• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 125
  • 27
  • 9
  • 8
  • 6
  • 3
  • 3
  • 2
  • 1
  • Tagged with
  • 231
  • 231
  • 48
  • 46
  • 45
  • 38
  • 32
  • 29
  • 28
  • 25
  • 22
  • 21
  • 21
  • 20
  • 20
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
201

Investigating the PI3K/AKT/ATM Pathway, Telomeric DNA Damage, T Cell Death, and CRISPR/Cas9-mediated Gene Editing During Acute and Chronic HIV Infection

Khanal, Sushant 01 December 2022 (has links)
Human Immunodeficiency Virus (HIV) infection initiates major metabolic and cell- survival complications. Anti-retroviral therapy (ART) is the current approach to suppress active HIV replication to a level of undetected viral load, but it is not a curative approach. Newer and sophisticated gene editing technologies could indeed be a potent antiviral therapy to achieve a clinical sterilization/cure of HIV infection. Chronic HIV patients, even under a successful ART regimen, exhibit a low-grade inflammation, immune senescence, premature aging, telomeric DNA attrition, T cell apoptosis, and cellular homeostasis. In this dissertation, we investigated CD4 T cell homeostasis, degree of T cell apoptosis, an associated telomeric DNA damage, DNA damage repair signaling, and the apoptotic pathways in CD4 T cells during HIV infection with or without ART treatment. Our data support a DNA damage accumulation, and impaired DNA damage repair in chromosome ends via recruitment of 53BP1 protein to the damaged foci. We found that a key player of DNA damage and repair enzyme, ATM, and its associated checkpoint proteins (CHK1, CKH2) are affected by HIV infection. HIV infection also altered another multifunctional master regulator protein AKT that is crucial in maintaining cellular homeostasis. Curing HIV is the ultimate redemption against HIV-associated complications. To explore the possibility of a functional cure, we investigated the use of a transient and a non-viral CRISPR/Cas9-based gene editing technology targeting the latently incorporated HIV provirus. After performing a nucleofection/electroporation using an in vitro formulated ribonucleoprotein (RNP) constituting a synthetic guide RNA (gRNA) and Cas9 nuclease protein, we demonstrated a significant (maximum 97%) reduction of HIV-mRNA and p24-capsid protein expression, upon stimulation (using PMA) and latency reactivation of latently HIV-infected CD4 T cells and latent-monocytes. Notably, the RNP treatment did not induce any cytotoxic effects, without affecting the abilility of cell proliferation. A sequence specific cleavage of HIV-provirus in two crucial gene locations (targeting vpr/tat genes) showed the most significant suppression of HIV reactivation or latency reversal. We have used DNA sequencing, and T7EI assay to confirm the target-site-specific cleavage of the HIV-proviral genome. Our data confirm the activation of non- homologous end joining (NHEJ) pathway to repair the double-stranded DNA break created by the CRISPR/Cas9 treatment. Taken together, this study provides a new gene therapeutic approach using synthetic gRNA/Cas9 targeting HIV genome, which warrant further in vivo animal and human studies.
202

Production of recombinant A1AT with human glycosylation profile In CHO cells and its interaction with asialoglycoprotein receptors

Koyuturk, Izel 08 1900 (has links)
L'alpha-1 antitrypsine (A1AT) est un inhibiteur de sérine protéase sécrété principalement par le foie et libéré dans la circulation où sa concentration physiologique est de 1,5 à 3,5 g/L. La principale fonction de l'A1AT est d'inhiber l'activité de l'élastase des neutrophiles (NE) afin de maintenir l'équilibre protéase/anti-protéase dans les poumons. Son déficit (A1ATD) touche plus de 3,4 millions d'individus dans le monde chez qui l'élastase des neutrophiles décompose l'élastine, provoquant ainsi une diminution de l'élasticité du poumon ainsi qu'une dégradation de son tissu conjonctif. En conséquence, l'A1ATD entraîne des troubles respiratoires tels que l'emphysème ou la maladie pulmonaire obstructive chronique et ceux qui en sont atteints nécessitent des injections fréquentes d'A1AT purifiée à partir du sang d'un donneur. Cependant, l'A1AT plasmatique est hétérogène dans son état de glycosylation et sa qualité varie d'un lot à l'autre. De plus, il y a un risque, même très faible, de transmission d'agents pathogènes avec l'administration d'A1AT purifiée par plasma. Par conséquent, il existe un besoin pour une version recombinante. La glycoprotéine mature possède trois sites de N-glycosylation comprenant principalement des structures de type complexe bi-antennaires afucosylées et α-2,6-di-sialylées, A2G2S2 (6,6). Bien que la glycosylation ne soit pas essentielle à l'activité inhibitrice de l'A1AT, il a été démontré qu'elle a un impact significatif sur sa demi-vie in vivo. Notamment, l'acide sialique, un monosaccharide terminal chargé négativement présent sur les N-glycanes, aide à prolonger la demi-vie de l'A1AT dans le sérum en empêchant l'interaction entre l'avant-dernier galactose (Gal) du N-glycane et les récepteurs hépatiques des asialoglycoprotéines (ASGPRs), composés de deux sous-unités appelées lectines hépatiques (HL) 1 et 2, qui se lient aux glycoprotéines asialylées contenant un Gal terminal et conduisent à leur dégradation. Par conséquent, il est important de produire A1AT dans un système d'expression qui peut effectuer les modifications post-traductionnelles (PTM) appropriées à des fins thérapeutiques. Jusqu'à présent, la production d'A1AT recombinante (rA1AT) a été tentée dans différents systèmes d'expression cellulaire avec un succès limité. Malgré la disponibilité de diverses lignées cellulaires, les cellules ovariennes de hamster chinois (CHO) ont été largement utilisées pour la production de glycoprotéines thérapeutiques car ces cellules sont compatibles avec des stratégies de glyco-ingénierie pour produire des glycoprotéines recombinantes composées de glycanes de type humain. Cependant, ces cellules synthétisent des N-glycanes de type complexe comprenant de la fucosylation centrale et de l'acide sialique lié en α-2,3. Par conséquent, dans ce projet, l'objectif était de développer une version recombinante d'A1AT avec un profil de glycosylation humaine exprimée en cellules CHO modifiées et qui se prête à des utilisations thérapeutiques. À cette fin, dans notre étude, nous avons d'abord empêché l'α-2,3 sialylation ainsi que la fucosylation centrale en éliminant les gènes responsables via la technologie CRISPR/Cas9, suivie de la surexpression de l'α-2,6‐sialyltransférase humaine à l'aide d'un système d'expression inductible au cumate. Nous avons ensuite montré la supériorité du promoteur inductible CR5 pour l’expression de A1AT par rapport à cinq promoteurs constitutifs forts couramment utilisés dans l'industrie. En utilisant le promoteur CR5, nous avons généré des populations de CHO stables modifiées par glyco-ingénierie produisant plus de 2,1 g/L pour la forme native et 2,8 g/L pour la version mutée d'A1AT avec des N-glycanes analogues au produit clinique dérivé du plasma, la Prolastin-C. L'effet bénéfique de la supplémentation en N‐acétylmannosamine du milieu de culture cellulaire sur la glycosylation de l'A1AT a également été démontré. Enfin, nous avons montré que l'activité anti‐élastase des rA1ATs est comparable à celle de la Prolastin-C, et que la substitution des résidus méthionines critiques par des valines rendait A1AT significativement plus résistante à l'oxydation. Nous avons ensuite étudié l'impact de la glycosylation d'A1AT sur son interaction avec les orthologues d'ASGPR. Pour cela, nous avons initialement utilisé un test d'internalisation cellulaire basé sur la lignée cellulaire hépatique humaine HepG2 connue pour exprimer les ASGPRs à sa surface et avons examiné leur interaction avec les rA1ATs possédant divers profils de glycosylation. Comme le test d'internalisation basé sur les cellules HepG2 a démontré un faible rapport signal sur bruit (SNR) ainsi qu'un niveau élevé de signal de fond d'internalisation, nous avons cherché à développer un nouveau test basé sur des cellules CHO surexprimant des orthologues ASGPR recombinants. Alors que la sous-unité HL-1 humaine seule était suffisante pour lier et internaliser l'A1AT asialylée, les sous-unités HL-1 et HL-2 étaient nécessaires pour former des récepteurs fonctionnels et ayant une forte affinité pour les ASGPR de rat et de souris. Afin d'améliorer le SNR de notre test cellulaire d'internalisation, le tri cellulaire activé par fluorescence (FACS) a été utilisé pour enrichir les populations de cellules CHO pour celles exprimant des niveaux élevés d'orthologues ASGPR. Enfin, en utilisant des structures de glycanes remodelés par voie enzymatique de Prolastin-C, nous n'avons observé aucune internalisation lorsque les glycanes sont terminés avec α-2,6-Neu5Ac ni α-2,8-Neu5Ac-α-2,6-Neu5Ac par l’ASGPR de l'humain, du rat et de la souris. D'autre part, l'absorption de Prolastin-C portant des glycanes bi-antennaires avec une branche terminée par de l'acide sialique α-2,3 et l'autre par du galactose terminal, par l'ASGPR de souris a été statistiquement plus élevée que celle de l'humain et du rat. En somme, l'A1AT recombinante résistante à l'oxydation décrite dans ce projet pourrait représenter un meilleur médicament biothérapeutique tout en offrant une alternative sûre et plus stable pour la thérapie d'augmentation. Nous avons également contribué à une meilleure compréhension de l'impact de la sialylation de l'A1AT sur son internalisation cellulaire par les orthologues ASGPR. / Alpha-1 antitrypsin (A1AT) is a serine protease inhibitor secreted primarily by the liver, and released in the circulation where its physiological concentration is 1.5-3.5 g/L. The main physiological function of A1AT is to inhibit the activity of neutrophil elastase (NE) to maintain the protease/anti-protease balance in the lung. The A1AT deficiency (A1ATD) is affecting more than 3.4 million individuals worldwide where neutrophil elastase breaks down elastin, thereby causing a decrease in the elasticity of the lung as well as a degradation of its connective tissue. As a result, A1ATD leads to respiratory disorders such as emphysema or chronic obstructive pulmonary disease. Treatment of this health condition requires frequent injections of A1AT purified from donor blood. However, plasma A1AT is heterogeneous in its glycosylation state and its quality varies from batch to batch. Moreover, there is a risk, however very low, of pathogen transmittance with plasma-purified A1AT administration. Therefore, there is a need for recombinant version. The mature glycoprotein has three N-glycosylation sites possessing mostly afucosylated, α-2,6-di-sialylated bi-antennary complex-type structures, A2G2S2 (6,6). Though glycosylation is not essential for A1AT's inhibitory activity, it has been shown to have a significant impact on its in vivo half-life. Notably, sialic acid, a terminal negatively charged monosaccharide present on N-glycans, helps to prolong the half-life of A1AT in serum by preventing the interaction between the penultimate galactose (Gal) of the N-glycan and the hepatic asialoglycoprotein receptors (ASGPRs), composed of two subunits termed hepatic lectin (HL) 1 and 2, which bind to asialylated glycoproteins containing terminal Gal and lead to their degradation. To this extend, it is important to produce A1AT in an expression system that can carry out the appropriate post-translational modifications (PTMs) for therapeutic purposes. Thus far, the production of recombinant A1AT (rA1AT) has been attempted in different cell expression systems with limited success. Despite the availability of various cell lines, Chinese hamster ovary (CHO) cells have been widely used to produce therapeutic glycoproteins as these cells can tolerate glycoengineering strategies to produce recombinant glycoproteins with human-like glycans. However, these cells synthesize complex-type N-glycans with core-fucosylation along with α-2,3-linked sialic acid. Therefore, in this research project, the aim was to develop a recombinant version of A1AT with human glycosylation pattern expressed in genetically engineered CHO cells that would be amenable to therapeutic uses. To this end, in our study, we first prevented α-2,3 sialylation as well as core-fucosylation by eliminating the corresponding genes via CRISPR/Cas9 technology, followed by overexpressed human α-2,6‐sialyltransferase using a cumate‐inducible CHO expression system. We then showed superiority of the CR5 inducible promoter compared to five strong constitutive promoters commonly used in the industry. Using the CR5 promoter, we generated glycoengineered stable CHO pools producing over 2.1 g/L of the wild-type and 2.8 g/L of the mutein forms of A1AT, with N‐glycans analogous to the plasma‐derived clinical product, Prolastin-C. The effect of N‐acetylmannosamine supplementation to the cell culture media on the A1AT glycosylation was also demonstrated. Finally, we showed that the anti‐elastase activity of rA1ATs is comparable to that of Prolastin-C, and that substitution of critical methionine residues with valines rendered A1AT significantly more resistant to oxidation. We then studied the impact of A1AT glycosylation on its interaction with ASGPR orthologs. For this, we initially used a cell-based internalization assay based on the human HepG2 hepatic cell line known to express ASGPRs at its surface and examined their interaction with rA1ATs possessing various glycosylation profiles. As HepG2 cell-based internalization assay demonstrated poor signal-to-noise ratio (SNR) as well as high level of background internalization signal, we then aimed at developing a new assay based on CHO cells overexpressing recombinant ASGPRs orthologs. While human HL-1 subunit alone was sufficient to bind and internalize asialylated A1AT, both HL-1 and HL-2 subunits were required to form functional and high affinity receptors for the rat and mouse ASGPRs. To enhance SNR of our cell-based uptake assay, fluorescence-activated cell sorting (FACS) was used to enrich the CHO pools for cells expressing high levels of ASGPR orthologs. Finally, using enzymatically remodelled glycan structures of Prolastin-C, we observed no uptake when glycans are terminated with α-2,6-Neu5Ac nor α-2,8-Neu5Ac-α-2,6-Neu5Ac by human, rat, and mouse ASGPR orthologs. On the other hand, the uptake of Prolastin-C bearing bi-antennary glycans with one branch terminated with α-2,3 sialic acid and the other with terminal galactose, by mouse ASGPR was observed to be statistically higher than that by human and rat ASGPR orthologs. Collectively, the oxidation-resistant recombinant A1AT described in this project could represent a viable biobetter drug while offering a safe and more stable alternative for augmentation therapy. We also contributed a better understanding of the impact of A1AT sialylation on its cellular uptake by ASGPR orthologs.
203

Novel Genetic Modifiers in a Monogenic Cardiac Arrhythmia

Chai, Shin Luen, Chai 31 May 2018 (has links)
No description available.
204

Mechanistic studies of enzymes involved in DNA transactions

Stephenson, Anthony Aaron 07 November 2018 (has links)
No description available.
205

Computational Modeling of Planktonic and Biofilm Metabolism

Guo, Weihua 16 October 2017 (has links)
Most of microorganisms are ubiquitously able to live in both planktonic and biofilm states, which can be applied to dissolve the energy and environmental issues (e.g., producing biofuels and purifying waste water), but can also lead to serious public health problems. To better harness microorganisms, plenty of studies have been implemented to investigate the metabolism of planktonic and/or biofilm cells via multi-omics approaches (e.g., transcriptomics and proteomics analysis). However, these approaches are limited to provide the direct description of intracellular metabolism (e.g., metabolic fluxes) of microorganisms. Therefore, in this study, I have applied computational modeling approaches (i.e., 13C assisted pathway and flux analysis, flux balance analysis, and machine learning) to both planktonic and biofilm cells for better understanding intracellular metabolisms and providing valuable biological insights. First, I have summarized recent advances in synergizing 13C assisted pathway and flux analysis and metabolic engineering. Second, I have applied 13C assisted pathway and flux analysis to investigate the intracellular metabolisms of planktonic and biofilm cells. Various biological insights have been elucidated, including the metabolic responses under mixed stresses in the planktonic states, the metabolic rewiring in homogenous and heterologous chemical biosynthesis, key pathways of biofilm cells for electricity generation, and mechanisms behind the electricity generation. Third, I have developed a novel platform (i.e., omFBA) to integrate multi-omics data with flux balance analysis for accurate prediction of biological insights (e.g., key flux ratios) of both planktonic and biofilm cells. Fourth, I have designed a computational tool (i.e., CRISTINES) for the advanced genome editing tool (i.e., CRISPR-dCas9 system) to facilitate the sequence designs of guide RNA for programmable control of metabolic fluxes. Lastly, I have also accomplished several outreaches in metabolic engineering. In summary, during my Ph.D. training, I have systematically applied computational modeling approaches to investigate the microbial metabolisms in both planktonic and biofilm states. The biological findings and computational tools can be utilized to guide the scientists and engineers to derive more productive microorganisms via metabolic engineering and synthetic biology. In the future, I will apply 13C assisted pathway analysis to investigate the metabolism of pathogenic biofilm cells for reducing their antibiotic resistance. / Ph. D.
206

Caractérisation des fonctions des modifications post-traductionnelles de PCNA à l'aide d'un nouvel outil génétique / Characterization of PCNA’s post-translational modification functions using a new genetic tool

Dietsch, Frank 09 April 2019 (has links)
PCNA est une protéine essentielle qui intervient dans de nombreux mécanismes cellulaires et qui possède de nombreuses modifications post-traductionnelles (MPTs) dont les fonctions de certaines, restent encore inconnues. Afin d’étudier la fonction de ces MPTs, nous avons développé un nouvel outil génétique permettant in cellulo, de substituer la protéine endogène PCNA par une version mutée de la protéine appelée version de complémentation. La technique consiste à cotransfecter des cellules en culture avec deux types de plasmides. Un premier plasmide permet l’invalidation du gène de PCNA endogène dans les cellules transfectées par le système CRISPR-Cas9. Le deuxième plasmide dit de complémentation permet l’expression d’une forme mutée de PCNA. Sur l’ensemble d’une banque de mutants testés, deux mutants de PCNA se sont avérés être létaux (D122A et E124A). Nous avons démontré que ces deux sites sont impliqués dans l’initiation d’une voie de dégradation ubiquitine dépendante CRL4Cdt2 essentielle pour la mise en place de la protéolyse d’un cocktail de protéines (cdt1, p21, set8) durant la phase S. Nous avons démontré que les cellules mutantes pour PCNA (D122A et E124A) accumulent la protéine p21. Ce défaut de dégradation de p21 provoque alors des évènements de re-réplication menant à terme à la mort des cellules mutantes. / PCNA is an essential protein that is involved in many cellular mechanisms and has many post-translational modifications (PTMs). The functions of some PTMs, still remain unknown. In order to study the function of these PTMs, we have developed a new genetic tool allowing, in cellulo, the substitution of endogenous PCNA protein with a mutated version of the protein named complementation version. The technique involves cotransfection of the cells in culture with two types of plasmids. A first plasmid allows invalidation of the endogenous PCNA gene in transfected cells by the CRISPR-Cas9 system. The second plasmid, named complementation plasmid allows the expression of a mutated form of PCNA. In the whole bank of tested mutants, two PCNA mutants were found to be lethal (D122A and E124A). We have demonstrated that these two sites are involved in the initiation of an ubiquitin-dependent protein degradation CRL4Cdt2 pathway essential for the proteolysis of a protein cocktail (cdt1, p21, set8) during the S phase. We demonstrated that PCNA mutant cells (D122A and E124A) accumulate p21 protein. This lack of degradation of p21 then causes re-replication events leading ultimately to the mutant cells death.
207

CRISPR-Cas9 versus Prime Editing : en metodjämförelse, kliniska prövningar och etiska aspekter / CRISPR-Cas9 versus Prime Editing : a method comparison, clinical trials and ethical aspects

Olsson, Anna January 2020 (has links)
Det finns idag flera tusen genetiska sjukdomar som inte kan botas med hjälp av dagens läkemedelsbehandlingar. Detta är något forskarna försöker finna en lösning på. Två nya potenta genredigeringsverktyg har utvecklats och tros kunna bota och behandla många av de idag kända genetiska sjukdomarna. Detta är clustered regularly interspaced short palindromic repeats med CRISPR-associerade proteiner, CRISPR/Cas9 och prime editing. Tekniker som utvecklats från det adaptiva immunförsvaret hos prokaryoter. Både CRISPR/Cas9 och prime editing är RNA-guidade system med DNA som mål, de är även möjliga att programmera. Syftet med denna litteratursökning var att: 1) Jämföra teknikerna CRISPR/Cas9 och prime editing, 2) Undersöka vilka idag pågående kliniska prövningar som finns där någon av teknikerna används vid behandling av sjukdom. 3) Undersöka vilka sjukdomstillstånd som tros kunna botas och/eller behandlas med hjälp av någon av teknikerna samt 4) undersöka hur forskare ser på de etiska aspekterna av dessa tekniker. Information har hämtats under arbetets gång, främst från PubMed, Google och clinicaltrials.gov. Det finns idag 16 pågående studier där CRISPR/Cas9 används som behandlingsmetod. För prime editing finns det inga pågående studier. Sjukdomarna som forskarna hoppas kunna behandla med hjälp av metoderna är många, men de har kommit längst i utvecklingen av läkemedel för cancer, blodsjukdomar och ögonsjukdomar. De etiska diskussionerna har varit många och den stora frågan som diskuteras är hur tekniken skall regleras för att inte utnyttjas till sådant som potentiellt kan vara skadligt. Detta är två tekniker med hopp om nya behandlingsmetoder för genetiska sjukdomar, dock är de endast i början av sin utveckling och mer forskning och förfining av metoderna krävs innan de kan tillämpas kliniskt. / Today, there are thousands of genetic diseases that cannot be cured with the help of today's drug treatments. This is something the researchers are trying to find a solution to. Two new potent gene editing tools have been developed and are believed to be able to treat or cure many of today's genetic diseases. These are Clustered regularly interspaced short palindromic repeats with CRISPR-associated proteins, CRISPR/Cas9 and prime editing. Techniques developed from the adaptive immune system of prokaryotes. Both CRISPR/Cas9 and prime editing are RNA-guided DNA-targeted systems that are programmable. The purpose of this literature search was to: 1) compare the CRISPR/Cas9 and prime editing techniques, 2) investigate the current clinical trials in which any of the techniques are used to treat disease. 3) investigate which diseases that are believed to be cured and/or treated by using one of the techniques, and 4) investigare how researchers view the ethical aspects of these techniques. Information was gathered during a period between January to May 2020, mainly from PubMed, Google and clinicaltrials.gov. There are currently 16 ongoing studies using CRISPR/Cas9 as a treatment method. For prime editing there are no ongoing studies. The diseases that the researchers hope to be able to treat using the methods are many, but they have come the farthest in the development of a drug for cancer, blood diseases and eye diseases. There have been many discussions about the ethical side, but the big question being discussed is how the technology should be regulated so that it may not be used to harm instead of treat. These two techniques give hope of new treatment methods of genetic diseases, however, they are in the early stages of their development and more research and refinement of the methods is required before they can be applied clinically.
208

O gene Aire pode controlar mRNAs bem como os lncRNAs em células tímicas epiteliais medulares como evidenciado pela edição do genoma por CRISPR-Cas9 / Aire gene can control mRNAs as well as lncRNAs in medullary thymic epitelial cells as evidentiated by genome editing by CRISPR-Cas9

Duarte, Max Jordan de Souza 26 November 2018 (has links)
O timo é um órgão linfoide primário essencial para a manutenção da tolerância central através da seleção e eliminação de células T autoreativas. Precursores de células T, oriundas da medula óssea, chegam ao timo e migram do córtex para região da medula. As células epiteliais medulares tímicas (mTECs) expressam em sua superfície antígenos de tecidos periféricos (em inglês tissue-restricted antigens ou TRAs) que representam autoantígenos de todos os tecidos do corpo. Atuando como um fator de transcrição não clássico em células mTEC, o gene Autoimmune Regulator (Aire) desempenha um papel na expressão dos TRAs, cuja proteína codificada libera a RNA polimerase II (RNA Pol II) ancorada na cromatina e regula a expressão de mRNAs na glândula timo. A função biológica deste gene está ligada à indução de tolerância imunológica central impedindo o aparecimento de doenças autoimunes. Isso é resultado da seleção negativa de timócitos (precursores de células T) autoreativos que interagem fisicamente com as mTECs. Os timócitos autoreativos que reconhecem os TRAs como elementos estranhos são eliminados por apoptose. O co-cultivo de mTECs com timócitos representa um sistema-modelo in vitro adequado para se aproximar da interação celular que ocorre dentro do timo. Os resultados anteriores do nosso laboratório demonstraram que além do controle de mRNA de TRAs, o gene Aire também participa da modulação de miRNAs em mTECs uma vez que estas espécies de RNA são transcritas pela RNA Pol II. Continuando com essa linha de estudos, neste trabalho nós demonstramos pela primeira vez que Aire também modula a expressão de long noncoding RNAs (lncRNAs) em mTECs. Para isto fizemos uso da estratégia da perda de função analisando a expressão dessa espécie de RNA, assim como de mRNAs, em células mTEC Aire +/+ e mTEC Aire nocautes (KO Aire -/-) obtidas pela edição gênica por Crispr-Cas9. O transcriptoma dessas células que passaram ou não por adesão com timócitos, foi então analisado por hibridizações com microarrays. Isso evidenciou que Aire e adesão celular influenciam a expressão tanto de mRNAs como de lncRNAs. A reconstrução de redes de interação lncRNAs-mRNAs possibilitou evidenciar uma nova via de regulação pós-transcricional em células mTEC. / The thymus is a primary lymphoid organ essential for the maintenance of central tolerance through the selection and elimination of autoreactive T cells. Precursors of T cells, originating from the bone marrow, reach the thymus and migrate from the thymic cortex to the medullary region. Thymic medullary epithelial cells (mTECs) express on their surface tissue-restricted antigens (TRAs) that represent autoantigens of all tissues in the body. Acting as a non-classical transcription factor in mTEC cells, the Autoimmune regulator (Aire) gene plays a role in the expression of TRAs, whose encoded protein releases the RNA polymerase II (RNA Pol II) anchored in the chromatin and regulates the expression of mRNAs in the thymus gland. The biological function of this gene is associated to the induction of central immune tolerance preventing the onset of autoimmune diseases. This is a result of negative selection of autoreactive thymocytes (T cell precursors) that interact physically with mTECs. Self-reactive thymocytes that recognize TRAs as foreign elements are eliminated by apoptosis. The co-culture of mTECs with thymocytes represents an appropriate in vitro model system to approximate the cellular interaction that occurs within the thymus. Previous results from our laboratory demonstrated that in addition to the control of TRA mRNAs, Aire also participates in the modulation of miRNAs in mTECs since these RNA species are transcribed by RNA Pol II. Continuing with this line of studies, in this study we demonstrate for the first time that Aire also modulates the expression of long non-coding RNAs (lncRNAs) in mTECs. For this, we used the loss-of-function strategy to analyze the expression of this RNA species, as well as mRNAs in mTEC Aire + / + or Aire knockout mTEC cells (KO Aire - / -) obtained by the gene editing by Crispr-Cas9. The transcriptome of these cells, whether or not adhered to thymocytes, was then analyzed by microarray hybridizations. This demonstrated that Aire and cell adhesion influence the expression of both mRNAs and lncRNAs. The reconstruction of lncRNAs-mRNAs interaction networks made possible to evidence a new post-transcriptional regulation pathway in mTEC cells.
209

World Automatic (((((The Really Real Project)))))

Mayer, Jonna January 2019 (has links)
In a prospective post-work society, a select group of citizens participate in a cross-temporal project. As we follow along their journeys, questions arise. How does time move when it is no longer a commodity? What is design in the age of rampant robotics? How do we define creativity? What is wasted potential, and is it possible to be a failure? Most importantly, can fiction teach us anything about reality?
210

Research towards the effective disruption of reproductive competence in Nile tilapia Oreochromis niloticus

Jin, Yehwa January 2018 (has links)
Reproductive containment in farmed fish is highly desired for sustainable aquaculture to prevent genetic introgression with wild conspecifics and enhance productivity by suppressing sexual maturation. A number of strategies have already been implemented or have been tested in commercially important fish (e.g. triploidy, monosexing, hormonal therapies); however, they either do not result in 100% containment, or they cannot be applied to all species. One promising new approach consists in disrupting primordial germ cells (PGCs), at the origin of germline cells, to induce sterility. The work carried out in this doctoral thesis aimed to investigate the genes involved in the survival of germ cells and subsequently conduct a functional analysis of candidate genes using CRISPR/Cas9 gene editing system to ultimately provide the basis for the development of a novel sterilisation technique. Nile tilapia was chosen as the experimental animal as it is a major aquaculture species worldwide and the control of reproduction plays a critical role in the farming productivity in this species. In addition, the species has clear advantages as its whole genome sequence is accessible, the generation time is relatively short and zygotes can be available all year round. Initially, a panel of 11 candidate genes with reported roles in survival of PGCs was investigated during the ontogenic development which led to the selection of piwi-like (piwil) gene as a target for genome editing. Then, high temperature was tested as a means to induce germ cell loss to better understand the mechanism underlying germ cell survival and apoptosis, and this study confirmed the functional importance of piwil genes in relation to germ cell loss and proliferation. In addition, the study suggested potential subfunctionalisation within the Bcl-2 gene family which requires further investigation. The next step aimed to optimise the CRISPR/Cas9 gene editing method by improving the microinjection system and testing different concentrations of sgRNAs. Over 95% of injected embryos showed on-target mutation in piwil2 via zygote injection of CRISPR/Cas9 reagents and complete KO larvae were shown in half of the mutants, producing putative sterile fish. However, there was no clear association between the phenotypes in PGCs and the mutation rate. Further comparative studies of mutant screening methods including T7E1, RGEN, HRMA, fragment analysis and NGS revealed that the genotypes of F0 are highly mosaic, suggesting that deep sequencing is recommended for accurate and high throughput F0 screening and further improvement for predictable genome editing is required for a reliable gene functional analysis in F0. In summary, the current thesis provided new scientific knowledge and supporting evidence for the use of the CRISPR/Cas9 gene editing platform to study gene function associated with sterility, with the ultimate goal to develop an alternative sterilisation method in fish.

Page generated in 0.0608 seconds