficacy of liposomal dexamethasone phosphate was evaluated in this model showing strong tumor growth inhibition while free drug being ineffective at an equivalent dose (4 mg/kg) regimen. The liposomal formulation slightly reduced total body weight of myeloma-bearing mice during the course of treatment, which appeared reversible when treatment was stopped. Liposomal dexamethasone could be further developed as monotherapy or could fit in with existing therapy regimens to improve therapeutic outcomes for multiple myeloma.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Multiple Myeloma/drug therapy , Animals , Body Weight/drug effects , Bone and Bones/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Liposomes , Mice, Knockout , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tissue Distribution , Tumor Burden/drug effects
18.
Haematologica ; 93(7): 1049-57, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18492693

ABSTRACT

BACKGROUND: The development and preclinical testing of novel immunotherapy strategies for multiple myeloma can benefit substantially from a humanized animal model that enables quantitative real-time monitoring of tumor progression. Here we have explored the feasibility of establishing such a model in immunodeficient RAG2(-/-)gammac(-/-) mice, by utilizing non-invasive bioluminescent imaging for real-time monitoring of multiple myeloma cell growth. DESIGN AND METHODS: Seven multiple myeloma cell lines, marked with a green fluorescent protein firefly luciferase fusion gene, were intravenously injected into RAG2(-/-)gammac(-/-) mice. Tumor localization and outgrowth was monitored by bioluminescent imaging. The sensitivity of this imaging technique was compared to that of free immumoglobulin light chain -based myeloma monitoring. Established tumors were treated with radiotherapy or with allogeneic peripheral blood mononuclear cell infusions to evaluate the application areas of the model. RESULTS: Five out of seven tested multiple myeloma cell lines progressed as myeloma-like tumors predominantly in the bone marrow; the two other lines showed additional growth in soft tissues. In our model bioluminescent imaging appeared superior to free light chain-based monitoring and also allowed semi-quantitative monitoring of individual foci of multiple myeloma. Tumors treated with radiotherapy showed temporary regression. However, infusion of allogeneic peripheral blood mononuclear cells resulted in the development of xenogeneic graft-versus-host-disease and a powerful cell dose-dependent graft-versus-myeloma effect, resulting in complete eradication of tumors, depending on the in vitro immunogenicity of the inoculated multiple myeloma cells. CONCLUSIONS: Our results indicate that this new model allows convenient and sensitive real-time monitoring of cellular approaches for immunotherapy of multiple myeloma-like tumors with different immunogenicities. This model, therefore, allows comprehensive preclinical evaluation of novel combination therapies for multiple myeloma.


Subject(s)
Graft vs Tumor Effect , Immunotherapy/methods , Multiple Myeloma/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Disease Progression , Humans , Luminescent Proteins/chemistry , Mice , Mice, Transgenic , Multiple Myeloma/immunology , Neoplasm Transplantation , Retroviridae/metabolism , Treatment Outcome
19.
Exp Hematol ; 35(1): 117-27, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198880

ABSTRACT

OBJECTIVE: Recent clinical trials of adoptive immunotherapy showed diminished reactivity of human T cells upon ex vivo manipulation. For a safe and effective clinical application of human T cells, it is necessary to improve ex vivo manipulation procedures and evaluate their impact on in vivo functionality. However, there is no preclinical model for quantitative assessment of in vivo functionality of human T cells. In this study, we investigated the feasibility of using the huPBMC- RAG2(-/-)gammac(-/-) xenogeneic mouse model. As a first example, we compared 3 different ex vivo culture conditions for human T cells. METHODS: RAG2(-/-)gammac(-/-) mice received cultured human T cells that were stimulated via CD3 alone or costimulated via CD28 (CD3/28) and/or human 4-1BB (CD3/28/4-1BB). Engraftment levels and survival of the cells were measured. The dynamics of the human T cell phenotypes were analyzed during culture and in vivo, as well as the mechanism of the xenoresponse. RESULTS: Engraftment potential was improved twofold for costimulation compared to CD3 alone (p < 0.001). Phenotypic analysis showed a strikingly similar pattern of development towards CD4(+) and CD8(+) effector and effector-memory cells, suggesting antigen-driven survival and expansion. All parameters used to analyze different effects on in vivo T-cell functionality, like culture condition, engraftment levels, survival of the cells over time, or xenogeneic graft-vs-host disease were absolutely independent of the distribution of the T cell population in vivo following contact with xeno-antigen. CONCLUSION: The huPBMC-RAG2(-/-)gammac(-/-) xenogeneic transplant model is the most sensitive to date for in vivo functional evaluation of human T cells.


Subject(s)
DNA-Binding Proteins/deficiency , Immunotherapy, Adoptive/methods , T-Lymphocytes/cytology , T-Lymphocytes/transplantation , Animals , Cell Culture Techniques/methods , Cell Survival , Graft Survival , Graft vs Host Disease , Humans , Immunoglobulin gamma-Chains/genetics , Lymphocyte Transfusion/methods , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets
20.
J Tissue Eng Regen Med ; 10(3): 233-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-23255230

ABSTRACT

The combination of scaffolds and mesenchymal stromal cells (MSCs) is a promising approach in bone tissue engineering (BTE). Knowledge on the survival, outgrowth and bone-forming capacity of MSCs in vivo is limited. Bioluminescence imaging (BLI), histomorphometry and immunohistochemistry were combined to study the fate of gene-marked goat and human MSCs (gMSCs, hMSCs) on scaffolds with different osteoinductive properties. Luciferase-GFP-labelled MSCs were seeded on hydroxyapatite (HA) or ß-tricalcium phosphate (TCP), cultured for 7 days in vitro in osteogenic medium, implanted subcutaneously in immunodeficient mice and monitored with BLI for 6 weeks. The constructs were retrieved and processed for histomorphometry and detection of luciferase-positive cells (LPCs). For gMSCs, BLI revealed doubling of signal after 1 week, declining to 60% of input after 3 weeks and remaining constant until week 6. hMSCs showed a constant decrease of BLI signal to 25% of input, indicating no further expansion. Bone formation of gMSCs was two-fold higher on TCP than HA. hMSCs and gMSCs control samples produced equal amounts of bone on TCP. Upon transduction, there was a four-fold reduction in bone formation compared with untransduced hMSCs, and no bone was formed on HA. LPCs were detected at day 14, but were much less frequent at day 42. Striking differences were observed in spatial distribution. MSCs in TCP were found to be aligned and interconnected on the surface but were scattered in an unstructured fashion in HA. In conclusion, the spatial distribution of MSCs on the scaffold is critical for cell-scaffold-based BTE.


Subject(s)
Calcium Phosphates/pharmacology , Durapatite/pharmacology , Mesenchymal Stem Cells/cytology , Animals , Cell Survival/drug effects , DNA/metabolism , Goats , Humans , Immunohistochemistry , Luciferases/metabolism , Luminescent Measurements , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Osteogenesis/drug effects , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL