Physical Extraction and Fast Protein Liquid Chromatography for Purifying Flagella Filament from Uropathogenic Escherichia Coli for Immune Assay

Total Page:16

File Type:pdf, Size:1020Kb

Physical Extraction and Fast Protein Liquid Chromatography for Purifying Flagella Filament from Uropathogenic Escherichia Coli for Immune Assay Bacterial Flagella as Triggers of the Innate Immune System and IL-10 Production During Acute Urinary Tract Infection Author Acharya, Dhruba Published 2020 Thesis Type Thesis (PhD Doctorate) School School of Medical Science DOI https://doi.org/10.25904/1912/3786 Copyright Statement The author owns the copyright in this thesis, unless stated otherwise. Downloaded from http://hdl.handle.net/10072/391068 Griffith Research Online https://research-repository.griffith.edu.au Bacterial Flagella as Triggers of the Innate Immune System and IL-10 Production During Acute Urinary Tract Infection Dhruba Acharya BSc. MLT, MSc. (Immunology) School of Medical Sciences Griffith University Submitted in fulfilment of the requirements of degree of Doctor of Philosophy June 2019 Abstract Urinary Tract Infections (UTIs) are a huge public health problem affecting 150 million people each year worldwide. Expenditures aimed at the management of UTI account for approximately $3.5 billion USD in medical costs annually. More than 80% of all UTIs are caused by uropathogenic Escherichia coli (UPEC). Studies have shown key roles for virulence factors of UPEC, such as flagella, autotransporters, capsule, fimbriae, toxins, lipopolysaccharide (LPS), and siderophores, in UTI disease pathogenesis. UPEC flagella contribute to the pathogenesis of UTI in several ways, such as through flagella-mediated motility, colonization, adhesion, and biofilm formation. The flagella filament is synthesized as a polymerized product of >20,000 protein monomers termed flagellin or FliC (encoded by the gene fliC). The contribution of flagella to innate immune stimulation is only partially defined, for example gaps exist in our understanding of how flagella engage host responses in the bladder during acute UTI. Extracellular FliC is recognized by Toll-Like Receptor 5 (TLR5), whereas intracellular FliC is recognized by Naip5 and Ipaf. Through MyD88-dependent and independent pathways, FliC causes upregulation of various proinflammatory, regulatory cytokines, and growth factors. A detailed understanding of how FliC from UPEC engages innate immunity in the bladder during UTI is lacking. A major focus of this thesis was the regulatory cytokine, IL-10. IL-10 plays pleiotropic roles in defence against infection; depending on the illness and the causal pathogen. Frequently, IL-10 facilitates immune suppression to moderate inflammatory mechanisms that can damage the host. IL-10 is rapidly produced in the bladder in response to UPEC early during UTI. However, the mechanism of engagement of innate immunity by UPEC that leads to elicitation of IL-10 in the bladder is unknown. One facet in understanding the role of IL-10 in infectious disease is the elucidation of microbial products that elicit production of this key regulator of host innate immune responses. The overarching aim of this thesis was to define the role of flagella (particularly FliC) in immune modulation during acute UTI. A plethora of methods have been previously described for the extraction and purification of FliC. However, there is limited literature utilizing highly purified, non-denatured FliC, offering any insight into host-pathogen interactions. In this study, UPEC CFT073, and a derivative strain containing four mutations in genes encoding fimbriae and pili (CFT073Δ4) were used to generate a deletion in the flagellar filament, fliC. This enabled the concurrent preparation of a iii suitable carrier control to be applied in downstream assays. The flhDC gene, which regulates flagella biosynthesis, was used in an IPTG inducible plasmid (pMG600) to overexpress flagella. Extraction and purification of FliC to homogeneity was performed using a sequential extraction method based on mechanical shearing, ultracentrifugation, size exclusion chromatography, protein concentration and endotoxin removal. Protein purity and integrity was assessed using SDS-PAGE, western blots with anti-flagellin antisera, and native-PAGE. This study established a new, carefully optimized method to extract and purify FliC from the reference UPEC strain CFT073 to be suitable for immune assays. The biological activity of FliC was assessed by measuring cytokine responses in both mouse macrophages (J774A.1) and human monocytes(U937). Macrophage and monocyte responses to purified FliC included significant levels of several cytokines, including TNF-α, IL-6, IL12, INF-γ, consistent with prior literature reports. This thesis sought to identify the role of the major UPEC flagella filament protein, FliC, in triggering IL-10 synthesis in the bladder. Analysis of IL-10 induction in response to various UPEC CFT073 derivatives and purified FliC showed a role for FliC in the induction of IL-10 in both human and mouse cell cultures. The effect of FliC on other cytokines, chemokines, and growth factors indicated several pro-inflammatory, and cell recruitment/ migratory factors are also shown to be induced by host cells in response to this protein. This study also explored the FliC induced immune responses, including IL- 10 induction in the bladder in vivo. ELISA and multiplex assays revealed that FliC, when injected via transurethral catheterization, induced a rapid IL-10 response in the mouse bladder. Characterization of the genome-wide innate immunological context of FliC- induced cytokines in the bladder using RNA-sequencing also identified 1400-gene network of transcriptional and antibacterial defences, that were differently expressed compared to carrier control. Of the FliC-responsive bladder transcriptome, the changes in expression of il10 and other genes were dependent on TLR5, according to the -/- comparative analysis of TLR5-deficient mice (Tlr5 ). Exploration of the potential of FliC, and its associated innate immune signature in the bladder, revealed a significant benefit for the control of infection in the mice that received purified FliC, prophylactically or therapeutically after transurethral UPEC infection. Collectively, this thesis shows that detection of FliC through TLR5 triggers rapid IL-10 synthesis in the bladder, and FliC therefore represents a potential immune modulator for the treatment or prevention of UTI. iv Statement of Originality This work has not previously been submitted for a degree or diploma in any university. To the best of my knowledge and belief, the thesis contains no material previously published or written by another person except where due reference is made in the thesis itself. Professional English language editing of this thesis was completed by a professional scientific editing company called Scribendi (www.scribendi.com). Thesis editing was done in accordance with the Griffith University HDR policy, ‘Guidelines for editing research thesis’. _____________ Dhruba Acharya v Acknowledgement of Assistance I am profoundly grateful to my primary supervisor, Professor Glen C. Ulett, for the continuous support during my PhD study. I appreciate his advice, motivation, enthusiasm, and guidance to develop and advance my research through different stages of this project. I would like to acknowledge my co-supervisor Dr. Matthew J. Sullivan for his constant guidance from bench to book. He has been a true mentor and a wonderful friend of mine throughout this degree. I would like to express my deepest gratitude to Dr. Harry Sakellaris, without whom, I would not have started my career in Australia. Many thanks to Professor Mark Schembri, (The University of Queensland) for his intellectual advice to shape and plan my experiments. I appreciate his insightful contribution in manuscript writing. To the fellow past and present members of my lab, Dr. Deepak S. Ipe, Dr. Kelvin Goh, Dean Gosling, Lahiru Katupitiya, Michelle Chamoun, and Darren Prince. They are all amazing colleagues and friends and I would like to thank them for creating an excellent environment around working stations. Dr. Prabin Gyawali; Heartfelt thanks to you brother for being there in my highs and lows, thick and thin, and pleasure and setbacks. I am deeply indebted to these beautiful souls: Dr. Narayan Gyawali and family, Nirajan Shrestha, Sabina Shrestha, Bimbishar Bhattarai, Shikha Karki, Indra Shrestha, Chandani Shrestha, Manasvi and Shriyans Shrestha. I would like to remember my father Late Shree Jhabindra Acharya. I take this opportunity to acknowledge my mother Saraswoti Devi Acharya and rest of “The Acharya’s”: Chandra Prakash Acharya, Bhawana Thapaliya Acharya, Emil and Milee Acharya, Narendra Acharya, Archana Pokharel Acharya, Nevan and Niyan Acharya. “My soulmate Kabita Kuikel Acharya, this would not have been possible without your support, love and care. You are the best thing that has ever happened in my life and our shared DNA, Adhvik Acharya, the greatest gift of all”. Finally, I would like to thank the Government of Australia and Griffith University for the scholarship, support and the people of Australia for giving us a home away from home. vi Table of Contents Abstract ...................................................................................................................... iii Statement of Originality ............................................................................................. v Acknowledgement of Assistance ................................................................................ vi Table of Contents ...................................................................................................... vii List of Figures ...........................................................................................................
Recommended publications
  • Transcriptome Analyses of Tumor-Adjacent Somatic Tissues Reveal Genes Co-Expressed with Transposable Elements Nicky Chung1†, G
    Chung et al. Mobile DNA (2019) 10:39 https://doi.org/10.1186/s13100-019-0180-5 RESEARCH Open Access Transcriptome analyses of tumor-adjacent somatic tissues reveal genes co-expressed with transposable elements Nicky Chung1†, G. M. Jonaid1†, Sophia Quinton1†, Austin Ross1†, Corinne E. Sexton1, Adrian Alberto2, Cody Clymer2, Daphnie Churchill2, Omar Navarro Leija 2 and Mira V. Han1,3* Abstract Background: Despite the long-held assumption that transposons are normally only expressed in the germ-line, recent evidence shows that transcripts of transposable element (TE) sequences are frequently found in the somatic cells. However, the extent of variation in TE transcript levels across different tissues and different individuals are unknown, and the co-expression between TEs and host gene mRNAs have not been examined. Results: Here we report the variation in TE derived transcript levels across tissues and between individuals observed in the non-tumorous tissues collected for The Cancer Genome Atlas. We found core TE co-expression modules consisting mainly of transposons, showing correlated expression across broad classes of TEs. Despite this co-expression within tissues, there are individual TE loci that exhibit tissue-specific expression patterns, when compared across tissues. The core TE modules were negatively correlated with other gene modules that consisted of immune response genes in interferon signaling. KRAB Zinc Finger Proteins (KZFPs) were over-represented gene members of the TE modules, showing positive correlation across multiple tissues. But we did not find overlap between TE-KZFP pairs that are co-expressed and TE-KZFP pairs that are bound in published ChIP-seq studies.
    [Show full text]
  • EVALUATION of BMP2/Mirna CO-EXPRESSION SYSTEMS for POTENT THERAPEUTIC EFFICACY in BONE-TISSUE REGENERATION
    EuropeanTK Brenner Cells et al. and Materials Vol. 41 2021 (pages 245-268) DOI: Non-viral 10.22203/eCM.v041a18 BMP2/miRNA co-expression ISSN 1473-2262 systems EVALUATION OF BMP2/miRNA CO-EXPRESSION SYSTEMS FOR POTENT THERAPEUTIC EFFICACY IN BONE-TISSUE REGENERATION T.K. Brenner1,§, K. Posa-Markaryan1,§, D. Hercher1,4, S. Sperger1,4, P. Heimel1,4, C. Keibl1, S. Nürnberger1,2,3,4, J. Grillari1,4,5, H. Redl1,4 and A. Hacobian1,4,* 1 Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Centre, The Austrian Cluster for Tissue Regeneration, European Institute of Excellence on Tissue Engineering and Regenerative Medicine Research (Expertissues EEIG), Donaueschingenstrasse 13, 1200 Vienna, Austria 2 Department of Orthopaedics and Trauma-Surgery, Division of Trauma-Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria 3 University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria 4 Austrian Cluster for Tissue Engineering 5 Institute for Molecular Biotechnology, Department of Biotechnology, BOKU – University of Natural Resources and Life Sciences, Vienna, Austria § These authors contributed equally to this work Abstract Reconstruction of bone defects and compensation of deficient repair mechanisms represent important goals within the field of regenerative medicine and require novel safe strategies for translation into the clinic. A non-viral osteogenic gene therapeutic vector system (‘hybrid vectors’) was generated, combining an improved bone morphogenetic protein 2 (BMP2) gene cassette and single pro-osteogenic microRNAs (miR-148b-3p, miR-20-5p, miR-590b-5p), driven by the U6 promoter. The vectors were tested in vitro for their osteogenic differentiation potential in C2C12 and C3H/10T1/2 cell lines, usingBMP2 alone as a control.
    [Show full text]
  • Reference Transcriptomes of Porcine Peripheral Immune Cells Created Through Bulk and Single-Cell RNA Sequencing
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.02.438107; this version posted April 4, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. 1 Reference transcriptomes of porcine peripheral immune cells created through bulk and 2 single-cell RNA sequencing 3 4 Juber Herrera-Uribe1†, Jayne E. Wiarda2,3,4†, Sathesh K. Sivasankaran2,5, Lance Daharsh1, Haibo 5 Liu1, Kristen A. Byrne2, Timothy P.L .Smith6, Joan K. Lunney7, CrystaL L. Loving2‡*, 6 Christopher K. Tuggle1‡* 7 8 1 Department of AnimaL Science, Iowa State University, Ames, IA, USA. 9 2 Food Safety and Enteric Pathogens Research Unit, NationaL AnimaL Disease Center, 10 AgriculturaL Research Service, United States Department of Agriculture, Ames, IA, USA 11 3 Immunobiology Graduate Program, Iowa State University, Ames, IA, USA 12 4 Oak Ridge Institute for Science and Education, AgriculturaL Research Service Participation 13 Program, Oak Ridge, TN, USA 14 5 Genome Informatics FaciLity, Iowa State University, Ames, IA, USA 15 6 USDA, ARS, U.S. Meat AnimaL Research Center, Clay Center, Nebraska, USA 16 7 USDA-ARS, BeLtsviLLe AgriculturaL Research Center, AnimaL Parasitic Diseases Laboratory, 17 BeLtsviLLe, MD, USA. 18 † These authors have contributed equaLLy to this work and share first authorshiP 19 ‡ These authors have contributed equaLLy to this work and share senior and last authorshiP 20 *Correspondence: 21 Corresponding authors: [email protected], [email protected] 22 23 Keywords: Pig, immune ceLLs, transcriptome, Single-ceLL RNA-seq, bulkRNA-seq, FAANG.
    [Show full text]
  • The Changing Chromatome As a Driver of Disease: a Panoramic View from Different Methodologies
    The changing chromatome as a driver of disease: A panoramic view from different methodologies Isabel Espejo1, Luciano Di Croce,1,2,3 and Sergi Aranda1 1. Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain 2. Universitat Pompeu Fabra (UPF), Barcelona, Spain 3. ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain *Corresponding authors: Luciano Di Croce ([email protected]) Sergi Aranda ([email protected]) 1 GRAPHICAL ABSTRACT Chromatin-bound proteins regulate gene expression, replicate and repair DNA, and transmit epigenetic information. Several human diseases are highly influenced by alterations in the chromatin- bound proteome. Thus, biochemical approaches for the systematic characterization of the chromatome could contribute to identifying new regulators of cellular functionality, including those that are relevant to human disorders. 2 SUMMARY Chromatin-bound proteins underlie several fundamental cellular functions, such as control of gene expression and the faithful transmission of genetic and epigenetic information. Components of the chromatin proteome (the “chromatome”) are essential in human life, and mutations in chromatin-bound proteins are frequently drivers of human diseases, such as cancer. Proteomic characterization of chromatin and de novo identification of chromatin interactors could thus reveal important and perhaps unexpected players implicated in human physiology and disease. Recently, intensive research efforts have focused on developing strategies to characterize the chromatome composition. In this review, we provide an overview of the dynamic composition of the chromatome, highlight the importance of its alterations as a driving force in human disease (and particularly in cancer), and discuss the different approaches to systematically characterize the chromatin-bound proteome in a global manner.
    [Show full text]
  • Signatures of Adaptive Evolution in Platyrrhine Primate Genomes 5 6 Hazel Byrne*, Timothy H
    1 2 Supplementary Materials for 3 4 Signatures of adaptive evolution in platyrrhine primate genomes 5 6 Hazel Byrne*, Timothy H. Webster, Sarah F. Brosnan, Patrícia Izar, Jessica W. Lynch 7 *Corresponding author. Email [email protected] 8 9 10 This PDF file includes: 11 Section 1: Extended methods & results: Robust capuchin reference genome 12 Section 2: Extended methods & results: Signatures of selection in platyrrhine genomes 13 Section 3: Extended results: Robust capuchins (Sapajus; H1) positive selection results 14 Section 4: Extended results: Gracile capuchins (Cebus; H2) positive selection results 15 Section 5: Extended results: Ancestral Cebinae (H3) positive selection results 16 Section 6: Extended results: Across-capuchins (H3a) positive selection results 17 Section 7: Extended results: Ancestral Cebidae (H4) positive selection results 18 Section 8: Extended results: Squirrel monkeys (Saimiri; H5) positive selection results 19 Figs. S1 to S3 20 Tables S1–S3, S5–S7, S10, and S23 21 References (94 to 172) 22 23 Other Supplementary Materials for this manuscript include the following: 24 Tables S4, S8, S9, S11–S22, and S24–S44 1 25 1) Extended methods & results: Robust capuchin reference genome 26 1.1 Genome assembly: versions and accessions 27 The version of the genome assembly used in this study, Sape_Mango_1.0, was uploaded to a 28 Zenodo repository (see data availability). An assembly (Sape_Mango_1.1) with minor 29 modifications including the removal of two short scaffolds and the addition of the mitochondrial 30 genome assembly was uploaded to NCBI under the accession JAGHVQ. The BioProject and 31 BioSample NCBI accessions for this project and sample (Mango) are PRJNA717806 and 32 SAMN18511585.
    [Show full text]
  • SETDB1-Mediated Silencing of Retroelements
    viruses Review SETDB1-Mediated Silencing of Retroelements Kei Fukuda * and Yoichi Shinkai * Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako 351-0198, Japan * Correspondence: [email protected] (K.F.); [email protected] (Y.S.) Received: 7 May 2020; Accepted: 28 May 2020; Published: 30 May 2020 Abstract: SETDB1 (SET domain bifurcated histone lysine methyltransferase 1) is a protein lysine methyltransferase and methylates histone H3 at lysine 9 (H3K9). Among other H3K9 methyltransferases, SETDB1 and SETDB1-mediated H3K9 trimethylation (H3K9me3) play pivotal roles for silencing of endogenous and exogenous retroelements, thus contributing to genome stability against retroelement transposition. Furthermore, SETDB1 is highly upregulated in various tumor cells. In this article, we describe recent advances about how SETDB1 activity is regulated, how SETDB1 represses various types of retroelements such as L1 and class I, II, and III endogenous retroviruses (ERVs) in concert with other epigenetic factors such as KAP1 and the HUSH complex and how SETDB1-mediated H3K9 methylation can be maintained during replication. Keywords: SETDB1; heterochromatin; H3K9me3 1. Introduction Transposable elements (TEs) comprise more than 40% of most extant mammalian genomes [1,2]. Among these, retroelements including short/long interspersed elements (SINEs/LINEs) and endogenous retroviruses (ERVs) are still active and capable of retrotransposition [3,4]. Although retrotransposition contributes to genome diversification and evolution, it can cause genome instability, insertional mutagenesis, or transcriptional perturbation and is often deleterious to host species [5–7]. Therefore, evolution has also driven the development of multiple defense mechanisms against retrotransposition. The first line of defense is transcriptional silencing of integrated retroelements, using various epigenetic modifications, such as DNA methylation and histone H3 lysine 9 tri-methylation (H3K9me3) [8,9].
    [Show full text]
  • Opposite Roles of BAP1 in Overall Survival of Uveal Melanoma and Cutaneous Melanoma
    Title: Opposite roles of BAP1 in overall survival of uveal melanoma and cutaneous melanoma Feng Liu-Smith Department of Epidemiology, Department of Medicine, Chao Family Comprehensive Cancer Center, School of Population Health, University of California Irvine, Irvine, CA 92697 Email: [email protected] Phone:949-824-2778 Running title: BAP1 in CM and UM Abstract Background: BAP1 germline mutations predispose individuals to a number of cancer types including uveal melanoma (UM) and cutaneous melanoma (CM) which are distinctively different in the oncogenic pathways. BAP1 loss was common in UM and was associated with a worse prognosis. BAP1 loss was rare in CM and the outcome was unclear. Methods: This study used TCGA UM and CM databases for survival analysis for patients with different BAP1 status and mRNA expression levels. Cox regression model was used for adjusting to known prognosis factors. Results: BAP1- (loss or low expression) predicted a poor overall survival in UM (Cox HR = 0.062, logrank p =0.007) but a contrasting better overall survival in CM (HR = 1.69, p =0.009). Multi-covariate Cox regression analysis indicated BAP1 was a significant predictor for overall survival after adjusting for age of diagnosis, presence of ulceration, Breslow depth and CM stages in patients older than 50 years but not in younger patients. Co-expression analysis revealed no shared genes in BAP1 altered UM and CM tumors, further supporting a completely distinctive role of BAP1 in CM and UM. Conclusions: low BAP1 mRNA was significantly associated with a better overall survival in CM patients, in sharp contrast to its tumor suppressor role in UM where low or loss of BAP1 indicated a worse overall survival.
    [Show full text]
  • Predicting Informative Spatio-Temporal Neurodevelopmental Windows and Gene Risk for Autism Spectrum Disorder
    PREDICTING INFORMATIVE SPATIO-TEMPORAL NEURODEVELOPMENTAL WINDOWS AND GENE RISK FOR AUTISM SPECTRUM DISORDER. a thesis submitted to the graduate school of engineering and science of bilkent university in partial fulfillment of the requirements for the degree of master of science in computer engineering By O˘guzhanKarakahya October 2020 Predicting informative spatio-temporal neurodevelopmental windows and gene risk for autism spectrum disorder. By O˘guzhanKarakahya October 2020 We certify that we have read this thesis and that in our opinion it is fully adequate, in scope and in quality, as a thesis for the degree of Master of Science. A. Erc¨ument C¸i¸cek(Advisor) Can Alkan Tunca Do˘gan Approved for the Graduate School of Engineering and Science: Ezhan Kara¸san Director of the Graduate School ii ABSTRACT PREDICTING INFORMATIVE SPATIO-TEMPORAL NEURODEVELOPMENTAL WINDOWS AND GENE RISK FOR AUTISM SPECTRUM DISORDER. O˘guzhanKarakahya M.S. in Computer Engineering Advisor: A. Erc¨ument C¸i¸cek October 2020 Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder with a strong genetic basis. Due to its intricate nature, only a fraction of the risk genes were identified despite the effort spent on large-scale sequencing studies. To perceive underlying mechanisms of ASD and predict new risk genes, a deep learning architecture is designed which processes mutational burden of genes and gene co-expression networks using graph convolutional networks. In addition, a mixture of experts model is employed to detect specific neurodevelopmental periods that are of particular importance for the etiology of the disorder. This end-to-end trainable model produces a posterior ASD risk probability for each gene and learns the importance of each network for this prediction.
    [Show full text]
  • Resf1 Supports Embryonic Stem Cell Self-Renewal and Effective Germline Entry
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.25.445589; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Resf1 supports ESC self-renewal and germline entry Resf1 supports embryonic stem cell self-renewal and effective germline entry Matúš Vojtek1 and Ian Chambers1 1Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, Scotland Correspondence: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.05.25.445589; this version posted May 25, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Resf1 supports ESC self-renewal and germline entry Summary Retroelement silencing factor 1 (Resf1) interacts with the key regulators of mouse embryonic stem cells (ESCs) Oct4 and Nanog, and its absence results in sterility of mice. However, the function of Resf1 in ESCs and germ line specification is poorly understood. In this study, we used Resf1 knockout cell lines to determine the requirements of RESF1 for ESCs self- renewal and for in vitro specification of ESCs into primordial germ cell-like cells (PGCLCs). We found that deletion of Resf1 in ESCs cultured in serum and LIF reduces self-renewal potential whereas episomal expression of RESF1 has a modest positive effect on ESC self- renewal.
    [Show full text]
  • Centro De Investigación Y De Estudios Avanzados Del
    CENTRO DE INVESTIGACIÓN Y DE ESTUDIOS AVANZADOS DEL INSTITUTO POLITÉCNICO NACIONAL UNIDAD ZACATENCO DEPARTAMENTO DE BIOMEDICINA MOLECULAR “La mutación A431E en PSEN1 causante de Enfermedad de Alzheimer Familiar altera el perfil proteómico de las células mesenquimales del epitelio olfatorio” T E S I S Que presenta Lory Jhenifer Rochín Hernández Para obtener el grado de MAESTRA EN CIENCIAS EN LA ESPECIALIDAD DE BIOMEDICINA MOLECULAR Director de la Tesis: Dr. Marco Antonio Meraz Ríos Ciudad de México Agosto, 2019. AGRADECIMIENTOS Primero que nada, me gustaría agradecer a mi familia, como se los he dicho siempre; mis logros serán sus logros, ya que sin ustedes esto no sería posible. A mis padres, Rodolfo y Gloria, quienes han sacrificado gran parte de su vida por educarnos y guiarnos, muchas gracias por todo, pero principalmente por su amor incondicional, soy muy afortunada de tenerlos como padres. A mis hermanos, por ser un gran ejemplo para mí; Rody gracias por enseñarme a ser perseverante y luchar por tus objetivos. A Fany, mi persona favorita, gracias por existir y estar siempre a mi lado, si no hubiera sido por ti yo no habría intentado hacer la maestría, gracias por darme la fuerza y apoyo necesarios para lograrlo eres mi máximo orgullo y ejemplo, simplemente lo mejor de mi vida. A mi director de tesis, el Dr. Marco Antonio Meraz Ríos, por permitirme ser parte de su grupo de trabajo, por depositar su confianza en mí, por su apoyo, paciencia, tiempo y enseñanzas, pero sobre todo por motivarme a seguir adelante y a ser mejor cada día, ¡lo quiero mucho! A la IBt.
    [Show full text]
  • Dfschmidt.Pdf
    AVALIAÇÃO DE MECANISMOS ENVOLVIDOS NA TRANSFERÊNCIA DA RESISTÊNCIA A 5-FU MEDIADOS POR VESÍCULAS EXTRACELULARES EM ADENOCARCINOMA GÁSTRICO DAYANE DE FÁTIMA SCHMIDT Dissertação apresentada à Fundação Antônio Prudente para obtenção de Título de Mestre em Ciências Área de concentração: Oncologia Orientadora: Dra. Vilma Regina Martins São Paulo 2020 FICHA CATALOGRÁFICA Preparada pelo Ensino Apoio ao aluno da Fundação Antônio Prudente* S349 Schmidt, Dayane de Fátima Avaliação de mecanismos envolvidos na transferência de resistência a 5-FU mediados por vesículas extracelulares em adenocarcinomas gástrico / Dayane de Fátima Schmidt – São Paulo, 2020. 75p. Dissertação (Mestrado)-Fundação Antônio Prudente. Curso de Pós-Graduação em Ciências - Área de concentração: Oncologia. Orientadora: Vilma Regina Martins Descritores: 1. Câncer Gástrico/Gastric Cancer . 2. Vesículas Extracelulares/Extracellular Vesicles. 3. Resistência a Quimioterapia/ Chemotherapy Resistance. 4. 5-fluoracil/5-fluorouracil. 5. Fascina/Fascin. Elaborado por Suely Francisco CRB 8/2207 *Todos os direitos reservados à FAP. A violação dos direitos autorais constitui crime, previsto no art. 184 do Código Penal, sem prejuízo de indenizações cabíveis, nos termos da Lei nº 9.610/08 SUPORTE À PESQUISA POR AGÊNCIA DE FOMENTO Este trabalho recebeu apoio da Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP), através de auxilio à Pesquisa - processo número 2014/50943-1. AGRADECIMENTOS Em primeiro lugar, à meus pais pelo apoio incondicional que foi essencial para tornar possível a realização deste trabalho. Agradeço por me incentivarem a alcançar meus objetivos, pela compreensão e por sempre estarem presentes mesmo distantes fisicamente; À Dra. Vilma Regina Martins, pela oportunidade e por tudo que me ensinou durante o desenvolvimento deste trabalho.
    [Show full text]
  • A CRISPR Knockout Screen Identifies SETDB1-Target Retroelement Silencing Factors in Embryonic Stem Cells
    Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Research A CRISPR knockout screen identifies SETDB1-target retroelement silencing factors in embryonic stem cells Kei Fukuda,1 Akihiko Okuda,2 Kosuke Yusa,3 and Yoichi Shinkai1 1Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; 2Division of Developmental Biology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane Hidaka Saitama 350-1241, Japan; 3Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom In mouse embryonic stem cells (mESCs), the expression of provirus and endogenous retroelements is epigenetically re- pressed. Although many cellular factors involved in retroelement silencing have been identified, the complete molecular mechanism remains elusive. In this study, we performed a genome-wide CRISPR screen to advance our understanding of retroelement silencing in mESCs. The Moloney murine leukemia virus (MLV)–based retroviral vector MSCV-GFP, which is repressed by the SETDB1/TRIM28 pathway in mESCs, was used as a reporter provirus, and we identified more than 80 genes involved in this process. In particular, ATF7IP and the BAF complex components are linked with the repression of most of the SETDB1 targets. We characterized two factors, MORC2A and RESF1, of which RESF1 is novel molecule in retroelement silencing. Although both factors are recruited to repress provirus, their roles in repression are different. MORC2A appears to function dependent on repressive epigenetic modifications, while RESF1 regulates repressive epigenet- ic modifications associated with SETDB1. Our genome-wide CRISPR screen cataloged genes which function at different levels in silencing of SETDB1-target retroelements and provides a useful resource for further molecular studies.
    [Show full text]