Phylogenetic History of the AMY Gene Cluster in Catarrhines

Total Page:16

File Type:pdf, Size:1020Kb

Phylogenetic History of the AMY Gene Cluster in Catarrhines City University of New York (CUNY) CUNY Academic Works School of Arts & Sciences Theses Hunter College 2-1-2019 Phylogenetic History of the AMY Gene Cluster in Catarrhines Christian M. Gagnon CUNY Hunter College How does access to this work benefit ou?y Let us know! More information about this work at: https://academicworks.cuny.edu/hc_sas_etds/377 Discover additional works at: https://academicworks.cuny.edu This work is made publicly available by the City University of New York (CUNY). Contact: [email protected] Phylogenetic History of the AMY Gene Cluster in Catarrhines By Christian Gagnon Submitted in partial fulfillment of the requirements for the degree of Master of Arts in Anthropology, Hunter College of the City University of New York 2018 Thesis Sponsor: _____01/02/2019_____ _____________________________ Date Signature Dr. Michael E. Steiper _____01/02/2019_____ _____________________________ Date Signature of Second Reader Dr. Jessica Rothman 1 Acknowledgements: I owe a great debt of gratitude to my academic advisor, Dr. Michael E. Steiper, whose efforts and cooperation made this project possible. Furthermore, Dr. Steiper’s encouragement and support have been instrumental in my development as both an undergraduate and graduate student, I feel fortunate to have benefitted from his knowledge and guidance over these past several years. I thank my second reader Dr. Jessica Rothman for her invaluable expertise in the fields of primatology, ecology, and nutrition without which the completion of this project would have been much more difficult. I am also grateful to Dr. George Perry of Penn State University for his support and input in the development of this project. I would also like to thank the developers of the many tools necessary for a project of this scope to be undertaken, including the creators of MAFFT, PAML, Mega, and Datamonkey, Special thanks to my lab mate, Natalia Grube, for always being a sounding board for my ideas and her unwavering support. I also would like to recognize Caley Johnson for her contributions to the project. Her insights into primate nutritional ecology were a valuable resource. Thank you to the faculty and Graduate Students of the Hunter College Anthropology Department and the New York Consortium of Evolutionary Primatology for providing a stimulating learning environment conducive to my success. 2 Table of Contents I. List of Figures ........................................................................................................................ 4 II. List of Tables ........................................................................................................................ 4 III. Abstract ............................................................................................................................... 4 IV. Introduction: AMY and Its Relevance to Human Evolution ........................................... 5 V. Materials and Methods ..................................................................................................... 14 A. Methods Summary ......................................................................................................... 14 B. Genomic Sequences ........................................................................................................ 15 C. Coding DNA Sequences ................................................................................................. 18 D. Genomic Sequence Alignment (MAFFT) ...................................................................... 19 E. Adaptive Branch-Site Rel Test (aBSREL) .................................................................... 20 F. Gene Conversion Analysis (GENECONV) .................................................................... 20 G. Phylogenetic Analysis (RAXML) .................................................................................. 21 VI. Results ............................................................................................................................... 21 A. Sequence Alignment ...................................................................................................... 22 B. GENECONV Results ..................................................................................................... 22 C. Phylogenetic Results ...................................................................................................... 26 D. aBSREL Results ............................................................................................................. 29 VII. Discussion ........................................................................................................................ 32 VIII. References ...................................................................................................................... 36 IX. Appendix ........................................................................................................................... 41 3 I. List of Figures Figure 1. Human AMY gene positions Pg. 10 Figure 2. Phylogenetic Tree (Whole Gene) Pg. 27 Figure 3. Phylogenetic Tree (Coding Regions Only) Pg. 28 Figure 4. aBSREL Tree Pg. 30 Figure 5. Phylogenetic Analysis Summary Pg. 33 II. List of Tables Table 1. Genome Coordinates for AMY Orthologs Pg. 16 Table 2. Coding Region Coordinates for Human AMY1A Pg. 19 Table 3. Gene Conversion Sites Pg. 24 Table 4. aBSREL Tree Summary Pg. 31 Table 5. aBSREL Positively-Selected Branches Pg. 31 III. Abstract The AMY gene family plays an essential role in the expression of ⍺-amylase, an enzyme critical to starch digestion. The importance of dietary starch in the evolution of human traits such as the brain is an ongoing point of debate in evolutionary biology because many studies have focused on the increase in animal proteins in hominin diets as the catalyst that made its expansion possible. Levels of amylase expression vary among primates, and although the regulatory mechanism is mostly unknown, evidence suggests that gene copy number variation (CNV) plays a role in humans (Perry et al., 2007). Previous studies indicate that humans who have traditionally 4 consumed high starch diets possess an increased number of AMY gene copies which correlated with increased amylase protein expression in saliva. However, it is important to note that Papio hamadryas and Theropithecus gelada show higher salivary amylase expression than Homo sapiens and Pan troglodytes despite having fewer gene copies. The finding that these primates have higher amylase expression in saliva suggests CNV cannot fully explain its regulation. In this study, I mined and phylogenetically analyzed 30 AMY-related genes from 11 species of haplorrhines. My research sheds light on the complex evolutionary history of this gene family in humans and other primates to further our understanding of our ecological past and the evolutionary pressures that drove these adaptive changes. These findings show that the ancestor of all anthropoids likely had a single AMY-like gene. This gene duplicated independently in New World monkeys and Old World monkeys. Assuming that the gibbon lost its “AMY1 like” gene, in the ancestor of the apes, there was a single AMY that duplicated into the AMY1 and AMY2 like genes. This AMY2 gene then duplicated into AMY2a and AMY2b. All apes have these three orthologs, but gibbons lost their AMY1. In gorillas, AMY2b duplicated again. In humans, AMY1 duplicated further resulting in a three salivary orthologs 1a, 1b, and 1c. These results suggest that the gradual expansion was driven by selective forces to allow humans and other primates to adapt to various ecological landscapes and maximize energy intake from starch-rich foods in periods of food scarcity or in some cases, a staple of their diet. IV. Introduction AMY and its Relevance to Human Evolution 5 There is an ongoing debate among evolutionary theorists regarding the relative importance of proteins derived from meat-based diets versus starch, a form of carbohydrates, from plant foods in the evolution of enlarged primate brains. Although there is support for the idea that the shift from a more plant-rich diet to a meat-rich diet drove the expansion of the primate brain (Aiello & Wheeler, 1995; Milton, 2003), others argued that digestible carbohydrates and cooking may have played equally important roles (Hardy et al., 2015; Wrangham, 2009). Glycogen production in the liver and muscle tissues, the result of glucose intake from carbohydrates, including dietary starch, is essential to human cognitive abilities (Suzuki et al., 2011). Human brains require a steady and reliable source of glycemic carbohydrates to maintain healthy brain function, which accounts for nearly 25% of our basal metabolic spending (Fonseca-Azevedo & Herculano-Houzel, 2012). Based on isotopic and craniodental fossil evidence, several studies suggest that the transition to open habitats led to a possible increase in starch-rich foods from underground storage organs (i.e. tubers, corms, roots and bulbs) and allow early hominins to optimize energy intake from their diet (Codron et al., 2007; Dominy et al., 2008; Laden & Wrangham, 2005; Sponheimer et al., 2013). Many living primate populations inhabit open habitats, and as research has shown, they consume these critical food sources. Savannah dwelling chimpanzees in Ugalla (Tanzania), use tools to uncover underground storage organs (USOs) during the rainy season when food is more abundant (Constantino & Wright, 2009; Hernandez-Aguilar et al., 2007). This evidence of tool use is in contrast some fallback
Recommended publications
  • Structural Forms of the Human Amylase Locus and Their Relationships to Snps, Haplotypes, and Obesity
    Structural Forms of the Human Amylase Locus and Their Relationships to SNPs, Haplotypes, and Obesity The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Usher, Christina Leigh. 2015. Structural Forms of the Human Amylase Locus and Their Relationships to SNPs, Haplotypes, and Obesity. Doctoral dissertation, Harvard University, Graduate School of Arts & Sciences. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:17467224 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA Structural forms of the human amylase locus and their relationships to SNPs, haplotypes, and obesity A dissertation presented by Christina Leigh Usher to The Division of Medical Sciences in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the subject of Genetics and Genomics Harvard University Cambridge, Massachusetts March 2015 © 2015 Christina Leigh Usher All rights reserved. Dissertation Advisor: Professor Steven McCarroll Christina Leigh Usher Structural forms of the human amylase locus and their relationships to SNPs, haplotypes, and obesity Abstract Hundreds of human genes reside in structurally complex loci that elude molecular analysis and assessment in genome-wide association studies (GWAS). One such locus contains the three different amylase genes (AMY2B, AMY2A, and AMY1) responsible for digesting starch into sugar. The copy number of AMY1 is reported to be the genome’s largest influence on obesity, yet has gone undetected in GWAS.
    [Show full text]
  • Chromosome 1 (Human Genome/Inkae) A
    Proc. Nati. Acad. Sci. USA Vol. 89, pp. 4598-4602, May 1992 Medical Sciences Integration of gene maps: Chromosome 1 (human genome/inkae) A. COLLINS*, B. J. KEATSt, N. DRACOPOLIt, D. C. SHIELDS*, AND N. E. MORTON* *CRC Research Group in Genetic Epidemiology, Department of Child Health, University of Southampton, Southampton, S09 4XY, United Kingdom; tDepartment of Biometry and Genetics, Louisiana State University Center, 1901 Perdido Street, New Orleans, LA 70112; and tCenter for Cancer Research, Massachusetts Institute of Technology, 40 Ames Street, Cambridge, MA 02139 Contributed by N. E. Morton, February 10, 1992 ABSTRACT A composite map of 177 locI has been con- standard lod tables extracted from the literature. Multiple structed in two steps. The first combined pairwise logarithm- pairwise analysis of these data was performed by the MAP90 of-odds scores on 127 loci Into a comprehensive genetic map. computer program (6), which can estimate an errorfrequency Then this map was projected onto the physical map through e (7) and a mapping parameter p such that map distance w is cytogenetic assignments, and the small amount ofphysical data a function of 0, e and p (8). It also includes a bootstrap to was interpolated for an additional 50 loci each of which had optimize order and a stepwise elimination of weakly sup- been assigned to an interval of less than 10 megabases. The ported loci to identify a conservative set of reliably ordered resulting composite map is on the physical scale with a reso- (framework) markers. The genetic map was combined with lution of 1.5 megabases.
    [Show full text]
  • Characterization of Genomic Copy Number Variation in Mus Musculus Associated with the Germline of Inbred and Wild Mouse Populations, Normal Development, and Cancer
    Western University Scholarship@Western Electronic Thesis and Dissertation Repository 4-18-2019 2:00 PM Characterization of genomic copy number variation in Mus musculus associated with the germline of inbred and wild mouse populations, normal development, and cancer Maja Milojevic The University of Western Ontario Supervisor Hill, Kathleen A. The University of Western Ontario Graduate Program in Biology A thesis submitted in partial fulfillment of the equirr ements for the degree in Doctor of Philosophy © Maja Milojevic 2019 Follow this and additional works at: https://ir.lib.uwo.ca/etd Part of the Genetics and Genomics Commons Recommended Citation Milojevic, Maja, "Characterization of genomic copy number variation in Mus musculus associated with the germline of inbred and wild mouse populations, normal development, and cancer" (2019). Electronic Thesis and Dissertation Repository. 6146. https://ir.lib.uwo.ca/etd/6146 This Dissertation/Thesis is brought to you for free and open access by Scholarship@Western. It has been accepted for inclusion in Electronic Thesis and Dissertation Repository by an authorized administrator of Scholarship@Western. For more information, please contact [email protected]. Abstract Mus musculus is a human commensal species and an important model of human development and disease with a need for approaches to determine the contribution of copy number variants (CNVs) to genetic variation in laboratory and wild mice, and arising with normal mouse development and disease. Here, the Mouse Diversity Genotyping array (MDGA)-approach to CNV detection is developed to characterize CNV differences between laboratory and wild mice, between multiple normal tissues of the same mouse, and between primary mammary gland tumours and metastatic lung tissue.
    [Show full text]
  • Renal Cell Neoplasms Contain Shared Tumor Type–Specific Copy Number Variations
    The American Journal of Pathology, Vol. 180, No. 6, June 2012 Copyright © 2012 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2012.01.044 Tumorigenesis and Neoplastic Progression Renal Cell Neoplasms Contain Shared Tumor Type–Specific Copy Number Variations John M. Krill-Burger,* Maureen A. Lyons,*† The annual incidence of renal cell carcinoma (RCC) has Lori A. Kelly,*† Christin M. Sciulli,*† increased steadily in the United States for the past three Patricia Petrosko,*† Uma R. Chandran,†‡ decades, with approximately 58,000 new cases diag- 1,2 Michael D. Kubal,§ Sheldon I. Bastacky,*† nosed in 2010, representing 3% of all malignancies. Anil V. Parwani,*†‡ Rajiv Dhir,*†‡ and Treatment of RCC is complicated by the fact that it is not a single disease but composes multiple tumor types with William A. LaFramboise*†‡ different morphological characteristics, clinical courses, From the Departments of Pathology* and Biomedical and outcomes (ie, clear-cell carcinoma, 82% of RCC ‡ Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania; cases; type 1 or 2 papillary tumors, 11% of RCC cases; † the University of Pittsburgh Cancer Institute, Pittsburgh, chromophobe tumors, 5% of RCC cases; and collecting § Pennsylvania; and Life Technologies, Carlsbad, California duct carcinoma, approximately 1% of RCC cases).2,3 Benign renal neoplasms are subdivided into papillary adenoma, renal oncocytoma, and metanephric ade- Copy number variant (CNV) analysis was performed on noma.2,3 Treatment of RCC often involves surgical resec- renal cell carcinoma (RCC) specimens (chromophobe, tion of a large renal tissue component or removal of the clear cell, oncocytoma, papillary type 1, and papillary entire affected kidney because of the relatively large size of type 2) using high-resolution arrays (1.85 million renal tumors on discovery and the availability of a life-sus- probes).
    [Show full text]
  • Salivary Alpha Amylase (AMY1C) (NM 001008219) Human Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RG215827 Salivary alpha amylase (AMY1C) (NM_001008219) Human Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: Salivary alpha amylase (AMY1C) (NM_001008219) Human Tagged ORF Clone Tag: TurboGFP Symbol: AMY1C Synonyms: AMY1 Vector: pCMV6-AC-GFP (PS100010) E. coli Selection: Ampicillin (100 ug/mL) Cell Selection: Neomycin This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 5 Salivary alpha amylase (AMY1C) (NM_001008219) Human Tagged ORF Clone – RG215827 ORF Nucleotide >RG215827 representing NM_001008219 Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGAAGCTCTTTTGGTTGCTTTTCACCATTGGGTTCTGCTGGGCTCAGTATTCCTCAAATACACAACAAG GACGAACATCTATTGTTCATCTGTTTGAATGGCGATGGGTTGATATTGCTCTTGAATGTGAGCGATATTT AGCTCCCAAGGGATTTGGAGGGGTTCAGGTCTCTCCACCAAATGAAAATGTTGCCATTCACAACCCTTTC AGACCTTGGTGGGAAAGATACCAACCAGTTAGCTATAAATTATGCACAAGATCTGGAAATGAAGATGAAT TTAGAAACATGGTGACTAGATGCAACAATGTTGGGGTTCGTATTTATGTGGATGCTGTAATTAATCATAT GTGTGGTAATGCTGTGAGTGCAGGAACAAGCAGTACCTGTGGAAGTTACTTCAACCCTGGAAGTAGGGAC TTTCCAGCAGTCCCATATTCTGGATGGGATTTTAATGATGGTAAATGTAAAACTGGAAGTGGAGATATCG AGAACTATAATGATGCTACTCAGGTCAGAGATTGTCGTCTGTCTGGTCTTCTCGATCTTGCACTGGGGAA
    [Show full text]
  • Differential Proteomic Analysis of the Pancreas of Diabetic Db/Db Mice Reveals the Proteins Involved in the Development of Complications of Diabetes Mellitus
    Int. J. Mol. Sci. 2014, 15, 9579-9593; doi:10.3390/ijms15069579 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article Differential Proteomic Analysis of the Pancreas of Diabetic db/db Mice Reveals the Proteins Involved in the Development of Complications of Diabetes Mellitus Victoriano Pérez-Vázquez 1,*, Juan M. Guzmán-Flores 1, Daniela Mares-Álvarez 1, Magdalena Hernández-Ortiz 2, Maciste H. Macías-Cervantes 1, Joel Ramírez-Emiliano 1 and Sergio Encarnación-Guevara 2 1 Depto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato 37320, Mexico; E-Mails: [email protected] (J.M.G.-F.); [email protected] (D.M.-A.); [email protected] (M.H.M.-C.); [email protected] (J.R.-E.) 2 Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico; E-Mails: [email protected] (M.H.-O.); [email protected] (S.E.-G.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +52-477-7143-812; Fax: +52-477-7167-623. Received: 4 April 2014; in revised form: 14 May 2014 / Accepted: 19 May 2014 / Published: 30 May 2014 Abstract: Type 2 diabetes mellitus is characterized by hyperglycemia and insulin-resistance. Diabetes results from pancreatic inability to secrete the insulin needed to overcome this resistance. We analyzed the protein profile from the pancreas of ten-week old diabetic db/db and wild type mice through proteomics. Pancreatic proteins were separated in two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and significant changes in db/db mice respect to wild type mice were observed in 27 proteins.
    [Show full text]
  • Chuanxiong Rhizoma Compound on HIF-VEGF Pathway and Cerebral Ischemia-Reperfusion Injury’S Biological Network Based on Systematic Pharmacology
    ORIGINAL RESEARCH published: 25 June 2021 doi: 10.3389/fphar.2021.601846 Exploring the Regulatory Mechanism of Hedysarum Multijugum Maxim.-Chuanxiong Rhizoma Compound on HIF-VEGF Pathway and Cerebral Ischemia-Reperfusion Injury’s Biological Network Based on Systematic Pharmacology Kailin Yang 1†, Liuting Zeng 1†, Anqi Ge 2†, Yi Chen 1†, Shanshan Wang 1†, Xiaofei Zhu 1,3† and Jinwen Ge 1,4* Edited by: 1 Takashi Sato, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of 2 Tokyo University of Pharmacy and Life Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China, Galactophore Department, The First 3 Sciences, Japan Hospital of Hunan University of Chinese Medicine, Changsha, China, School of Graduate, Central South University, Changsha, China, 4Shaoyang University, Shaoyang, China Reviewed by: Hui Zhao, Capital Medical University, China Background: Clinical research found that Hedysarum Multijugum Maxim.-Chuanxiong Maria Luisa Del Moral, fi University of Jaén, Spain Rhizoma Compound (HCC) has de nite curative effect on cerebral ischemic diseases, *Correspondence: such as ischemic stroke and cerebral ischemia-reperfusion injury (CIR). However, its Jinwen Ge mechanism for treating cerebral ischemia is still not fully explained. [email protected] †These authors share first authorship Methods: The traditional Chinese medicine related database were utilized to obtain the components of HCC. The Pharmmapper were used to predict HCC’s potential targets. Specialty section: The CIR genes were obtained from Genecards and OMIM and the protein-protein This article was submitted to interaction (PPI) data of HCC’s targets and IS genes were obtained from String Ethnopharmacology, a section of the journal database.
    [Show full text]
  • Role of Amylase in Ovarian Cancer Mai Mohamed University of South Florida, [email protected]
    University of South Florida Scholar Commons Graduate Theses and Dissertations Graduate School July 2017 Role of Amylase in Ovarian Cancer Mai Mohamed University of South Florida, [email protected] Follow this and additional works at: http://scholarcommons.usf.edu/etd Part of the Pathology Commons Scholar Commons Citation Mohamed, Mai, "Role of Amylase in Ovarian Cancer" (2017). Graduate Theses and Dissertations. http://scholarcommons.usf.edu/etd/6907 This Dissertation is brought to you for free and open access by the Graduate School at Scholar Commons. It has been accepted for inclusion in Graduate Theses and Dissertations by an authorized administrator of Scholar Commons. For more information, please contact [email protected]. Role of Amylase in Ovarian Cancer by Mai Mohamed A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy Department of Pathology and Cell Biology Morsani College of Medicine University of South Florida Major Professor: Patricia Kruk, Ph.D. Paula C. Bickford, Ph.D. Meera Nanjundan, Ph.D. Marzenna Wiranowska, Ph.D. Lauri Wright, Ph.D. Date of Approval: June 29, 2017 Keywords: ovarian cancer, amylase, computational analyses, glycocalyx, cellular invasion Copyright © 2017, Mai Mohamed Dedication This dissertation is dedicated to my parents, Ahmed and Fatma, who have always stressed the importance of education, and, throughout my education, have been my strongest source of encouragement and support. They always believed in me and I am eternally grateful to them. I would also like to thank my brothers, Mohamed and Hussien, and my sister, Mariam. I would also like to thank my husband, Ahmed.
    [Show full text]
  • Early Growth Response 1 Regulates Hematopoietic Support and Proliferation in Human Primary Bone Marrow Stromal Cells
    Hematopoiesis SUPPLEMENTARY APPENDIX Early growth response 1 regulates hematopoietic support and proliferation in human primary bone marrow stromal cells Hongzhe Li, 1,2 Hooi-Ching Lim, 1,2 Dimitra Zacharaki, 1,2 Xiaojie Xian, 2,3 Keane J.G. Kenswil, 4 Sandro Bräunig, 1,2 Marc H.G.P. Raaijmakers, 4 Niels-Bjarne Woods, 2,3 Jenny Hansson, 1,2 and Stefan Scheding 1,2,5 1Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden; 2Lund Stem Cell Center, Depart - ment of Laboratory Medicine, Lund University, Lund, Sweden; 3Division of Molecular Medicine and Gene Therapy, Department of Labora - tory Medicine, Lund University, Lund, Sweden; 4Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands and 5Department of Hematology, Skåne University Hospital Lund, Skåne, Sweden ©2020 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol. 2019.216648 Received: January 14, 2019. Accepted: July 19, 2019. Pre-published: August 1, 2019. Correspondence: STEFAN SCHEDING - [email protected] Li et al.: Supplemental data 1. Supplemental Materials and Methods BM-MNC isolation Bone marrow mononuclear cells (BM-MNC) from BM aspiration samples were isolated by density gradient centrifugation (LSM 1077 Lymphocyte, PAA, Pasching, Austria) either with or without prior incubation with RosetteSep Human Mesenchymal Stem Cell Enrichment Cocktail (STEMCELL Technologies, Vancouver, Canada) for lineage depletion (CD3, CD14, CD19, CD38, CD66b, glycophorin A). BM-MNCs from fetal long bones and adult hip bones were isolated as reported previously 1 by gently crushing bones (femora, tibiae, fibulae, humeri, radii and ulna) in PBS+0.5% FCS subsequent passing of the cell suspension through a 40-µm filter.
    [Show full text]
  • Marker Identification of the Grade of Dysplasia of Intraductal Papillary
    cancers Article Marker Identification of the Grade of Dysplasia of Intraductal Papillary Mucinous Neoplasm in Pancreatic Cyst Fluid by Quantitative Proteomic Profiling 1, 2, 1 3 4 Misol Do y , Hongbeom Kim y, Dongyoon Shin , Joonho Park , Haeryoung Kim , Youngmin Han 2, Jin-Young Jang 2,* and Youngsoo Kim 1,3,* 1 Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea; [email protected] (M.D.); [email protected] (D.S.) 2 Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea; [email protected] (H.K.); [email protected] (Y.H.) 3 Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Korea; [email protected] 4 Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea; [email protected] * Correspondence: [email protected] (J.-Y.J.); [email protected] (Y.K.); Tel.: +82-10-8338-6719 (J.-Y.J.); +82-2-740-8073 (Y.K.) The first two authors contributed equally to this work. y Received: 10 August 2020; Accepted: 20 August 2020; Published: 23 August 2020 Abstract: The incidence of patients with pancreatic cystic lesions, particularly intraductal papillary mucinous neoplasm (IPMN), is increasing. Current guidelines, which primarily consider radiological features and laboratory data, have had limited success in predicting malignant IPMN. The lack of a definitive diagnostic method has led to low-risk IPMN patients undergoing unnecessary surgeries. To address this issue, we discovered IPMN marker candidates by analyzing pancreatic cystic fluid by mass spectrometry. A total of 30 cyst fluid samples, comprising IPMN dysplasia and other cystic lesions, were evaluated.
    [Show full text]
  • Improved Detection of Gene Fusions by Applying Statistical Methods Reveals New Oncogenic RNA Cancer Drivers
    bioRxiv preprint doi: https://doi.org/10.1101/659078; this version posted June 3, 2019. 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. Improved detection of gene fusions by applying statistical methods reveals new oncogenic RNA cancer drivers Roozbeh Dehghannasiri1, Donald Eric Freeman1,2, Milos Jordanski3, Gillian L. Hsieh1, Ana Damljanovic4, Erik Lehnert4, Julia Salzman1,2,5* Author affiliation 1Department of Biochemistry, Stanford University, Stanford, CA 94305 2Department of Biomedical Data Science, Stanford University, Stanford, CA 94305 3Department of Computer Science, University of Belgrade, Belgrade, Serbia 4Seven Bridges Genomics, Cambridge, MA 02142 5Stanford Cancer Institute, Stanford, CA 94305 *Corresponding author [email protected] Short Abstract: The extent to which gene fusions function as drivers of cancer remains a critical open question. Current algorithms do not sufficiently identify false-positive fusions arising during library preparation, sequencing, and alignment. Here, we introduce a new algorithm, DEEPEST, that uses statistical modeling to minimize false-positives while increasing the sensitivity of fusion detection. In 9,946 tumor RNA-sequencing datasets from The Cancer Genome Atlas (TCGA) across 33 tumor types, DEEPEST identifies 31,007 fusions, 30% more than identified by other methods, while calling ten-fold fewer false-positive fusions in non-transformed human tissues. We leverage the increased precision of DEEPEST to discover new cancer biology. For example, 888 new candidate oncogenes are identified based on over-representation in DEEPEST-Fusion calls, and 1,078 previously unreported fusions involving long intergenic noncoding RNAs partners, demonstrating a previously unappreciated prevalence and potential for function.
    [Show full text]
  • Research Article Sex Difference of Ribosome in Stroke-Induced Peripheral Immunosuppression by Integrated Bioinformatics Analysis
    Hindawi BioMed Research International Volume 2020, Article ID 3650935, 15 pages https://doi.org/10.1155/2020/3650935 Research Article Sex Difference of Ribosome in Stroke-Induced Peripheral Immunosuppression by Integrated Bioinformatics Analysis Jian-Qin Xie ,1,2,3 Ya-Peng Lu ,1,3 Hong-Li Sun ,1,3 Li-Na Gao ,2,3 Pei-Pei Song ,2,3 Zhi-Jun Feng ,3 and Chong-Ge You 2,3 1Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China 2Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China 3The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, China Correspondence should be addressed to Chong-Ge You; [email protected] Received 13 April 2020; Revised 8 October 2020; Accepted 18 November 2020; Published 3 December 2020 Academic Editor: Rudolf K. Braun Copyright © 2020 Jian-Qin Xie 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. Ischemic stroke (IS) greatly threatens human health resulting in high mortality and substantial loss of function. Recent studies have shown that the outcome of IS has sex specific, but its mechanism is still unclear. This study is aimed at identifying the sexually dimorphic to peripheral immune response in IS progression, predicting potential prognostic biomarkers that can lead to sex- specific outcome, and revealing potential treatment targets. Gene expression dataset GSE37587, including 68 peripheral whole blood samples which were collected within 24 hours from known onset of symptom and again at 24-48 hours after onset (20 women and 14 men), was downloaded from the Gene Expression Omnibus (GEO) datasets.
    [Show full text]