Identification of Potential Proteases for Abdominal Aortic Aneurysm by Weighted Gene Coexpression Network Analysis

Total Page:16

File Type:pdf, Size:1020Kb

Identification of Potential Proteases for Abdominal Aortic Aneurysm by Weighted Gene Coexpression Network Analysis Genome Identification of potential proteases for abdominal aortic aneurysm by weighted gene coexpression network analysis Journal: Genome Manuscript ID gen-2020-0041.R1 Manuscript Type: Article Date Submitted by the 28-Jun-2020 Author: Complete List of Authors: Zhang, Hui; Peking Union Medical College Hospital, Department of Vascular Surgery Yang, Dan; Chinese Academy of Medical Sciences and Peking Union Medical College, Department of Computational Biology and Bioinformatics,Draft Institute of Medicinal Plant Development Chen, Siliang; Peking Union Medical College Hospital, Department of Vascular Surgery Li, Fangda; Peking Union Medical College Hospital, Department of Vascular Surgery Cui, Liqiang; Peking Union Medical College Hospital, Department of Vascular Surgery Liu, Zhili; Peking Union Medical College Hospital, Department of Vascular Surgery Shao, Jiang; Peking Union Medical College Hospital, Department of Vascular Surgery Chen, Yuexin; Peking Union Medical College Hospital, Department of Vascular Surgery Liu, Bao; Peking Union Medical College Hospital, Department of Vascular Surgery Zheng, Yuehong; Peking Union Medical College Hospital, Department of Vascular Surgery Abdominal aortic aneurysm, next-generation sequencing, WGCNA, Keyword: proteases, matrix metalloproteinase Is the invited manuscript for consideration in a Special Not applicable (regular submission) Issue? : https://mc06.manuscriptcentral.com/genome-pubs Page 1 of 35 Genome 1 Identification of potential proteases for abdominal aortic aneurysm by weighted gene 2 coexpression network analysis 3 Short title: WGCNA identifies crucial proteases in AAA 4 5 Hui Zhang1, Dan Yang2, Siliang Chen1, Fangda Li1, Liqiang Cui1, Zhili Liu1, Jiang Shao1, Yuexin 6 Chen1, Bao Liu1, Yuehong Zheng1. 7 1Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing 100730, PR 8 China; 2Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant 9 Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 10 100730, PR China. 11 Correspondence: Draft 12 Yuehong Zheng, MD, Department of Vascular Surgery, Peking Union Medical College Hospital, 13 Shuaifuyuan #1, Dongcheng District, Beijing 100730, China. 14 Phone #: +8613811015811 15 Email: [email protected] 1 https://mc06.manuscriptcentral.com/genome-pubs Genome Page 2 of 35 16 Abstract 17 Proteases are involved in the degradation of the extracellular matrix, which contributes to the 18 formation of abdominal aortic aneurysm (AAA). To identify new disease targets in addition to the 19 results of previous microarray studies, we performed next-generation sequencing (NGS) of the 20 whole transcriptome of Angiotensin II-treated Apo E-/- male mice (n=4) and control mice (n=4) to 21 obtain differentially expressed genes (DEGs). Identified DEGs of proteases were analyzed using 22 weighted gene coexpression network analysis (WGCNA). RT-qPCR was conducted to validate 23 the differential expression of selected hub genes. We found that 43 DEGs were correlated with 24 the expression of the protease profile, and most were clustered in immune response module. 25 Among 26 hub genes, we found that Mmp16 and Mmp17 were significantly downregulated in AAA 26 mice, while Ctsa, Ctsc and Ctsw were Draftupregulated. Our functional annotation analysis of genes 27 coexpressed with the five hub genes indicated that Ctsw and Mmp17 were involved in T cell 28 regulation and Cell adhesion molecule pathway, respectively, and that both were involved in 29 general regulation of the cell cycle and gene expression. Overall, our data suggest that these 30 ectopic genes are potentially crucial to AAA formation and may act as biomarkers for the diagnosis 31 of AAA. 32 Key words: Abdominal aortic aneurysm, next-generation sequencing, transcriptome, WGCNA, 33 extracellular matrix, proteases, matrix metalloproteinase, cathepsin. 2 https://mc06.manuscriptcentral.com/genome-pubs Page 3 of 35 Genome 34 Introduction 35 Abdominal aortic aneurysm (AAA) is defined as a 50% increase in the diameter of the infrarenal 36 aorta and is an age-related degenerative disease(B Timothy, Terrin, & Dalman, 2008). The only 37 treatment option for AAA is surgical repair(Schermerhorn et al., 2008). It is necessary to explore 38 suitable noninvasive pharmacological treatments for this disease from a health economics 39 perspective. The pathogenesis of AAA formation, development and rupture is not fully understood. 40 Aortic wall inflammation is considered one of the major causes of AAA development and 41 progression(Goldstone, Malone, & Moore, 1978). After infiltration of immune inflammatory cells, 42 a wide range of immunoreactive mediators are released, which leads to direct degradation of the 43 aortic wall. Similarly, the development of intraluminal thrombi in the aortic wall is a source of 44 immune-derived proteases that exacerbateDraft pathogenesis(Jean-Baptiste et al., 2011). Collectively, 45 these events result in significant changes in the medial layer, including degradation of 46 extracellular matrix (ECM) structural proteins such as elastin and collagen fibers, which ultimately 47 lead to aortic wall expansion and rupture(Jean-Baptiste et al., 2011). 48 Proteases are known to be involved in AAA by degradation of the ECM during tissue 49 remodeling(Haiying, Sasaki, Jin, Kuzuya, & Cheng, 2018). The role of ECM proteases and their 50 role in AAA development has been extensively studied because they may serve as suitable 51 candidates for pharmacological treatments and can be specifically targeted without impairing 52 overall immunity. Key proteases that play pathological roles in AAA include elastase, chymase, 53 tryptase, matrix metalloproteinases (MMPs), cathepsins, granzyme B, etc.(Alon, Lisa S, & David 54 J, 2012). Elastases induce aneurysms via cleavage of elastin and destruction of elastic lamellae, 55 as well as generating proinflammatory elastin fragments (elastokines)(Folkesson, Silveira, 56 Eriksson, & Swedenborg, 2011). Chymases and tryptases can activate MMPs and cathepsins, 57 resulting in elastic lamellae fragmentation. They also promote immune infiltration and smooth 58 muscle cell apoptosis(Jiusong et al., 2009; Zhang, Sun, Lindholt, Sukhova, & Shi, 2011). MMPs 3 https://mc06.manuscriptcentral.com/genome-pubs Genome Page 4 of 35 59 are a large class of endopeptidases that are essential in physiological homeostasis and 60 remodeling processes in the ECM. Deciphering the specific contribution of ECM proteases may 61 provide valuable insights into the development of therapeutic approaches for AAA(Alon et al., 62 2012). There are supporting data that MMP-1, 2, 3, 9, 12, and 13 are increased in the aortic wall 63 of AAA(Rabkin, 2017). Cathepsins, or cysteine proteases, are a family of lysosomal proteases 64 that act effectively as elastolytic and collagenolytic enzymes and therefore play a significant role 65 in the degradation of the ECM(Schulte et al.; Sun et al.). Increased activity of cathepsins B, H, L, 66 and S has been detected in the aortic wall with AAA(Abisi et al., 2007). In addition, cathepsin C, 67 a protease responsible for activating zymogen granzyme, may also contribute to the development 68 of AAA by increasing the activation and release of granzyme and promoting the recruitment of 69 neutrophils to the affected aorta(Pagano et al., 2007). The data are weaker or insufficient for other 70 MMPs or cathepsins. Therefore, further Draftresearch is necessary to explore other possibly important 71 but neglected proteases. 72 The rapidly increasing availability of transcriptomics data generated by RNA sequencing (RNA- 73 seq) provides us with the opportunity to use this information to generate testable hypotheses to 74 understand the molecular mechanisms that control gene expression and biological processes. 75 With the latest developments in transcriptomics and next-generation sequencing technology, 76 coexpression networks constructed from RNA-seq data can also identify or infer the functional 77 status of genes from a systematic perspective(Sipko, Thomas, & Pedro, 2014). One way to infer 78 gene function and gene-disease associations from genome-wide gene expression is weighted 79 gene coexpression network analysis (WGCNA), which builds a gene network with a tendency to 80 coactivate across a group of samples and analyzes this network(van Dam, Võsa, van der Graaf, 81 Franke, & de Magalhães, 2018). The application of the above technology will help us discover 82 new AAA-related proteases and potential biomarkers associated with the diagnosis and treatment 83 of AAA. 4 https://mc06.manuscriptcentral.com/genome-pubs Page 5 of 35 Genome 84 In this study, we first identified novel genes that might underlie the aneurysm formation of AAA in 85 ApoE-/- mice via high-throughput or next-generation sequencing (NGS) of the transcriptome. Then 86 we explored the gene expression modules that correlate with protease activity in AAA progression 87 by WGCNA based on the RNA-seq data. Furthermore, we selected specific genes from protease 88 family for RT-qPCR analyses to examine whether the differences in gene expression associated 89 with aneurysm formation translated into similar changes in protein expression. 90 Materials and Methods 91 Animals and treatment 92 Male Apo E-/- mice were obtained from the Jackson Laboratory (Bar Harbor, ME). All mice were 93 bred as littermate controls and housedDraft in pathogen-free barrier cages. They were fed a normal 94 laboratory diet. Mice at 10-12 weeks
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]
  • SUPPLEMENTARY MATERIAL Human Engineered Skeletal Muscle
    SUPPLEMENTARY MATERIAL Human engineered skeletal muscle of hypaxial origin from pluripotent stem cells with advanced function and regenerative capacity Mina Shahriyari1,2, Md Rezaul Islam3, M. Sadman Sakib3, Anastasia Rika1,2, Dennis Krüger3, Lalit Kaurani3, Harithaa Anandakumar1,2, Orr Shomroni4, Matthias Schmidt5, Jana Zschüntzsch5, Jens Schmidt5, Gabriela Salinas-Riester4, Andreas Unger6, Wolfgang A. Linke6, André Fischer3,7, Wolfram-Hubertus Zimmermann1,2,3,7,8*, Malte Tiburcy1,2* 1 Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany. 2 DZHK (German Center for Cardiovascular Research), partner site Göttingen. 3 Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany 4 NGS Integrative Genomics Core Unit, Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany 5 Department of Neurology, University Medical Center Göttingen, Göttingen, Germany 6 Institute of Physiology II, University of Münster, D-48149 Münster, Germany 7 Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; 8 Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany Supplementary table 1 related to Figure 2: Biological process annotation of coexpression clusters. Cluster Biological process annotated Enriched GO terms identifier Black Migrating limb progenitors Muscle
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Gen-2020-0136.Pdf
    Genome Gut microbiome mediated epigenetic regulation of brain disorder and application of machine learning for multi- omics data analysis Journal: Genome Manuscript ID gen-2020-0136.R1 Manuscript Type: Mini Review Date Submitted by the 25-Sep-2020 Author: Complete List of Authors: Kaur, Harpreet; University of North Dakota School of Medicine and Health Sciences, Department of Biomedical Science Singh, Yuvraj; University of Calgary Faculty of Science, Department of Biological SciencesDraft Singh, Surjeet; University of Lethbridge, Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN) Singh, Raja; University of Alberta, Faculty of Medicine and Dentistry; University of Calgary Cumming School of Medicine Gut-brain axis, epigenetics, neurodegenerative diseases, Machine Keyword: learning, Microbiota Is the invited manuscript for consideration in a Special Genome Biology Issue? : © The Author(s) or their Institution(s) Page 1 of 46 Genome 1 Gut microbiome mediated epigenetic regulation of brain disorder and application of 2 machine learning for multi-omics data analysis 3 4 Harpreet Kaur1*#, Yuvraj Singh2#, Surjeet Singh3, Raja B Singh4,5 5 1Department of Biomedical Sciences, School of Medicine and Health Sciences, University of 6 North Dakota, Grand Forks, ND, USA 7 2Department of Biological Sciences, Faculty of Science, University of Calgary, Alberta, Canada 8 3Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), 9 University of Lethbridge, Lethbridge, AB, Canada 10 4Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada 11 5Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada 12 13 # equal contribution 14 15 Draft 16 17 18 19 20 21 *Corresponding author: 22 Harpreet Kaur, PhD 23 Department of Biomedical Sciences 24 School of Medicine and Health Sciences 25 University of North Dakota 26 Grand Forks, ND, 58202, USA.
    [Show full text]
  • Maintenance of Mammary Epithelial Phenotype by Transcription Factor Runx1 Through Mitotic Gene Bookmarking Joshua Rose University of Vermont
    University of Vermont ScholarWorks @ UVM Graduate College Dissertations and Theses Dissertations and Theses 2019 Maintenance Of Mammary Epithelial Phenotype By Transcription Factor Runx1 Through Mitotic Gene Bookmarking Joshua Rose University of Vermont Follow this and additional works at: https://scholarworks.uvm.edu/graddis Part of the Biochemistry Commons, and the Genetics and Genomics Commons Recommended Citation Rose, Joshua, "Maintenance Of Mammary Epithelial Phenotype By Transcription Factor Runx1 Through Mitotic Gene Bookmarking" (2019). Graduate College Dissertations and Theses. 998. https://scholarworks.uvm.edu/graddis/998 This Thesis is brought to you for free and open access by the Dissertations and Theses at ScholarWorks @ UVM. It has been accepted for inclusion in Graduate College Dissertations and Theses by an authorized administrator of ScholarWorks @ UVM. For more information, please contact [email protected]. MAINTENANCE OF MAMMARY EPITHELIAL PHENOTYPE BY TRANSCRIPTION FACTOR RUNX1 THROUGH MITOTIC GENE BOOKMARKING A Thesis Presented by Joshua Rose to The Faculty of the Graduate College of The University of Vermont In Partial Fulfillment of the Requirements for the Degree of Master of Science Specializing in Cellular, Molecular, and Biomedical Sciences January, 2019 Defense Date: November 12, 2018 Thesis Examination Committee: Sayyed Kaleem Zaidi, Ph.D., Advisor Gary Stein, Ph.D., Advisor Seth Frietze, Ph.D., Chairperson Janet Stein, Ph.D. Jonathan Gordon, Ph.D. Cynthia J. Forehand, Ph.D. Dean of the Graduate College ABSTRACT Breast cancer arises from a series of acquired mutations that disrupt normal mammary epithelial homeostasis and create multi-potent cancer stem cells that can differentiate into clinically distinct breast cancer subtypes. Despite improved therapies and advances in early detection, breast cancer remains the leading diagnosed cancer in women.
    [Show full text]
  • Identification of Genetic Factors Underpinning Phenotypic Heterogeneity in Huntington’S Disease and Other Neurodegenerative Disorders
    Identification of genetic factors underpinning phenotypic heterogeneity in Huntington’s disease and other neurodegenerative disorders. By Dr Davina J Hensman Moss A thesis submitted to University College London for the degree of Doctor of Philosophy Department of Neurodegenerative Disease Institute of Neurology University College London (UCL) 2020 1 I, Davina Hensman Moss confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Collaborative work is also indicated in this thesis. Signature: Date: 2 Abstract Neurodegenerative diseases including Huntington’s disease (HD), the spinocerebellar ataxias and C9orf72 associated Amyotrophic Lateral Sclerosis / Frontotemporal dementia (ALS/FTD) do not present and progress in the same way in all patients. Instead there is phenotypic variability in age at onset, progression and symptoms. Understanding this variability is not only clinically valuable, but identification of the genetic factors underpinning this variability has the potential to highlight genes and pathways which may be amenable to therapeutic manipulation, hence help find drugs for these devastating and currently incurable diseases. Identification of genetic modifiers of neurodegenerative diseases is the overarching aim of this thesis. To identify genetic variants which modify disease progression it is first necessary to have a detailed characterization of the disease and its trajectory over time. In this thesis clinical data from the TRACK-HD studies, for which I collected data as a clinical fellow, was used to study disease progression over time in HD, and give subjects a progression score for subsequent analysis. In this thesis I show blood transcriptomic signatures of HD status and stage which parallel HD brain and overlap with Alzheimer’s disease brain.
    [Show full text]
  • Targeting Mlst8 in Mtorc2-Dependent Cancers by Laura Chonghae Kim Dissertation Submitted to the Faculty of the Graduate School
    Targeting mLST8 in mTORC2-dependent cancers By Laura Chonghae Kim Dissertation Submitted to the Faculty of the Graduate School of Vanderbilt University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY in Cancer Biology June 30th, 2020 Nashville, Tennessee Approved: Albert B. Reynolds, Ph.D. (chair) Mark R. Boothby, M.D., Ph.D. Christine M. Lovly, M.D. Ph.D. William P. Tansey, Ph.D. Jin Chen, M.D., Ph.D. (adviser) DEDICATION This dissertation is dedicated to all cancer patients and their loved ones for their strength and courage in the fight against this disease. This is also dedicated to my family (Kalhee and Jenny Kim, Stephanie Kim) for their unwavering love and support. ii ACKNOWLEDGEMENTS Completion of this work would have been impossible without the support of many, and I would like to express my sincere gratitude here. First and foremost, I must thank my mentor, Jin Chen. Her support, mentorship, and high-fives have been invaluable in my development as a scientist, and I plan to continue seeking her guidance even when I am many years beyond graduate school. I would also like to acknowledge my committee members Al Reynolds, Bill Tansey, Mark Boothby, Rebecca Cook, and Christine Lovly, who continuously challenged me and provided scientific guidance. Much of this work stems from their insightful questions, and I am extremely appreciative of the time they put into thinking about my projects. I would also like to thank the past and present members of the Chen Lab: Shan Wang, Deanna Edwards, Wenqiang Song, Dana Brantley-Sieders, Katherine Hastings, Victoria Youngblood, Tammy Sobolik, Yoonha Hwang, Eileen Shiuan, Kalin Wilson, Verra Ngwa, Ashwin Inala, and Chris Rhee.
    [Show full text]
  • Identification of Macrophage Polarization-Related Genes As Biomarkers of Chronic Obstructive Pulmonary Disease Based on Bioinformatics Analyses
    Hindawi BioMed Research International Volume 2021, Article ID 9921012, 17 pages https://doi.org/10.1155/2021/9921012 Research Article Identification of Macrophage Polarization-Related Genes as Biomarkers of Chronic Obstructive Pulmonary Disease Based on Bioinformatics Analyses Yalin Zhao , Meihua Li, Yanxia Yang, Tao Wu, Qingyuan Huang, Qinghua Wu, and Chaofeng Ren Respiratory and Critical Care Medicine, Kunming First People’s Hospital, Kunming, Yunnan Province, China Correspondence should be addressed to Yalin Zhao; [email protected] and Chaofeng Ren; [email protected] Received 31 March 2021; Accepted 4 June 2021; Published 21 June 2021 Academic Editor: Nagarajan Raju Copyright © 2021 Yalin Zhao et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Objectives. Chronic obstructive pulmonary disease (COPD) is characterized by lung inflammation and remodeling. Macrophage polarization is associated with inflammation and tissue remodeling, as well as immunity. Therefore, this study attempts to investigate the diagnostic value and regulatory mechanism of macrophage polarization-related genes for COPD by bioinformatics analysis and to provide a new theoretical basis for experimental research. Methods. The raw gene expression profile dataset (GSE124180) was collected from the Gene Expression Omnibus (GEO) database. Next, a weighted gene coexpression network analysis (WGCNA) was conducted to screen macrophage polarization-related genes. The differentially expressed genes (DEGs) between the COPD and normal samples were generated using DESeq2 v3.11 and overlapped with the macrophage polarization-related genes. Moreover, functional annotations of overlapped genes were conducted by Database for Annotation, Visualization and Integrated Discovery (DAVID) Bioinformatics Resource.
    [Show full text]
  • An Interstitial Deletion-Insertion Involving Chromosomes 2P25.3 and Xq27.1, Near SOX3, Causes X-Linked Recessive Hypoparathyroidism Michael R
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2005 An interstitial deletion-insertion involving chromosomes 2p25.3 and Xq27.1, near SOX3, causes X-linked recessive hypoparathyroidism Michael R. Bowl University of Oxford Andrew Nesbit University of Oxford Brian Harding University of Oxford Elaine Levy University of Oxford Andrew Jefferson University of Oxford See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Bowl, Michael R.; Nesbit, Andrew; Harding, Brian; Levy, Elaine; Jefferson, Andrew; Volpi, Emanuela; Rizzoti, Karine; Lovell-Badge, Robin; Schlessinger, David; Whyte, Michael P.; and Thakker, Rajesh V., ,"An interstitial deletion-insertion involving chromosomes 2p25.3 and Xq27.1, near SOX3, causes X-linked recessive hypoparathyroidism." The ourJ nal of Clinical Investigation.,. 2822-2831. (2005). https://digitalcommons.wustl.edu/open_access_pubs/1524 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Michael R. Bowl, Andrew Nesbit, Brian Harding, Elaine Levy, Andrew Jefferson, Emanuela Volpi, Karine Rizzoti, Robin Lovell-Badge, David Schlessinger, Michael P. Whyte, and Rajesh V. Thakker This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/1524 Downloaded on July 12, 2013. The Journal of Clinical Investigation. More information at www.jci.org/articles/view/24156 3FTFBSDIBSUJDMF "OJOUFSTUJUJBMEFMFUJPOJOTFSUJPOJOWPMWJOH DISPNPTPNFTQBOE9R OFBS409 DBVTFT9MJOLFESFDFTTJWFIZQPQBSBUIZSPJEJTN Michael R. Bowl,1 M. Andrew Nesbit,1 Brian Harding,1 Elaine Levy,2 Andrew Jefferson,2 Emanuela Volpi,2 Karine Rizzoti,3 Robin Lovell-Badge,3 David Schlessinger,4 Michael P.
    [Show full text]
  • (PKD2), Eccentric (XNTA), and Meckelin (MKS3) in the Ciliated Model Organism Paramecium Tetraurelia Megan Smith Valentine University of Vermont
    University of Vermont ScholarWorks @ UVM Graduate College Dissertations and Theses Dissertations and Theses 2015 Polycystin-2 (PKD2), Eccentric (XNTA), and Meckelin (MKS3) in the Ciliated Model Organism Paramecium tetraurelia Megan Smith Valentine University of Vermont Follow this and additional works at: https://scholarworks.uvm.edu/graddis Part of the Biology Commons, and the Cell Biology Commons Recommended Citation Valentine, Megan Smith, "Polycystin-2 (PKD2), Eccentric (XNTA), and Meckelin (MKS3) in the Ciliated Model Organism Paramecium tetraurelia" (2015). Graduate College Dissertations and Theses. 419. https://scholarworks.uvm.edu/graddis/419 This Dissertation is brought to you for free and open access by the Dissertations and Theses at ScholarWorks @ UVM. It has been accepted for inclusion in Graduate College Dissertations and Theses by an authorized administrator of ScholarWorks @ UVM. For more information, please contact [email protected]. POLYCYSTIN-2 ( PKD2 ), ECCENTRIC ( XNTA ), AND MECKELIN ( MKS3 ) IN THE CILIATED MODEL ORGANISM PARAMECIUM TETRAURELIA A Dissertation Presented by Megan Smith Valentine to The Faculty of the Graduate College of The University of Vermont In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy Specializing in Biology October, 2015 Defense Date: June 4 th , 2015 Dissertation Examination Committee: Judith Van Houten, Ph.D., Advisor Alan Howe, Ph.D., Chairperson Anthony Morielli, Ph.D. Jim O. Vigoreaux, Ph.D. Cynthia J. Forehand, Ph.D., Dean of the Graduate College Abstract Paramecium tetraurelia is a ciliated single cell used as a model organism for the study of ciliopathies. Ciliopathies are mammalian diseases involving the dysfunction of cilia, including cilia maintenance, construction, and signaling.
    [Show full text]
  • Pre-Implantation Alcohol Exposure Induces Lasting Sex-Specific DNA
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.07.415208; this version posted June 30, 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-NC 4.0 International license. 1 Pre-Implantation Alcohol Exposure Induces Lasting Sex-Specific DNA 2 Methylation Programming Errors in the Developing Forebrain 3 4 5 6 Legault LM1,2, Doiron K1, Breton-Larrivée M1,2, Langford-Avelar A1,2, Lemieux A1,2, Caron M1, 7 Jerome-Majewska LA3,4, Sinnett D1,5, McGraw S1,2,6, #. 8 9 1CHU Sainte-Justine Research Center, Montreal, Canada. 10 2Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada. 11 3McGill University Health Centre Glen Site, Montreal, Canada. 12 4Department of Pediatrics, McGill University, Montreal, Canada. 13 5Department of Pediatrics, Université de Montréal, Montreal, Canada. 14 6Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Canada. 15 16 #Corresponding author: [email protected] 17 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.12.07.415208; this version posted June 30, 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-NC 4.0 International license. 1 ABSTRACT 2 Background 3 Prenatal alcohol exposure is recognized for altering DNA methylation profiles of brain cells during 4 development, and to be part of the molecular basis underpinning Fetal Alcohol Spectrum Disorder 5 (FASD) etiology.
    [Show full text]