Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Leukemia ; 38(2): 291-301, 2024 02.
Article in English | MEDLINE | ID: mdl-38182819

ABSTRACT

Internal tandem duplication mutations in fms-like tyrosine kinase 3 (FLT3-ITD) are recurrent in acute myeloid leukemia (AML) and increase the risk of relapse. Clinical responses to FLT3 inhibitors (FLT3i) include myeloid differentiation of the FLT3-ITD clone in nearly half of patients through an unknown mechanism. We identified enhancer of zeste homolog 2 (EZH2), a component of polycomb repressive complex 2 (PRC2), as a mediator of this effect using a proteomic-based screen. FLT3i downregulated EZH2 protein expression and PRC2 activity on H3K27me3. FLT3-ITD and loss-of-function mutations in EZH2 are mutually exclusive in human AML. We demonstrated that FLT3i increase myeloid maturation with reduced stem/progenitor cell populations in murine Flt3-ITD AML. Combining EZH1/2 inhibitors with FLT3i increased terminal maturation of leukemic cells and reduced leukemic burden. Our data suggest that reduced EZH2 activity following FLT3 inhibition promotes myeloid differentiation of FLT3-ITD leukemic cells, providing a mechanistic explanation for the clinical observations. These results demonstrate that in addition to its known cell survival and proliferation signaling, FLT3-ITD has a second, previously undefined function to maintain a myeloid stem/progenitor cell state through modulation of PRC2 activity. Our findings support exploring EZH1/2 inhibitors as therapy for FLT3-ITD AML.


Subject(s)
Leukemia, Myeloid, Acute , Protein-Tyrosine Kinases , Humans , Animals , Mice , Protein-Tyrosine Kinases/genetics , Polycomb Repressive Complex 2/genetics , Proteomics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/therapeutic use
2.
Genes Dev ; 36(11-12): 752-763, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35738678

ABSTRACT

Self-renewal of spermatogonial stem cells is vital to lifelong production of male gametes and thus fertility. However, the underlying mechanisms remain enigmatic. Here, we show that DOT1L, the sole H3K79 methyltransferase, is required for spermatogonial stem cell self-renewal. Mice lacking DOT1L fail to maintain spermatogonial stem cells, characterized by a sequential loss of germ cells from spermatogonia to spermatids and ultimately a Sertoli cell only syndrome. Inhibition of DOT1L reduces the stem cell activity after transplantation. DOT1L promotes expression of the fate-determining HoxC transcription factors in spermatogonial stem cells. Furthermore, H3K79me2 accumulates at HoxC9 and HoxC10 genes. Our findings identify an essential function for DOT1L in adult stem cells and provide an epigenetic paradigm for regulation of spermatogonial stem cells.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Spermatogonia , Stem Cells , Animals , Cell Differentiation , Male , Mice , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism
3.
Mol Cell ; 81(11): 2332-2348.e9, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33974912

ABSTRACT

Meningioma-1 (MN1) overexpression in AML is associated with poor prognosis, and forced expression of MN1 induces leukemia in mice. We sought to determine how MN1 causes AML. We found that overexpression of MN1 can be induced by translocations that result in hijacking of a downstream enhancer. Structure predictions revealed that the entire MN1 coding frame is disordered. We identified the myeloid progenitor-specific BAF complex as the key interaction partner of MN1. MN1 over-stabilizes BAF on enhancer chromatin, a function directly linked to the presence of a long polyQ-stretch within MN1. BAF over-stabilization at binding sites of transcription factors regulating a hematopoietic stem/progenitor program prevents the developmentally appropriate decommissioning of these enhancers and results in impaired myeloid differentiation and leukemia. Beyond AML, our data detail how the overexpression of a polyQ protein, in the absence of any coding sequence mutation, can be sufficient to cause malignant transformation.


Subject(s)
Carcinogenesis/genetics , DNA Helicases/genetics , Intrinsically Disordered Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Base Sequence , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Chromatin/pathology , DNA Helicases/metabolism , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Leukemic , Gene Regulatory Networks , Humans , Intrinsically Disordered Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Protein Interaction Mapping , Protein Stability , Protein Transport , Signal Transduction , Survival Analysis , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
4.
Leukemia ; 35(5): 1405-1417, 2021 05.
Article in English | MEDLINE | ID: mdl-33542482

ABSTRACT

Translocations of Meningioma-1 (MN1) occur in a subset of acute myeloid leukemias (AML) and result in high expression of MN1, either as a full-length protein, or as a fusion protein that includes most of the N-terminus of MN1. High levels of MN1 correlate with poor prognosis. When overexpressed in murine hematopoietic progenitors, MN1 causes an aggressive AML characterized by an aberrant myeloid precursor-like gene expression program that shares features of KMT2A-rearranged (KMT2A-r) leukemia, including high levels of Hoxa and Meis1 gene expression. Compounds that target a critical KMT2A-Menin interaction have proven effective in KMT2A-r leukemia. Here, we demonstrate that Menin (Men1) is also critical for the self-renewal of MN1-driven AML through the maintenance of a distinct gene expression program. Genetic inactivation of Men1 led to a decrease in the number of functional leukemia-initiating cells. Pharmacologic inhibition of the KMT2A-Menin interaction decreased colony-forming activity, induced differentiation programs in MN1-driven murine leukemia and decreased leukemic burden in a human AML xenograft carrying an MN1-ETV6 translocation. Collectively, these results nominate Menin inhibition as a promising therapeutic strategy in MN1-driven leukemia.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Leukemic/genetics , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Mice, Knockout
5.
Blood Adv ; 4(13): 3109-3122, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32634241

ABSTRACT

Understanding mechanisms of cooperation between oncogenes is critical for the development of novel therapies and rational combinations. Acute myeloid leukemia (AML) cells with KMT2A-fusions and KMT2A partial tandem duplications (KMT2APTD) are known to depend on the histone methyltransferase DOT1L, which methylates histone 3 lysine 79 (H3K79). About 30% of KMT2APTD AMLs carry mutations in IDH1/2 (mIDH1/2). Previous studies showed that 2-hydroxyglutarate produced by mIDH1/2 increases H3K79 methylation, and mIDH1/2 patient samples are sensitive to DOT1L inhibition. Together, these findings suggested that stabilization or increases in H3K79 methylation associated with IDH mutations support the proliferation of leukemias dependent on this mark. However, we found that mIDH1/2 and KMT2A alterations failed to cooperate in an experimental model. Instead, mIDH1/2 and 2-hydroxyglutarate exert toxic effects, specifically on KMT2A-rearranged AML cells (fusions/partial tandem duplications). Mechanistically, we uncover an epigenetic barrier to efficient cooperation; mIDH1/2 expression is associated with high global histone 3 lysine 79 dimethylation (H3K79me2) levels, whereas global H3K79me2 is obligate low in KMT2A-rearranged AML. Increasing H3K79me2 levels, specifically in KMT2A-rearrangement leukemias, resulted in transcriptional downregulation of KMT2A target genes and impaired leukemia cell growth. Our study details a complex genetic and epigenetic interaction of 2 classes of oncogenes, IDH1/2 mutations and KMT2A rearrangements, that is unexpected based on the high percentage of IDH mutations in KMT2APTD AML. KMT2A rearrangements are associated with a trend toward lower response rates to mIDH1/2 inhibitors. The substantial adaptation that has to occur for 2 initially counteracting mutations to be tolerated within the same leukemic cell may provide at least a partial explanation for this observation.


Subject(s)
Gene Rearrangement , Leukemia, Myeloid, Acute , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Methylation , Oncogenes
6.
Exp Hematol ; 85: 57-69, 2020 05.
Article in English | MEDLINE | ID: mdl-32437908

ABSTRACT

Inhibition of the H3K79 histone methyltransferase DOT1L has exhibited encouraging preclinical and early clinical activity in KMT2A (MLL)-rearranged leukemia, supporting the development of combinatorial therapies. Here, we investigated two novel combinations: dual inhibition of the histone methyltransferases DOT1L and EZH2, and the combination with a protein synthesis inhibitor. EZH2 is the catalytic subunit in the polycomb repressive complex 2 (PRC2), and inhibition of EZH2 has been reported to have preclinical activity in KMT2A-r leukemia. When combined with DOT1L inhibition, however, we observed both synergistic and antagonistic effects. Interestingly, antagonistic effects were not due to PRC2-mediated de-repression of HOXA9. HOXA cluster genes are key canonical targets of both KMT2A and the PRC2 complex. The independence of the HOXA cluster from PRC2 repression in KMT2A-r leukemia thus affords important insights into leukemia biology. Further studies revealed that EZH2 inhibition counteracted the effect of DOT1L inhibition on ribosomal gene expression. We thus identified a previously unrecognized role of DOT1L in regulating protein production. Decreased translation was one of the earliest effects measurable after DOT1L inhibition and specific to KMT2A-rearranged cell lines. H3K79me2 chromatin immunoprecipitation sequencing patterns over ribosomal genes were similar to those of the canonical KMT2A-fusion target genes in primary AML patient samples. The effects of DOT1L inhibition on ribosomal gene expression prompted us to evaluate the combination of EPZ5676 with a protein translation inhibitor. EPZ5676 was synergistic with the protein translation inhibitor homoharringtonine (omacetaxine), supporting further preclinical/clinical development of this combination. In summary, we discovered a novel epigenetic regulation of a metabolic process-protein synthesis-that plays a role in leukemogenesis and affords a combinatorial therapeutic opportunity.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Gene Rearrangement , Leukemia, Myeloid, Acute/metabolism , Protein Biosynthesis , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
7.
Front Immunol ; 11: 593321, 2020.
Article in English | MEDLINE | ID: mdl-33584657

ABSTRACT

Acute graft-versus-host disease (aGvHD) is a severe and often life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). AGvHD is mediated by alloreactive donor T-cells targeting predominantly the gastrointestinal tract, liver, and skin. Recent work in mice and patients undergoing allo-HCT showed that alloreactive T-cells can be identified by the expression of α4ß7 integrin on T-cells even before manifestation of an aGvHD. Here, we investigated whether the detection of a combination of the expression of T-cell surface markers on peripheral blood (PB) CD8+ T-cells would improve the ability to predict aGvHD. To this end, we employed two independent preclinical models of minor histocompatibility antigen mismatched allo-HCT following myeloablative conditioning. Expression profiles of integrins, selectins, chemokine receptors, and activation markers of PB donor T-cells were measured with multiparameter flow cytometry at multiple time points before the onset of clinical aGvHD symptoms. In both allo-HCT models, we demonstrated a significant upregulation of α4ß7 integrin, CD162E, CD162P, and conversely, a downregulation of CD62L on donor T-cells, which could be correlated with the development of aGvHD. Other surface markers, such as CD25, CD69, and CC-chemokine receptors were not found to be predictive markers. Based on these preclinical data from mouse models, we propose a surface marker panel on peripheral blood T-cells after allo-HCT combining α4ß7 integrin with CD62L, CD162E, and CD162P (cutaneous lymphocyte antigens, CLA, in humans) to identify patients at risk for developing aGvHD early after allo-HCT.


Subject(s)
Biomarkers , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Acute Disease , Animals , Antigens, CD/metabolism , Biopsy , Disease Models, Animal , Disease Susceptibility , Female , Graft vs Host Disease/diagnosis , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Prognosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation, Homologous
8.
Exp Hematol ; 44(7): 596-602.e3, 2016 07.
Article in English | MEDLINE | ID: mdl-27033163

ABSTRACT

Transient potential receptor melastatin-2 (TRPM2) is a nonselective cationic, Ca(2+)-permeable transmembrane pore that is preferentially expressed in cells of the myeloid lineage and modulates signaling pathways converging into NF-kB. This is of potential interest for acute myeloid leukemia (AML) therapy, as NF-κB signaling is emerging as a key pathway, mediating drug resistance and leukemia-initiating cell survival in AML. Inhibition of NF-κB signaling has been found to be synergistic with chemotherapy. TRPM2 is overexpressed in AML compared with normal bone marrow, with the highest levels in the FAB M3-6 subtypes. To determine the effect of TRPM2 depletions in a defined genetic model, we established MLL-AF9-driven AML on a Trpm2(-/-) genetic background. Trpm2(-/-) MLL-AF9 leukemias displayed reduced NF-κB phosphorylation as well as nuclear translocation. In vivo, primary and secondary recipients of Trpm2(-/-) MLL-AF9 leukemias exhibit increased latency compared with recipients of wild-type leukemia cells. However, the difference in latency was small and was lost in tertiary transplants. The effect of loss of Trpm2 in a BCR-ABL/NUP98-HOXA9 fusion model was even smaller. Given reports that loss or inhibition of TRPM2 enhanced killing by DNA-damaging agents in neuroblastoma, breast cancer, and prostate cancer cell lines, we exposed Trpm2(-/-) and Trpm2(wt) primary MLL-AF9 leukemias to doxorubicin, cytarabine, and etoposide, but found no difference in IC50 values. The in vitro response to decitabine was also unaffected. In summary, Trpm2 does not seem to play a major role in myeloid leukemogenesis. Additionally, loss of Trpm2 does not augment the cytotoxicity of standard AML chemotherapeutic agents.


Subject(s)
Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , TRPM Cation Channels/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Prognosis , TRPM Cation Channels/metabolism
9.
J Clin Invest ; 126(4): 1438-50, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26927674

ABSTRACT

Meningioma-1 (MN1) overexpression is frequently observed in patients with acute myeloid leukemia (AML) and is predictive of poor prognosis. In murine models, forced expression of MN1 in hematopoietic progenitors induces an aggressive myeloid leukemia that is strictly dependent on a defined gene expression program in the cell of origin, which includes the homeobox genes Hoxa9 and Meis1 as key components. Here, we have shown that this program is controlled by two histone methyltransferases, MLL1 and DOT1L, as deletion of either Mll1 or Dot1l in MN1-expressing cells abrogated the cell of origin-derived gene expression program, including the expression of Hoxa cluster genes. In murine models, genetic inactivation of either Mll1 or Dot1l impaired MN1-mediated leukemogenesis. We determined that HOXA9 and MEIS1 are coexpressed with MN1 in a subset of clinical MN1hi leukemia, and human MN1hi/HOXA9hi leukemias were sensitive to pharmacologic inhibition of DOT1L. Together, these data point to DOT1L as a potential therapeutic target in MN1hi AML. In addition, our findings suggest that epigenetic modulation of the interplay between an oncogenic lesion and its cooperating developmental program has therapeutic potential in AML.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/metabolism , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Female , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Methyltransferases/genetics , Mice , Mice, Knockout , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins/genetics , Trans-Activators , Tumor Suppressor Proteins/genetics
10.
Cell Rep ; 14(8): 1953-65, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26904942

ABSTRACT

Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) is an aggressive subtype of ALL distinguished by stem-cell-associated and myeloid transcriptional programs. Inactivating alterations of Polycomb repressive complex 2 components are frequent in human ETP-ALL, but their functional role is largely undefined. We have studied the involvement of Ezh2 in a murine model of NRASQ61K-driven leukemia that recapitulates phenotypic and transcriptional features of ETP-ALL. Homozygous inactivation of Ezh2 cooperated with oncogenic NRASQ61K to accelerate leukemia onset. Inactivation of Ezh2 accentuated expression of genes highly expressed in human ETP-ALL and in normal murine early thymic progenitors. Moreover, we found that Ezh2 contributes to the silencing of stem-cell- and early-progenitor-cell-associated genes. Loss of Ezh2 also resulted in increased activation of STAT3 by tyrosine 705 phosphorylation. Our data mechanistically link Ezh2 inactivation to stem-cell-associated transcriptional programs and increased growth/survival signaling, features that convey an adverse prognosis in patients.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Leukemic , Genes, ras , Polycomb Repressive Complex 2/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , STAT3 Transcription Factor/genetics , Animals , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein/deficiency , Histones/genetics , Histones/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Mice , Mice, Transgenic , Phosphorylation , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/metabolism , Precursor Cells, T-Lymphoid/metabolism , Precursor Cells, T-Lymphoid/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic
11.
Pharmacol Ther ; 154: 87-109, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26210403

ABSTRACT

While DNA abnormalities have long been recognized as the cause of cancer, the contribution of chromatin is a relatively recent discovery. Excitement in the field of cancer epigenetics is driven by 3 key elements: 1. Chromatin may play an active and often critical role in controlling gene expression, DNA stability and cell identity. 2. Chromatin modifiers are frequent targets of DNA aberrations, in some cancers reaching near 100%. Particularly in cancers with low rates of DNA mutations, the key "driver" of malignancy is often a chromatin modifier. 3. Cancer-associated aberrant chromatin is amenable to pharmacologic modulation. This has sparked the rapidly expanding development of small molecules targeting chromatin modifiers or reader domains, several of which have shown promise in clinical trials. In parallel, technical advances have greatly enhanced our ability to perform comprehensive chromatin/histone profiling. Despite the discovery that distinct histone profiles are associated with prognostic subgroups, and in some instances may point towards an underlying aberration that can be targeted, histone profiling has not entered clinical diagnostics. Even eligibility for clinical trials targeting chromatin hinges on traditional histologic or DNA-based molecular criteria rather than chromatin profiles. This review will give an overview of the philosophical debate around the role of histones in controlling or modulating gene expression and discuss the most common techniques for histone profiling. In addition, we will provide prominent examples of aberrantly expressed or mutated chromatin modifiers that result in either globally or locally aberrant histone profiles, and that may be promising therapeutic targets.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Histones/genetics , Histones/metabolism , Neoplasms/genetics , Chromatin Assembly and Disassembly/physiology , Epigenesis, Genetic/physiology , Gene Expression , Humans , Immunohistochemistry , Mass Spectrometry , Models, Biological , Polycomb Repressive Complex 2/metabolism , Sequence Analysis, Protein , Transcription Factors/metabolism , Transcription, Genetic
12.
MAbs ; 6(1): 297-308, 2014.
Article in English | MEDLINE | ID: mdl-24135629

ABSTRACT

Expression of fibroblast growth factor (FGF)-inducible 14 (Fn14), a member of the tumor necrosis factor receptor superfamily, is typically low in healthy adult organisms, but strong Fn14 expression is induced in tissue injury and tissue remodeling. High Fn14 expression is also observed in solid tumors, which is why this receptor is under consideration as a therapeutic target in oncology. Here, we describe various novel mouse-human cross-reactive llama-derived recombinant Fn14-specific antibodies (5B6, 18D1, 4G5) harboring the human IgG1 Fc domain. In contrast to recombinant variants of the established Fn14-specific antibodies PDL192 and P4A8, all three llama-derived antibodies efficiently bound to the W42A and R56P mutants of human Fn14. 18D1 and 4G5, but not 5B6, efficiently blocked TNF-like weak inducer of apoptosis(TWEA K) binding at low concentrations (0.2­2 µg/ml). Oligomerization and Fcγ receptor (FcγR) binding converted all antibodies into strong Fn14 agonists. Variants of 18D1 with enhanced and reduced antibody-dependent cell-mediated cytotoxicity (ADCC) activity were further analyzed in vivo with respect to their effect on metastasis. In a xenogeneic model using human colon carcinoma cancer cells, both antibody variants were effective in reducing metastasis to the liver. In contrast, only the 18D1 variant with enhanced ADCC activity, but not its ADCC-defective counterpart, suppressed lung metastasis in the RE NCA model. In sum, this suggests that Fn14 targeting might primarily act by triggering of antibody effector functions, but also by blockade of TWEA K-Fn14 interaction in some cases


Subject(s)
Antibodies, Neoplasm/pharmacology , Camelids, New World/immunology , Colonic Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Neoplasm Proteins/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antibodies, Neoplasm/immunology , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , HEK293 Cells , Heterografts , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Neoplasm Metastasis , Neoplasm Transplantation , TWEAK Receptor , Xenograft Model Antitumor Assays
13.
J Surg Res ; 185(1): 143-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23998649

ABSTRACT

BACKGROUND: Bioluminescence imaging (BLI) is an ideal tool for noninvasive, quantitative monitoring of tumor progression/regression in animal models. The effectiveness of different treatment strategies is displayed by an altered intensity of bioluminescence, demonstrating a change of the tumor burden. The aim of this study was to establish a reliable, reproducible colorectal hepatic metastases cancer animal model. METHODS: Cells of the human colon carcinoma cell line HCT-116 Luc(pos) expressing the firefly luciferase enzyme gene were used. HCT-116 Luc(pos) cells (2.5 × 10(6)) were injected through the portal vein into the liver of immunoincompetent nude mice. BLI was used to analyze intrahepatic tumor burden and growth kinetic. RESULTS: HCT-116 Luc(pos) cells demonstrated a progressive and reproducible growth in the liver after intraportal injection. Four days after injection, the animals were analyzed for tumor growth by BLI, and mice without or too low bioluminescence signals were excluded (between 10% and 20% animals). HCT-116 Luc(pos) intrahepatic tumors responded successfully to different dosages (5 and 10 mg/kg) of 5-fluorouracil. CONCLUSIONS: BLI is an important tool with many potential advantages for investigators. The measurement of intrahepatic tumor growth by imaging luciferase activity noninvasively provides valuable information on tumor burden and effectiveness of therapy. Thus, the presented intrahepatic metastases model based on the growth of HCT-116 Luc(pos) cells is suitable for in vivo testing of different cancer therapy strategies.


Subject(s)
Colorectal Neoplasms/pathology , Liver Neoplasms, Experimental/secondary , Luminescent Measurements/methods , Tumor Burden , Animals , Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/drug therapy , Disease Models, Animal , Disease Progression , Female , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Liver/pathology , Liver Neoplasms, Experimental/drug therapy , Luciferases/genetics , Mice , Mice, Nude , Xenograft Model Antitumor Assays
14.
BMC Med ; 11: 134, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23692886

ABSTRACT

BACKGROUND: Acute graft-versus-host disease (aGVHD) poses a major limitation for broader therapeutic application of allogeneic hematopoietic cell transplantation (allo-HCT). Early diagnosis of aGVHD remains difficult and is based on clinical symptoms and histopathological evaluation of tissue biopsies. Thus, current aGVHD diagnosis is limited to patients with established disease manifestation. Therefore, for improved disease prevention it is important to develop predictive assays to identify patients at risk of developing aGVHD. Here we address whether insights into the timing of the aGVHD initiation and effector phases could allow for the detection of migrating alloreactive T cells before clinical aGVHD onset to permit for efficient therapeutic intervention. METHODS: Murine major histocompatibility complex (MHC) mismatched and minor histocompatibility antigen (miHAg) mismatched allo-HCT models were employed to assess the spatiotemporal distribution of donor T cells with flow cytometry and in vivo bioluminescence imaging (BLI). Daily flow cytometry analysis of peripheral blood mononuclear cells allowed us to identify migrating alloreactive T cells based on homing receptor expression profiles. RESULTS: We identified a time period of 2 weeks of massive alloreactive donor T cell migration in the blood after miHAg mismatch allo-HCT before clinical aGVHD symptoms appeared. Alloreactive T cells upregulated α4ß7 integrin and P-selectin ligand during this migration phase. Consequently, targeted preemptive treatment with rapamycin, starting at the earliest detection time of alloreactive donor T cells in the peripheral blood, prevented lethal aGVHD. CONCLUSIONS: Based on this data we propose a critical time frame prior to the onset of aGVHD symptoms to identify alloreactive T cells in the peripheral blood for timely and effective therapeutic intervention.


Subject(s)
Disease Models, Animal , Graft vs Host Disease/diagnosis , Graft vs Host Disease/surgery , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocyte Subsets/transplantation , Acute Disease , Animals , Blood Group Incompatibility/immunology , Female , Forecasting , Graft vs Host Disease/immunology , HLA Antigens/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology , Time Factors , Transplantation, Homologous , Treatment Outcome
15.
Carcinogenesis ; 34(6): 1296-303, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23385062

ABSTRACT

The cytokine tumor necrosis factor (TNF) has pleiotropic functions both in normal physiology and disease. TNF signals by the virtue of two cell surface receptors, TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Exogenous TNF promotes experimental metastasis in some models, yet the underlying mechanisms are poorly understood. To study the contribution of host TNFR1 and TNFR2 on tumor cell progression and metastasis, we employed a syngeneic B16F10 melanoma mouse model of lung metastasis combined with in vivo bioluminescence imaging. Treatment of tumor-bearing mice with recombinant human TNF resulted in a significant increase in tumor burden and metastatic foci. This correlated with an increase in pulmonary regulatory CD4(+)/Foxp3(+) T cells. TNF caused an expansion of regulatory T (Treg) cells in vitro in a TNFR2-dependent manner. To assess the contribution of immune cell expression of endogenous TNF and its two receptors on B16F10 metastasis, we generated bone marrow chimeras by reconstituting wild-type mice with bone marrow from different knockout mice. Loss of either TNF or TNFR2 on immune cells resulted in decreased B16F10 metastasis and lower numbers of Treg cells within the lungs of these animals. Selective depletion of Treg cells attenuated metastasis even in conjunction with TNF treatment. We propose a novel mechanism in which TNF activates TNFR2 on Treg cells and thereby expands this immunosuppressive immune cell population. Loss of either TNF or TNFR2 prevents the accumulation of Treg cells and results in a less tolerogenic environment, enabling the immune system to control B16F10 tumor metastasis and growth.


Subject(s)
Lung Neoplasms/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , CD4 Antigens/biosynthesis , Cell Line, Tumor , Cell Proliferation , Forkhead Transcription Factors/biosynthesis , Lung Neoplasms/secondary , Melanoma , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Receptors, Tumor Necrosis Factor, Type I/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
J Clin Invest ; 122(12): 4439-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143304

ABSTRACT

Understanding the spatiotemporal changes of cellular and molecular events within an organism is crucial to elucidate the complex immune processes involved in infections, autoimmune disorders, transplantation, and neoplastic transformation and metastasis. Here we introduce a novel multicolor light sheet fluorescence microscopy (LSFM) approach for deciphering immune processes in large tissue specimens on a single-cell level in 3 dimensions. We combined and optimized antibody penetration, tissue clearing, and triple-color illumination to create a method for analyzing intact mouse and human tissues. This approach allowed us to successfully quantify changes in expression patterns of mucosal vascular addressin cell adhesion molecule-1 (MAdCAM-1) and T cell responses in Peyer's patches following stimulation of the immune system. In addition, we employed LSFM to map individual T cell subsets after hematopoietic cell transplantation and detected rare cellular events. Thus, we present a versatile imaging technology that should be highly beneficial in biomedical research.


Subject(s)
Adaptive Immunity , Imaging, Three-Dimensional/methods , Animals , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Single-Cell Analysis , Whole Body Imaging
17.
PLoS One ; 7(12): e52398, 2012.
Article in English | MEDLINE | ID: mdl-23300660

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is a B-cell malignancy, where malignant plasma cells clonally expand in the bone marrow of older people, causing significant morbidity and mortality. Typical clinical symptoms include increased serum calcium levels, renal insufficiency, anemia, and bone lesions. With standard therapies, MM remains incurable; therefore, the development of new drugs or immune cell-based therapies is desirable. To advance the goal of finding a more effective treatment for MM, we aimed to develop a reliable preclinical MM mouse model applying sensitive and reproducible methods for monitoring of tumor growth and metastasis in response to therapy. MATERIAL AND METHODS: A mouse model was created by intravenously injecting bone marrow-homing mouse myeloma cells (MOPC-315.BM) that expressed luciferase into BALB/c wild type mice. The luciferase in the myeloma cells allowed in vivo tracking before and after melphalan treatment with bioluminescence imaging (BLI). Homing of MOPC-315.BM luciferase+ myeloma cells to specific tissues was examined by flow cytometry. Idiotype-specific myeloma protein serum levels were measured by ELISA. In vivo measurements were validated with histopathology. RESULTS: Strong bone marrow tropism and subsequent dissemination of MOPC-315.BM luciferase(+) cells in vivo closely mimicked the human disease. In vivo BLI and later histopathological analysis revealed that 12 days of melphalan treatment slowed tumor progression and reduced MM dissemination compared to untreated controls. MOPC-315.BM luciferase(+) cells expressed CXCR4 and high levels of CD44 and α4ß1 in vitro which could explain the strong bone marrow tropism. The results showed that MOPC-315.BM cells dynamically regulated homing receptor expression and depended on interactions with surrounding cells. CONCLUSIONS: This study described a novel MM mouse model that facilitated convenient, reliable, and sensitive tracking of myeloma cells with whole body BLI in living animals. This model is highly suitable for monitoring the effects of different treatment regimens.


Subject(s)
Disease Progression , Molecular Imaging/methods , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Hematopoiesis/drug effects , Humans , Luciferases/genetics , Melphalan/pharmacology , Melphalan/therapeutic use , Mice , Mice, Inbred BALB C , Multiple Myeloma/genetics , Neoplasm Invasiveness , Spatio-Temporal Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL