Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cancers (Basel) ; 13(8)2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33921898

ABSTRACT

The prognosis of multiple myeloma (MM), an incurable B-cell malignancy, has significantly improved through the introduction of novel therapeutic modalities. Myeloma prognosis is essentially determined by cytogenetics, both at diagnosis and at disease progression. However, for a large cohort of patients, cytogenetic analysis is not always available. In addition, myeloma patients with favorable cytogenetics can display an aggressive clinical course. Therefore, it is necessary to develop additional prognostic and predictive markers for this disease to allow for patient risk stratification and personalized clinical decision-making. Genomic instability is a prominent characteristic in MM, and we have previously shown that the three-dimensional (3D) nuclear organization of telomeres is a marker of both genomic instability and genetic heterogeneity in myeloma. In this study, we compared in a longitudinal prospective study blindly the 3D telomeric profiles from bone marrow samples of 214 initially treatment-naïve patients with either monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), or MM, with a minimum follow-up of 5 years. Here, we report distinctive 3D telomeric profiles correlating with disease aggressiveness and patient response to treatment in MM patients, and also distinctive 3D telomeric profiles for disease progression in smoldering multiple myeloma patients. In particular, lower average intensity (telomere length, below 13,500 arbitrary units) and increased number of telomere aggregates are associated with shorter survival and could be used as a prognostic factor to identify high-risk SMM and MM patients.

2.
Sci Rep ; 10(1): 8532, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444772

ABSTRACT

Oncogenic transformation impacts cancer cell interactions with their stroma, including through formation of abnormal blood vessels. This influence is often attributed to angiogenic growth factors, either soluble, or associated with tumor cell-derived extracellular vesicles (EVs). Here we examine some of the cancer-specific components of EV-mediated tumor-vascular interactions, including the impact of genetic driver mutations and genetic instability. Cancer cells expressing mutant HRAS oncogene exhibit aberrations of chromatin architecture, aneuploidy, cytoplasmic chromatin deposition and formation of micronuclei with a non-random chromosome content. EVs released from such HRAS-driven cells carry genomic DNA, including oncogenic sequences, and transfer this material to endothelial cells while inducing abnormal formation of micronuclei, along with cell migration and proliferation. Micronuclei were also triggered following treatment with EVs derived from glioma cells (and stem cells) expressing EGFRvIII oncogene, and in both endothelial cells and astrocytes. EVs from HRAS and EGFRvIII-driven cancer cells carry 19 common proteins while EVs from indolent control cells exhibit more divergent proteomes. Immortalized endothelial cell lines with disrupted TP53 pathway were refractory to EV-mediated micronuclei induction. We suggest that oncogenic transformation and intercellular trafficking of cancer-derived EVs may contribute to pathological vascular responses in cancer due to intercellular transmission of genomic instability.


Subject(s)
Cell Transformation, Neoplastic/pathology , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Glioblastoma/pathology , Micronuclei, Chromosome-Defective , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Proteome , Tumor Cells, Cultured
3.
Viruses ; 10(12)2018 12 19.
Article in English | MEDLINE | ID: mdl-30572664

ABSTRACT

Influenza A virus (IAV) infections remain a major human health threat. IAV has enormous genetic plasticity and can rapidly escape virus-targeted anti-viral strategies. Thus, there is increasing interest to identify host proteins and processes the virus requires for replication and maturation. The IAV non-structural protein 1 (NS1) is a critical multifunctional protein that is expressed to high levels in infected cells. Host proteins that interact with NS1 may serve as ideal targets for attenuating IAV replication. We previously developed and characterized broadly cross-reactive anti-NS1 monoclonal antibodies. For the current study, we used these mAbs to co-immunoprecipitate native IAV NS1 and interacting host proteins; 183 proteins were consistently identified in this NS1 interactome study, 124 of which have not been previously reported. RNAi screens identified 11 NS1-interacting host factors as vital for IAV replication. Knocking down one of these, nuclear mitotic apparatus protein 1 (NUMA1), dramatically reduced IAV replication. IAV genomic transcription and translation were not inhibited but transport of viral structural proteins to the cell membrane was hindered during maturation steps in NUMA1 knockdown (KD) cells.


Subject(s)
Antigens, Nuclear/metabolism , Host-Pathogen Interactions , Influenza A virus/physiology , Nuclear Matrix-Associated Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Assembly , A549 Cells , Antigens, Nuclear/genetics , Cell Cycle Proteins , Cell Line , Gene Knockdown Techniques , Humans , Immunoprecipitation , Influenza A virus/genetics , Nuclear Matrix-Associated Proteins/genetics , RNA Interference , Viral Nonstructural Proteins/genetics , Virus Replication
4.
Cancers (Basel) ; 10(9)2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30149530

ABSTRACT

Classical Hodgkin's lymphoma (cHL) is a B-Cell lymphoma comprised of mononuclear Hodgkin cells (H) and bi- to multi-nucleated Reed-Sternberg (RS) cells. Previous studies revealed that H and RS cells express lamin A/C, a component of the lamina of the nuclear matrix. Since no information was available about the three-dimensional (3D) expression patterns of lamin A/C in H and RS cells, we analyzed the 3D spatial organization of lamin in such cells, using 3D fluorescent microscopy. H and RS cells from cHL derived cell lines stained positive for lamin A/C, in contrast to peripheral blood lymphocytes (PBLs), in which the lamin A/C protein was not detected or weak, although its presence could be transiently increased with lymphocyte activation by lipopolysaccharide (LPS). Most importantly, in H and RS cells, the regular homogeneous and spherically shaped lamin A/C pattern, identified in activated lymphocytes, was absent. Instead, in H and RS cells, lamin staining showed internal lamin A/C structures, subdividing the nuclei into two or more smaller compartments. Analysis of pre-treatment cHL patients' samples replicated the lamin patterns identified in cHL cell lines. We conclude that the investigation of lamin A/C protein could be a useful tool for understanding nuclear remodeling in cHL.

5.
Nucleic Acids Res ; 45(8): e56, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28082388

ABSTRACT

Advanced light microscopy is an important tool for nanostructure analysis of chromatin. In this report we present a general concept for Single Molecule localization Microscopy (SMLM) super-resolved imaging of DNA-binding dyes based on modifying the properties of DNA and the dye. By careful adjustment of the chemical environment leading to local, reversible DNA melting and hybridization control over the fluorescence signal of the DNA-binding dye molecules can be introduced. We postulate a transient binding as the basis for our variation of binding-activated localization microscopy (BALM). We demonstrate that several intercalating and minor-groove binding DNA dyes can be used to register (optically isolate) only a few DNA-binding dye signals at a time. To highlight this DNA structure fluctuation-assisted BALM (fBALM), we applied it to measure, for the first time, nanoscale differences in nuclear architecture in model ischemia with an anticipated structural resolution of approximately 50 nm. Our data suggest that this approach may open an avenue for the enhanced microscopic analysis of chromatin nano-architecture and hence the microscopic analysis of nuclear structure aberrations occurring in various pathological conditions. It may also become possible to analyse nuclear nanostructure differences in different cell types, stages of development or environmental stress conditions.


Subject(s)
Chromatin/ultrastructure , DNA/ultrastructure , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Ascorbic Acid/pharmacology , Benzoxazoles/chemistry , Binding Sites , Cell Hypoxia , Cell Line , Cell Line, Tumor , Chromatin/metabolism , DNA/metabolism , Glucose/deficiency , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Hydrogen-Ion Concentration , Intercalating Agents/chemistry , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Nucleic Acid Conformation , Nucleic Acid Denaturation , Quinolinium Compounds/chemistry
6.
Mol Biol Cell ; 28(6): 817-824, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28077620

ABSTRACT

Stimulation of the MAPK pathway results in mitogen- and stress-activated protein kinase 1/2 (MSK1/2)-catalyzed phosphorylation of histone H3 at serine 10 or 28 and expression of immediate-early (IE) genes. In 10T1/2 mouse fibroblasts, phosphorylation of H3S10 and H3S28 occurs on different H3 molecules and in different nuclear regions. Similarly, we show that mitogen-induced H3S10 and H3S28 phosphorylation occurs in separate pools in human primary fibroblasts. High-resolution imaging studies on both cell types reveal that H3S10 and H3S28 phosphorylation events can be induced in a single cell but on different alleles, giving rise to H3S10ph and H3S28ph epialleles. Coimmunoprecipitation and inhibition studies demonstrate that CBP/p300-mediated H3K27 acetylation is required for MSK1/2 to phosphorylate S28. Although the K9ac and S10ph marks coexist on H3, S10 phosphorylation is not dependent on K9 acetylation by PCAF. We propose that random targeting of H3S10 or H3S28 results from the stochastic acetylation of H3 by CBP/p300 or PCAF, a process comparable to transcriptional bursting causing temporary allelic imbalance. In 10T1/2 cells expressing Jun, at least two of three alleles per cell were induced, a sign of high expression level. The redundant roles of H3S10ph and H3S28ph might enable rapid and efficient IE gene induction.


Subject(s)
Histones/genetics , Acetylation , Animals , Cell Culture Techniques , Fibroblasts , Histones/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Phosphorylation , Polymorphism, Single Nucleotide/genetics , Protein Processing, Post-Translational , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Serine , Transcriptional Activation
7.
Int J Cancer ; 140(2): 400-410, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27711972

ABSTRACT

The consistent appearance of specific chromosomal translocations in multiple myeloma has suggested that the positioning of chromosomes in the interphase nucleus might play a role in the occurrence of particular chromosomal rearrangements associated with malignant transformation. Using fluorescence in situ hybridization, we have determined the positions of selected chromosome pairs (18 and 19, 9 and 22, 4 and 14, 14 and 16, 11 and 14) in interphase nuclei of myeloma cells compared to normal lymphocytes of treatment-naïve patients. All chromosome pairs were arranged in a nonrandom pattern. Chromosomes commonly involved in myeloma-associated translocations (4 and 14, 14 and 16, 11 and 14) were found in close spatial proximity, and this is correlated with the occurrence of overlapping chromosome territories. The spatial distribution of chromosomes may increase the possibility of chromosomal translocations in multiple myeloma.


Subject(s)
Chromosomes/genetics , Lymphocytes/pathology , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Paraproteinemias/genetics , Aged , Cell Nucleus/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/pathology , Paraproteinemias/pathology , Pilot Projects , Translocation, Genetic/genetics
8.
J Cell Biochem ; 116(5): 704-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25501803

ABSTRACT

The mammalian nucleus has a distinct substructure that cannot be visualized directly by conventional microscopy. In this study, the organization of the DNA within the nucleus of multiple myeloma (MM) cells, their precursor cells (monoclonal gammopathy of undetermined significance; MGUS) and control lymphocytes of the representative patients is visualized and quantified by superresolution microscopy. Three-dimensional structured illumination microscopy (3D-SIM) increases the spatial resolution beyond the limits of conventional widefield fluorescence microscopy. 3D-SIM reveals new insights into the nuclear architecture of cancer as we show for the first time that it resolves organizational differences in intranuclear DNA organization of myeloma cells in MGUS and in MM patients. In addition, we report a significant increase in nuclear submicron DNA structure and structure of the DNA-free space in myeloma nuclei compared to normal lymphocyte nuclei. Our study provides previously unknown details of the nanoscopic DNA architecture of interphase nuclei of the normal lymphocytes, MGUS and MM cells. This study opens new avenues to understanding the disease progression from MGUS to MM.


Subject(s)
Cell Nucleus/ultrastructure , DNA/ultrastructure , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/pathology , Aged , Aged, 80 and over , Cell Line, Tumor , Humans , Lymphocytes/ultrastructure , Microscopy , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics
9.
Transl Oncol ; 6(6): 749-56, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24466378

ABSTRACT

Multiple myeloma (MM) is preceded by monoclonal gammopathy of undetermined significance (MGUS). Up to date, it is difficult to predict an individual's time to disease progression and the treatment response. To examine whether the nuclear telomeric architecture will unravel some of these questions, we carried out. Three-dimensional (3D) telomere analysis on samples from patients diagnosed with MGUS and MM, as well as from patients who went into relapse. Telomere signal intensity, number of telomere aggregates, nuclear volume, and the overall nuclear telomere distribution (a/c ratio) were analyzed. The telomeric profiles allowed for the differentiation of the disease stages. The telomeric profiles of myeloma cells obtained from blood and bone marrow aspirates were identical. Based on this study, we discuss the use of 3D telomere profiling as a potential future tool for risk stratification and personalized treatment decisions.

10.
Clin Cancer Res ; 18(12): 3293-304, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22539801

ABSTRACT

PURPOSE: Myelodysplastic syndromes (MDS) are a group of disorders characterized by cytopenias, with a propensity for evolution into acute myeloid leukemias (AML). This transformation is driven by genomic instability, but mechanisms remain unknown. Telomere dysfunction might generate genomic instability leading to cytopenias and disease progression. EXPERIMENTAL DESIGN: We undertook a pilot study of 94 patients with MDS (56 patients) and AML (38 patients). The MDS cohort consisted of refractory cytopenia with multilineage dysplasia (32 cases), refractory anemia (12 cases), refractory anemia with excess of blasts (RAEB)1 (8 cases), RAEB2 (1 case), refractory anemia with ring sideroblasts (2 cases), and MDS with isolated del(5q) (1 case). The AML cohort was composed of AML-M4 (12 cases), AML-M2 (10 cases), AML-M5 (5 cases), AML-M0 (5 cases), AML-M1 (2 cases), AML-M4eo (1 case), and AML with multidysplasia-related changes (1 case). Three-dimensional quantitative FISH of telomeres was carried out on nuclei from bone marrow samples and analyzed using TeloView. RESULTS: We defined three-dimensional nuclear telomeric profiles on the basis of telomere numbers, telomeric aggregates, telomere signal intensities, nuclear volumes, and nuclear telomere distribution. Using these parameters, we blindly subdivided the MDS patients into nine subgroups and the AML patients into six subgroups. Each of the parameters showed significant differences between MDS and AML. Combining all parameters revealed significant differences between all subgroups. Three-dimensional telomeric profiles are linked to the evolution of telomere dysfunction, defining a model of progression from MDS to AML. CONCLUSIONS: Our results show distinct three-dimensional telomeric profiles specific to patients with MDS and AML that help subgroup patients based on the severity of telomere dysfunction highlighted in the profiles.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Telomere/ultrastructure , Adolescent , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/pathology , Cell Nucleus/pathology , Cohort Studies , Disease Progression , Female , Genomic Instability , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Pilot Projects , Telomere/metabolism , Young Adult
11.
Adv Cancer Res ; 112: 77-126, 2011.
Article in English | MEDLINE | ID: mdl-21925302

ABSTRACT

This chapter focuses on the three-dimensional organization of the nucleus in normal, early genomically unstable, and tumor cells. A cause-consequence relationship is discussed between nuclear alterations and the resulting genomic rearrangements. Examples are presented from studies on conditional Myc deregulation, experimental tumorigenesis in mouse plasmacytoma, nuclear remodeling in Hodgkin's lymphoma, and in adult glioblastoma. A model of nuclear remodeling is proposed for cancer progression in multiple myeloma. Current models of nuclear remodeling are described, including our model of altered nuclear architecture and the onset of genomic instability.


Subject(s)
Cell Nucleus/genetics , Genomic Instability , Neoplasms/genetics , Neoplasms/pathology , Animals , Humans , Mice
12.
Cytometry A ; 79(2): 159-66, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21265009

ABSTRACT

Telomeres, the end of chromosomes, are organized in a nonoverlapping fashion and form microterritories in nuclei of normal cells. Previous studies have shown that normal and tumor cell nuclei differ in their 3D telomeric organization. The differences include a change in the spatial organization of the telomeres, in telomere numbers and sizes and in the presence of telomeric aggregates. Previous attempts to identify the above parameters of 3D telomere organization were semi-automated. Here we describe the automation of 3D scanning for telomere signatures in interphase nuclei based on three-dimensional fluorescent in situ hybridization (3D-FISH) and, for the first time, define its sensitivity in tumor cell detection. The data were acquired with a high-throughput scanning/acquisition system that allows to measure cells and acquire 3D images of nuclei at high resolution with 40 × or 60 × oil and at a speed of 10,000-15,000 cells h(-1) , depending on the cell density on the slides. The automated scanning, TeloScan, is suitable for large series of samples and sample sizes. We define the sensitivity of this automation for tumor cell detection. The data output includes 3D telomere positions, numbers of telomeric aggregates, telomere numbers, and telomere signal intensities. We were able to detect one aberrant cell in 1,000 normal cells. In conclusions, we are able to detect tumor cells based on 3D architectural profiles of the genome. This new tool could, in the future, assist in patient diagnosis, in the detection of minimal residual disease, in the analysis of treatment response and in treatment decisions.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy/methods , Telomere/ultrastructure , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Genome , In Situ Hybridization, Fluorescence/methods , Interphase/genetics , Lymphocytes/ultrastructure , Male , Mice , Neoplasms/genetics , Neoplasms/ultrastructure , Plasmacytoma/ultrastructure , Sensitivity and Specificity , Telomere/genetics
13.
Haematologica ; 95(12): 2038-46, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20823137

ABSTRACT

BACKGROUND: Hodgkin's lymphoma is characterized by the presence of mono-nucleated Hodgkin cells and bi- to multi-nucleated Reed-Sternberg cells. We have recently shown telomere dysfunction and aberrant synchronous/asynchronous cell divisions during the transition of Hodgkin cells to Reed-Sternberg cells.1 DESIGN AND METHODS: To determine whether overall changes in nuclear architecture affect genomic instability during the transition of Hodgkin cells to Reed-Sternberg cells, we investigated the nuclear organization of chromosomes in these cells. RESULTS: Three-dimensional fluorescent in situ hybridization revealed irregular nuclear positioning of individual chromosomes in Hodgkin cells and, more so, in Reed-Sternberg cells. We characterized an increasingly unequal distribution of chromosomes as mono-nucleated cells became multi-nucleated cells, some of which also contained chromosome-poor 'ghost' cell nuclei. Measurements of nuclear chromosome positions suggested chromosome overlaps in both types of cells. Spectral karyotyping then revealed both aneuploidy and complex chromosomal rearrangements: multiple breakage-bridge-fusion cycles were at the origin of the multiple rearranged chromosomes. This conclusion was challenged by super resolution three-dimensional structured illumination imaging of Hodgkin and Reed-Sternberg nuclei. Three-dimensional super resolution microscopy data documented inter-nuclear DNA bridges in multi-nucleated cells but not in mono-nucleated cells. These bridges consisted of chromatids and chromosomes shared by two Reed-Sternberg nuclei. The complexity of chromosomal rearrangements increased as Hodgkin cells developed into multi-nucleated cells, thus indicating tumor progression and evolution in Hodgkin's lymphoma, with Reed-Sternberg cells representing the highest complexity in chromosomal rearrangements in this disease. CONCLUSIONS: This is the first study to demonstrate nuclear remodeling and associated genomic instability leading to the generation of Reed-Sternberg cells of Hodgkin's lymphoma. We defined nuclear remodeling as a key feature of Hodgkin's lymphoma, highlighting the relevance of nuclear architecture in cancer.


Subject(s)
Cell Nucleus/metabolism , Chromosome Aberrations , Chromosome Breakage , Hodgkin Disease/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 9/genetics , Hodgkin Disease/pathology , Humans , Reed-Sternberg Cells/metabolism , Reed-Sternberg Cells/pathology , Spectral Karyotyping/methods , Translocation, Genetic
14.
Neoplasia ; 12(2): 183-91, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20126476

ABSTRACT

The absence of biological markers allowing for the assessment of the evolution and prognosis of glioblastoma (GBM) is a major impediment to the clinical management of GBM patients. The observed variability in patients' treatment responses and in outcomes implies biological heterogeneity and the existence of unidentified patient categories. Here, we define for the first time three GBM patient categories with distinct and clinically predictive three-dimensional nuclear-telomeric architecture defined by telomere number, size, and frequency of telomeric aggregates. GBM patient samples were examined by three-dimensional fluorescent in situ hybridization of telomeres using two independent three-dimensional telomere-measurement tools (TeloView program [P(1)] and SpotScan system [P(2)]). These measurements identified three patients categories (categories 1-3), displaying significant differences in telomere numbers/nucleus (P(1) = .0275; P(2)

Subject(s)
Brain Neoplasms/pathology , Cell Nucleus/pathology , Glioblastoma/pathology , Telomere/pathology , Adult , Aged , Brain Neoplasms/mortality , Disease Progression , Disease-Free Survival , Female , Glioblastoma/mortality , Humans , Image Interpretation, Computer-Assisted , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...