321 resultados para Rnai
Resumo:
The switch from within-hive activities to foraging behavior is a major transition in the life cycle of a honeybee (Apis mellifera) worker. A prominent regulatory role in this switch has long been attributed to juvenile hormone (JH), but recent evidence also points to the yolk precursor protein vitellogenin as a major player in behavioral development. In the present study, we injected vitellogenin double-stranded RNA (dsVg) into newly emerged worker bees of Africanized genetic origin and introduced them together with controls into observation hives to record flight behavior. RNA interference-mediated silencing of vitellogenin gene function shifted the onset of long-duration flights (> 10 min) to earlier in life (by 3-4 days) when compared with sham and untreated control bees. In fact, dsVg bees were observed conducting such flights extremely precociously, when only 3 days old. Short-duration flights (< 10 min), which bees usually perform for orientation and cleaning, were not affected. Additionally, we found that the JH titer in dsVg bees collected after 7 days was not significantly different from the controls. The finding that depletion of the vitellogenin titer can drive young bees to become extremely precocious foragers could imply that vitellogenin is the primary switch signal. At this young age, downregulation of vitellogenin gene activity apparently had little effect on the JH titer. As this unexpected finding stands in contrast with previous results on the vitellogenin/JH interaction at a later age, when bees normally become foragers, we propose a three-step sequence in the constellation of physiological parameters underlying behavioral development.
Resumo:
In insects, the steroid hormone 20-hydroxyecdysone (20E) coordinates major developmental transitions. While the first and the final steps of 20E biosynthesis are characterized, the pathway from 7-dehydrocholesterol to 5β-ketodiol, commonly referred as the "black box", remains hypothetical and whether there are still unidentified enzymes is unknown. The black box would include some oxidative steps, which are believed to be mediated by P450 enzymes. To identify new enzyme(s) involved in steroid synthesis, we analyzed by small-scale microarray the expression of all the genes encoding P450 enzymes of the malaria mosquito Anopheles gambiae in active steroidogenic organs of adults, ovaries from blood-fed females and male reproductive tracts, compared to inactive steroidogenic organs, ovaries from non-blood-fed females. Some genes encoding P450 enzymes were specifically overexpressed in female ovaries after a blood-meal or in male reproductive tracts but only three genes were found to be overexpressed in active steroidogenic organs of both females and males: cyp307a1, cyp4g16 and cyp6n1. Among these genes, only cyp307a1 has an expression pattern similar to other mosquito steroidogenic genes. Moreover, loss-of-function by transient RNAi targeting cyp307a1 disrupted ecdysteroid production demonstrating that this gene is required for ecdysteroid biosynthesis in Anopheles gambiae.
Resumo:
Tartraatti-resistentin happaman fosfataasin hiljentäminen RNAi menetelmällä: odottamaton vaikutus monosyytti-makrofagi linjan soluissa RNA interferenssi (RNAi) eli RNA:n hiljentyminen löydettiin ensimmäisenä kasveissa, ja 2000-luvulla RNAi menetelmä on otettu käyttöön myös nisäkässoluissa. RNAi on mekanismi, jossa lyhyet kaksi juosteiset RNA molekyylit eli siRNA:t sitoutuvat proteiinikompleksiin ja sitoutuvat komplementaarisesti proteiinia koodaavaan lähetti RNA:han katalysoiden lähetti RNA:n hajoamisen. Tällöin RNA:n koodaamaa proteiinia ei solussa tuoteta. Tässä työssä on RNA interferenssi menetelmän avuksi kehitetty uusi siRNA molekyylien suunnittelualgoritmi siRNA_profile, joka etsii lähetti RNA:sta geenin hiljentämiseen sopivia kohdealueita. Optimaalisesti suunnitellulla siRNA molekyylillä voi olla mahdollista saavuttaa pitkäaikainen geenin hiljeneminen ja spesifinen kohdeproteiinin määrän aleneminen solussa. Erilaiset kemialliset modifikaatiot, mm. 2´-Fluoro-modifikaatio, siRNA molekyylin riboosirenkaassa lisäsivät siRNA molekyylin stabiilisuutta veren plasmassa sekä siRNA molekyylin tehokkuutta. Nämä ovat tärkeitä siRNA molekyylien ominaisuuksia kun RNAi menetelmää sovelletaan lääketieteellisiin tarkoituksiin. Tartraatti-resistentti hapan fosfataasi (TRACP) on entsyymi, joka esiintyy luunsyöjäsoluissa eli osteoklasteissa, antigeenejä esittelevissä dendiriittisissä soluissa sekä eri kudosten makrofageissa, jotka ovat syöjäsoluja. TRACP entsyymin biologista tehtävää ei ole saatu selville, mutta oletetaan että TRACP entsyymin kyvyllä tuottaa reaktiivisia happiradikaaleja on tehtävä sekä luuta hajoittavissa osteoklasteissa sekä antigeenia esittelevissä dendriittisissä soluissa. Makrofageilla, jotka yliekpressoivat TRACP entsyymiä, on myös solunsisäinen reaktiivisten happiradikaalien tuotanto sekä bakteerin tappokyky lisääntynyt. TRACP-geenin hiljentämiseen tarkoitetut spesifiset DNA ja siRNA molekyylit aiheuttivat monosyytti-makrofagilinjan soluviljelymallissa TRACP entsyymin tuoton lisääntymistä odotusten vastaisesti. DNA ja RNA molekyylien vaikutusta TRACP entsyymin tuoton lisääntymiseen tutkittiin myös Tolllike reseptori 9 (TLR9) poistogeenisestä hiirestä eristetyissä monosyyttimakrofaagisoluissa. TRACP entsyymin tuoton lisääntyminen todettiin sekvenssistä ja TLR9:stä riippumattomaksi vasteeksi solun ulkopuolisia DNA ja RNA molekyylejä vastaan. Havainto TRACP entsyymin tuoton lisääntymisestä viittaa siihen, että TRACP entsyymillä on tehtävä solun immuunipuolustusjärjestelmässä.
Resumo:
Searching for effective Smad3 gene-based gene therapies for hepatic fibrosis, we constructed siRNA expression plasmids targeting the rat Smad3 gene and then delivered these plasmids into hepatic stellate cells (HSCs). The effect of siRNAs on the mRNA levels of Smad2, Smad3, Smad4, and collagens I-α1, III-α1 and IV-α1 (Colα1, Col3α1, Col4α1, respectively) was determined by RT-PCR. Eighty adult male Sprague-Dawley rats were randomly divided into three groups. Twice a week for 8 weeks, the untreated hepatic fibrosis model (N = 30) and the treated group (N = 20) were injected subcutaneously with 40% (v/v) carbon tetrachloride (CCl4)-olive oil (3 mL/kg), and the normal control group (N = 30) was injected with olive oil (3 mL/kg). In the 4th week, the treated rats were injected subcutaneously with liposome-encapsulated plasmids (150 µg/kg) into the right liver lobe under general anesthesia once every 2 weeks, and the untreated rats were injected with the same volume of buffer. At the end of the 6th and 8th weeks, liver tissue and sera were collected. Pathological changes were assessed by a semi-quantitative scoring system (SSS), and a radioimmunoassay was used to establish a serum liver fibrosis index (type III procollagen, type IV collagen, laminin, and hyaluronic acid). The mRNA expression levels of the above cited genes were reduced in the HSCs transfected with the siRNA expression plasmids. Moreover, in the treated group, fibrosis evaluated by the SSS was significantly reduced (P < 0.05) and the serum indices were greatly improved (P < 0.01). These results suggest that Smad3 siRNA expression plasmids have an anti-fibrotic effect.
Resumo:
Pituitary tumor-transforming gene-1 (PTTG1) is a proto-oncogene that promotes tumorigenesis and metastasis in numerous cell types and is overexpressed in a variety of human tumors. We have demonstrated that PTTG1 expression was up-regulated in both human prostate cancer specimens and prostate cancer cell lines. For a more direct assessment of the function of PTTG1 in prostate tumorigenesis, RNAi-mediated knockdown was used to selectively decrease PTTG1 expression in PC3 human prostate tumor cells. After three weeks of selection, colonies stably transfected with PTTG1-targeted RNAi (the knockdown PC3 cell line) or empty vector (the control PC3 cell line) were selected and expanded to investigate the role of PTTG1 expression in PC3 cell growth and invasion. Cell proliferation rate was significantly slower (28%) in the PTTG1 knockdown line after 6 days of growth as indicated by an MTT cell viability assay (P < 0.05). Similarly, a soft agar colony formation assay revealed significantly fewer (66.7%) PTTG1 knockdown PC3 cell colonies than control colonies after three weeks of growth. In addition, PTTG1 knockdown resulted in cell cycle arrest at G1 as indicated by fluorescence-activated cell sorting. The PTTG1 knockdown PC3 cell line also exhibited significantly reduced migration through Matrigel in a transwell assay of invasive potential, and down-regulation of PTTG1 could lead to increased sensitivity of these prostate cancer cells to a commonly used anticancer drug, taxol. Thus, PTTG1 expression is crucial for PC3 cell proliferation and invasion, and could be a promising new target for prostate cancer therapy.
Resumo:
Fanconi anemia complementation group F protein (FANCF) is a key factor, which maintains the function of FA/BRCA, a DNA damage response pathway. However, the functional role of FANCF in breast cancer has not been elucidated. We performed a specific FANCF-shRNA knockdown of endogenous FANCF in vitro. Cell viability was measured with a CCK-8 assay. DNA damage was assessed with an alkaline comet assay. Apoptosis, cell cycle, and drug accumulation were measured by flow cytometry. The expression levels of protein were determined by Western blot using specific antibodies. Based on these results, we used cell migration and invasion assays to demonstrate a crucial role for FANCF in those processes. FANCF shRNA effectively inhibited expression of FANCF. We found that proliferation of FANCF knockdown breast cancer cells (MCF-7 and MDA-MB-435S) was significantly inhibited, with cell cycle arrest in the S phase, induction of apoptosis, and DNA fragmentation. Inhibition of FANCF also resulted in decreased cell migration and invasion. In addition, FANCF knockdown enhanced sensitivity to doxorubicin in breast cancer cells. These results suggest that FANCF may be a potential target for molecular, therapeutic intervention in breast cancer.
Resumo:
Current studies find that degenerated cartilage endplates (CEP) of vertebrae, with fewer diffusion areas, decrease nutrient supply and accelerate intervertebral disc degeneration. Many more apoptotic cells have been identified in degenerated than in normal endplates, and may be responsible for the degenerated grade. Previous findings suggest that inhibition of apoptosis is one possible approach to improve disc regeneration. It is postulated that inhibition of CEP cell apoptosis may be responsible for the regeneration of endplates. Caspase-3, involved in the execution phase of apoptosis, is a candidate for regulating the apoptotic process. In the present study, CEP cells were incubated in 1% fetal bovine serum. Activated caspases were detected to identify the apoptotic pathway, and apoptosis was quantified by flow cytometry. Lentiviral caspase-3 short hairpin RNA (shRNA) was employed to study its protective effects against serum deprivation. Silencing of caspase-3 expression was quantified by reverse transcription-polymerase chain reaction and Western blots, and inhibition of apoptosis was quantified by flow cytometry. Serum deprivation increased apoptosis of rat CEP cells through activation of a caspase cascade. Lentiviral caspase-3 shRNA was successfully transduced into CEP cells, and specifically silenced endogenous caspase-3 expression. Surviving cells were protected by the downregulation of caspase-3 expression and activation. Thus, lentiviral caspase-3 shRNA-mediated RNAi successfully silenced endogenous caspase-3 expression, preventing inappropriate or premature apoptosis.
Resumo:
Tesis (Maestría en Ciencias con Acentuación en Inmunolobiología) UANL, 2012.
Resumo:
About 80 years ago, the neurosecretory eyestalk structures and their role in endocrine regulation was recognized in crustaceans. After the recognition it took half a century to identify the first peptide hormone. Till date a large number of homologous peptides of crustacean hyperglycaemic hormone and moult-inhibiting hormone have been identified, consequently they are called the CHH family hormones. This family comprises of highly multifunctional peptides which according to sequences and precursor structures can be divided into two subfamilies, type-I (CHH/ITP) and II (MIH, MOIH, VIH/GIH) (Webster et al., 2012). The XO-SG complex has been the major site of the two subfamilies. The advent of molecular techniques resulted in the characterization of different precursors of CHH, MIH and GIH; these hormones consist of a signal peptide, but only the preprohormone of CHHs contain a precursor- related peptide (CPRP) located between the signal and the mature hormone (Weidemann et al., 1989; Klein et al., 1993b; De Kleijn and Van Herp, 1995). The essentialities of the gene structure comply with the functions of the CHH family hormones. The CHH family hormone functions are inhibitory as well as stimulatory in the process of reproduction and maturation
Resumo:
RNA interference (RNAi) is a recently discovered process, in which double stranded RNA (dsRNA) triggers the homology-dependant degradation of cognate messenger RNA (mRNA). In a search for new components of the RNAi machinery in Dictyostelium, a new gene was identified, which was called helF. HelF is a putative RNA helicase, which shows a high homology to the helicase domain of Dicer, to the helicase domain of Dictyostelium RdRP and to the C. elegans gene drh-1, that codes for a dicer related DExH-box RNA helicase, which is required for RNAi. The aim of the present Ph.D. work was to investigate the role of HelF in PTGS, either induced by RNAi or asRNA. A genomic disruption of the helF gene was performed, which resulted in a distinct mutant morphology in late development. The cellular localization of the protein was elucidated by creating a HelF-GFP fusion protein, which was found to be localized in speckles in the nucleus. The involvement of HelF in the RNAi mechanism was studied. For this purpose, RNAi was induced by transformation of RNAi hairpin constructs against four endogenous genes in wild type and HelF- cells. The silencing efficiency was strongly enhanced in the HelF K.O. strain in comparison with the wild type. One gene, which could not be silenced in the wild type background, was successfully silenced in HelF-. When the helF gene was disrupted in a secondary transformation in a non-silenced strain, the silencing efficiency was strongly improved, a phenomenon named here “retrosilencing”. Transcriptional run-on experiments revealed that the enhanced gene silencing in HelF- was a posttranscriptional event, and that the silencing efficiency depended on the transcription levels of hairpin RNAs. In HelF-, the threshold level of hairpin transcription required for efficient silencing was dramatically lowered. The RNAi-mediated silencing was accompanied by the production of siRNAs; however, their amount did not depend on the level of hairpin transcription. These results indicated that HelF is a natural suppressor of RNAi in Dictyostelium. In contrast, asRNA mediated gene silencing was not enhanced in the HelF K.O, as shown for three tested genes. These results confirmed previous observations (H. Martens and W. Nellen, unpublished) that although similar, RNAi and asRNA mediated gene silencing mechanisms differ in their requirements for specific proteins. In order to characterize the function of the HelF protein on a molecular level and to study its interactions with other RNAi components, in vitro experiments were performed. Besides the DEAH-helicase domain, HelF contains a double-stranded RNA binding domain (dsRBD) at its N-terminus, which showed high similarity to the dsRBD domain of Dicer A from Dictyostelium. The ability of the recombinant dsRBDs from HelF and Dicer A to bind dsRNA was examined and compared. It was shown by gel-shift assays that both HelF-dsRBD and Dicer-dsRBD could bind directly to long dsRNAs. However, HelF-dsRBD bound more efficiently to dsRNA with imperfect matches than to perfect dsRNA. Both dsRBDs bound specifically to a pre-miRNA substrate (pre-let-7). The results suggested that most probably there were two binding sites for the proteins on the pre-miRNA substrate. Moreover, it was shown that HelF-dsRBD and Dicer-dsRBD have siRNA-binding activity. The affinities of the two dsRBDs to the pre-let-7 substrate were also examined by plasmon surface resonance analyses, which revealed a 9-fold higher binding affinity of the Dicer-dsRBD to pre-let-7 compared to that of the HelF-dsRBD. The binding of HelF-dsRBD to the pre-let-7 was impaired in the presence of Mg2+, while the Dicer-dsRBD interaction with pre-let-7 was not influenced by the presence of Mg2+. The results obtained in this thesis can be used to postulate a model for HelF function. In this, HelF acts as a nuclear suppressor of RNAi in wild type cells by recognition and binding of dsRNA substrates. The protein might act as a surveillance system to avoid RNAi initiation by fortuitous dsRNA formation or low abundance of dsRNA trigger. If the protein acts as an RNA helicase, it could unwind fold-back structures in the nucleus and thus lead to decreased RNAi efficiency. A knock-out of HelF would result in initiation of the RNAi pathway even by low levels of dsRNA. The exact molecular function of the protein in the RNAi mechanism still has to be elucidated. RNA interferenz (RNAi) ist ein in jüngster Zeit entdeckter Mechanismus, bei dem doppelsträngige RNA Moleküle (dsRNA) eine Homologie-abhängige Degradation einer verwandten messenger-RNA (mRNA) auslösen. Auf der Suche nach neuen Komponenten der RNAi-Maschinerie in Dictyostelium konnte ein neues Gen (helF) identifiziert werden. HelF ist eine putative RNA-Helikase mit einer hohen Homologie zur Helikasedomäne der bekannten Dicerproteine, der Helikasedomäne der Dictyostelium RdRP und zu dem C. elegans Gen drh-1, welches für eine Dicer-bezogene DExH-box RNA Helikase codiert, die am RNAi-Mechanismus beteiligt ist. Das Ziel dieser Arbeit war es, die Funktion von HelF im Zusammenhang des RNAi oder asRNA induzierten PTGS zu untersuchen. Es wurde eine Unterbrechung des helF-Gens auf genomischer Ebene (K.O.) vorgenommen, was bei den Mutanten zu einer veränderten Morphologie in der späten Entwicklung führte. Die Lokalisation des Proteins in der Zelle konnte mit Hilfe einer GFP-Fusion analysiert werden und kleinen Bereichen innerhalb des Nukleus zugewiesen werden. Im Weiteren wurde der Einfluss von HelF auf den RNAi-Mechanismus untersucht. Zu diesem Zweck wurde RNAi durch Einbringen von RNAi Hairpin-Konstrukten gegen vier endogene Gene im Wiltypstamm und der HelF--Mutante induziert. Im Vergleich zum Wildtypstamm konnte im HelF--Mutantenstamm eine stark erhöhte „Silencing“-Effizienz nachgewiesen werden. Ein Gen, welches nach RNAi Initiation im Wildtypstamm unverändert blieb, konnte im HelF--Mutantenstamm erfolgreich stillgelegt werden. Durch sekundäres Einführen einer Gendisruption im helF-Locus in einen Stamm, in welchem ein Gen nicht stillgelegt werden konnte, wurde die Effizienz des Stilllegens deutlich erhöht. Dieses Phänomen wurde hier erstmals als „Retrosilencing“ beschrieben. Mit Hilfe von transkriptionellen run-on Experimenten konnte belegt werden, dass es sich bei dieser erhöhten Stilllegungseffizienz um ein posttranskriptionelles Ereignis handelte, wobei die Stillegungseffizienz von der Transkriptionsstärke der Hairpin RNAs abhängt. Für die HelF--Mutanten konnte gezeigt werden, dass der Schwellenwert zum Auslösen eines effizienten Stillegens dramatisch abgesenkt war. Obwohl die RNAi-vermittelte Genstilllegung immer mit der Produktion von siRNAs einhergeht, war die Menge der siRNAs nicht abhängig von dem Expressionsniveau des Hairpin-Konstruktes. Diese Ergebnisse legen nahe, dass es sich bei der HelF um einen natürlichen Suppressor des RNAi-Mechanismus in Dictyostelium handelt. Im Gegensatz hierzu war die as-vermittelte Stilllegung von drei untersuchten Genen im HelF-K.O. im Vergleich zum Wildyp unverändert. Diese Ergebnisse bestätigten frühere Beobachtungen (H. Martens und W. Nellen, unveröffentlicht), wonach die Mechanismen für RNAi und asRNA-vermittelte Genstilllegung unterschiedliche spezifische Proteine benötigen. Um die Funktion des HelF-Proteins auf der molekularen Ebene genauer zu charakterisieren und die Interaktion mit anderen RNAi-Komponenten zu untersuchen, wurden in vitro Versuche durchgeführt. Das HelF-Protein enthält, neben der DEAH-Helikase-Domäne eine N-terminale Doppelstrang RNA bindende Domäne (dsRBD) mit einer hohen Ähnlichkeit zu der dsRBD des Dicer A aus Dictyostelium. Die dsRNA-Bindungsaktivität der beiden dsRBDs aus HelF und Dicer A wurde analysiert und verglichen. Es konnte mithilfe von Gel-Retardationsanalysen gezeigt werden, dass sowohl HelF-dsRBD als auch Dicer-dsRBD direkt an lange dsRNAs binden können. Hierbei zeigte sich, dass die HelF-dsRBD eine höhere Affinität zu einem imperfekten RNA-Doppelstrang besitzt, als zu einer perfekt gepaarten dsRNA. Für beide dsRBDs konnte eine spezifische Bindung an ein pre-miRNA Substrat nachgewiesen werden (pre-let-7). Dieses Ergebnis legt nah, dass es zwei Bindestellen für die Proteine auf dem pre-miRNA Substrat gibt. Überdies hinaus konnte gezeigt werden, dass die dsRBDs beider Proteine eine siRNA bindende Aktivität besitzen. Die Affinität beider dsRBDs an das pre-let-7 Substrat wurde weiterhin mit Hilfe der Plasmon Oberflächen Resonanz untersucht. Hierbei konnte eine 9-fach höhere Bindeaffinität der Dicer-dsRBD im Vergleich zur HelF-dsRBD nachgewiesen werden. Während die Bindung der HelF-dsRBD an das pre-let-7 durch die Anwesenheit von Mg2+ beeinträchtigt war, zeigte sich kein Einfluß von Mg2+ auf das Bindeverhalten der Dicer-dsRBD. Mit Hilfe der in dieser Arbeit gewonnen Ergebnisse lässt sich ein Model für die Funktion von HelF postulieren. In diesem Model wirkt HelF durch Erkennen und Binden von dsRNA Substraten als Suppressor von der RNAi im Kern. Das Protein kann als Überwachungsystem gegen eine irrtümliche Auslösung von RNAi wirken, die durch zufällige dsRNA Faltungen oder eine zu geringe Häufigkeit der siRNAs hervorgerufen sein könnte. Falls das Protein eine Helikase-Aktivität besitzt, könnte es rückgefaltete RNA Strukturen im Kern auflösen, was sich in einer verringerten RNAi-Effizienz wiederspiegelt. Durch Ausschalten des helF-Gens würde nach diesem Modell eine erfolgreiche Auslösung von RNAi schon bei sehr geringer Mengen an dsRNA möglich werden. Das Modell erlaubt, die exakte molekulare Funktion des HelF-Proteins im RNAi-Mechanismus weiter zu untersuchen.