Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Arthritis Res Ther ; 23(1): 202, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321071

ABSTRACT

OBJECTIVES: Hyperuricemia is a metabolic condition central to gout pathogenesis. Urate exposure primes human monocytes towards a higher capacity to produce and release IL-1ß. In this study, we assessed the epigenetic processes associated to urate-mediated hyper-responsiveness. METHODS: Freshly isolated human peripheral blood mononuclear cells or enriched monocytes were pre-treated with solubilized urate and stimulated with LPS with or without monosodium urate (MSU) crystals. Cytokine production was determined by ELISA. Histone epigenetic marks were assessed by sequencing immunoprecipitated chromatin. Mice were injected intraarticularly with MSU crystals and palmitate after inhibition of uricase and urate administration in the presence or absence of methylthioadenosine. DNA methylation was assessed by methylation array in whole blood of 76 participants with normouricemia or hyperuricemia. RESULTS: High concentrations of urate enhanced the inflammatory response in vitro in human cells and in vivo in mice, and broad-spectrum methylation inhibitors reversed this effect. Assessment of histone 3 lysine 4 trimethylation (H3K4me3) and histone 3 lysine 27 acetylation (H3K27ac) revealed differences in urate-primed monocytes compared to controls. Differentially methylated regions (e.g. HLA-G, IFITM3, PRKAB2) were found in people with hyperuricemia compared to normouricemia in genes relevant for inflammatory cytokine signaling. CONCLUSION: Urate alters the epigenetic landscape in selected human monocytes or whole blood of people with hyperuricemia compared to normouricemia. Both histone modifications and DNA methylation show differences depending on urate exposure. Subject to replication and validation, epigenetic changes in myeloid cells may be a therapeutic target in gout.


Subject(s)
Gout , Uric Acid , Animals , Epigenesis, Genetic , Gout/genetics , Humans , Leukocytes, Mononuclear , Membrane Proteins , Mice , Monocytes , RNA-Binding Proteins
2.
Mucosal Immunol ; 11(5): 1512-1523, 2018 09.
Article in English | MEDLINE | ID: mdl-30038215

ABSTRACT

The tissue dendritic cell (DC) compartment is heterogeneous, and the ontogeny and functional specialization of human tissue conventional DC (cDC) subsets and their relationship with monocytes is unresolved. Here we identify monocyte-related CSF1R+Flt3- antigen presenting cells (APCs) that constitute about half of the cells classically defined as SIRPα+ DCs in the steady-state human small intestine. CSF1R+Flt3- APCs express calprotectin and very low levels of CD14, are transcriptionally related to monocyte-derived cells, and accumulate during inflammation. CSF1R+Flt3- APCs show typical macrophage characteristics functionally distinct from their Flt3+ cDC counterparts: under steady-state conditions they excel at antigen uptake, have a lower migratory potential, and are inefficient activators of naïve T cells. These results have important implications for the understanding of the ontogenetic and functional heterogeneity within human tissue DCs and their relation to the monocyte lineage.


Subject(s)
Dendritic Cells/physiology , Intestines/physiology , Macrophages/physiology , Monocytes/physiology , Transcription, Genetic/physiology , Transcriptome/physiology , Aged , Aged, 80 and over , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , Cell Lineage/physiology , Dendritic Cells/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/physiopathology , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , fms-Like Tyrosine Kinase 3/metabolism
3.
Mol Psychiatry ; 23(5): 1356-1367, 2018 05.
Article in English | MEDLINE | ID: mdl-28416808

ABSTRACT

Synapse development and neuronal activity represent fundamental processes for the establishment of cognitive function. Structural organization as well as signalling pathways from receptor stimulation to gene expression regulation are mediated by synaptic activity and misregulated in neurodevelopmental disorders such as autism spectrum disorder (ASD) and intellectual disability (ID). Deleterious mutations in the PTCHD1 (Patched domain containing 1) gene have been described in male patients with X-linked ID and/or ASD. The structure of PTCHD1 protein is similar to the Patched (PTCH1) receptor; however, the cellular mechanisms and pathways associated with PTCHD1 in the developing brain are poorly determined. Here we show that PTCHD1 displays a C-terminal PDZ-binding motif that binds to the postsynaptic proteins PSD95 and SAP102. We also report that PTCHD1 is unable to rescue the canonical sonic hedgehog (SHH) pathway in cells depleted of PTCH1, suggesting that both proteins are involved in distinct cellular signalling pathways. We find that Ptchd1 deficiency in male mice (Ptchd1-/y) induces global changes in synaptic gene expression, affects the expression of the immediate-early expression genes Egr1 and Npas4 and finally impairs excitatory synaptic structure and neuronal excitatory activity in the hippocampus, leading to cognitive dysfunction, motor disabilities and hyperactivity. Thus our results support that PTCHD1 deficiency induces a neurodevelopmental disorder causing excitatory synaptic dysfunction.


Subject(s)
Cognitive Dysfunction/metabolism , Membrane Proteins/deficiency , Synapses/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cognition/physiology , Cognitive Dysfunction/genetics , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Hippocampus/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Signal Transduction , Synapses/genetics , Synaptic Transmission
4.
Leukemia ; 32(3): 828-836, 2018 03.
Article in English | MEDLINE | ID: mdl-28871137

ABSTRACT

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Subject(s)
Gene Rearrangement , Leukemia, Myeloid, Acute/genetics , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Transcriptional Activation , Translocation, Genetic , Cell Line , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , Epigenesis, Genetic , Exons , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Introns , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic
6.
Leukemia ; 31(11): 2315-2325, 2017 11.
Article in English | MEDLINE | ID: mdl-28216661

ABSTRACT

Deregulation of epigenetic mechanisms, including microRNA, contributes to leukemogenesis and drug resistance by interfering with cancer-specific molecular pathways. Here, we show that the balance between miR-194-5p and its newly discovered target BCL2-associated transcription factor 1 (BCLAF1) regulates differentiation and survival of normal hematopoietic progenitors. In acute myeloid leukemias this balance is perturbed, locking cells into an immature, potentially 'immortal' state. Enhanced expression of miR-194-5p by treatment with the histone deacetylase inhibitor SAHA or by exogenous miR-194-5p expression re-sensitizes cells to differentiation and apoptosis by inducing BCLAF1 to shuttle between nucleus and cytosol. miR-194-5p/BCLAF1 balance was found commonly deregulated in 60 primary acute myeloid leukemia patients and was largely restored by ex vivo SAHA treatment. Our findings link treatment responsiveness to re-instatement of miR-194-5p/BCLAF1 balance.


Subject(s)
Gene Expression Regulation , Leukemia, Myeloid, Acute/pathology , MicroRNAs/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Apoptosis , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Down-Regulation , Humans , Leukemia, Myeloid, Acute/genetics
7.
Oncogene ; 36(23): 3346-3356, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28114278

ABSTRACT

In 11q23 leukemias, the N-terminal part of the mixed lineage leukemia (MLL) gene is fused to >60 different partner genes. In order to define a core set of MLL rearranged targets, we investigated the genome-wide binding of the MLL-AF9 and MLL-AF4 fusion proteins and associated epigenetic signatures in acute myeloid leukemia (AML) cell lines THP-1 and MV4-11. We uncovered both common as well as specific MLL-AF9 and MLL-AF4 target genes, which were all marked by H3K79me2, H3K27ac and H3K4me3. Apart from promoter binding, we also identified MLL-AF9 and MLL-AF4 binding at specific subsets of non-overlapping active distal regulatory elements. Despite this differential enhancer binding, MLL-AF9 and MLL-AF4 still direct a common gene program, which represents part of the RUNX1 gene program and constitutes of CD34+ and monocyte-specific genes. Comparing these data sets identified several zinc finger transcription factors (TFs) as potential MLL-AF9 co-regulators. Together, these results suggest that MLL fusions collaborate with specific subsets of TFs to deregulate the RUNX1 gene program in 11q23 AMLs.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Adult , Core Binding Factor Alpha 2 Subunit/genetics , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Prognosis , Promoter Regions, Genetic
8.
Oncogene ; 35(15): 1965-76, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26148230

ABSTRACT

The ETS transcription factor ERG has been implicated as a major regulator of both normal and aberrant hematopoiesis. In acute myeloid leukemias harboring t(16;21), ERG function is deregulated due to a fusion with FUS/TLS resulting in the expression of a FUS-ERG oncofusion protein. How this oncofusion protein deregulates the normal ERG transcription program is unclear. Here, we show that FUS-ERG acts in the context of a heptad of proteins (ERG, FLI1, GATA2, LYL1, LMO2, RUNX1 and TAL1) central to proper expression of genes involved in maintaining a stem cell hematopoietic phenotype. Moreover, in t(16;21) FUS-ERG co-occupies genomic regions bound by the nuclear receptor heterodimer RXR:RARA inhibiting target gene expression and interfering with hematopoietic differentiation. All-trans retinoic acid treatment of t(16;21) cells as well as FUS-ERG knockdown alleviate the myeloid-differentiation block. Together, the results suggest that FUS-ERG acts as a transcriptional repressor of the retinoic acid signaling pathway.


Subject(s)
Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 21/genetics , Gene Expression Regulation, Neoplastic/genetics , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein FUS/physiology , Signal Transduction/physiology , Translocation, Genetic , Tretinoin/physiology , Amino Acid Motifs , Cell Line, Tumor , Chromosomes, Human, Pair 16/ultrastructure , Chromosomes, Human, Pair 21/ultrastructure , Dimerization , Enhancer Elements, Genetic , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/physiopathology , Leukemia, Myelomonocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/physiopathology , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/antagonists & inhibitors , RNA-Binding Protein FUS/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , U937 Cells
9.
Leukemia ; 28(4): 770-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24002588

ABSTRACT

Different mechanisms for CBFß-MYH11 function in acute myeloid leukemia with inv(16) have been proposed such as tethering of RUNX1 outside the nucleus, interference with transcription factor complex assembly and recruitment of histone deacetylases, all resulting in transcriptional repression of RUNX1 target genes. Here, through genome-wide CBFß-MYH11-binding site analysis and quantitative interaction proteomics, we found that CBFß-MYH11 localizes to RUNX1 occupied promoters, where it interacts with TAL1, FLI1 and TBP-associated factors (TAFs) in the context of the hematopoietic transcription factors ERG, GATA2 and PU.1/SPI1 and the coregulators EP300 and HDAC1. Transcriptional analysis revealed that upon fusion protein knockdown, a small subset of the CBFß-MYH11 target genes show increased expression, confirming a role in transcriptional repression. However, the majority of CBFß-MYH11 target genes, including genes implicated in hematopoietic stem cell self-renewal such as ID1, LMO1 and JAG1, are actively transcribed and repressed upon fusion protein knockdown. Together these results suggest an essential role for CBFß-MYH11 in regulating the expression of genes involved in maintaining a stem cell phenotype.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 16 , Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Leukemia, Myeloid, Acute/genetics , Myosin Heavy Chains/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Binding Sites , GATA2 Transcription Factor/physiology , Histone Deacetylases/physiology , Humans , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/physiology , Proto-Oncogene Proteins/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcriptional Activation
10.
Mol Ecol Resour ; 11(4): 662-74, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21676196

ABSTRACT

Next-generation sequencing (NGS) technologies are increasingly applied in many organisms, including nonmodel organisms that are important for ecological and conservation purposes. Illumina and 454 sequencing are among the most used NGS technologies and have been shown to produce optimal results at reasonable costs when used together. Here, we describe the combined application of these two NGS technologies to characterize the transcriptome of a plant species of ecological and conservation relevance for which no genomic resource is available, Scabiosa columbaria. We obtained 528,557 reads from a 454 GS-FLX run and a total of 28,993,627 reads from two lanes of an Illumina GAII single run. After read trimming, the de novo assembly of both types of reads produced 109,630 contigs. Both the contigs and the >75 bp remaining singletons were blasted against the Uniprot/Swissprot database, resulting in 29,676 and 10,515 significant hits, respectively. Based on sequence similarity with known gene products, these sequences represent at least 12,516 unique genes, most of which are well covered by contig sequences. In addition, we identified 4320 microsatellite loci, of which 856 had flanking sequences suitable for PCR primer design. We also identified 75,054 putative SNPs. This annotated sequence collection and the relative molecular markers represent a main genomic resource for S. columbaria which should contribute to future research in conservation and population biology studies. Our results demonstrate the utility of NGS technologies as starting point for the development of genomic tools in nonmodel but ecologically important species.


Subject(s)
Dipsacaceae/genetics , Gene Expression Profiling , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Genome, Plant
11.
Br J Cancer ; 104(4): 554-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21245861

ABSTRACT

PML-RAR (retinoic acid receptor)α is the hallmark protein of acute promyelocytic leukaemia, a highly malignant subtype of acute myeloid leukaemia that accounts for approximately 10% of all AML cases. Recently, several studies have been set out to obtain a comprehensive genome-wide view of the molecular actions of this chimeric protein. In this review, we highlight the new insights that arose from these studies, in particular focussing on newly identified PML-RARα target genes, its interplay with RXR and deregulation of epigenetic modifications.


Subject(s)
Genome, Human , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/physiology , Animals , Epigenesis, Genetic/physiology , Humans , Models, Biological , Oncogene Proteins, Fusion/genetics , Signal Transduction/genetics , Signal Transduction/physiology
12.
Nucleic Acids Res ; 29(16): 3424-32, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11504880

ABSTRACT

An important model system for studying the process leading to productive transcription is provided by the superfamily of nuclear receptors, which are for the most part ligand-controlled transcription factors. Over the past years several 'orphan' nuclear receptors have been isolated for which no ligand has yet been identified. Very little is known about how these 'orphan' receptors regulate transcription. In this study we have analysed the biochemical and transcriptional properties of the neuronally expressed orphan nuclear receptor RORbeta (NR1F2) and compared them with the retinoic acid receptor heterodimer RXRalpha-RARalpha (NR2B1-NR1B1) and Gal-VP16 in vitro. Although RORbeta binds to its DNA-binding sites with comparatively low affinity, it efficiently directs transcription in nuclear extracts derived from a neuronal cell line, Neuro2A, but not in nuclear extracts from non-neuronal HeLa cells. In contrast, RXRalpha-RARalpha and the acidic transcription factor Gal-VP16 support transcription in Neuro2A and HeLa nuclear extracts equally efficiently. These observations point to a different (co)factor requirement for transactivation by members of the NR1 subfamily of nuclear receptors.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Neurons/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear , Transcription, Genetic/genetics , Base Sequence , Binding Sites , Cell Extracts , DNA/genetics , DNA/metabolism , Dimerization , Genetic Vectors/genetics , HeLa Cells , Herpes Simplex Virus Protein Vmw65/metabolism , Humans , Neurons/cytology , Nuclear Receptor Subfamily 1, Group F, Member 2 , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors , Thermodynamics , Transcription Factors/metabolism , Transcriptional Activation , Tumor Cells, Cultured , Vaccinia virus/genetics
13.
Oncogene ; 20(24): 3100-9, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11420726

ABSTRACT

Transcriptional regulation at the level of chromatin plays crucial roles during eukaryotic development and differentiation. A plethora of studies revealed that the acetylation status of histones is controlled by multi-protein complexes containing (de)acetylase activities. In the current model, histone deacetylases and acetyltransferases are recruited to chromatin by DNA-bound repressors and activators, respectively. Shifting the balance between deacetylation, i.e. repressive chromatin and acetylation, i.e. active chromatin can lead to aberrant gene transcription and cancer. In human acute promyelocytic leukemia (APL) and avian erythroleukemia (AEL), chromosomal translocations and/or mutations in nuclear hormone receptors, RARalpha [NR1B1] and TRalpha [NR1A1], yielded oncoproteins that deregulate transcription and alter chromatin structure. The oncogenic receptors are locked in their 'off' mode thereby constitutively repressing transcription of genes that are critical for differentiation of hematopoietic cells. AEL involves an oncogenic version of the chicken TRalpha, v-ErbA. Apart from repression by v-ErbA via recruitment of corepressor complexes, other repressors and corepressors appear to be involved in repression of v-ErbA target genes, such as carbonic anhydrase II (CAII). Reactivation of repressed genes in APL and AEL by chromatin modifying agents such as inhibitors of histone deacetylase or of methylation provides new therapeutic strategies in the treatment of acute myeloid leukemia.


Subject(s)
Avian Leukosis/genetics , Retroviridae Infections/genetics , Alpharetrovirus , Animals , Avian Leukosis/virology , Chickens , Gene Expression Regulation , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/virology , Models, Biological , Neoplasms/genetics , Repressor Proteins/physiology , Retroviridae Infections/virology
15.
Oncogene ; 20(7): 775-87, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11314012

ABSTRACT

v-ErbA is a mutated variant of thyroid hormone receptor (TRalpha/NR1A1) borne by the Avian Erythroblastosis virus causing erythroleukemia. TRalpha is known to activate transcription of specific genes in the presence of its cognate ligand, T3 hormone, while in its absence it represses it. v-ErbA is unable to bind ligand, and hence is thought to contribute to leukemogenesis by actively repressing erythroid-specific genes such as the carbonic anhydrase II gene (CA II). In the prevailing model, v-ErbA occludes liganded TR from binding to its cognate elements and constitutively interacts with the corepressors NCoR/SMRT. We previously identified a v-ErbA responsive element (VRE) within a DNase I hypersensitive region (HS2) located in the second intron of the CA II gene. We now show that HS2 fulfils all the requirements for a genuine enhancer that functions independent of its orientation and position with a profound erythroid-specific activity in normal erythroid progenitors (T2ECs) and in leukemic erythroid cell lines. We find that the HS2 enhancer activity is governed by two adjacent GATA-factor binding sites. v-ErbA as well as unliganded TR prevent HS2 activity by nullifying the positive function of factors bound to GATA-sites. However, v-ErbA, in contrast to TR, does not convey active repression to silence the transcriptional activity intrinsic to a heterologous tk promoter. We propose that depending on the sequence and context of the binding site, v-ErbA contributes to leukemogenesis by occluding liganded TR as well as unliganded TR thereby preventing activation or repression, respectively.


Subject(s)
Carbonic Anhydrases/genetics , Enhancer Elements, Genetic , Leukemia, Erythroblastic, Acute/genetics , Oncogene Proteins v-erbA/genetics , Receptors, Thyroid Hormone/genetics , Animals , Base Sequence , Binding Sites , Chickens , Erythroid Precursor Cells , Gene Expression Regulation, Leukemic , Humans , Mice , Models, Genetic , Molecular Sequence Data , Protein Binding , Tumor Cells, Cultured
16.
Cell ; 104(1): 153-64, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11163248

ABSTRACT

Fertilization and zygote development are obligate features of the malaria parasite life cycle and occur during parasite transmission to mosquitoes. The surface protein PFS48/45 is expressed by male and female gametes of Plasmodium falciparum and PFS48/45 antibodies prevent zygote development and transmission. Here, gene disruption was used to show that Pfs48/45 and the ortholog Pbs48/45 from a rodent malaria parasite P. berghei play a conserved and important role in fertilization. p48/45- parasites had a reduced capacity to produce oocysts in mosquitoes due to greatly reduced zygote formation. Unexpectedly, only male gamete fertility of p48/45- parasites was affected, failing to penetrate otherwise fertile female gametes. P48/45 is shown to be a surface protein of malaria parasites with a demonstrable role in fertilization.


Subject(s)
Malaria/physiopathology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Antibodies , Culicidae , Female , Fertility/physiology , Gametogenesis/physiology , Genome, Protozoan , Malaria/immunology , Malaria/prevention & control , Malaria Vaccines , Male , Molecular Sequence Data , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Zygote
17.
Nat Struct Biol ; 7(12): 1100-4, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101889

ABSTRACT

Sin3A or Sin3B are components of a corepressor complex that mediates repression by transcription factors such as the helix-loop-helix proteins Mad and Mxi. Members of the Mad/Mxi family of repressors play important roles in the transition between proliferation and differentiation by down-regulating the expression of genes that are activated by the proto-oncogene product Myc. Here, we report the solution structure of the second paired amphipathic helix (PAH) domain (PAH2) of Sin3B in complex with a peptide comprising the N-terminal region of Mad1. This complex exhibits a novel interaction fold for which we propose the name 'wedged helical bundle'. Four alpha-helices of PAH2 form a hydrophobic cleft that accommodates an amphipathic Mad1 alpha-helix. Our data further show that, upon binding Mad1, secondary structure elements of PAH2 are stabilized. The PAH2-Mad1 structure provides the basis for determining the principles of protein interaction and selectivity involving PAH domains.


Subject(s)
Carrier Proteins , Nuclear Proteins , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transcription Factors , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins , Conserved Sequence , Humans , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Mas , Sequence Alignment , Solutions , Substrate Specificity
18.
Biochim Biophys Acta ; 1494(3): 236-41, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11121580

ABSTRACT

Nuclear receptors are ligand-inducible transcription factors that can be classified into two major groups according to their DNA-binding properties. Members of the first group bind to DNA as dimers, either homo- or heterodimers; members of the second group are also able to bind as monomers. While the first group has been extensively studied biochemically, very little is known about nuclear receptors that bind and act as monomers. In this study, we compared the binding and transcriptional behaviour of ROR alpha (NR1F1) and ROR beta (NR1F2), two representatives of the subgroup of monomer-binding receptors. We show that although they are highly related in their amino acid structures, they display remarkably different binding behaviours. Furthermore, we provide evidence that ROR beta can efficiently activate transcription in vitro as a monomer.


Subject(s)
Receptors, Cell Surface/genetics , Receptors, Cytoplasmic and Nuclear , Binding Sites , Escherichia coli/genetics , Gene Expression , Nuclear Receptor Subfamily 1, Group F, Member 1 , Nuclear Receptor Subfamily 1, Group F, Member 2 , Receptor Protein-Tyrosine Kinases , Receptor Tyrosine Kinase-like Orphan Receptors , Receptors, Cell Surface/metabolism , Trans-Activators , Transcription, Genetic , Vaccinia virus/genetics
19.
Mol Cell ; 6(3): 527-37, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11030333

ABSTRACT

Transcription of TATA box-containing genes by RNA polymerase II is mediated by TBP-containing and TBP-free multisubunit complexes consisting of common and unique components. We have identified a highly stable TBP-TFIIA-containing complex, TAC, which is detectable in embryonal carcinoma (EC) cells but not in differentiated cells. TAC contains the TFIIAgamma subunit and the unprocessed form of TFIIAalphabeta, although the processed TFIIAalpha and TFIIAbeta subunits are present in EC cells. TAC mediates transcriptional activation by RNA polymerase II in vivo, even though it does not contain classical TAFs. Formaldehyde cross-linking revealed that in EC but not in differentiated cells, association of TBP with chromatin is strongly enhanced when complexed with TFIIA in vivo. Remarkably, the TFIIAalphabeta precursor is preferentially, if not exclusively, associated with chromatin as compared to the processed subunits present in "free" TFIIA in EC cells.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , COS Cells , Carcinoma, Embryonal , Chromatin/genetics , Chromatin/metabolism , Cross-Linking Reagents , DNA/metabolism , Formaldehyde , Gene Expression/physiology , Humans , Protein Precursors/genetics , Protein Precursors/metabolism , TATA Box/physiology , TATA-Box Binding Protein , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription Factors/chemistry , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Transcriptional Activation/physiology , Transfection , Tumor Cells, Cultured
20.
Vaccine ; 18(14): 1402-11, 2000 Jan 31.
Article in English | MEDLINE | ID: mdl-10618538

ABSTRACT

Pfs48/45 is an important transmission-blocking vaccine candidate antigen of the human malaria parasite Plasmodium falciparum. This study was aimed at synthesis of recombinant Pfs48/45 containing conformation-constrained epitopes of the native antigen in yeast. Since in the yeast Saccharomyces cerevisiae induction of gene-expression led to prematurely terminated transcripts, an entirely synthetic gene of higher GC content was assembled. Replacement of the AT rich natural gene by the synthetic gene relieved the observed premature transcription termination. Nevertheless, recombinant protein expression could not be detected. In contrast, in the yeast Pichia pastoris low levels of recombinant Pfs48/45 were produced upon induction of synthetic gene expression. The recombinant protein was shown to be disulphide-bridge constrained, but was not recognised by transmission-blocking antibodies and did not induce transmission-blocking sera in mice.


Subject(s)
Membrane Glycoproteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Protozoan/genetics , Genes, Synthetic , Humans , Malaria/parasitology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Membrane Glycoproteins/isolation & purification , Molecular Sequence Data , Pichia/genetics , Pichia/metabolism , Protozoan Proteins/biosynthesis , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...