Identification and Functional Validation of Novel Foxp2 Regulatory Transcription Factors in Human Cell Lines

Total Page:16

File Type:pdf, Size:1020Kb

Identification and Functional Validation of Novel Foxp2 Regulatory Transcription Factors in Human Cell Lines IDENTIFICATION AND FUNCTIONAL VALIDATION OF NOVEL FOXP2 REGULATORY TRANSCRIPTION FACTORS IN HUMAN CELL LINES Quimbaya, Mauricio1 and España, Jina Marcela1 1Pontificia Universidad Javeriana Cali, Colombia ABSTRACT Different types of genetic analyzes based on individuals with different hereditary forms of speech disorders, such as verbal dyspraxia and some types of autism, have allowed the identification of specific genes closely associated with the disorders mentioned above. In humans, the transcription factor FOXP2 has been widely studied. Mutations in FOXP2 are directly associated with inherited verbal dyspraxia and speech disorders. However, there is little experimental evidence on which genes or transcription factors directly regulate FOXP2, from this point of view, it is essential to determine the transcriptional regulators that act upstream of FOXP2. The identification of new transcription factors associated with FOXP2 regulation will allow the construction of more holistic and inclusive networks, providing an important advance in the identification of molecular pathways involved in the establishment of neural circuit associated with the evolution of vertebrate communication skills. In the same way, the identification of specific regulatory elements that control FOXP2 expression could be of great importance for the appearance of new therapies to prevent or control nervous system development pathologies, specifically those related to speech and language. Here, four transcription factors were chose to evaluate their regulation function with FOXP2. POU3F2 and TBR1 showed great significance in the study since both confirmed a positive FOXP2 regulation and have special roles in the nervous system development. INTRODUCTION Language and communication are inherent characteristics of the human brain with great importance in the establishment of social relations. Complications during language and communication development relay on specific dysfunctions at neuronal and nervous system scales. These inabilities have not been extensively analyzed from a genetics perspective. During the 80s a case of study was followed in three generations within a single family showing verbal disorders. The genetic analysis identified a mutation in the FOXP2 gene, awakening the interest for many neuroscientists to study it thoroughly, trying to understand how only one particular gene might contribute to human communication. This approach opened a new era for language and communication understanding from a genetics perspective. Known as the communication gene, the transcription factor FOXP2 has been extensively studied in the nervous system development. FOXP2 expression is confined to specific brain regions and its expression is highly conserved in vertebrates. Numerous disruptions and alterations in this gene have been associated with deficits in human language development. Similarly, the understanding of FOXP2 control and signaling, has allowed the understanding of synaptic plasticity, the motor ability of mice and the mechanism of songbirds learning. The disorders associated with FOXP2 are due to 1) gene deletion (52% of the affected individuals), 2) specific variant sequences within the gene (~ 29%), 3) maternal uniparental disomy of chromosome 7 that reduces the expression of FOXP2 (~ 11%) and 4) structural variants of the gene (translocations and inversion) (~ 8%) [1]. One of the most common disorders associated with FOXP2 alterations is speech apraxia, characterized by disturbances in speech planning, affecting the production, pronunciation and timing of sounds, syllables and words [2]; apraxia that hinders the transformation of sounds into syllables, syllables into words and words into matching phrases, affects 10% of the population [3]. Other FOXP2 disorders include oral motor dyspraxia, which obstructs the planning of oral movements [4]; dysarthria, which interferes with the quality of the voice and affect the neuromuscular side of speech [5]; the difficulty in receiving and expressing the language (Watkins et al., 2002), deficiency in reading and writing [6]; the difficulty of developing fine motor skills [7] among others. The aim of the present research project is to propose a new genetic approach to identify and validate novel FOXP2 regulatory transcription factors. The identification of these regulatory elements will contribute to the generation of new information for the scientific community, being of high significance in the elucidation of new therapies to prevent pathologies associated with the nervous system development. THEORIC FRAME The 114076877-positional substitution in FOXP2 eighth intron, significantly affects the expression of the gene since it is located in a conserved sequence fundamental for POU3F2 binding, an important neural transcription factor [8]. The direct interaction between POU3F2 and FOXP2 is also supported by their expression pattern since both are exclusively expressed in postmitotic neurons, so it is possible to assume that FOXP2 regulation by POU3F2 occurs precisely in neuronal tissue [8]. Similarly, Bao and collaborators [9] reported, that the CTCF protein binds upstream and downstream to the FOXP2 gene, moreover, Sakai and colleagues [10] and Deriziotis teamwork [11] reported in a protein interaction study based on autism disorders, that the transcription factor TBR1 interacts physically with FOXP2. It is important to highlight the importance of TBR1 in the development of the nervous system, since it is a neuron-specific transcription factor with key roles in nervous system patterning, regulating neuronal identity in cortical development, with numerous implications in the autism disorder [12]. Morelli and colleagues [13] reported a young woman case with severe language disorders who presented a chromosomal rearrangement involving an inversion of chromosome 7 that was translocated to chromosome 11. The karyotype of the young woman, together with the clinical history that included sever complications in neurodevelopment and language abilities, indicated that the different defects were probably associated with the FOXP2 gene. The breaking point was located at 7q31 position, more specifically 200 Kb downstream to the 3'UTR of FOXP2. In this region, numerous miRNA bind, influencing not only FOXP2 expression, but also, other genes such as ROBO2, a gene associated with pathologies of the nervous system, such as autism and dyslexia [14]. Additionally, it was possible to identify and characterize a localized regulatory element 205kb downstream FOXP2, which includes an open chromatin region with specific histone modifications, typical from an enhancer region, suggesting that the 7q31 breakpoint could interfere with FOXP2 expression. The same region has been also documented as the genomic binding site of transcription factors such as PPP1R3A, MDFIC and TFEC [15]. METHODS AND RESULTS 1. Identification of novel transcription factors, involved in nervous system development that might interact with FOXP2. For the selection of FOXP2 potential interactors, different filters were applied using four bioinformatics databases: MotifMap, Uniprot, COXPRESdb and Cytoscape. From the four, two of them were relevant in the study. Table 1. Transcription factors potentially associated with FOXP2 according to MotifMap database. Name Protein name Name Protein name Name Protein name Pancreas/duodenum Forkhead box protein TBR1 T-Box, Brain 1 ipf1 FOXF2 homeobox protein 1 F2 POU domain, class Homeobox protein Cdx-2 FOXD1 Forkhead box protein D1 POU3F2 3, transcription CDX-2 factor 2 ARP- Homeobox protein Pituitary homeobox HOXB9 FOXO1 Forkhead box protein O1 1(COUP- Hox-B9 2 TF2) Hepatocyte nuclear factor Homeobox protein HOXA9 Homeobox A9 HNF3beta NKX25 3-beta Nkx-2.5 TAL BHLH Homeobox protein TAL1 Transcription Factor Freac-2 Forkhead box protein F2 NKX22 Nkx-2.2 1 Forkhead box Homeobox protein FOXM1 Freac-4 Forkhead box protein D1 Nkx3-2 protein M1 Nkx-3.2 Homeobox protein Forkhead box protein Barx1 Meis1 Homeobox protein Meis1 Freac-3 BarH-like 1 C1 vasoconstriction gliogenesis muscle organ development generation of neurons vascular smooth muscle contraction nervous system development regulation of blood vessel size smooth muscle contraction skeletal system development vascular process in circulatory neurogenesis system organ development negative regulation of locomotion vasculature development blood circulation muscle hypertrophy neuron differentiation tissue development muscle contraction system development muscle structure development muscle tissue development circulatory system processmuscle system process blood vessel development regulation of locomotion cell differentiation blood vessel remodeling anatomical structure development system process cell development multicellular organismal process angiogenesis positive regulation of locomotion neuron development blood vessel morphogenesis neuron projection morphogenesis regulation of cell migration negative regulation of biological process cellular developmental process positive regulation of biological cell projection morphogenesis anatomical structure morphogenesis process positive regulation of cell migration multicellular organismal neuron projection development regulation of biological process development negative regulation of angiogenesis cell projection organization regulation of multicellular organismal process developmental process regulation of cell morphogenesis cellular process regulation of anatomical structure regulation of developmental process morphogenesis activation
Recommended publications
  • 1 Evidence for Gliadin Antibodies As Causative Agents in Schizophrenia
    1 Evidence for gliadin antibodies as causative agents in schizophrenia. C.J.Carter PolygenicPathways, 20 Upper Maze Hill, Saint-Leonard’s on Sea, East Sussex, TN37 0LG [email protected] Tel: 0044 (0)1424 422201 I have no fax Abstract Antibodies to gliadin, a component of gluten, have frequently been reported in schizophrenia patients, and in some cases remission has been noted following the instigation of a gluten free diet. Gliadin is a highly immunogenic protein, and B cell epitopes along its entire immunogenic length are homologous to the products of numerous proteins relevant to schizophrenia (p = 0.012 to 3e-25). These include members of the DISC1 interactome, of glutamate, dopamine and neuregulin signalling networks, and of pathways involved in plasticity, dendritic growth or myelination. Antibodies to gliadin are likely to cross react with these key proteins, as has already been observed with synapsin 1 and calreticulin. Gliadin may thus be a causative agent in schizophrenia, under certain genetic and immunological conditions, producing its effects via antibody mediated knockdown of multiple proteins relevant to the disease process. Because of such homology, an autoimmune response may be sustained by the human antigens that resemble gliadin itself, a scenario supported by many reports of immune activation both in the brain and in lymphocytes in schizophrenia. Gluten free diets and removal of such antibodies may be of therapeutic benefit in certain cases of schizophrenia. 2 Introduction A number of studies from China, Norway, and the USA have reported the presence of gliadin antibodies in schizophrenia 1-5. Gliadin is a component of gluten, intolerance to which is implicated in coeliac disease 6.
    [Show full text]
  • Detection of Interacting Transcription Factors in Human Tissues Using
    Myšičková and Vingron BMC Genomics 2012, 13(Suppl 1):S2 http://www.biomedcentral.com/1471-2164/13/S1/S2 PROCEEDINGS Open Access Detection of interacting transcription factors in human tissues using predicted DNA binding affinity Alena Myšičková*, Martin Vingron From The Tenth Asia Pacific Bioinformatics Conference (APBC 2012) Melbourne, Australia. 17-19 January 2012 Abstract Background: Tissue-specific gene expression is generally regulated by combinatorial interactions among transcription factors (TFs) which bind to the DNA. Despite this known fact, previous discoveries of the mechanism that controls gene expression usually consider only a single TF. Results: We provide a prediction of interacting TFs in 22 human tissues based on their DNA-binding affinity in promoter regions. We analyze all possible pairs of 130 vertebrate TFs from the JASPAR database. First, all human promoter regions are scanned for single TF-DNA binding affinities with TRAP and for each TF a ranked list of all promoters ordered by the binding affinity is created. We then study the similarity of the ranked lists and detect candidates for TF-TF interaction by applying a partial independence test for multiway contingency tables. Our candidates are validated by both known protein-protein interactions (PPIs) and known gene regulation mechanisms in the selected tissue. We find that the known PPIs are significantly enriched in the groups of our predicted TF-TF interactions (2 and 7 times more common than expected by chance). In addition, the predicted interacting TFs for studied tissues (liver, muscle, hematopoietic stem cell) are supported in literature to be active regulators or to be expressed in the corresponding tissue.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Genetic Variability in the Italian Heavy Draught Horse from Pedigree Data and Genomic Information
    Supplementary material for manuscript: Genetic variability in the Italian Heavy Draught Horse from pedigree data and genomic information. Enrico Mancin†, Michela Ablondi†, Roberto Mantovani*, Giuseppe Pigozzi, Alberto Sabbioni and Cristina Sartori ** Correspondence: [email protected] † These two Authors equally contributed to the work Supplementary Figure S1. Mares and foal of Italian Heavy Draught Horse (IHDH; courtesy of Cinzia Stoppa) Supplementary Figure S2. Number of Equivalent Generations (EqGen; above) and pedigree completeness (PC; below) over years in Italian Heavy Draught Horse population. Supplementary Table S1. Descriptive statistics of homozygosity (observed: Ho_obs; expected: Ho_exp; total: Ho_tot) in 267 genotyped individuals of Italian Heavy Draught Horse based on the number of homozygous genotypes. Parameter Mean SD Min Max Ho_obs 35,630.3 500.7 34,291 38,013 Ho_exp 35,707.8 64.0 35,010 35,740 Ho_tot 50,674.5 93.8 49,638 50,714 1 Definitions of the methods for inbreeding are in the text. Supplementary Figure S3. Values of BIC obtained by analyzing values of K from 1 to 10, corresponding on the same amount of clusters defining the proportion of ancestry in the 267 genotyped individuals. Supplementary Table S2. Estimation of genomic effective population size (Ne) traced back to 18 generations ago (Gen. ago). The linkage disequilibrium estimation, adjusted for sampling bias was also included (LD_r2), as well as the relative standard deviation (SD(LD_r2)). Gen. ago Ne LD_r2 SD(LD_r2) 1 100 0.009 0.014 2 108 0.011 0.018 3 118 0.015 0.024 4 126 0.017 0.028 5 134 0.019 0.031 6 143 0.021 0.034 7 156 0.023 0.038 9 173 0.026 0.041 11 189 0.029 0.046 14 213 0.032 0.052 18 241 0.036 0.058 Supplementary Table S3.
    [Show full text]
  • Roles of Id3 and IL-13 in a Mouse Model of Autoimmune Exocrinopathy
    Roles of Id3 and IL-13 in a Mouse Model of Autoimmune Exocrinopathy by Ian Lawrence Belle Department of Immunology Duke University Date:_______________________ Approved: ___________________________ Yuan Zhuang, Supervisor ___________________________ Michael Krangel, Chair ___________________________ Qi-jing Li ___________________________ Richard Lee Reinhardt ___________________________ Arno Greenleaf Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Immunology in the Graduate School of Duke University 2015 ABSTRACT Roles of Id3 and IL-13 in a Mouse Model of Autoimmune Exocrinopathy by Ian Lawrence Belle Department of Immunology Duke University Date:_______________________ Approved: ___________________________ Yuan Zhuang, Supervisor ___________________________ Michael Krangel, Chair ___________________________ Qi-jing Li ___________________________ Richard Lee Reinhardt ___________________________ Arno Greenleaf An abstract of a dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Immunology in the Graduate School of Duke University 2015 Copyright by Ian Lawrence Belle 2015 Abstract Within the field of immunology, the existence of autoimmune diseases presents a unique set of challenges. The immune system typically protects the host by identifying foreign pathogens and mounting an appropriate response to eliminate them. Great strides have been made in understanding how foreign pathogens are identified and responded to, leading to the development of powerful immunological tools, such as vaccines and a myriad of models used to study infectious diseases and processes. However, it is occasionally possible for host tissues themselves to be inappropriately identified as foreign, prompting an immune response that attempts to eliminate the host tissue. The immune system has processes in place, referred to as selection, designed to prevent the development of cells capable of recognizing the self as foreign.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Based Mechanism Study on the Beneficial Effects of Vitamin D
    www.nature.com/scientificreports OPEN Network Systems Pharmacology- Based Mechanism Study on the Benefcial Efects of Vitamin D against Psychosis in Alzheimer’s Disease Peihao Fan1, Xiguang Qi1, Robert A. Sweet2,3* & Lirong Wang1* Alzheimer’s disease (AD) is a chronic neurodegenerative disease with signifcant fnancial costs and negative impacts on quality of life. Psychotic symptoms, i.e., the presence of delusions and/ or hallucinations, is a frequent complication of AD. About 50% of AD patients will develop psychotic symptoms (AD with Psychosis, or AD + P) and these patients will experience an even more rapid cognitive decline than AD patients without psychosis (AD-P). In a previous analysis on medication records of 776 AD patients, we had shown that use of Vitamin D was associated with delayed time to psychosis in AD patients and Vitamin D was used more by AD-P than AD + P patients. To explore the potential molecular mechanism behind our fndings, we applied systems pharmacology approaches to investigate the crosstalk between AD and psychosis. Specifcally, we built protein-protein interaction (PPI) networks with proteins encoded by AD- and psychosis-related genes and Vitamin D-perturbed genes. Using network analysis we identifed several high-impact genes, including NOTCH4, COMT, CACNA1C and DRD3 which are related to calcium homeostasis. The new fndings highlight the key role of calcium-related signaling pathways in AD + P development and may provide a new direction and facilitate hypothesis generation for future drug development. Alzheimer’s disease (AD) is a chronic neurodegenerative disease commonly seen in the aging population, and the presence of AD is responsible for a signifcant decrease in the quality of life1.
    [Show full text]
  • Investigating the Role of the ETS Transcription Factor ELK1 in Stem Cell Transcription
    Investigating the role of the ETS transcription factor ELK1 in stem cell transcription A thesis submitted to the University of Manchester for the degree of Doctor of Philosophy in the Faculty of Biology, Medicine and Health 2017 Ian E. Prise Division of Molecular & Cellular Function School of Biological Sciences I. Table of Contents II. List of Figures ...................................................................................................................................... 5 III. Abstract .............................................................................................................................................. 7 IV. Declaration ......................................................................................................................................... 8 V. Copyright Statement ........................................................................................................................... 8 VI. Experimental Contributions ............................................................................................................... 9 VII. Acknowledgments .......................................................................................................................... 10 1. Introduction ...................................................................................................................................... 12 1.I Pluripotency ................................................................................................................................. 12 1.II Chromatin
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • The Cytokine FAM3B/PANDER Is an FGFR Ligand That Promotes Posterior Development in Xenopus
    The cytokine FAM3B/PANDER is an FGFR ligand that promotes posterior development in Xenopus Fangfang Zhanga,b,1, Xuechen Zhuc,d,1, Pan Wange,f,1, Qing Hea,b, Huimei Huangg, Tianrui Zhengf, Yongyu Lif, Hong Jiab, Linping Xub, Huaxiang Zhaoh, Gabriele Colozzai, Qinghua Taod,f,2, Edward M. De Robertisi,2, and Yi Dinga,b,2 aInstitute of Neuroscience, Translational Medicine Institute, Health Science Center, Xi’an Jiaotong University, 710061 Xi’an, China; bDepartment of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, 710061 Xi’an, China; cKey Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024 Hangzhou, China; dBeijing Advanced Innovation Center for Structural Biology, 100084 Beijing, China; eTsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China; fMinistry of Education (MOE) Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China; gDepartment of Nephrology, Xi’an Children’s Hospital, The Affiliated Children’s Hospital of Xi’an Jiaotong University, 710061 Xi’an, China; hDepartment of Orthodontics, College of Stomatology, Xi’an Jiaotong University, 710061 Xi’an, China; and iDepartment of Biological Chemistry, University of California, Los Angeles, CA 90095-1662 Contributed by Edward M. De Robertis, April 8, 2021 (sent for review January 7, 2021; reviewed by Enrique Amaya, Makoto Asashima, and Edgar M. Pera) Fibroblast growth factor (FGF)/extracellular signal-regulated ki- processes during vertebrate early embryogenesis, including gas- nase (ERK) signaling plays a crucial role in anterior–posterior trulation, mesoderm formation, and A–P axis specification (6).
    [Show full text]
  • Impact of the Transcription Factor Hoxa9 on Toll-Like Receptor-Mediated Innate Immune Responses and Development of Dendritic Cells and Macrophages
    From the Institut for Immunology Center for Infection, Inflammation, and Immunity Executive Director: Prof. Dr. Stefan Bauer of the Faculty of Medicine of the Philipps-Universität Marburg Impact of the Transcription Factor HoxA9 on Toll-like Receptor-Mediated Innate Immune Responses and Development of Dendritic Cells and Macrophages Inaugural-Dissertation For the Degree Doctor of Medicine Submitted to the Faculty of Medicine of the Philipps-Universität Marburg by Tobias Mischa Bleyl from Erlabrunn Marburg, 2018 Aus dem Institut für Immunologie Zentrum für Infektion, Entzündung und Immunität Geschäftsführender Direktor: Prof. Dr. Stefan Bauer des Fachbereichs Medizin der Philipps-Universität Marburg Einfluss des Transkriptionsfaktors HoxA9 auf Toll-like Rezeptor-vermittelte Reaktionen des angeborenen Immunsystems und die Entwicklung von dendritischen Zellen und Makrophagen Inaugural-Dissertation zur Erlangung des Doktorgrades der Humanmedizin dem Fachbereich Medizin der Philipps-Universität Marburg vorgelegt von Tobias Mischa Bleyl aus Erlabrunn Marburg, 2018 Angenommen vom Fachbereich Medizin der Philipps-Universität Marburg am: 10.07.2018 Gedruckt mit Genehmigung des Fachbereichs Dekan: Prof. Dr. Helmut Schäfer Referent: Prof. Dr. Stefan Bauer 1. Korreferentin: Prof. Dr. Adriana del Rey Dedicated to my beloved parents Karla & Fritz Bleyl and my wonderful wife Jessica Bleyl ABSTRACT Purpose of this work: Toll-like receptors (TLRs) are sensors of the innate immune system that perceive evolutionary conserved microbial structures and act as first line defense mechanisms against bacteria, viruses, fungi, and parasites. As a subset of the TLR family, intracellular TLRs reside in endosomal compartments to encounter their ligands comprising different types of nucleic acids. Important members are TLR3, 7, 8, and 9 recognizing double-stranded RNA (dsRNA), single-stranded RNA (ssRNA), again ssRNA, and unmethylated CpG-motif containing DNA, respectively.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]