Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35
Filtrer
1.
Viruses ; 16(4)2024 04 10.
Article de Anglais | MEDLINE | ID: mdl-38675925

RÉSUMÉ

The interferon-induced host cell protein Shiftless (SFL) inhibits -1 programmed ribosomal frameshifting (-1PRF) required for the expression of HIV-1 Gal-Pol and the formation of infectious HIV-1 particles. However, the specific regions in SFL required for antiviral activity and the mechanism by which SFL inhibits -1PRF remain unclear. Employing alanine scanning mutagenesis, we found that basic amino acids in the predicted zinc ribbon motif of SFL are essential for the suppression of Gag-Pol expression but dispensable for anti-HIV-1 activity. We have shown that SFL inhibits the expression of the murine leukemia virus (MLV) Gag-Pol polyprotein and the formation of infectious MLV particles, although Gag-Pol expression of MLV is independent of -1PRF but requires readthrough of a stop codon. These findings indicate that SFL might inhibit HIV-1 infection by more than one mechanism and that SFL might target programmed translational readthrough as well as -1PRF signals, both of which are regulated by mRNA secondary structure elements.


Sujet(s)
Protéines de fusion gag-pol , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Protéines de liaison à l'ARN , Humains , Décalage ribosomique , Protéines de fusion gag-pol/génétique , Protéines de fusion gag-pol/métabolisme , Régulation de l'expression des gènes viraux , Cellules HEK293 , Infections à VIH/virologie , Infections à VIH/génétique , Infections à VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Virus de la leucémie murine/génétique , Virus de la leucémie murine/physiologie , Réplication virale , Protéines de liaison à l'ARN/métabolisme
2.
Appl Biosaf ; 29(1): 26-34, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38434099

RÉSUMÉ

Introduction: The current global situation with increasing zoonotic transmissions of pathogens, rapidly changing ecosystems due to the climate change and with it the distribution of potential vectors, demands new ways of teaching and educating students in the field of infectious disease research. Methods: The international master program "Infectious Diseases and One Health-IDOH" started its second application period in 2019. Biosafety is an integral part of IDOH, exemplified by a biosafety level 3 hands-on training at the Animal Health Research Center IRTA-Centre de Recerca en Sanitat Animal (CReSA), Barcelona. At Hanover Medical School, biosafety is expanded to a comprehensive biorisk management approach with focus on risk assessment, bioethics, and training in a mobile containment laboratory. This article illustrates in depth the intention and the individual teaching units of the biorisk management module within the third semester of the IDOH master. Risk assessment is taught based on the new WHO Laboratory Biosafety Manual 4th edition, which represents a paradigm shift toward a risk-based approach instead of a prescriptive definition of biosafety levels. This methodology will enable the international IDOH students to cope with different national legislations and to provide guidance on biosafety in their home countries. In the final unit, the students operate a mobile containment laboratory and simulated a viral hemorrhagic fever outbreak in Africa. Conclusion: In sum, this module combines theoretical risk assessment and its practical implementation in the mobile laboratory as a future direction for training infectiologists. In addition, our report may serve as a blue print for others to amend their education with the herewith mentioned pillars of biosafety teaching.

3.
Nat Commun ; 13(1): 5215, 2022 09 05.
Article de Anglais | MEDLINE | ID: mdl-36064805

RÉSUMÉ

Hemochromatosis is one of the most common inherited metabolic diseases among white populations and predominantly originates from a homozygous C282Y mutation in the HFE gene. The G > A transition at position c.845 of the gene causes misfolding of the HFE protein, ultimately resulting in its absence at the cell membrane. Consequently, the lack of interaction with the transferrin receptors 1 and 2 leads to systemic iron overload. We screened potential gRNAs in a highly precise cell culture assay and applied an AAV8 split-vector expressing the adenine base editor ABE7.10 and our candidate gRNA in 129-Hfetm.1.1Nca mice. Here we show that a single injection of our therapeutic vector leads to a gene correction rate of >10% and improved iron metabolism in the liver. Our study presents a proof-of-concept for a targeted gene correction therapy for one of the most frequent hereditary diseases affecting humans.


Sujet(s)
Adénine , Protéine de l'hémochromatose , Hémochromatose , Adénine/métabolisme , Animaux , Ferritines/génétique , Hémochromatose/génétique , Hémochromatose/métabolisme , Hémochromatose/thérapie , Protéine de l'hémochromatose/génétique , Protéine de l'hémochromatose/métabolisme , Antigènes d'histocompatibilité de classe I/métabolisme , Homozygote , Fer/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Mutation , Transferrine/métabolisme
5.
PLoS Biol ; 19(11): e3001423, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34735435

RÉSUMÉ

Herpesviruses cause severe diseases particularly in immunocompromised patients. Both genome packaging and release from the capsid require a unique portal channel occupying one of the 12 capsid vertices. Here, we report the 2.6 Å crystal structure of the pentameric pORF19 of the γ-herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) resembling the portal cap that seals this portal channel. We also present the structure of its ß-herpesviral ortholog, revealing a striking structural similarity to its α- and γ-herpesviral counterparts despite apparent differences in capsid association. We demonstrate pORF19 pentamer formation in solution and provide insights into how pentamerization is triggered in infected cells. Mutagenesis in its lateral interfaces blocked pORF19 pentamerization and severely affected KSHV capsid assembly and production of infectious progeny. Our results pave the way to better understand the role of pORF19 in capsid assembly and identify a potential novel drug target for the treatment of herpesvirus-induced diseases.


Sujet(s)
Herpèsvirus humain de type 8/physiologie , Cadres ouverts de lecture/génétique , Multimérisation de protéines , Protéines virales/métabolisme , Assemblage viral/physiologie , Animaux , Capside/composition chimique , Séquence conservée , Cristallographie aux rayons X , Empaquetage de l'ADN , ADN viral/génétique , Drosophila , Cellules HEK293 , Herpèsvirus humain de type 8/ultrastructure , Humains , Modèles moléculaires , Mutagenèse/génétique , Protéines mutantes/métabolisme , Protéines virales/composition chimique
6.
Viruses ; 13(6)2021 05 27.
Article de Anglais | MEDLINE | ID: mdl-34071819

RÉSUMÉ

Alternative splicing and the expression of intron-containing mRNAs is one hallmark of HIV gene expression. To facilitate the otherwise hampered nuclear export of non-fully processed mRNAs, HIV encodes the Rev protein, which recognizes its intronic response element and fuels the HIV RNAs into the CRM-1-dependent nuclear protein export pathway. Both alternative splicing and Rev-dependency are regulated by the primary HIV RNA sequence. Here, we show that these processes are extremely sensitive to sequence alterations in the 5'coding region of the HIV genomic RNA. Increasing the GC content by insertion of either GFP or silent mutations activates a cryptic splice donor site in gag, entirely deregulates the viral splicing pattern, and lowers infectivity. Interestingly, an adaptation of the inserted GFP sequence toward an HIV-like nucleotide bias reversed these phenotypes completely. Of note, the adaptation yielded completely different primary sequences although encoding the same amino acids. Thus, the phenotypes solely depend on the nucleotide composition of the two GFP versions. This is a strong indication of an HIV-specific mRNP code in the 5' gag region wherein the primary RNA sequence bias creates motifs for RNA-binding proteins and controls the fate of the HIV-RNA in terms of viral gene expression and infectivity.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Épissage des ARN , Réplication virale/génétique , Produits du gène gag du virus de l'immunodéficience humaine/génétique , Épissage alternatif , Cellules HEK293 , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/pathogénicité , Humains , Sites d'épissage d'ARN , ARN messager , ARN viral/génétique
8.
Wiley Interdiscip Rev RNA ; 12(1): e1616, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32633083

RÉSUMÉ

Noncoding sequences constitute the major part of the human genome and also of pre-mRNAs. Single nucleotide variants in these regions are often overlooked, but may be responsible for much of the variation of phenotypes observed. Mutations in the noncoding part of pre-mRNAs often reveal new and meaningful insights into the regulation of cellular gene expression. Thus, the mechanistic analysis of the pathological mechanism of such mutations will both foster a deeper understanding of the disease and the underlying cellular pathways. Even synonymous mutations can cause diseases, since the primary mRNA sequence not only encodes amino acids, but also encrypts information on RNA-binding proteins and secondary structure. In fact, the RNA sequence directs assembly of a specific mRNP complex, which in turn dictates the fate of the mRNA or regulates its biogenesis. The accumulation of genomic sequence information is increasing at a rapid pace. However, much of the diversity uncovered may not explain the phenotype of a certain syndrome or disease. For this reason, we also emphasize the value of mechanistic studies on pathological mechanisms being complementary to genome-wide studies and bioinformatic approaches. This article is categorized under: RNA Processing > Splicing Regulation/Alternative Splicing RNA Processing > 3' End Processing RNA in Disease and Development > RNA in Disease.


Sujet(s)
Régulation de l'expression des gènes , Mutation , Épissage des ARN , ARN non traduit/génétique , Épissage alternatif , Humains
9.
PLoS Genet ; 16(4): e1008690, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32267853

RÉSUMÉ

Loss-of-function mutations in the human coagulation factor 9 (F9) gene lead to hemophilia B. Here, we dissected the consequences and the pathomechanism of a non-coding mutation (c.2545A>G) in the F9 3' untranslated region. Using wild type and mutant factor IX (FIX) minigenes we revealed that the mutation leads to reduced F9 mRNA and FIX protein levels and to lower coagulation activity of cell culture supernatants. The phenotype could not be compensated by increased transcription. The pathomechanism comprises the de novo creation of a binding site for the spliceosomal component U1snRNP, which is able to suppress the nearby F9 poly(A) site. This second, splicing-independent function of U1snRNP was discovered previously and blockade of U1snRNP restored mutant F9 mRNA expression. In addition, we explored the vice versa approach and masked the mutation by antisense oligonucleotides resulting in significantly increased F9 mRNA expression and coagulation activity. This treatment may transform the moderate/severe hemophilia B into a mild or subclinical form in the patients. This antisense based strategy is applicable to other mutations in untranslated regions creating deleterious binding sites for cellular proteins.


Sujet(s)
Facteur IX/génétique , Hémophilie B/génétique , Mutation perte de fonction , ARN messager/génétique , Suppression génétique , Régions 3' non traduites , Animaux , Cellules CHO , Cricetinae , Cricetulus , Facteur IX/métabolisme , Cellules HEK293 , Cellules HeLa , Humains , Oligonucléotides antisens/génétique , Phénotype , ARN messager/métabolisme , Petit ARN nucléaire/génétique
10.
iScience ; 23(1): 100764, 2020 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-31887661

RÉSUMÉ

Adeno-associated virus (AAV)-based vectors are considered efficient and safe gene delivery systems in gene therapy. We combined two guide RNA genes, Cas9, and a self-linearizing repair template in one vector (AIO-SL) to correct fumarylacetoacetate hydrolase (FAH) deficiency in mice. The vector genome of 5.73 kb was packaged into VP2-depleted AAV particles (AAV2/8ΔVP2), which, however, did not improve cargo capacity. Reprogrammed hepatocytes were treated with AIO-SL.AAV2ΔVP2 and subsequently transplanted, resulting in large clusters of FAH-positive hepatocytes. Direct injection of AIO-SL.AAV8ΔVP2 likewise led to FAH expression and long-term survival. The AIO-SL vector achieved an ∼6-fold higher degree of template integration than vectors without template self-linearization. Subsequent analysis revealed that AAV8 particles, in contrast to AAV2, incorporate oversized genomes distinctly greater than 5.2 kb. Finally, our AAV8-based vector represents a promising tool for gene editing strategies to correct monogenic liver diseases requiring (large) fragment removal and/or simultaneous sequence replacement.

11.
J Virol ; 93(24)2019 12 15.
Article de Anglais | MEDLINE | ID: mdl-31597782

RÉSUMÉ

When expressed in virus-producing cells, the cellular multipass transmembrane protein SERINC5 reduces the infectivity of HIV-1 particles and is counteracted by HIV-1 Nef. Due to the unavailability of an antibody of sufficient specificity and sensitivity, investigation of SERINC5 protein expression and subcellular localization has been limited to heterologously expressed SERINC5. We generated, via CRISPR/Cas9-assisted gene editing, Jurkat T-cell clones expressing endogenous SERINC5 bearing an extracellularly exposed hemagglutinin (HA) epitope [Jurkat SERINC5(iHA knock-in) T cells]. This modification enabled quantification of endogenous SERINC5 protein levels and demonstrated a predominant localization in lipid rafts. Interferon alpha (IFN-α) treatment enhanced cell surface levels of SERINC5 in a ruxolitinib-sensitive manner in the absence of modulation of mRNA and protein quantities. Parental and SERINC5(iHA knock-in) T cells shared the ability to produce infectious wild-type HIV-1 but not an HIV-1 Δnef mutant. SERINC5-imposed reduction of infectivity involved a modest reduction of virus fusogenicity. An association of endogenous SERINC5 protein with HIV-1 Δnef virions was consistently detectable as a 35-kDa species, as opposed to heterologous SERINC5, which presented as a 51-kDa species. Nef-mediated functional counteraction did not correlate with virion exclusion of SERINC5, arguing for the existence of additional counteractive mechanisms of Nef that act on virus-associated SERINC5. In HIV-1-infected cells, Nef triggered the internalization of SERINC5 in the absence of detectable changes of steady-state protein levels. These findings establish new properties of endogenous SERINC5 expression and subcellular localization, challenge existing concepts of HIV-1 Nef-mediated antagonism of SERINC5, and uncover an unprecedented role of IFN-α in modulating SERINC5 through accumulation at the cell surface.IMPORTANCE SERINC5 is the long-searched-for antiviral factor that is counteracted by the HIV-1 accessory gene product Nef. Here, we engineered, via CRISPR/Cas9 technology, T-cell lines that express endogenous SERINC5 alleles tagged with a knocked-in HA epitope. This genetic modification enabled us to study basic properties of endogenous SERINC5 and to verify proposed mechanisms of HIV-1 Nef-mediated counteraction of SERINC5. Using this unique resource, we identified the susceptibility of endogenous SERINC5 protein to posttranslational modulation by type I IFNs and suggest uncoupling of Nef-mediated functional antagonism from SERINC5 exclusion from virions.


Sujet(s)
Agents antiVIH/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/pharmacologie , Systèmes CRISPR-Cas , Lignée cellulaire , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Édition de gène , Régulation de l'expression des gènes , Techniques de knock-out de gènes , Génotype , Cellules HEK293 , Infections à VIH/virologie , Interactions hôte-pathogène/physiologie , Humains , Interféron alpha , Protéines membranaires/génétique , Nitriles , Pyrazoles/pharmacologie , Pyrimidines , Lymphocytes T/virologie , Virion/métabolisme , Produits du gène nef du virus de l'immunodéficience humaine/génétique , Produits du gène nef du virus de l'immunodéficience humaine/métabolisme
12.
J Clin Immunol ; 39(2): 207-215, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30903457

RÉSUMÉ

Inborn errors in interleukin 2 receptor, gamma (IL2RG) perturb signaling of the common gamma chain family cytokines and cause severe combined immunodeficiency (SCID). Here, we report two brothers suffering from chronic cryptosporidiosis, severe diarrhea, and cholangitis. Pan T, B, and NK cell numbers were normal, but immunophenotyping revealed defective B cell differentiation. Using whole exome sequencing, we identified a base pair deletion in the first exon of IL2RG predicted to cause a frameshift and premature stop. However, flow cytometry revealed normal surface expression of the IL-2Rγ chain. While IL-2, IL-7, and IL-15 signaling showed only mild defects of STAT5 phosphorylation in response to the respective cytokines, IL-4- and IL-21-induced phosphorylation of STAT3 and STAT6 was markedly reduced. Examination of RNA isoforms detected alternative splicing downstream of IL2RG exon 1 in both patients resulting in resolution of the predicted frameshift and 16 mutated amino acids. In silico modeling suggested that the IL-2Rγ mutation reduces the stabilization of IL-4 and IL-21 cytokine binding by affecting the N-terminal domain of the IL-2Rγ. Thus, our study shows that IL2RG deficiency can be associated with differential signaling defects. Confounding effects of alternative splicing may partially rescue genetic defects and should be considered in patients with inborn errors of immunity.


Sujet(s)
Sous-unité alpha du récepteur à l'interleukine 21/génétique , Immunodéficience combinée grave/génétique , Épissage alternatif , Lymphocytes B/immunologie , Enfant d'âge préscolaire , Angiocholite/génétique , Angiocholite/immunologie , Croatie , Cryptosporidiose/génétique , Cryptosporidiose/immunologie , Diarrhée/génétique , Diarrhée/immunologie , Humains , Sous-unité alpha du récepteur à l'interleukine 21/déficit , Sous-unité alpha du récepteur à l'interleukine 21/immunologie , Mâle , Infections de l'appareil respiratoire/génétique , Infections de l'appareil respiratoire/immunologie , Immunodéficience combinée grave/immunologie
13.
Eur J Hum Genet ; 27(6): 879-887, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30723317

RÉSUMÉ

Wilson disease (WD) is an autosomal recessive disease of copper excess due to pathogenic variants in the ATP7B gene coding for a copper-transporting ATPase. We present a 5-year-old girl with the homozygous frame shift variant NM_000053.3: c.19_20del in exon 1 of ATP7B (consecutive exon numbering with c.1 as first nucleotide of exon 1), detected by whole-exome sequencing as a secondary finding. The variant leads to a premature termination codon in exon 2. The girl exhibited no WD symptoms and no abnormalities in liver biopsy. ATP7B liver mRNA expression was comparable to healthy controls suggesting that nonsense-mediated mRNA decay (NMD) could be bypassed by the mechanism of translation reinitiation. To verify this hypothesis, a CMV-driven ATP7B minigene (pcDNA3) was equipped with the authentic ATP7B 5' untranslated region  and a truncated intron 2. We introduced c.19_20del by site-directed mutagenesis and overexpressed the constructs in HEK293T cells. We analyzed ATP7B expression by qRT-PCR, northern and western blot, and examined protein function by copper export capacity assays. Northern blot, qRT-PCR, and western blot revealed that c.19_20del ATP7B mRNA and protein is expressed in size and amount comparable to wild-type. Copper export capacity was also comparable to wild-type. Our results indicate that c.19_20del in ATP7B is able to bypass NMD by translation reinitiation, demonstrating that the classification of truncating variants as pathogenic without additional investigations should be done carefully.


Sujet(s)
Copper-transporting ATPases , Cuivre/métabolisme , Exons , Mutation avec décalage du cadre de lecture , Dégénérescence hépatolenticulaire , Homozygote , Dégradation des ARNm non-sens , Enfant d'âge préscolaire , Copper-transporting ATPases/génétique , Copper-transporting ATPases/métabolisme , Femelle , Cellules HEK293 , Dégénérescence hépatolenticulaire/génétique , Dégénérescence hépatolenticulaire/métabolisme , Humains , Transport des ions/génétique
14.
Nucleic Acids Res ; 46(22): 11968-11979, 2018 12 14.
Article de Anglais | MEDLINE | ID: mdl-30321376

RÉSUMÉ

A number of viruses remodel the cellular gene expression landscape by globally accelerating messenger RNA (mRNA) degradation. Unlike the mammalian basal mRNA decay enzymes, which largely target mRNA from the 5' and 3' end, viruses instead use endonucleases that cleave their targets internally. This is hypothesized to more rapidly inactivate mRNA while maintaining selective power, potentially though the use of a targeting motif(s). Yet, how mRNA endonuclease specificity is achieved in mammalian cells remains largely unresolved. Here, we reveal key features underlying the biochemical mechanism of target recognition and cleavage by the SOX endonuclease encoded by Kaposi's sarcoma-associated herpesvirus (KSHV). Using purified KSHV SOX protein, we reconstituted the cleavage reaction in vitro and reveal that SOX displays robust, sequence-specific RNA binding to residues proximal to the cleavage site, which must be presented in a particular structural context. The strength of SOX binding dictates cleavage efficiency, providing an explanation for the breadth of mRNA susceptibility observed in cells. Importantly, we establish that cleavage site specificity does not require additional cellular cofactors, as had been previously proposed. Thus, viral endonucleases may use a combination of RNA sequence and structure to capture a broad set of mRNA targets while still preserving selectivity.


Sujet(s)
Endonucleases/génétique , Herpèsvirus humain de type 8/enzymologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines à domaine LIM/métabolisme , Protéines virales/génétique , Animaux , Sites de fixation , Endonucleases/métabolisme , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/génétique , Humains , Insectes , Cinétique , Mutation , Clivage de l'ARN , Stabilité de l'ARN/génétique , ARN messager/métabolisme , Spécificité du substrat , Protéines virales/métabolisme
15.
Virus Genes ; 53(5): 714-722, 2017 Oct.
Article de Anglais | MEDLINE | ID: mdl-28762206

RÉSUMÉ

After 30 years of retroviral vector research it became clear that the parental viruses can be both friend and foe. Especially human immunodeficiency virus sparked a global pandemic, but could be converted into a versatile tool for cell therapy. For all retroviral genera, the way from virus to vector was similar resulting in split-vector systems based on the separation of the genes needed for vector particle formation and transgene expression. The first gene therapy trials, although clinically effective, revealed the genotoxicity of retroviral vectors caused by insertional mutagenesis. This issue was solved using self-inactivating vectors carrying weaker cellular promoters. Further fine-tuning was able to generate inducible systems. The current toolbox also contains vectors for the generation of induced pluripotent stem cells or efficient RNA interference. More recently the application of CRISPR-Cas9-mediated gene editing led to the development of genome-wide small guide RNA libraries targeting all human genes and single lentiviral vectors for an easy delivery of Cas9.


Sujet(s)
Vecteurs génétiques/génétique , Retroviridae/génétique , Animaux , Thérapie génétique/méthodes , Humains , Lentivirus/génétique , Régions promotrices (génétique)/génétique , Interférence par ARN/physiologie , Transgènes/génétique
16.
Wiley Interdiscip Rev RNA ; 7(2): 173-85, 2016.
Article de Anglais | MEDLINE | ID: mdl-26769399

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes ORF57, which enhances the expression of intron-less KSHV genes on multiple post-transcriptional levels mainly affecting RNA stability and export to the cytoplasm. Yet, it remains elusive how ORF57 recognizes viral RNAs and discriminates them from cellular messenger RNAs (mRNAs). Although one common binding motif on three separate KSHV RNAs has been described, most other lytic genes lack this sequence element. In this article we will review the sequence requirements for ORF57 to enhance RNA expression and discuss a model how ORF57 achieves specificity for viral RNAs. Finally, the role of ORF57 is integrated into the viral life cycle as a complex interplay with other viral and host factors and with implications for cellular gene expression.


Sujet(s)
Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/physiologie , ARN viral/génétique , ARN viral/métabolisme , Protéines virales régulatrices ou accessoires/métabolisme , Humains , Liaison aux protéines , Maturation post-transcriptionnelle des ARN , ARN viral/composition chimique , Protéines de liaison à l'ARN/métabolisme , Éléments de réponse , Protéines virales régulatrices ou accessoires/composition chimique , Réplication virale
17.
Biochim Biophys Acta ; 1849(9): 1145-54, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26170143

RÉSUMÉ

The heteromeric transcription factor GA-binding protein (GABP) consists of two subunits, the alpha subunit (GABPA) carrying the DNA-binding ETS domain, and the beta subunit (GABPB1) harbouring the transcriptional activation domain. GABP is involved in haematopoietic stem cell maintenance and differentiation of myeloid and lymphoid lineages in mice. To elucidate the molecular function of GABP in human haematopoiesis, the present study addressed effects of ectopic overexpression of GABP focussing on the myeloid compartment. Combined overexpression of GABPA and GABPB1 caused a proliferation block in cell lines and drastically reduced the colony-forming capacity of murine lineage-negative cells. Impaired proliferation resulted from perturbed cellular cycling and induction of myeloid differentiation shown by surface markers and myelomonocytic morphology of U937 cells. Depending on the dosage and functional integrity of GABP, ITGAM expression was induced. ITGAM encodes CD11b, the alpha subunit of integrin Mac-1, whose beta subunit, ITGB2/CD18, was already described to be regulated by GABP. Finally, Shield1-dependent proteotuning, luciferase reporter assays and chromatin immunoprecipitation showed that GABP activates the ITGAM/CD11b promoter via three binding sites close to the translational start site. In conclusion, the present study supports the crucial role of GABP in myeloid cell differentiation and identified ITGAM/CD11b as a novel GABP target gene.


Sujet(s)
Antigènes CD11b/génétique , Différenciation cellulaire/physiologie , Facteur de transcription GABP/physiologie , Cellules myéloïdes/cytologie , Régions promotrices (génétique) , Animaux , Lignée cellulaire , Facteur de transcription GABP/génétique , Dosage génique , Humains , Souris
18.
J Virol ; 89(9): 5097-109, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25694606

RÉSUMÉ

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) encodes ORF57, which enhances the expression of intronless KSHV genes on multiple posttranscriptional levels. However, it remains elusive how ORF57 recognizes viral RNAs. Here, we demonstrate that ORF57 also increases the expression of the multiple intron-containing K15 gene. The nucleotide bias of the K15 cDNA revealed an unusual high AT content. Thus, we optimized the K15 cDNA by raising the frequency of GC nucleotides, yielding an ORF57-independent version. To further prove the importance of the sequence bias of ORF57-dependent RNAs, we grouped KSHV mRNAs according to their AT content and found a correlation between AT-richness and ORF57 dependency. More importantly, latent genes, which have to be expressed in the absence of ORF57, have a low AT content and are indeed ORF57 independent. The nucleotide composition of K15 resembles that of HIV gag, which cannot be expressed unless RNA export is facilitated by the HIV Rev protein. Interestingly, ORF57 can partially rescue HIV Gag expression. Thus, the KSHV target RNAs of ORF57 and HIV gag RNA may share certain motifs based on the nucleotide bias. A bioinformatic comparison between wild-type and sequence-optimized K15 revealed a higher density for hnRNP-binding motifs in the former. We speculate that binding of particular hnRNPs to KSHV lytic transcripts is the prerequisite for ORF57 to enhance their expression. IMPORTANCE: The mostly intronless genes of KSHV are only expressed in the presence of the viral regulator protein ORF57, but how ORF57 recognizes viral RNAs remains elusive. We focused on the multiple intron-containing KSHV gene K15 and revealed that its expression is also increased by ORF57. Moreover, sequences in the K15 cDNA mediate this enhancement. The quest for a target sequence or a response element for ORF57 in the lytic genes was not successful. Instead, we found the nucleotide bias to be the critical determinant of ORF57 dependency. Based on the fact that ORF57 has only a weak affinity for nucleic acids, we speculate that a cellular RNA-binding protein provides the sequence preference for ORF57. This study provides evidence that herpesviral RNA regulator proteins use the sequence bias of lytic genes and the resulting composition of the viral mRNP to distinguish between viral and cellular mRNAs.


Sujet(s)
Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/physiologie , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Biosynthèse des protéines , Protéines virales régulatrices ou accessoires/métabolisme , Réplication virale , Humains , Liaison aux protéines , ARN messager/métabolisme , ARN viral/métabolisme
19.
J Clin Immunol ; 34(3): 331-9, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24519095

RÉSUMÉ

PURPOSE: Loss-of-function mutations in IL10 and IL10R cause very early onset inflammatory bowel disease (VEO-IBD). Here, we investigated the molecular pathomechanism of a novel intronic IL10RA mutation and describe a new therapeutic approach of T cell replete haploidentical hematopoietic stem cell transplantation (HSCT). METHODS: Clinical data were collected by chart review. Genotypes of IL10 and IL10R genes were determined by Sanger sequencing. Expression and function of mutated IL-10R1 were assessed by quantitative PCR, Western blot analysis, enzyme-linked immunosorbent assays, confocal microscopy, and flow cytometry. RESULTS: We identified a novel homozygous point mutation in intron 3 of the IL10RA (c.368-10C > G) in three related children with VEO-IBD. Bioinformatical analysis predicted an additional 3' splice site created by the mutation. Quantitative PCR analysis showed normal mRNA expression of mutated IL10RA. Sequencing of the patient's cDNA revealed an insertion of the last nine nucleotides of intron 3 as a result of aberrant splicing. Structure-based modeling suggested misfolding of mutated IL-10R1. Western blot analysis demonstrated a different N-linked glycosylation pattern of mutated protein. Immunofluorescence and FACS analysis revealed impaired expression of mutated IL-10R1 at the plasma membrane. In the absence of HLA-identical donors, T cell replete haploidentical HSCT was successfully performed in two patients. CONCLUSIONS: Our findings expand the spectrum of IL10R mutations in VEO-IBD and emphasize the need for genetic diagnosis of mutations in conserved non-coding sequences of candidate genes. Transplantation of haploidentical stem cells represents a curative therapy in IL-10R-deficient patients, but may be complicated by non-engraftment.


Sujet(s)
Transplantation de moelle osseuse , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/thérapie , Sous-unité alpha du récepteur de l'interleukine-10/métabolisme , Âge de début , Épissage alternatif , Séquence d'acides aminés , Lignée cellulaire , Membrane cellulaire/métabolisme , Enfant , Enfant d'âge préscolaire , Consanguinité , Analyse de mutations d'ADN , Femelle , Génotype , Glycosylation , Transplantation de cellules souches hématopoïétiques , Humains , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/immunologie , Sous-unité alpha du récepteur de l'interleukine-10/composition chimique , Sous-unité alpha du récepteur de l'interleukine-10/génétique , Introns , Mâle , Modèles moléculaires , Données de séquences moléculaires , Mutation , Pedigree , Phénotype , Conformation des protéines , Transport des protéines , Alignement de séquences , Transduction du signal , Lymphocytes T/immunologie , Résultat thérapeutique
20.
Retrovirology ; 10: 55, 2013 May 29.
Article de Anglais | MEDLINE | ID: mdl-23718736

RÉSUMÉ

BACKGROUND: During reverse transcription, retroviruses duplicate the long terminal repeats (LTRs). These identical LTRs carry both promoter regions and functional polyadenylation sites. To express full-length transcripts, retroviruses have to suppress polyadenylation in the 5'LTR and activate polyadenylation in the 3'LTR. Foamy viruses have a unique LTR structure with respect to the location of the major splice donor (MSD), which is located upstream of the polyadenylation signal. RESULTS: Here, we describe the mechanisms of foamy viruses regulating polyadenylation. We show that binding of the U1 small nuclear ribonucleoprotein (U1snRNP) to the MSD suppresses polyadenylation at the 5'LTR. In contrast, polyadenylation at the 3'LTR is achieved by adoption of a different RNA structure at the MSD region, which blocks U1snRNP binding and furthers RNA cleavage and subsequent polyadenylation. CONCLUSION: Recently, it was shown that U1snRNP is able to suppress the usage of intronic cryptic polyadenylation sites in the cellular genome. Foamy viruses take advantage of this surveillance mechanism to suppress premature polyadenylation at the 5'end of their RNA. At the 3'end, Foamy viruses use a secondary structure to presumably block access of U1snRNP and thereby activate polyadenylation at the end of the genome. Our data reveal a contribution of U1snRNP to cellular polyadenylation site selection and to the regulation of gene expression.


Sujet(s)
Poly A/métabolisme , Petit ARN nucléaire/métabolisme , ARN viral/composition chimique , ARN viral/métabolisme , Spumavirus/physiologie , Animaux , Lignée cellulaire , Cricetinae , Conformation d'acide nucléique , Polyadénylation , Signaux de polyadénylation , Sites d'épissage d'ARN , Séquences répétées terminales
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE