Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Waste Manag Res ; : 734242X241241601, 2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38616533

ABSTRACT

According to the state of the art, most of the mixed copper and copper alloy scrap and residues are processed in a copper smelter. Despite the environmental and economic advantages relative to primary production, the recycling of copper and its alloying elements (zinc, tin, lead, nickel, etc.) requires significantly more energy and cost than remelting unmixed or pure scrap fractions such as separate collected material or production scrap. To date, however, less attention has been given to the mechanical purification of mixed scrap. Therefore, sorting by alloy-specific components (SBASC) using an industrial X-ray fluorescence (XRF) sorting system was tested on the coarse metallic fraction (10-32 mm) of mixed foundry residues. The findings show that XRF-SBASC can recover higher-grade copper concentrates (reaching 98.3% Cu), leaded brass and complex alloys, such as aluminium bronze and red brass with high purities, for the use in the production of new materials. XRF-SBASC can therefore contribute to a more resource efficient metal recycling, mainly by reducing the energy consumption and loss levels in copper metallurgy.

2.
Nucleic Acids Res ; 49(22): 12622-12633, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871435

ABSTRACT

The design of high-affinity, RNA-binding ligands has proven very challenging. This is due to the unique structural properties of RNA, often characterized by polar surfaces and high flexibility. In addition, the frequent lack of well-defined binding pockets complicates the development of small molecule binders. This has triggered the search for alternative scaffolds of intermediate size. Among these, peptide-derived molecules represent appealing entities as they can mimic structural features also present in RNA-binding proteins. However, the application of peptidic RNA-targeting ligands is hampered by a lack of design principles and their inherently low bio-stability. Here, the structure-based design of constrained α-helical peptides derived from the viral suppressor of RNA silencing, TAV2b, is described. We observe that the introduction of two inter-side chain crosslinks provides peptides with increased α-helicity and protease stability. One of these modified peptides (B3) shows high affinity for double-stranded RNA structures including a palindromic siRNA as well as microRNA-21 and its precursor pre-miR-21. Notably, B3 binding to pre-miR-21 inhibits Dicer processing in a biochemical assay. As a further characteristic this peptide also exhibits cellular entry. Our findings show that constrained peptides can efficiently mimic RNA-binding proteins rendering them potentially useful for the design of bioactive RNA-targeting ligands.


Subject(s)
Peptides/chemistry , RNA Interference , RNA, Double-Stranded/chemistry , RNA-Binding Proteins/chemistry , Viral Proteins/chemistry , Cell Membrane Permeability , Cucumovirus , Endopeptidase K , Humans , K562 Cells , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Mimicry , Peptides/metabolism , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Double-Stranded/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism
3.
Front Immunol ; 12: 798087, 2021.
Article in English | MEDLINE | ID: mdl-35058934

ABSTRACT

The generation and expansion of functionally competent NK cells in vitro is of great interest for their application in immunotherapy of cancer. Since CD33 constitutes a promising target for immunotherapy of myeloid malignancies, NK cells expressing a CD33-specific chimeric antigen receptor (CAR) were generated. Unexpectedly, we noted that CD33-CAR NK cells could not be efficiently expanded in vitro due to a fratricide-like process in which CD33-CAR NK cells killed other CD33-CAR NK cells that had upregulated CD33 in culture. This upregulation was dependent on the stimulation protocol and encompassed up to 50% of NK cells including CD56dim NK cells that do generally not express CD33 in vivo. RNAseq analysis revealed that upregulation of CD33+ NK cells was accompanied by a unique transcriptional signature combining features of canonical CD56bright (CD117high, CD16low) and CD56dim NK cells (high expression of granzyme B and perforin). CD33+ NK cells exhibited significantly higher mobilization of cytotoxic granula and comparable levels of cytotoxicity against different leukemic target cells compared to the CD33- subset. Moreover, CD33+ NK cells showed superior production of IFNγ and TNFα, whereas CD33- NK cells exerted increased antibody-dependent cellular cytotoxicity (ADCC). In summary, the study delineates a novel functional divergence between NK cell subsets upon in vitro stimulation that is marked by CD33 expression. By choosing suitable stimulation protocols, it is possible to preferentially generate CD33+ NK cells combining efficient target cell killing and cytokine production, or alternatively CD33- NK cells, which produce less cytokines but are more efficient in antibody-dependent applications.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Cytokines/immunology , Killer Cells, Natural/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Flow Cytometry/methods , Gene Expression Profiling/methods , Humans , K562 Cells , Killer Cells, Natural/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/metabolism , Up-Regulation
4.
Cells ; 9(4)2020 04 17.
Article in English | MEDLINE | ID: mdl-32316635

ABSTRACT

Direct acting antivirals (DAAs) revolutionized the therapy of chronic hepatitis C infection. However, unexpected high recurrence rates of hepatocellular carcinoma (HCC) after DAA treatment became an issue in patients with advanced cirrhosis and fibrosis. In this study, we aimed to investigate an impact of DAA treatment on the molecular changes related to HCC development and progression in hepatoma cell lines and primary human hepatocytes. We found that treatment with sofosbuvir (SOF), a backbone of DAA therapy, caused an increase in EGFR expression and phosphorylation. As a result, enhanced translocation of EGFR into the nucleus and transactivation of factors associated with cell cycle progression, B-MYB and Cyclin D1, was detected. Serine/threonine kinase profiling identified additional pathways, especially the MAPK pathway, also activated during SOF treatment. Importantly, the blocking of EGFR kinase activity by erlotinib during SOF treatment prevented all downstream events. Altogether, our findings suggest that SOF may have an impact on pathological processes in the liver via the induction of EGFR signaling. Notably, zidovudine, another nucleoside analogue, exerted a similar cell phenotype, suggesting that the observed effects may be induced by additional members of this drug class.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Liver/drug effects , Sofosbuvir/therapeutic use , Antiviral Agents/pharmacology , Humans , Liver/pathology , Risk Factors , Sofosbuvir/pharmacology
5.
Front Immunol ; 11: 586168, 2020.
Article in English | MEDLINE | ID: mdl-33584651

ABSTRACT

Cellular immunotherapy using chimeric antigen receptors (CARs) so far has almost exclusively used autologous peripheral blood-derived T cells as immune effector cells. However, harvesting sufficient numbers of T cells is often challenging in heavily pre-treated patients with malignancies and perturbed hematopoiesis and perturbed hematopoiesis. Also, such a CAR product will always be specific for the individual patient. In contrast, NK cell infusions can be performed in non-HLA-matched settings due to the absence of alloreactivity of these innate immune cells. Still, the infused NK cells are subject to recognition and rejection by the patient's immune system, thereby limiting their life-span in vivo and undermining the possibility for multiple infusions. Here, we designed genome editing and advanced lentiviral transduction protocols to render primary human NK cells unsusceptible/resistant to an allogeneic response by the recipient's CD8+ T cells. After knocking-out surface expression of HLA class I molecules by targeting the B2M gene via CRISPR/Cas9, we also co-expressed a single-chain HLA-E molecule, thereby preventing NK cell fratricide of B2M-knockout (KO) cells via "missing self"-induced lysis. Importantly, these genetically engineered NK cells were functionally indistinguishable from their unmodified counterparts with regard to their phenotype and their natural cytotoxicity towards different AML cell lines. In co-culture assays, B2M-KO NK cells neither induced immune responses of allogeneic T cells nor re-activated allogeneic T cells which had been expanded/primed using irradiated PBMNCs of the respective NK cell donor. Our study demonstrates the feasibility of genome editing in primary allogeneic NK cells to diminish their recognition and killing by mismatched T cells and is an important prerequisite for using non-HLA-matched primary human NK cells as readily available, "off-the-shelf" immune effectors for a variety of immunotherapy indications in human cancer.


Subject(s)
Allogeneic Cells/immunology , Gene Editing/methods , Histocompatibility Antigens Class I/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Gene Knockout Techniques/methods , Humans , Killer Cells, Natural/transplantation
6.
J Immunol ; 202(6): 1895-1903, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30700588

ABSTRACT

Comprehensive knockout of HLA class II (HLA-II) ß-chain genes is complicated by their high polymorphism. In this study, we developed CRISPR/Cas9 genome editing to simultaneously target HLA-DRB, -DQB1, and -DPB1 through a single guide RNA recognizing a conserved region in exon 2. Abrogation of HLA-II surface expression was achieved in five different HLA-typed, human EBV-transformed B lymphoblastoid cell lines (BLCLs). Next-generation sequencing-based detection confirmed specific genomic insertion/deletion mutations with 99.5% penetrance in sorted cells for all three loci. No alterations were observed in HLA-I genes, the HLA-II peptide editor HLA-DMB, or its antagonist HLA-DOB, showing high on-target specificity. Transfection of full-length HLA-DPB1 mRNA into knockout BLCLs fully restored HLA-DP surface expression and recognition by alloreactive human CD4 T cells. The possibility to generate single HLA-II-expressing BLCLs by one-shot genome editing opens unprecedented opportunities for mechanistically dissecting the interaction of individual HLA variants with the immune system.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Gene Editing/methods , Gene Knockout Techniques/methods , HLA-DR beta-Chains , RNA, Guide, Kinetoplastida , Cell Line, Tumor , HLA-DR beta-Chains/genetics , Humans
7.
Stem Cell Reports ; 10(3): 875-889, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29456178

ABSTRACT

Generation of hematopoietic stem cells (HSCs) from pluripotent stem cells, in vitro, holds great promise for regenerative therapies. Primarily, this has been achieved in mouse cells by overexpression of the homeotic selector protein HOXB4. The exact cellular stage at which HOXB4 promotes hematopoietic development, in vitro, is not yet known. However, its identification is a prerequisite to unambiguously identify the molecular circuits controlling hematopoiesis, since the activity of HOX proteins is highly cell and context dependent. To identify that stage, we retrovirally expressed HOXB4 in differentiating mouse embryonic stem cells (ESCs). Through the use of Runx1(-/-) ESCs containing a doxycycline-inducible Runx1 coding sequence, we uncovered that HOXB4 promoted the formation of hemogenic endothelium cells without altering endothelial cell development. Whole-transcriptome analysis revealed that its expression mediated the upregulation of transcription of core transcription factors necessary for hematopoiesis, culminating in the formation of blood progenitors upon initiation of Runx1 expression.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium/metabolism , Endothelium/physiology , Hematopoiesis/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Gene Expression Profiling/methods , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Transcription, Genetic/physiology , Up-Regulation/physiology
8.
Front Immunol ; 9: 2978, 2018.
Article in English | MEDLINE | ID: mdl-30619335

ABSTRACT

Viruses and hosts are situated in a molecular arms race. To avoid morbidity and mortality, hosts evolved antiviral restriction factors. These restriction factors exert selection pressure on the viruses and drive viral evolution toward increasingly efficient immune antagonists. Numerous viruses exploit cellular DNA damage-binding protein 1 (DDB1)-containing Cullin RocA ubiquitin ligases (CRLs) to induce the ubiquitination and subsequent proteasomal degradation of antiviral factors expressed by their hosts. To establish a comprehensive understanding of the underlying protein interaction networks, we performed immuno-affinity precipitations for a panel of DDB1-interacting proteins derived from viruses such as mouse cytomegalovirus (MCMV, Murid herpesvirus [MuHV] 1), rat cytomegalovirus Maastricht MuHV2, rat cytomegalovirus English MuHV8, human cytomegalovirus (HCMV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Cellular interaction partners were identified and quantified by mass spectrometry (MS) and validated by classical biochemistry. The comparative approach enabled us to separate unspecific interactions from specific binding partners and revealed remarkable differences in the strength of interaction with DDB1. Our analysis confirmed several previously described interactions like the interaction of the MCMV-encoded interferon antagonist pM27 with STAT2. We extended known interactions to paralogous proteins like the interaction of the HBV-encoded HBx with different Spindlin proteins and documented interactions for the first time, which explain functional data like the interaction of the HIV-2-encoded Vpr with Bax. Additionally, several novel interactions were identified, such as the association of the HIV-2-encoded Vpx with the transcription factor RelA (also called p65). For the latter interaction, we documented a functional relevance in antagonizing NF-κB-driven gene expression. The mutation of the DDB1 binding interface of Vpx significantly impaired NF-κB inhibition, indicating that Vpx counteracts NF-κB signaling by a DDB1- and CRL-dependent mechanism. In summary, our findings improve the understanding of how viral pathogens hijack cellular DDB1 and CRLs to ensure efficient replication despite the expression of host restriction factors.


Subject(s)
HIV-2/immunology , Protein Binding/immunology , Transcription Factor RelA/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Diseases/immunology , Animals , Cytomegalovirus/immunology , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Fibroblasts , Gene Expression Regulation/immunology , HEK293 Cells , HIV-2/genetics , HIV-2/metabolism , Hepatitis B virus/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunoprecipitation/methods , Mass Spectrometry/methods , Mice , Muromegalovirus/immunology , NIH 3T3 Cells , Primary Cell Culture , Protein Interaction Mapping/methods , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/immunology , Virus Diseases/virology
10.
Cell Chem Biol ; 24(8): 958-968.e5, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28757184

ABSTRACT

The Wnt signaling pathway plays a critical role in cell proliferation and differentiation, thus it is often associated with diseases such as cancers. Unfortunately, although attractive, developing anti-cancer strategy targeting Wnt signaling has been challenging given that the most attractive targets are involved in protein-protein interactions (PPIs). Here, we develop a stapled peptide inhibitor that targets the interaction between ß-catenin and T cell factor/lymphoid enhancer-binding factor transcription factors, which are crucially involved in Wnt signaling. Our integrative approach combines peptide stapling to optimize proteolytic stability, with lessons learned from cell-penetrating peptide (CPP) design to maximize cellular uptake resulting in NLS-StAx-h, a selective, cell permeable, stapled peptide inhibitor of oncogenic Wnt signaling that efficiently inhibits ß-catenin-transcription factor interactions. We expect that this type of integrative strategy that endows stapled peptides with CPP features will be generally useful for developing inhibitors of intracellular PPIs.


Subject(s)
Cell-Penetrating Peptides/metabolism , beta Catenin/metabolism , Amino Acid Sequence , Axin Protein/genetics , Axin Protein/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Movement , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Gene Expression/drug effects , Genes, Reporter , HeLa Cells , Humans , Microscopy, Confocal , Protein Interaction Domains and Motifs , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors
11.
Elife ; 2: e00825, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23878725

ABSTRACT

A common deleted region (CDR) in both myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN) affects the long arm of chromosome 20 and has been predicted to harbor a tumor suppressor gene. Here we show that MYBL2, a gene within the 20q CDR, is expressed at sharply reduced levels in CD34+ cells from most MDS cases (65%; n = 26), whether or not they harbor 20q abnormalities. In a murine competitive reconstitution model, Mybl2 knockdown by RNAi to 20-30% of normal levels in multipotent hematopoietic progenitors resulted in clonal dominance of these 'sub-haploinsufficient' cells, which was reflected in all blood cell lineages. By 6 months post-transplantation, the reconstituted mice had developed a clonal myeloproliferative/myelodysplastic disorder originating from the cells with aberrantly reduced Mybl2 expression. We conclude that downregulation of MYBL2 activity below levels predicted by classical haploinsufficiency underlies the clonal expansion of hematopoietic progenitors in a large fraction of human myeloid malignancies. DOI:http://dx.doi.org/10.7554/eLife.00825.001.


Subject(s)
Cell Cycle Proteins/genetics , Genes, Tumor Suppressor , Haploinsufficiency/genetics , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Trans-Activators/genetics , Dosage Compensation, Genetic , Humans , Karyotyping
12.
J Clin Invest ; 122(7): 2369-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22684105

ABSTRACT

Natural killer (NK) cells are primary effectors of innate immunity directed against transformed tumor cells. In response, tumor cells have developed mechanisms to evade NK cell-mediated lysis through molecular mechanisms that are not well understood. In the present study, we used a lentiviral shRNA library targeting more than 1,000 human genes to identify 83 genes that promote target cell resistance to human NK cell-mediated killing. Many of the genes identified in this genetic screen belong to common signaling pathways; however, none of them have previously been known to modulate susceptibility of human tumor cells to immunologic destruction. Gene silencing of two members of the JAK family (JAK1 and JAK2) increased the susceptibility of a variety of tumor cell types to NK-mediated lysis and induced increased secretion of IFN-γ by NK cells. Treatment of tumor cells with JAK inhibitors also increased susceptibility to NK cell activity. These findings may have important clinical implications and suggest that small molecule inhibitors of tyrosine kinases being developed as therapeutic antitumor agents may also have significant immunologic effects in vivo.


Subject(s)
Janus Kinase 1/genetics , Janus Kinase 2/genetics , Killer Cells, Natural/immunology , Tumor Escape/genetics , Apoptosis , Cell Line, Tumor , Coculture Techniques , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Interferon-gamma/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Killer Cells, Natural/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Tyrphostins/pharmacology
13.
Blood ; 115(21): 4157-61, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20304806

ABSTRACT

Comprehensive analysis of the cancer genome has become a standard approach to identifying new disease loci, and ultimately will guide therapeutic decisions. A key technology in this effort, single nucleotide polymorphism arrays, has been applied in hematologic malignancies to detect deletions, amplifications, and loss of heterozygosity (LOH) at high resolution. An inherent challenge of such studies lies in correctly distinguishing somatically acquired, cancer-specific lesions from patient-specific inherited copy number variations or segments of homozygosity. Failure to include appropriate normal DNA reference samples for each patient in retrospective or prospective studies makes it difficult to identify small somatic deletions not evident by standard cytogenetic analysis. In addition, the lack of proper controls can also lead to vastly overestimated frequencies of LOH without accompanying loss of DNA copies, so-called copy-neutral LOH. Here we use examples from patients with myeloid malignancies to demonstrate the superiority of matched tumor and normal DNA samples (paired studies) over multiple unpaired samples with respect to reducing false discovery rates in high-resolution single nucleotide polymorphism array analysis. Comparisons between matched tumor and normal samples will continue to be critical as the field moves from high resolution array analysis to deep sequencing to detect abnormalities in the cancer genome.


Subject(s)
Hematologic Neoplasms/genetics , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Single Nucleotide , DNA Mutational Analysis , DNA, Neoplasm/genetics , False Positive Reactions , Gene Dosage , Genes, Neoplasm , Humans , Leukemia, Myeloid, Acute/genetics , Loss of Heterozygosity , Myelodysplastic Syndromes/genetics , Sequence Deletion
14.
Mol Cell Biol ; 29(21): 5911-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19704007

ABSTRACT

A growing body of evidence indicates that early mitotic inhibitor 1 (Emi1) is essential for genomic stability, but how this function relates to embryonic development and cancer pathogenesis remains unclear. We have identified a zebrafish mutant line in which deficient emi1 gene expression results in multilineage hematopoietic defects and widespread developmental defects that are p53 independent. Cell cycle analyses of Emi1-depleted zebrafish or human cells showed chromosomal rereplication, and metaphase preparations from mutant zebrafish embryos revealed rereplicated, unsegregated chromosomes and polyploidy. Furthermore, EMI1-depleted mammalian cells relied on topoisomerase II alpha-dependent mitotic decatenation to progress through metaphase. Interestingly, the loss of a single emi1 allele in the absence of p53 enhanced the susceptibility of adult fish to neural sheath tumorigenesis. Our results cast Emi1 as a critical regulator of genomic fidelity during embryogenesis and suggest that the factor may act as a tumor suppressor.


Subject(s)
Cell Cycle Proteins/metabolism , Embryonic Development/genetics , Genome/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animals , Apoptosis , Cell Cycle , Cell Size , DNA Damage , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/pathology , Hematopoiesis , Mutation/genetics , Myeloid Cells/pathology , Phenotype
15.
Genome Biol ; 8(7): 219, 2007.
Article in English | MEDLINE | ID: mdl-17666119

ABSTRACT

Recent studies using single-nucleotide polymorphism arrays have pinpointed novel oncogenes and tumor suppressors involved in specific types of human cancers.


Subject(s)
Carcinogens , Chromosome Aberrations , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Single Nucleotide , Humans
16.
J Biol Phys ; 32(5): 383-92, 2006 Nov.
Article in English | MEDLINE | ID: mdl-19669444

ABSTRACT

The statistical properties of heart beat intervals of 130 long-term surface electrocardiogram recordings during atrial fibrillation (AF) are investigated. We find that the distribution of interbeat intervals exhibits a characteristic exponential tail, which is absent during sinus rhythm, as tested in a corresponding control study with 72 healthy persons. The rate gamma of the exponential decay lies in the range 3-12 Hz and shows diurnal variations. It equals, up to statistical uncertainties, the level of the previously uncovered white noise part of the power spectrum, which is also characteristic for AF. The overall statistical features can be described by decomposing the intervals into two statistically independent times, where the first one is associated with a correlated process with 1/f noise characteristics, while the second one belongs to an uncorrelated process and is responsible for the exponential tail. It is suggested to use gamma as a further parameter for a better classification of AF and for the medical diagnosis. The relevance of the findings with respect to a general understanding of AF is discussed.

17.
Cell ; 123(4): 641-53, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16286009

ABSTRACT

In response to DNA damage, the p53 tumor suppressor can elicit either apoptosis or cell-cycle arrest and repair, but how this critical decision is made in specific cell types remains largely undefined. We investigated the mechanism by which the transcriptional repressor Slug specifically rescues hematopoietic progenitor cells from lethal doses of gamma radiation. We show that Slug is transcriptionally induced by p53 upon irradiation and then protects the damaged cell from apoptosis by directly repressing p53-mediated transcription of puma, a key BH3-only antagonist of the antiapoptotic Bcl-2 proteins. We established the physiologic significance of Slug-mediated repression of puma by demonstrating that mice deficient in both genes survive doses of total-body irradiation that lethally deplete hematopoietic progenitor populations in mice lacking only slug. Thus, Slug functions downstream of p53 in developing blood cells as a critical switch that prevents their apoptosis by antagonizing the trans-activation of puma by p53.


Subject(s)
Apoptosis/physiology , Hematopoietic Stem Cells/physiology , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/physiology , Bone Marrow/radiation effects , Bone Marrow Transplantation , Cell Line, Transformed , Cell Line, Tumor , Chromatin Immunoprecipitation , Gamma Rays , Gene Expression/genetics , Gene Expression/radiation effects , Hematopoietic Stem Cells/radiation effects , Humans , Introns/genetics , Mice , Mice, Knockout , Mitochondria/physiology , Mitochondria/radiation effects , Models, Biological , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/radiation effects , Myelopoiesis/radiation effects , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Snail Family Transcription Factors , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
18.
Br J Haematol ; 130(1): 83-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15982348

ABSTRACT

Overexpression of HOXA9 is linked to the molecular pathogenesis of acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS), conferring a poor prognosis. HOXA9 expression levels were analysed in the diagnostic bone marrow (BM) samples of 13 MDS patients. HOXA9 was expressed by CD34(+) BM cells at median levels 3.1-fold higher than in CD34(-) cells from the same patient and at median levels 4.3-fold higher than in CD34(+) cells from healthy donors. These results indicate that CD34(+) cell selection is required to accurately assess the expression levels of HOXA9 and related genes in the multipotential malignant progenitor cells of MDS patients.


Subject(s)
Antigens, CD34/immunology , Homeodomain Proteins/genetics , Myelodysplastic Syndromes/diagnosis , T-Lymphocytes/immunology , Aged , Aged, 80 and over , Bone Marrow Examination , Case-Control Studies , Cell Separation/methods , Female , Gene Expression Regulation, Leukemic , Humans , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
19.
Oncogene ; 22(4): 555-71, 2003 Jan 30.
Article in English | MEDLINE | ID: mdl-12555069

ABSTRACT

Activation of the tumor suppressor p53 after genotoxic insults may result in two different responses: growth arrest or apoptosis. In this study, we analysed how mitogenic stimulation of primary mouse lymphocytes influences p53 signaling upon gamma-irradiation. We found that G(0) lymphocytes rapidly went into p53-dependent apoptosis, whereas stimulated lymphocytes went into a p53-dependent, p21-mediated growth arrest. The switch in p53 response upon stimulation did neither result from a switch in transcriptional activation of major p53 target genes, nor from the high level of p21 expressed in stimulated, irradiated cells. Growth stimulation, however, led to the upregulation of the antiapoptotic factors Bcl-x(L) and Bfl-1. In resting cells, p53 induced apoptosis after gamma-irradiation was accompanied by a breakdown of the mitochondrial membrane potential (psi(m)) that was counteracted by growth stimulation. We propose that growth stimulation intercepted p53 proapoptotic signaling at the level of mitochondrial integrity, most likely by upregulating the antiapoptotic factors Bcl-x(L) and Bfl-1. Upregulation of Bcl-x(L) and of Bfl-1 upon growth stimulation was mediated by the PKC-dependent activation of NF-kappaB. Consequently, blocking PKC activity restored apoptosis in stimulated, irradiated splenocytes. The inherent coupling of growth stimulation with antiapoptotic signaling in primary lymphocytes might provide hints as to how precancerous lymphocytes bypass the need for mutational inactivation of p53. Thus, our findings might explain the relatively low frequency of p53 mutations in lymphomas in comparison to other tumor entities.


Subject(s)
Apoptosis/physiology , Cell Division/physiology , Signal Transduction , Tumor Suppressor Protein p53/physiology , Animals , Base Sequence , DNA Primers , Immunophenotyping , In Situ Nick-End Labeling , Membrane Potentials , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Spleen/cytology , Spleen/radiation effects , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...