Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nature ; 551(7680): 327-332, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144448

ABSTRACT

Junctional epidermolysis bullosa (JEB) is a severe and often lethal genetic disease caused by mutations in genes encoding the basement membrane component laminin-332. Surviving patients with JEB develop chronic wounds to the skin and mucosa, which impair their quality of life and lead to skin cancer. Here we show that autologous transgenic keratinocyte cultures regenerated an entire, fully functional epidermis on a seven-year-old child suffering from a devastating, life-threatening form of JEB. The proviral integration pattern was maintained in vivo and epidermal renewal did not cause any clonal selection. Clonal tracing showed that the human epidermis is sustained not by equipotent progenitors, but by a limited number of long-lived stem cells, detected as holoclones, that can extensively self-renew in vitro and in vivo and produce progenitors that replenish terminally differentiated keratinocytes. This study provides a blueprint that can be applied to other stem cell-mediated combined ex vivo cell and gene therapies.


Subject(s)
Epidermal Cells , Epidermolysis Bullosa, Junctional/therapy , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Transgenes/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Lineage , Cell Self Renewal , Cell Tracking , Child , Clone Cells/cytology , Clone Cells/metabolism , Dermis/cytology , Dermis/pathology , Epidermis/pathology , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/metabolism , Epidermolysis Bullosa, Junctional/pathology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/transplantation , Male , Proviruses/genetics , Kalinin
2.
Blood ; 127(5): 572-81, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26660426

ABSTRACT

The sequential activation of distinct developmental gene networks governs the ultimate identity of a cell, but the mechanisms involved in initiating downstream programs are incompletely understood. The pre-B-cell receptor (pre-BCR) is an important checkpoint of B-cell development and is essential for a pre-B cell to traverse into an immature B cell. Here, we show that activation of myocyte enhancer factor 2 (Mef2) transcription factors (TFs) by the pre-BCR is necessary for initiating the subsequent genetic network. We demonstrate that B-cell development is blocked at the pre-B-cell stage in mice deficient for Mef2c and Mef2d TFs and that pre-BCR signaling enhances the transcriptional activity of Mef2c/d through phosphorylation by the Erk5 mitogen-activating kinase. This activation is instrumental in inducing Krüppel-like factor 2 and several immediate early genes of the AP1 and Egr family. Finally, we show that Mef2 proteins cooperate with the products of their target genes (Irf4 and Egr2) to induce secondary waves of transcriptional regulation. Our findings uncover a novel role for Mef2c/d in coordinating the transcriptional network that promotes early B-cell development.


Subject(s)
B-Lymphocytes/metabolism , Precursor Cells, B-Lymphoid/metabolism , Animals , B-Lymphocytes/cytology , Cell Line , Gene Expression Regulation , Gene Knockout Techniques , Gene Regulatory Networks , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation , Precursor Cells, B-Lymphoid/cytology , Signal Transduction , Transcriptional Activation
3.
J Infect Dis ; 212 Suppl 2: S316-21, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26185094

ABSTRACT

Ebola virus (EBOV) protein 24 antagonizes the host interferon (IFN) response by hijacking select nuclear importin-α isoforms. Thereby, it blocks STAT1-mediated IFN-α/ß and IFN-γ synthesis. However, owing to the lack of importin-α knockout animal models in the past, their role in EBOV pathogenesis remained largely unknown. Here, we demonstrate that importin-α7 is involved in the formation of EBOV inclusion bodies and replication. However, deletion of the gene encoding importin-α7 was not sufficient to increase survival rates among mice infected with EBOV.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Inclusion Bodies, Viral/physiology , Virulence/physiology , alpha Karyopherins/metabolism , Animals , Cell Line , Chlorocebus aethiops , DNA Replication/genetics , Ebolavirus/genetics , Ebolavirus/metabolism , Mice , Mice, Inbred C57BL , Vero Cells , Viral Proteins/metabolism , Virulence/genetics , Virus Replication/genetics
4.
Blood ; 122(3): 413-23, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23704093

ABSTRACT

The t(12;21) chromosomal translocation, targeting the gene encoding the RUNX1 transcription factor, is observed in 25% of pediatric acute lymphoblastic leukemia (ALL) and is an initiating event in the disease. To elucidate the mechanism by which RUNX1 disruption initiates leukemogenesis, we investigated its normal role in murine B-cell development. This study revealed 2 critical functions of Runx1: (1) to promote survival and development of progenitors specified to the B-cell lineage, a function that can be substituted by ectopic Bcl2 expression, and (2) to enable the developmental transition through the pre-B stage triggered by the pre-B-cell antigen receptor (pre-BCR). Gene expression analysis and genomewide Runx1 occupancy studies support the hypothesis that Runx1 reinforces the transcription factor network governing early B-cell survival and development and specifically regulates genes encoding members of the Lyn kinase subfamily (key integrators of interleukin-7 and pre-BCR signaling) and the stage-specific transcription factors SpiB and Aiolos (critical downstream effectors of pre-BCR signaling). Interrogation of expression databases of 257 ALL samples demonstrated the specific down-regulation of the SPIB and IKZF3 genes (the latter encoding AIOLOS) in t(12;21) ALL, providing novel insight into the mechanism by which the translocation blocks B-cell development and promotes leukemia.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Core Binding Factor Alpha 2 Subunit/metabolism , Animals , Apoptosis/genetics , Binding Sites , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 21/genetics , Core Binding Factor Alpha 2 Subunit/deficiency , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Gene Targeting , Genome/genetics , Humans , Ikaros Transcription Factor , Mice , Mice, Inbred C57BL , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Translocation, Genetic
5.
PLoS One ; 19(1): e0295641, 2024.
Article in English | MEDLINE | ID: mdl-38215076

ABSTRACT

Brain metastasis leads to increased mortality and is a major site of relapse for several cancers, yet the molecular mechanisms of brain metastasis are not well understood. In this study, we established and characterized a new leukemic cell line, FIA10, that metastasizes into the central nervous system (CNS) following injection into the tail vein of syngeneic mice. Mice injected with FIA10 cells developed neurological symptoms such as loss of balance, tremor, ataxic gait and seizures, leading to death within 3 months. Histopathology coupled with PCR analysis clearly showed infiltration of leukemic FIA10 cells into the brain parenchyma of diseased mice, with little involvement of bone marrow, peripheral blood and other organs. To define pathways that contribute to CNS metastasis, global transcriptome and proteome analysis was performed on FIA10 cells and compared with that of the parental stem cell line FDCP-Mix and the related FIA18 cells, which give rise to myeloid leukemia without CNS involvement. 188 expressed genes (RNA level) and 189 proteins were upregulated (log2 ratio FIA10/FIA18 ≥ 1) and 120 mRNAs and 177 proteins were downregulated (log2 ratio FIA10/FIA18 ≤ 1) in FIA10 cells compared with FIA18 cells. Major upregulated pathways in FIA10 cells revealed by biofunctional analyses involved immune response components, adhesion molecules and enzymes implicated in extracellular matrix remodeling, opening and crossing the blood-brain barrier (BBB), molecules supporting migration within the brain parenchyma, alterations in metabolism necessary for growth within the brain microenvironment, and regulators for these functions. Downregulated RNA and protein included several tumor suppressors and DNA repair enzymes. In line with the function of FIA10 cells to specifically infiltrate the brain, FIA10 cells have acquired a phenotype that permits crossing the BBB and adapting to the brain microenvironment thereby escaping immune surveillance. These data and our model system FIA10 will be valuable resources to study the occurrence of brain metastases and may help in the development of potential therapies against brain invasion.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Mice , Animals , Transcriptome , Proteomics , Brain/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System Neoplasms/pathology , Brain Neoplasms/pathology , Gene Expression Profiling , RNA/metabolism , Cell Line , Tumor Microenvironment
6.
Oncogene ; 24(51): 7579-91, 2005 Nov 17.
Article in English | MEDLINE | ID: mdl-16044150

ABSTRACT

The t(12;21) translocation, generating the TEL/AML1 fusion protein, is the most common genetic lesion in childhood cancer. Using a bone marrow transplantation model, we demonstrate that TEL/AML1 expression impinges on normal hematopoietic differentiation, leading to the in vivo accumulation and persistence of an early progenitor compartment with a Sca1(+)/Kit(hi)/CD11b(+) phenotype and an increased self-renewal capacity, as documented by replating assays in vitro. Differentiation of these cells is not blocked, but the frequency of mature blood cells arising from TEL/AML1-transduced progenitors is low. Impaired differentiation is prominently observed in the pro-B-cell compartment, resulting in an proportional increase in early progenitors in vivo, consistent with the t(12;21) ALL phenotype. Despite the accumulation of both multipotent and B-cell progenitors in vivo, no leukemia induction was observed during an observation period of over 1 year. These results are consistent with findings in twins with concordant ALL, showing that TEL/AML1 generates a preleukemic clone in utero that persists for several years in a clinically covert fashion. Furthermore, our studies showed that the pointed domain of TEL/AML1, which recruits transcriptional repressors and directs oligomerization with either TEL/AML1 or wild-type TEL, was essential for the observed differentiation impairment and could not be replaced with another oligomerization domain.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Preleukemia/genetics , Animals , B-Lymphocytes , Bone Marrow Transplantation , Cell Differentiation , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Preleukemia/physiopathology , Translocation, Genetic
7.
PLoS One ; 7(12): e52247, 2012.
Article in English | MEDLINE | ID: mdl-23284953

ABSTRACT

BACKGROUND: Tyrosine kinase inhibitors (TKIs) are effective in treating malignant disorders and were lately suggested to have an impact on non-malignant diseases. However, in some inflammatory conditions like rheumatoid arthritis (RA) the in vivo effect seemed to be moderate. As most TKIs are taken up actively into cells by cell membrane transporters, this study aimed to evaluate the role of such transporters for the accumulation of the TKI Imatinib mesylates in RA synovial fibroblasts as well as their regulation under inflammatory conditions. METHODOLOGY/PRINCIPAL FINDINGS: The transport and accumulation of Imatinib was investigated in transporter-transfected HEK293 cells and human RA synovial fibroblasts (hRASF). Transporter expression was quantified by qRT-PCR. In transfection experiments, hMATE1 showed the highest apparent affinity for Imatinib among all known Imatinib transporters. Experiments quantifying the Imatinib uptake in the presence of specific transporter inhibitors and after siRNA knockdown of hMATE1 indeed identified hMATE1 to mediate Imatinib transport in hRASF. The anti-proliferative effect of Imatinib on PDGF stimulated hRASF was quantified by cell counting and directly correlated with the uptake activity of hMATE1. Expression of hMATE1 was investigated by Western blot and immuno-fluorescence. Imatinib transport under disease-relevant conditions, such as an altered pH and following stimulation with different cytokines, was also investigated by HPLC. The uptake was significantly reduced by an acidic extracellular pH as well as by the cytokines TNFα, IL-1ß and IL-6, which all decreased the expression of hMATE1-mRNA and protein. CONCLUSION/SIGNIFICANCE: The regulation of Imatinib uptake via hMATE1 in hRASF and resulting effects on their proliferation may explain moderate in vivo effects on RA. Moreover, our results suggest that investigating transporter mediated drug processing under normal and pathological conditions is important for developing intracellular acting drugs used in inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Benzamides/pharmacology , Cell Line , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Cytokines/pharmacology , Humans , Hydrogen-Ion Concentration , Imatinib Mesylate , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors , Pyrimidines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
8.
Dev Comp Immunol ; 34(11): 1160-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600277

ABSTRACT

The genome of Caenorhabditis elegans includes six homologs of matrix metalloproteinases (MMPs). The C. elegans MMP gene zmp-1 has recently been shown to be involved in anchor cell invasion during post-embryonic vulval development. Here, we identified H19M22.3 (zmp-2) as a pleiotropic MMP gene regulating disease resistance, molting, larval development, and fecundity. Zmp-2(RNAi) nematodes showed significant lifespan reduction during infection with pathogenic Photorhabdus luminescence. Moreover, we observed molting defects indicating a direct or regulative role in extracellular matrix degradation during ecdysis, delayed larval to adult development, and reduced offspring production in hermaphrodite adults. GFP-expressing nematodes revealed predominant expression of zmp-2 in multiple cells during embryogenesis; in hypodermal, muscle, and somatic gonad cells during larval development; and in developing and mature spermathecae in the L4 larval stage and adults. These results give evidence for pleiotropic roles of zmp-2 and provide novel insights into evolutionarily conserved and derived MMP functions in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans , Enterobacteriaceae Infections/immunology , Matrix Metalloproteinases/metabolism , Photorhabdus/immunology , Animals , Animals, Genetically Modified , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Enterobacteriaceae Infections/genetics , Fertility/genetics , Gene Expression Regulation, Developmental/immunology , Humans , Immunity/genetics , Jurkat Cells , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/immunology , Metalloendopeptidases , Molting/genetics , Photorhabdus/pathogenicity , Protein Engineering , RNA, Small Interfering/genetics
9.
Blood Cells Mol Dis ; 40(2): 211-8, 2008.
Article in English | MEDLINE | ID: mdl-17920312

ABSTRACT

The RUNX1 gene encodes the alpha subunit of the core binding factor (CBF) and is a common target of genetic mutations in acute leukemia. We propose that RUNX1 is a gatekeeper gene, the disruption of which leads to the exodus of a subset of hematopoietic progenitors with increased self-renewal potential from the normal environmental controls of homeostasis. This pool of "escaped" cells is the target of secondary mutations, accumulating over time to induce the aggressive manifestation of acute leukemia. Evidence from patient and animal studies supports the concept that RUNX1 mutations are the initiating event in different leukemia subtypes, but also suggests that diverse mechanisms are used to subvert RUNX1 function. One common result is the inhibition of differentiation-but its effect impinges on different lineages and stages of differentiation, depending on the mutation or fusion partner. A number of different approaches have led to the identification of secondary events that lead to the overt acute phase; however, the majority is unknown. Finally, the concept of the "leukemia stem cell" and its therapeutic importance is discussed in light of the RUNX1 gatekeeper function.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Transcription, Genetic
10.
J Virol ; 81(2): 732-42, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17079317

ABSTRACT

Murine leukemia viruses (MuLV) induce leukemia through a multistage process, a critical step being the activation of oncogenes through provirus integration. Transcription elements within the long terminal repeats (LTR) are prime determinants of cell lineage specificity; however, the influence of other factors, including the Env protein that modulates cell tropism through receptor recognition, has not been rigorously addressed. The ability of 10A1-MuLV to use both PiT1 and PiT2 receptors has been implicated in its induction of blast cell leukemia. Here we show that restricting receptor usage of 10A1-MuLV to PiT2 results in loss of blast cell transformation capacity. However, the pathogenicity was unaltered when the env gene is exchanged with Moloney MuLV, which uses the Cat1 receptor. Significantly, the leukemic blasts express erythroid markers and consistently contain proviral integrations in the Fli1 locus, a target of Friend MuLV (F-MuLV) during erythroleukemia induction. Furthermore, an NB-tropic variant of 10A1 was unable to induce blast cell leukemia in C57BL/6 mice, which are also resistant to F-MuLV transformation. We propose that 10A1- and F-MuLV actually induce identical (erythro)blastic leukemia by a mechanism involving Fli1 activation and cooperation with inherent genetic mutations in susceptible mouse strains. Furthermore, we demonstrate that deletion of the Icsbp tumor suppressor gene in C57BL/6 mice is sufficient to confer susceptibility to 10A1-MuLV leukemia induction but with altered specificity. In summary, we validate the significance of the env gene in leukemia specificity and underline the importance of a complex interplay of cooperating oncogenes and/or tumor suppressors in determining the pathogenicity of MuLV variants.


Subject(s)
Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Leukemia Virus, Murine/pathogenicity , Proto-Oncogene Protein c-fli-1/metabolism , Receptors, Virus/metabolism , Animals , Cells, Cultured , Fibroblasts , Gene Products, env/genetics , Gene Products, env/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Leukemia, Experimental/pathology , Leukemia, Experimental/virology , Mice , Mice, Inbred C57BL , Proto-Oncogene Protein c-fli-1/genetics , Retroviridae Infections/pathology , Retroviridae Infections/virology , Species Specificity , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
11.
Blood ; 103(7): 2744-52, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14656889

ABSTRACT

The CCAAT/enhancer binding protein alpha (C/EBPalpha) is an essential transcription factor for granulocytic differentiation. C/EBPalpha mutations are found in approximately 8% of acute myeloid leukemia (AML) patients. Most of these mutations occur in the N-terminal coding region, resulting in a frame shift and the enhanced translation of a dominant-negative 30-kDa protein, which may be responsible for the differentiation block observed in AML. To test this hypothesis, we introduced a cDNA encoding an N-terminal mutated C/EBPalpha (mut10) into primary hematopoietic progenitors using a retroviral vector. Expression of mut10 in human CD34+ cord blood cells dramatically inhibited differentiation of both myeloid and erythroid lineages. Immunohistochemical analysis demonstrated coexpression of both myeloid and erythroid markers in the immature transformed cells. Surprisingly, mut10 did not block myelocytic differentiation in murine progenitors but did alter their differentiation kinetics and clonogenicity. Experiments were performed to confirm that the differential effect of mut10 on murine and human progenitors was not due to species-specific differences in C/EBPalpha protein sequences, expression levels, or inefficient targeting of relevant cells. Taken together, our results underline the intrinsic differences between hematopoietic controls in mouse and human and support the hypothesis that mutations in CEBPA are critical events in the disruption of myeloid differentiation in AMLs.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/pharmacology , Erythroid Precursor Cells/drug effects , Leukemia, Myeloid, Acute/genetics , Mutation , Myeloid Progenitor Cells/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Cloning, Molecular , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/pathology , Female , Fetal Blood/cytology , Genes, Dominant , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Infant, Newborn , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/pathology , Recombinant Proteins/pharmacology , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL