Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 518(7539): 409-12, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25470045

ABSTRACT

Angelman syndrome is a single-gene disorder characterized by intellectual disability, developmental delay, behavioural uniqueness, speech impairment, seizures and ataxia. It is caused by maternal deficiency of the imprinted gene UBE3A, encoding an E3 ubiquitin ligase. All patients carry at least one copy of paternal UBE3A, which is intact but silenced by a nuclear-localized long non-coding RNA, UBE3A antisense transcript (UBE3A-ATS). Murine Ube3a-ATS reduction by either transcription termination or topoisomerase I inhibition has been shown to increase paternal Ube3a expression. Despite a clear understanding of the disease-causing event in Angelman syndrome and the potential to harness the intact paternal allele to correct the disease, no gene-specific treatment exists for patients. Here we developed a potential therapeutic intervention for Angelman syndrome by reducing Ube3a-ATS with antisense oligonucleotides (ASOs). ASO treatment achieved specific reduction of Ube3a-ATS and sustained unsilencing of paternal Ube3a in neurons in vitro and in vivo. Partial restoration of UBE3A protein in an Angelman syndrome mouse model ameliorated some cognitive deficits associated with the disease. Although additional studies of phenotypic correction are needed, we have developed a sequence-specific and clinically feasible method to activate expression of the paternal Ube3a allele.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/therapy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , Alleles , Angelman Syndrome/complications , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Fathers , Female , Gene Silencing/drug effects , Genomic Imprinting/genetics , Male , Memory Disorders/complications , Memory Disorders/genetics , Memory Disorders/therapy , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Obesity/complications , Obesity/genetics , Obesity/therapy , Oligonucleotides, Antisense/pharmacology , Phenotype , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/deficiency , RNA, Antisense/genetics , Time Factors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Gen Physiol Biophys ; 39(4): 355-362, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32902404

ABSTRACT

Extensive scarring normally causes hypertrophic or keloid scars. This intuitively results in psychosocial distress and reduction in life quality. Tripterine is a bioactive pentacyclic triterpenoid compound while it is still poorly understood whether it possesses an anti-scarring function. NIH/3T3 cells were administrated with tripterine at increased concentrations (0-10 µM). Antisense RNA to INK4 locus (ANRIL) was transformed into NIH/3T3 cells, and the cells transfected with empty vector (mock transfection) were used as negative control. Then, cell viability and migration were profiled by cell counting kit-8 (CCK-8) and 24-Transwell assay. Protein expression was analyzed by Western blot assay. ANRIL was quantified by quantitative reverse transcription-PCR (qRT-PCR). Tripterine administration induced the growth inhibition of NIH/3T3 cells indicated by a trend toward the decreased expression of matrix metallopeptidase (MMPs), vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). This process was accompanied by the decreased phosphorylation of p65, inhibitor of nuclear factor kappa-B alpha (IκBα) and the downregulation of ß-catenin. Moreover, ANRIL expression was notably repressed by tripterine. By contrast, in ANRIL-transfected cells, the effect of tripterine was abolished. Tripterine exhibited an anti-scarring bioactivity in NIH/3T3 cells by inhibiting ANRIL, and this process was accompanied by the blockade of nuclear factor-kappa B (NF-κB) and ß-catenin cascades.


Subject(s)
Cicatrix , RNA, Antisense/antagonists & inhibitors , Signal Transduction , Triterpenes/pharmacology , 3T3 Cells , Animals , Mice , Pentacyclic Triterpenes , Vascular Endothelial Growth Factor A
3.
J Cell Physiol ; 234(10): 17494-17504, 2019 08.
Article in English | MEDLINE | ID: mdl-30912129

ABSTRACT

Growing evidence has shown that long noncoding RNAs (lncRNAs) play crucial roles in cervical cancer. Dy000sregulation of lncRNA SOX21 antisense RNA 1 (SOX21-AS1) has been reported in several tumors. However, its expression pattern and potential biological function in cervical cancer (CC) have not been investigated. In this study, we first reported that SOX21-AS1 expression was significantly upregulated in both CC tissues and cell lines. High expression of SOX21-AS1 was found to be significantly correlated with Federation of Gynecology and Obstetrics (FIGO) stage, lymph node metastasis and depth of cervical invasion. Further clinical assay confirmed that high SOX21-AS1 expression was associated with shorter overall survival and could be used as a potential prognostic biomarker for CC patients. Functional investigation showed that knockdown of SOX21-AS1 suppressed CC cells proliferation, migration, and invasion, as well as epithelial to mesenchymal transition progress. Furthermore, our data showed that microRNA-7 (miR-7) interacted with SOX21-AS1 by directly targeting the miRNA-binding site in the SOX21-AS1 sequence, and quantitative real-time polymerase chain reaction results showed overexpression of SOX21-AS1 decreased the levels of miR-7 in CC cells. Moreover, we confirmed that miR-7 directly targeted the 3'-untranslated region of voltage dependent anion channel 1 (VDAC1). Final in vitro assay suggested that in CC cells with SOX21-AS1, VDAC1 overexpression resulted in an increase of cell proliferation, migration, and invasion. Overall, our findings illuminate how SOX21-AS1 formed a regulatory network to confer an oncogenic function in CC and SOX21-AS1 could be regarded as an efficient therapeutic target and potential biomarker for CC patients.


Subject(s)
MicroRNAs/genetics , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , SOXB2 Transcription Factors/genetics , Uterine Cervical Neoplasms/genetics , Voltage-Dependent Anion Channel 1/genetics , 3' Untranslated Regions , Adult , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Gene Knockdown Techniques , HeLa Cells , Humans , MicroRNAs/metabolism , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , SOXB2 Transcription Factors/antagonists & inhibitors , Up-Regulation , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
4.
Gynecol Obstet Invest ; 84(2): 136-144, 2019.
Article in English | MEDLINE | ID: mdl-30253398

ABSTRACT

AIM: To investigate whether long non-coding RNA (lncRNA) ZEB1 antisense 1 (ZEB1-AS1) affects cell migration and invasion of cervical cancer by regulating epithelial-mesenchymal transition (EMT) via the p38MAPK pathway. METHODS: Human cervical cancer cell line Hela was classified into Control, NC siRNA, ZEB1-AS1 siRNA, SB203580 (p38MAPK pathway inhibitor) and ZEB1-AS1 siRNA + Anisomycin (p38MAPK pathway activator) groups. Quantitative real-time polymerase chain reaction was performed for ZEB1-AS1 expression, Western blotting to measure p38MAPK signaling pathway-/EMT-related proteins, and Wound-healing and Transwell assays to evaluate cell migration and invasion respectively. RESULTS: ZEB1-AS1 was upregulated in cancer tissues and related to major clinicopathological features of cervical cancer. Besides, patients with lower-ZEB1-AS1-expression had a higher 5-year survival rate than those patients with higher-ZEB1-AS1-expression. High ZEB1-AS1 expression and advanced Federation of Gynecology and Obstetrics stage were independent risk factors for patients' prognosis. Both ZEB1-AS1 siRNA and SB203580 effectively reduced p-p38 expression and the migration and invasion of Hela cells, with elevation of E-cadherin and reduction of Vimentin and N-cadherin. However, inhibitory effects of ZEB1-AS1 siRNA on EMT as well as cell migration and invasion of the Hela cell were reversed by Anisomycin. CONCLUSION: Inhibition of ZEB1-AS1 can block the p38MAPK signaling pathway, ultimately restricting the EMT and suppressing cell migration and invasion of cervical cancer cells.


Subject(s)
Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , MAP Kinase Signaling System/physiology , RNA, Long Noncoding/physiology , Uterine Cervical Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/genetics , Antigens, CD/analysis , Cadherins/analysis , Female , Gene Expression , HeLa Cells , Humans , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/genetics , RNA, Antisense/physiology , RNA, Small Interfering/physiology , Transfection , Vimentin/analysis , Zinc Finger E-box-Binding Homeobox 1/antagonists & inhibitors
5.
Nature ; 461(7262): 419-22, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19693008

ABSTRACT

Eukaryotic transcriptomes are characterized by widespread transcription of noncoding and antisense RNAs, which is linked to key chromosomal processes, such as chromatin remodelling, gene regulation and heterochromatin assembly. However, these transcripts can be deleterious, and their accumulation is suppressed by several mechanisms including degradation by the nuclear exosome. The mechanisms by which cells differentiate coding RNAs from transcripts targeted for degradation are not clear. Here we show that the variant histone H2A.Z, which is loaded preferentially at the 5' ends of genes by the Swr1 complex containing a JmjC domain protein, mediates suppression of antisense transcripts in the fission yeast Schizosaccharomyces pombe genome. H2A.Z is partially redundant in this regard with the Clr4 (known as SUV39H in mammals)-containing heterochromatin silencing complex that is also distributed at euchromatic loci, and with RNA interference component Argonaute (Ago1). Loss of Clr4 or Ago1 alone has little effect on antisense transcript levels, but cells lacking either of these factors and H2A.Z show markedly increased levels of antisense RNAs that are normally degraded by the exosome. These analyses suggest that as well as performing other functions, H2A.Z is a component of a genome indexing mechanism that cooperates with heterochromatin and RNAi factors to suppress read-through antisense transcripts.


Subject(s)
Gene Expression Regulation, Fungal , Heterochromatin/metabolism , Histones/metabolism , RNA Interference , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/genetics , Schizosaccharomyces/genetics , Argonaute Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Exosomes/metabolism , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase , Histones/deficiency , Histones/genetics , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , RNA, Antisense/biosynthesis , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
6.
Nucleic Acids Res ; 34(20): 5915-22, 2006.
Article in English | MEDLINE | ID: mdl-17065468

ABSTRACT

Short regulatory RNAs are widespread in bacteria, and many function through antisense recognition of mRNA. Among the best studied antisense transcripts are RNA antitoxins that repress toxin mRNA translation. The hok/sok locus of plasmid R1 from Escherichia coli is an established model for RNA antitoxin action. Base-pairing between hok mRNA and Sok-antisense-RNA increases plasmid maintenance through post-segregational-killing of plasmid-free progeny cells. To test the model and the idea that sequestration of Sok-RNA activity could provide a novel antimicrobial strategy, we designed anti Sok peptide nucleic acid (PNA) oligomers that, according to the model, would act as competitive inhibitors of hok mRNA::Sok-RNA interactions. In hok/sok-carrying cells, anti Sok PNAs were more bactericidal than rifampicin. Also, anti Sok PNAs induced ghost cell morphology and an accumulation of mature hok mRNA, consistent with cell killing through synthesis of Hok protein. The results support the sense/antisense model for hok mRNA repression by Sok-RNA and demonstrate that antisense agents can be used to out-compete RNA::RNA interactions in bacteria. Finally, BLAST analyses of approximately 200 prokaryotic genomes revealed that many enteric bacteria have multiple hok/sok homologous and analogous RNA-regulated toxin-antitoxin loci. Therefore, it is possible to activate suicide in bacteria by targeting antitoxins.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Peptide Nucleic Acids/pharmacology , RNA, Antisense/antagonists & inhibitors , RNA/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Toxins/metabolism , Binding, Competitive , Enterobacteriaceae/genetics , Escherichia coli/cytology , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Kinetics , Peptide Nucleic Acids/chemistry , RNA/genetics , RNA, Antisense/physiology , RNA, Bacterial , RNA, Messenger/metabolism
7.
Pigment Cell Melanoma Res ; 31(1): 64-72, 2018 01.
Article in English | MEDLINE | ID: mdl-28707763

ABSTRACT

The family of non-coding mitochondrial RNAs (ncmtRNA) is differentially expressed according to proliferative status. Normal proliferating cells express sense (SncmtRNA) and antisense ncmtRNAs (ASncmtRNAs), whereas tumor cells express SncmtRNA and downregulate ASncmtRNAs. Knockdown of ASncmtRNAs with oligonucleotides induces apoptotic cell death of tumor cells, leaving normal cells unaffected, suggesting a potential application for developing a novel cancer therapy. In this study, we knocked down the ASncmtRNAs in melanoma cell lines with a lentiviral-encoded shRNA approach. Transduction with lentiviral constructs targeted to the ASncmtRNAs induced apoptosis in murine B16F10 and human A375 melanoma cells in vitro and significantly retarded B16F10 primary tumor growth in vivo. Moreover, the treatment drastically reduced the number of lung metastatic foci in a tail vein injection assay, compared to controls. These results provide additional proof of concept to the knockdown of ncmtRNAs for cancer therapy and validate lentiviral-shRNA vectors for gene therapy.


Subject(s)
Lentivirus/genetics , Lung Neoplasms/therapy , Melanoma/therapy , RNA, Antisense/antagonists & inhibitors , RNA, Mitochondrial/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Untranslated/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , RNA, Antisense/genetics , RNA, Mitochondrial/genetics , RNA, Untranslated/genetics
8.
Biomed Pharmacother ; 96: 1103-1108, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29239821

ABSTRACT

Long non-coding RNAs (lncRNAs) have been demonstrated that it plays very important role in development and progression of carcinomas. LncRNA FEZF1 antisense RNA 1 (FEZF1-AS1) has been proved to be implicated in tumor initiation and progression of various cancers, recently. Nevertheless, the biological function and clinical significance of lncRNA FEZF1-AS1 in gastric cancer (GC) are not clear enough. Here, we concentrated on the association of FEZF1-AS1 expression and clinicopathological factors in GC tissues and cells. Moreover, we explored the potential regulatory mechanisms. The results showed that lncRNA FEZF1-AS1 was observably upregulated in human GC tissues and GC cell lines, compared with the adjacent non-tumor tissues and human gastric epithelial cell line (GES-1). Moreover, high expression of lncRNA FEZF1-AS1 was significantly associated with later stage and higher grade. Furthermore, Kaplan-Meier survival analysis was conducted, indicating that lncRNA FEZF1-AS1 may be an independent prognostic factor in GC. Additionally, the area under the receiver operating characteristic (ROC) curve of lncRNA FEZF1-AS1 exhibited its diagnostic value in GC. Notably, whenever the lncRNA FEZF1-AS1 was silenced, the proliferation of GC cells were significantly inhibited and the cell cycle was arrested at a G0/G1 stage in GC cells. Furthermore, downregulation of lncRNA FEZF1-AS1 could suppress the activation of the Wnt/ß-catenin signaling pathway. Conclusively, our findings indicated that lncRNA FEZF1-AS1 could be considered as a novel biomarker for the treatment of GC.


Subject(s)
Carcinogenesis/metabolism , RNA, Antisense/biosynthesis , RNA, Long Noncoding/biosynthesis , Stomach Neoplasms/metabolism , Transcription Factors/biosynthesis , Wnt Signaling Pathway/physiology , Adult , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Prognosis , RNA, Antisense/antagonists & inhibitors , RNA, Long Noncoding/antagonists & inhibitors , Repressor Proteins , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate/trends , Transcription Factors/antagonists & inhibitors
9.
Biochimie ; 138: 43-49, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28363791

ABSTRACT

BACKGROUND: Hypoxia/reoxygenation (H/R)-induced cardiomyocyte damage in vitro bears many similarities to myocardial infraction in vivo. In this study, we investigated whether long noncoding RNA (lncRNA) BDNF-AS has functional role in rescuing H/R-induced damage in cardiomyocyte in vitro. METHOD: Murine neonatal cardiomyocytes were cultured in vitro, and treated with either long (Hypoxia (L)) or short (Hypoxia (S)) H/R conditions. Viability and TUNEL assays were conducted to assess cardiomyocyte death or apoptosis. QRT-PCR was used to examine BDNF-AS mRNA expression. Endogenous BDNF-AS was downregulated by siRNA in cardiomyocyte. Its effect on H/R-induced cardiomyocyte damage was then examined. In addition, survival-associated proteins, including BDNF, VEGF and Akt were examined by western blot in siRNA-transfected cardiomyocytes. RESULTS: Hypoxia (S) induced apoptosis whereas Hypoxia (L) induced cell death in murine neonatal cardiomyocyte in vitro. Under both conditions, endogenous cardiomyocyte BDNF-AS was significantly upregulated. SiRNA-mediated BDNF-AS downregulation rescued cardiomyocyte death under Hypoxia (L) condition, and reduced cardiomyocyte apoptosis under Hypoxia (L) condition. Moreover, BDNF-AS downregulation activated BDNF and VEGF through upregulation, and activated Akt through phosphorylation in H/R-damaged cardiomyocyte. CONCLUSION: Inhibition of lncRNA BDNF-AS may be an efficient way to rescue cardiomyocyte from H/R-induced apoptosis or cell death.


Subject(s)
Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/genetics , Hypoxia , Myocytes, Cardiac/drug effects , RNA, Long Noncoding/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/metabolism , RNA, Long Noncoding/metabolism
10.
FEBS Lett ; 590(24): 4586-4593, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27861797

ABSTRACT

Interleukin-6 (IL-6) is widely expressed in a variety of malignant tumors; thus, targeting the IL-6/STAT3 pathway represents a promising therapeutic strategy for malignant cancers. In this study, we identified a noncoding RNA, AS-IL6, which is transcribed antisense to IL6 and induces IL6 expression in glioma cells. Knockdown of AS-IL6 attenuates LPS-induced IL6 transcription. Interestingly, AS-IL6 does not change IL6 mRNA stability, but induces the enrichment of histone H3 acetylated at lysine 27 (H3K27Ac) at the IL6 promoter. In addition, we found that depletion of AS-IL6 inhibits the invasive ability of glioblastoma cells, while treatment of cells with recombinant IL6 reverses this effect. Our results reveal a novel mechanism of IL6 regulation and demonstrate an oncogenic role for AS-IL6 in glioma cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Histones/genetics , Interleukin-6/genetics , Neuroglia/metabolism , RNA, Antisense/genetics , RNA, Untranslated/genetics , Acetylation , Cell Line, Tumor , Cell Movement/drug effects , Histones/metabolism , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Neuroglia/drug effects , Neuroglia/pathology , Promoter Regions, Genetic , RNA Stability , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/antagonists & inhibitors , RNA, Untranslated/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcriptional Activation
11.
BMC Mol Biol ; 6: 3, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15691375

ABSTRACT

BACKGROUND: The parasitic protozoa belonging to Leishmania (L.) donovani complex possess abundant, developmentally regulated cathepsin L-like cysteine proteases. Previously, we have reported the isolation of cysteine protease gene, Ldccys2 from Leishmania (L.) chagasi. Here, we have further characterized this cysteine protease gene and demonstrated its role during infection and survival of Leishmania (L.) chagasi within the U937 macrophage cells. RESULTS: The amastigote specific Ldccys2 genes of L. (L.) chagasi and L. (L.) donovani have identical gene organization, as determined by southern blots. In vivo expression analyses by Northern blots showed that Ldccys2 is amastigote specific. Western blot using anti-Ldccys2 antibody confirmed the amastigote specific protein expression. Recombinant expression of Ldccys2, a 30 kDA protein, was functionally active in a gelatin assay. Results from Ldccys2 heterozygous knockout mutants showed its role during macrophage infection and in intra-macrophage survival of the parasites. Since attempts to generate null mutants failed, we used antisense RNA inhibition to regulate Ldcccys2 gene expression. Not surprisingly, the results from antisense studies further confirmed the results from heterozygous knockout mutants, reiterating the importance of amastigote specific cysteine proteases in Leishmania infection and pathogenesis. CONCLUSIONS: The study shows that Ldccys2 is a developmentally regulated gene and that Ldccys2 is expressed only in infectious amastigote stages of the parasite. The collective results from both the heterozygous knockout mutants and antisense mRNA inhibition studies shows that Ldccys2 helps in infection and survival of L. (L.) chagasi amastigotes within the macrophage cells. Finally, antisense RNA technique can be used as an alternate approach to gene knockout, for silencing gene expression in L. (L.) chagasi, especially in cases such as this, where a null mutant cannot be achieved by homologous recombination.


Subject(s)
Cysteine Endopeptidases/genetics , Gene Expression/genetics , Genes, Protozoan , Leishmania infantum/genetics , Leishmania infantum/physiology , Protozoan Proteins/genetics , Animals , Blotting, Northern , Blotting, Western/methods , Cysteine Endopeptidases/metabolism , DNA Mutational Analysis , Humans , Leishmania infantum/enzymology , Macrophages/parasitology , Proteins/analysis , Protozoan Proteins/metabolism , RNA, Antisense/antagonists & inhibitors , RNA, Messenger/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Survival , U937 Cells/cytology , U937 Cells/parasitology
12.
Peptides ; 24(11): 1829-36, 2003 Nov.
Article in English | MEDLINE | ID: mdl-15019216

ABSTRACT

The quorum-sensing interfering RNAIII-inhibiting peptide (RIP) YSPXTNF and its synthetic analogues YSPWTNF and YSPITNF have been shown to prevent and suppress diseases caused by Staphylococcus aureus at different body sites in different animal models. This study was designed to investigate histopathologically the therapeutic efficacy of lead peptide RIP YSPWTNF-NH(2) in the subcutaneous air sac murine model of acquired S. aureus sepsis. Two experimental protocols were evaluated: an infection/therapy protocol, for which twenty BALB/c mice per group were infected with a subcutaneous inoculum of S. aureus strain ATCC 25923 ( [Formula: see text] colony forming units) that were either pretreated or not with 150microg of peptide RIP, and a safety protocol, for which three uninfected mice per group received treatment with either 150microg of peptide RIP or saline. Therapeutic efficacy was assessed by clinical examination for a period of 20 days and histopathology at 12, 24, 36, 48, 96 and 168h after inoculation. Treatment safety was assessed histopathologically at 24, 48 and 264h after inoculation. Subcutaneous administration of uninfected control mice with a single dose of peptide RIP YSPWTNF caused no significant histopathology in most organs examined, except for slight to moderate lung and liver congestions. In contrast to the situation with the untreated infected control group mice that presented with histopathological alterations consistent with the diagnosis of rapidly progressive and highly erosive disease (100% mortality by day 3), treatment of infected animals with peptide RIP YSPWTNF had a profound therapeutic effect on survival rate (67% by day 20) and on disease progression. The histopathological examination confirmed the clinical findings showing that extensive tissue damage at the site of the infection and in organs were greatly suppressed in the peptide RIP-treated animals.


Subject(s)
Oligopeptides/pharmacology , Oligopeptides/therapeutic use , RNA, Antisense/antagonists & inhibitors , RNA, Bacterial/antagonists & inhibitors , Sepsis/drug therapy , Sepsis/pathology , Staphylococcus aureus , Animals , Erythrocytes/drug effects , Female , Hemolysis/drug effects , Humans , Mice , Mice, Inbred BALB C , Oligopeptides/adverse effects , Sepsis/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Survival Rate
14.
Science ; 345(6201): 1139-45, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25081482

ABSTRACT

Many RNA regulatory proteins controlling pre-messenger RNA splicing contain serine:arginine (SR) repeats. Here, we found that these SR domains bound hydrogel droplets composed of fibrous polymers of the low-complexity domain of heterogeneous ribonucleoprotein A2 (hnRNPA2). Hydrogel binding was reversed upon phosphorylation of the SR domain by CDC2-like kinases 1 and 2 (CLK1/2). Mutated variants of the SR domains changing serine to glycine (SR-to-GR variants) also bound to hnRNPA2 hydrogels but were not affected by CLK1/2. When expressed in mammalian cells, these variants bound nucleoli. The translation products of the sense and antisense transcripts of the expansion repeats associated with the C9orf72 gene altered in neurodegenerative disease encode GRn and PRn repeat polypeptides. Both peptides bound to hnRNPA2 hydrogels independent of CLK1/2 activity. When applied to cultured cells, both peptides entered cells, migrated to the nucleus, bound nucleoli, and poisoned RNA biogenesis, which caused cell death.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Cell Nucleolus/metabolism , Dipeptides/metabolism , Frontotemporal Dementia/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Proteins/genetics , Alternative Splicing , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , C9orf72 Protein , Cell Death , Cells, Cultured , Dipeptides/genetics , Dipeptides/pharmacology , Excitatory Amino Acid Transporter 2 , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Glutamate Plasma Membrane Transport Proteins/genetics , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , RNA, Antisense/antagonists & inhibitors , RNA, Antisense/biosynthesis , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Ribosomal/antagonists & inhibitors , RNA, Ribosomal/biosynthesis , Repetitive Sequences, Amino Acid , Transcription, Genetic
15.
Mol Microbiol ; 26(2): 311-20, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9383156

ABSTRACT

The hok/sok system of plasmid R1, which mediates plasmid stabilization by the killing of plasmid-free cells, codes for two RNA species, Sok antisense RNA and hok mRNA. Sok RNA, which is unstable, inhibits translation of the stable hok mRNA. The 64nt Sok RNA folds into a single stem-loop domain with an 11 nt unstructured 5' domain. The initial recognition reaction between Sok RNA and hok mRNA takes place between the 5' domain and the complementary region in hok mRNA. In this communication we examine the metabolism of Sok antisense RNA. We find that RNase E cleaves the RNA 6nt from its 5' end and that this cleavage initiates Sok RNA decay. The RNase E cleavage occurs in the part of Sok RNA that is responsible for the initial recognition of the target loop in hok mRNA and thus leads to functional inactivation of the antisense. The major RNase E cleavage product (denoted pSok-6) is rapidly degraded by polynucleotide phosphorylase (PNPase). Thus, the RNase E cleavage tags pSok-6 for further rapid degradation by PNPase from its 3' end. We also show that Sok RNA is polyadenylated by poly(A) polymerase I (PAP I), and that the poly(A)-tailing is prerequisite for the rapid 3'-exonucleolytic degradation by PNPase.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins , Endoribonucleases/genetics , Escherichia coli Proteins , Plasmids/genetics , RNA, Antisense/genetics , RNA, Bacterial/genetics , Base Sequence , Endoribonucleases/pharmacology , Escherichia coli/genetics , Molecular Sequence Data , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , RNA, Antisense/antagonists & inhibitors , RNA, Bacterial/metabolism
16.
J Immunol ; 167(1): 327-35, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418667

ABSTRACT

To examine a role of DNA polymerase zeta in somatic hypermutation, we generated transgenic mice that express antisense RNA to a portion of mouse REV3, the gene encoding this polymerase. These mice express high levels of antisense RNA, significantly reducing the levels of endogenous mouse REV3 transcript. Following immunization to a hapten-protein complex, transgenic mice mounted vigorous Ab responses, accomplished the switch to IgG, and formed numerous germinal centers. However, in most transgenic animals, the generation of high affinity Abs was delayed. In addition, accumulation of somatic mutations in the V(H) genes of memory B cells from transgenic mice was decreased, particularly among those that generate amino acid replacements that enhance affinity of the B cell receptor to the hapten. These data implicate DNA polymerase zeta, a nonreplicative polymerase, in the process of affinity maturation, possibly through a role in somatic hypermutation, clonal selection, or both.


Subject(s)
Antibody Affinity/genetics , DNA-Directed DNA Polymerase/genetics , Germinal Center/enzymology , Germinal Center/immunology , Mutation , RNA, Antisense/biosynthesis , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Clone Cells , DNA-Directed DNA Polymerase/biosynthesis , Down-Regulation/genetics , Germinal Center/metabolism , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin lambda-Chains/biosynthesis , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleic Acid Synthesis Inhibitors , RNA, Antisense/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL