SI Correction

Total Page:16

File Type:pdf, Size:1020Kb

SI Correction SI Correction SYSTEMS BIOLOGY Correction to Supporting Information for “miRNA proxy approach issue 23, June 9, 2015, of Proc Natl Acad Sci USA (112:7327–7332; reveals hidden functions of glycosylation,” by Tomasz Kurcon, first published May 26, 2015; 10.1073/pnas.1502076112). Zhongyin Liu, Anika V. Paradkar, Christopher A. Vaiana, Sujeethraj The authors note that Table S1, Table S3, and Table S4 ap- Koppolu, Praveen Agrawal, and Lara K. Mahal, which appeared in peared incorrectly. The SI has been corrected online. Table S1. Classification of miR-200f predicted glycogenes according to the type of glycan synthesized (related to Table S3) N-linked O-linked NC-O-linked GAGs Glycolipid Other/terminal ALG2 C1GALT1 B3GAT1 CSGALNACT2 B3GALT2 B3GALNT2 ALG5 GALNT2 B3GLCT B3GALT6 B3GALNT1 B3GNT1 ALG6 GALNT3 GXYLT1 CHST2 B3GNT4 B3GNT2 ALG8 GALNT4 LARGE CHST7 B3GNT5 FUT2 ALG11 GALNT10 LFNG CHST12 B4GALT6 FUT4 ALG14 GALNT11 OGT CHSY1 GAL3ST1 HAS2 DDOST GALNT12 POFUT1 CHSY3 ST3GAL5 HAS3 DPAGT1 GALNT14 EXTL2 ST3GAL6 ST3GAL2 MGAT2 GALNTL2 HS2ST1 ST6GALNAC3 ST3GAL3 MGAT3 GCNT2 HS3ST1 ST6GALNAC5 ST8SIA2 MGAT4A GCNT4 HS3ST3A1 UGCG ST8SIA4 MGAT4C HS3ST5 HS6ST1 HS6ST2 HS6ST3 NDST3 Count 12 11 7 16 11 11 % Total 17.6 16.2 10.3 23.5 16.2 16.2 miR-200f predicted glycogenes were classified according to the glycan they synthesize using the Kyoto Encyclopedia of Genes and Genomes. The % total represents the distribution of one type of glycan among all predicted targets. GAGs, glycosaminoglycans; NC O-linked, noncanonical O-linked. E4632–E4635 | PNAS | August 18, 2015 | vol. 112 | no. 33 www.pnas.org Downloaded by guest on September 28, 2021 Table S3. miR-200f target glycogene transcript expression profile in MDA-MB-231 cells treated with miR-200a, miR-200b, or scramble mimics miR-200a treatment miR-200b treatment Glycogene Fold change P values Glycogene Fold change P values Glycan type Shared predicted targets MGAT4A 1.06 0.74 MGAT4A 1.17 0.31 N-linked C1GALT1 −1.19 0.050 C1GALT1 −1.10 0.40 O-linked GCNT2 1.04 0.86 GCNT2 1.20 0.36 B3GLCT −1.06 0.69 B3GLCT −1.61 4.18E-03 NC O-linked ST3GAL5 −1.61 6.37E-03 ST3GAL5 −2.13 2.11E-04 Glycolipid ST6GALNAC3 1.62 0.20 ST6GALNAC3 1.11 0.76 B3GNT2 1.07 0.66 B3GNT2 1.22 0.15 Other/terminal miR-200b/-200c/-429 predicted targets MGAT2 −1.08 0.72 MGAT2 1.01 0.98 N-linked MGAT3 −1.86 0.12 MGAT3 1.30 0.15 MGAT4C 1.93 0.20 MGAT4C −1.00 1.00 GALNT2 −1.09 0.52 GALNT2 −1.40 0.022 O-linked GALNT3 1.03 0.71 GALNT3 2.30 4.52E-04 GALNT10 −1.14 0.11 GALNT10 −1.68 1.22E-03 GALNT11 −1.11 0.29 GALNT11 −1.26 0.011 GALNT12 −1.40 7.38E-03 GALNT12 −1.52 6.32E-03 GALNT14 −1.49 0.011 GALNT14 −1.94 5.92E-03 GCNT4 −1.18 0.16 GCNT4 −1.05 0.79 LFNG −1.49 0.21 LFNG −2.14 0.074 NC O-linked CHST12 1.09 0.81 CHST12 1.22 0.66 GAGs CHSY3 1.15 0.36 CHSY3 −1.04 0.68 EXTL2 −1.05 0.78 EXTL2 1.07 0.71 HS3ST1 1.99 0.044 HS3ST1 3.28 0.014 HS3ST3A1 −1.11 0.74 HS3ST3A1 −1.24 0.39 NDST3 −2.87 3.16E-03 NDST3 −6.96 4.58E-04 B3GALNT1 −1.11 0.56 B3GALNT1 −1.16 0.46 Glycolipid ST6GALNAC5 2.23 0.033 ST6GALNAC5 4.79 0.010 B3GNT1 −1.04 0.80 B3GNT1 −1.47 0.039 Other/terminal ST3GAL2 −1.18 0.20 ST3GAL2 −1.59 1.23E-03 ST8SIA4 −1.23 0.013 ST8SIA4 −1.15 0.22 miR−/−200a/-141 predicted targets GALNT4 −1.16 0.20 GALNT4 −1.17 0.077 O-linked OGT −1.09 0.50 OGT −1.02 0.88 NC O-linked POFUT1 −1.00 0.98 POFUT1 −1.01 0.90 B3GALT6 1.05 0.83 B3GALT6 1.00 0.99 GAGs HS6ST1 −1.19 0.46 HS6ST1 −1.14 0.56 HS6ST3 1.13 0.59 HS6ST3 4.37 1.41E-03 B3GNT4 1.29 0.075 B3GNT4 2.42 6.44E-04 Glycolipid B3GNT5 1.25 0.11 B3GNT5 1.70 2.62E-03 ST3GAL6 −1.18 0.30 ST3GAL6 1.01 0.91 ST3GAL3 −1.42 0.051 ST3G AL3 −1.43 0.035 Other/terminal MDA-MB-231 cells were treated with miR-200a, miR-200b. or scramble mimics (50 nM) for 7 d. The qRT-PCR was performed using RT2 Profiler PCR Array Human Glycosylation (Qiagen) or custom primers for genes not repre- sented on the array (Dataset S1). Fold change (miR-treated/scramble) was calculated by taking the average relative expression of miR-treated samples (n = 4 independent experiments, normalized to GAPDH) over the average SI CORRECTION relative expression of the scramble treated cells (n = 4 independent experiments, normalized to GAPDH). Where the fold change was less than 1, the negative inverse is shown. P values for treated vs. scramble were calculated using Student’s t test. Statistically significant (P ≤ 0.05) down-regulation is in bold; up-regulation is underlined. GAGs, glycosaminoglycans; NC O-linked, noncanonical O-linked; See Table S1 for glycan type distribution. *Genes for which luciferase data were obtained (Fig. 1). PNAS | August 18, 2015 | vol. 112 | no. 33 | E4633 Downloaded by guest on September 28, 2021 Table S4. Expression profile of glycogenes not predicted to be targets of the miR-200 family in MDA-MB-231 cells treated with miR-200a, miR-200b, or scramble mimics miR-200a treatment miR-200b treatment Glycogene Fold change P values Glycogene Fold change P values A4GNT −1.41 7.17E-03 A4GNT −1.81 1.14E-03 AGA −1.37 2.50E-03 AGA −1.17 0.24 B3GNT3 13.51 5.78E-06 B3GNT3 67.80 1.35E-05 B3GNT8 −1.14 0.42 B3GNT8 1.01 0.95 B4GALT1 −1.17 0.20 B4GALT1 −1.15 0.16 B4GALT2 1.01 0.97 B4GALT2 −1.02 0.92 B4GALT3 1.21 0.43 B4GALT3 1.32 0.19 B4GALT5 1.11 0.62 B4GALT5 1.61 0.021 C1GALT1C1 −1.01 0.94 C1GALT1C1 1.02 0.80 EDEM1 −1.07 0.59 EDEM1 −1.05 0.65 EDEM2 −1.06 0.60 EDEM2 1.04 0.66 EDEM3 1.02 0.79 EDEM3 −1.10 0.23 FUCA1 −1.09 0.31 FUCA1 −1.12 0.19 FUCA2 −1.09 0.24 FUCA2 −1.03 0.61 FUT8 1.20 0.19 FUT8 −1.07 0.45 FUT11 1.03 0.79 FUT11 1.28 0.12 GALNT1 1.15 0.29 GALNT1 −1.12 0.30 GALNT6 2.14 3.74E-04 GALNT6 1.79 9.89E-05 GALNT7 −1.07 0.60 GALNT7 −1.09 0.39 GALNT8 −1.69 0.21 GALNT8 −2.07 0.18 GALNT9 −2.19 0.12 GALNT9 −1.90 0.17 GALNT13 −1.16 0.024 GALNT13 1.03 0.63 GALNTL1 −1.61 0.013 GALNTL1 −2.02 3.34E-03 GALNTL5 1.41 0.36 GALNTL5 1.08 0.36 GALNTL6 −2.06 0.12 GALNTL6 −3.08 0.048 GANAB −1.01 0.92 GANAB −1.02 0.80 GCNT1 −1.61 8.08E-05 GCNT1 −1.63 3.98E-03 GCNT3 −1.18 0.35 GCNT3 1.49 0.23 GLB1 −1.06 0.73 GLB1 −1.17 0.26 GNPTAB −1.15 0.19 GNPTAB −1.03 0.84 GNPTG −1.15 0.26 GNPTG −1.13 0.22 HEXA −1.11 0.36 HEXA −1.14 0.091 HEXB −1.13 0.24 HEXB −1.17 0.11 MAN1A1 1.09 0.54 MAN1A1 −1.14 0.45 MAN1A2 1.02 0.88 MAN1A2 −1.15 0.25 MAN1B1 −1.06 0.56 MAN1B1 −1.08 0.41 MAN1C1 −1.07 0.76 MAN1C1 −1.39 0.039 MAN2A1 −1.06 0.23 MAN2A1 1.13 0.23 MAN2A2 1.24 0.41 MAN2A2 1.43 0.12 MAN2B1 1.04 0.89 MAN2B1 1.01 0.97 MANBA −1.21 5.54E-04 MANBA −1.22 0.015 MGAT1 1.18 0.36 MGAT1 1.37 0.019 MGAT4B 1.13 0.39 MGAT4B 1.13 0.17 MGAT5 1.27 0.32 MGAT5 1.31 0.25 MGAT5B −1.29 0.15 MGAT5B −2.24 3.69E-03 MGEA5 −1.06 0.58 MGEA5 1.02 0.71 MOGS 1.10 0.63 MOGS 1.32 0.092 NAGPA −1.33 0.32 NAGPA −1.40 0.20 NEU1 −1.08 0.63 NEU1 1.25 0.10 NEU2 1.43 0.24 NEU2 1.62 0.032 NEU3 −1.05 0.63 NEU3 1.24 0.074 NEU4 −1.35 0.51 NEU4 −1.63 0.36 POFUT2 −1.14 0.18 POFUT2 −1.25 0.014 POMGNT1 1.06 0.71 POMGNT1 −1.05 0.59 POMT1 −1.18 0.33 POMT1 −1.07 0.61 POMT2 1.07 0.66 POMT2 1.02 0.85 PRKCSH 1.02 0.86 PRKCSH 1.07 0.39 ST3GAL1 1.19 0.056 ST3GAL1 1.21 0.027 ST6GAL1 −1.60 8.50E-03 ST6GAL1 −2.08 3.80E-04 ST6GALNAC1 4.57 0.044 ST6GALNAC1 9.10 6.11E-03 E4634 | www.pnas.org Downloaded by guest on September 28, 2021 Table S4. Cont. miR-200a treatment miR-200b treatment Glycogene Fold change P values Glycogene Fold change P values ST8SIA3 −1.11 0.91 ST8SIA3 −1.06 0.95 ST8SIA6 −1.72 0.017 ST8SIA6 −3.13 1.77E-03 UGGT1 −1.03 0.87 UGGT1 −1.15 0.30 UGGT2 −1.09 0.44 UGGT2 −1.18 0.093 MDA-MB-231 cells were treated with miR-200a, miR-200b, or scramble mimics (50 nM) for 7 d.
Recommended publications
  • Investigating the Genetic Basis of Cisplatin-Induced Ototoxicity in Adult South African Patients
    --------------------------------------------------------------------------- Investigating the genetic basis of cisplatin-induced ototoxicity in adult South African patients --------------------------------------------------------------------------- by Timothy Francis Spracklen SPRTIM002 SUBMITTED TO THE UNIVERSITY OF CAPE TOWN In fulfilment of the requirements for the degree MSc(Med) Faculty of Health Sciences UNIVERSITY OF CAPE TOWN University18 December of Cape 2015 Town Supervisor: Prof. Rajkumar S Ramesar Co-supervisor: Ms A Alvera Vorster Division of Human Genetics, Department of Pathology, University of Cape Town 1 The copyright of this thesis vests in the author. No quotation from it or information derived from it is to be published without full acknowledgement of the source. The thesis is to be used for private study or non- commercial research purposes only. Published by the University of Cape Town (UCT) in terms of the non-exclusive license granted to UCT by the author. University of Cape Town Declaration I, Timothy Spracklen, hereby declare that the work on which this dissertation/thesis is based is my original work (except where acknowledgements indicate otherwise) and that neither the whole work nor any part of it has been, is being, or is to be submitted for another degree in this or any other university. I empower the university to reproduce for the purpose of research either the whole or any portion of the contents in any manner whatsoever. Signature: Date: 18 December 2015 ' 2 Contents Abbreviations ………………………………………………………………………………….. 1 List of figures …………………………………………………………………………………... 6 List of tables ………………………………………………………………………………….... 7 Abstract ………………………………………………………………………………………… 10 1. Introduction …………………………………………………………………………………. 11 1.1 Cancer …………………………………………………………………………….. 11 1.2 Adverse drug reactions ………………………………………………………….. 12 1.3 Cisplatin …………………………………………………………………………… 12 1.3.1 Cisplatin’s mechanism of action ……………………………………………… 13 1.3.2 Adverse reactions to cisplatin therapy ……………………………………….
    [Show full text]
  • Broad and Thematic Remodeling of the Surface Glycoproteome on Isogenic
    bioRxiv preprint doi: https://doi.org/10.1101/808139; this version posted October 17, 2019. 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-ND 4.0 International license. Broad and thematic remodeling of the surface glycoproteome on isogenic cells transformed with driving proliferative oncogenes Kevin K. Leung1,5, Gary M. Wilson2,5, Lisa L. Kirkemo1, Nicholas M. Riley2,4, Joshua J. Coon2,3, James A. Wells1* 1Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA Departments of Chemistry2 and Biomolecular Chemistry3, University of Wisconsin- Madison, Madison, WI, 53706, USA 4Present address Department of Chemistry, Stanford University, Stanford, CA, 94305, USA 5These authors contributed equally *To whom correspondence should be addressed bioRxiv preprint doi: https://doi.org/10.1101/808139; this version posted October 17, 2019. 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-ND 4.0 International license. Abstract: The cell surface proteome, the surfaceome, is the interface for engaging the extracellular space in normal and cancer cells. Here We apply quantitative proteomics of N-linked glycoproteins to reveal how a collection of some 700 surface proteins is dramatically remodeled in an isogenic breast epithelial cell line stably expressing any of six of the most prominent proliferative oncogenes, including the receptor tyrosine kinases, EGFR and HER2, and downstream signaling partners such as KRAS, BRAF, MEK and AKT.
    [Show full text]
  • 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]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Down Regulation of Membrane-Bound Neu3 Constitutes a New
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Publications of the IAS Fellows IJC International Journal of Cancer Down regulation of membrane-bound Neu3 constitutes a new potential marker for childhood acute lymphoblastic leukemia and induces apoptosis suppression of neoplastic cells Chandan Mandal1, Cristina Tringali2, Susmita Mondal1, Luigi Anastasia2, Sarmila Chandra3, Bruno Venerando2 and Chitra Mandal1 1 Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, A Unit of Council of Scientific and Industrial Research, Govt of India, 4, Raja S. C. Mullick Road, Kolkata 700032, India 2 Department of Medical Chemistry, Biochemistry and Biotechnology, University of Milan, and IRCCS Policlinico San Donato, San Donato, Milan, Italy 3 Department of Hematology, Kothari Medical Centre, Kolkata 700027, India Membrane-linked sialidase Neu3 is a key enzyme for the extralysosomal catabolism of gangliosides. In this respect, it regulates pivotal cell surface events, including trans-membrane signaling, and plays an essential role in carcinogenesis. In this report, we demonstrated that acute lymphoblastic leukemia (ALL), lymphoblasts (primary cells from patients and cell lines) are characterized by a marked down-regulation of Neu3 in terms of both gene expression (230 to 40%) and enzymatic activity toward ganglioside GD1a (225.6 to 30.6%), when compared with cells from healthy controls. Induced overexpression of Neu3 in the ALL-cell line, MOLT-4, led to a significant increase of ceramide (166%) and to a parallel decrease of lactosylceramide (255%). These events strongly guided lymphoblasts to apoptosis, as we assessed by the decrease in Bcl2/ Bax ratio, the accumulation of Neu3 transfected cells in the sub G0–G1 phase of the cell cycle, the enhanced annexin-V positivity, the higher cleavage of procaspase-3.
    [Show full text]
  • Download This Article PDF Format
    Food & Function View Article Online PAPER View Journal | View Issue Human milk oligosaccharides and non-digestible carbohydrates prevent adhesion of specific Cite this: Food Funct., 2021, 12, 8100 pathogens via modulating glycosylation or inflammatory genes in intestinal epithelial cells Chunli Kong, *a,b Martin Beukema, b Min Wang,c Bart J. de Haanb and Paul de Vosb Human milk oligosaccharides (hMOs) and non-digestible carbohydrates (NDCs) are known to inhibit the adhesion of pathogens to the gut epithelium, but the mechanisms involved are not well understood. Here, the effects of 2’-FL, 3-FL, DP3–DP10, DP10–DP60 and DP30–DP60 inulins and DM7, DM55 and DM69 pectins were studied on pathogen adhesion to Caco-2 cells. As the growth phase influences viru- lence, E. coli ET8, E. coli LMG5862, E. coli O119, E. coli WA321, and S. enterica subsp. enterica LMG07233 Creative Commons Attribution-NonCommercial 3.0 Unported Licence. from both log and stationary phases were tested. Specificity for enteric pathogens was tested by including the lung pathogen K. pneumoniae LMG20218. Expression of the cell membrane glycosylation genes of galectin and glycocalyx and inflammatory genes was studied in the presence and absence of 2’-FL or NDCs. Inhibition of pathogen adhesion was observed for 2’-FL, inulins, and pectins. Pre-incubation with 2’-FL downregulated ICAM1, and pectins modified the glycosylation genes. In contrast, K. pneumoniae LMG20218 downregulated the inflammatory genes, but these were restored by pre-incubation with Received 22nd March 2021, pectins, which reduced the adhesion of K. pneumoniae LMG20218. In addition, DM69 pectin significantly Accepted 21st June 2021 upregulated the inflammatory genes.
    [Show full text]
  • Novel Cardiovascular Findings in Association with a POMT2
    European Journal of Human Genetics (2014) 22, 486–491 & 2014 Macmillan Publishers Limited All rights reserved 1018-4813/14 www.nature.com/ejhg ARTICLE Novel cardiovascular findings in association with a POMT2 mutation: three siblings with a-dystroglycanopathy Hugo R Martinez*,1, William J Craigen2, Monika Ummat3, Adekunle M Adesina4, Timothy E Lotze3 and John L Jefferies5 Dystroglycanopathies are a genetically heterogeneous subset of congenital muscular dystrophies that exhibit autosomal recessive inheritance and are characterized by abnormal glycosylation of a-dystroglycan. In particular, POMT2 (protein O-mannosyltransferase-2) mutations have been identified in congenital muscular dystrophy patients with a wide range of clinical involvement, ranging from the severe muscle-eye-brain disease and Walker–Warburg syndrome to limb girdle muscular dystrophy without structural brain or ocular involvement. Cardiovascular disease is thought to be uncommon in congenital muscular dystrophy, with rare reports of cardiac involvement. We describe three brothers aged 21, 19, and 17 years with an apparently homozygous POMT2 mutation who all presented with congenital muscular dystrophy, intellectual disabilities, and distinct cardiac abnormalities. All three brothers were homozygous for a p.Tyr666Cys missense mutation in exon 19 of the POMT2 gene. On screening echocardiograms, all siblings demonstrated significant dilatation of the aortic root and depressed left ventricular systolic function and/or left ventricular wall motion abnormalities. Our report is the first to document an association between POMT2 mutations and aortopathy with concomitant depressed left ventricular systolic function. On the basis of our findings, we suggest patients with POMT2 gene mutations be screened not only for myocardial dysfunction but also for aortopathy.
    [Show full text]
  • Congenital Disorders of Glycosylation from a Neurological Perspective
    brain sciences Review Congenital Disorders of Glycosylation from a Neurological Perspective Justyna Paprocka 1,* , Aleksandra Jezela-Stanek 2 , Anna Tylki-Szyma´nska 3 and Stephanie Grunewald 4 1 Department of Pediatric Neurology, Faculty of Medical Science in Katowice, Medical University of Silesia, 40-752 Katowice, Poland 2 Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland; [email protected] 3 Department of Pediatrics, Nutrition and Metabolic Diseases, The Children’s Memorial Health Institute, W 04-730 Warsaw, Poland; [email protected] 4 NIHR Biomedical Research Center (BRC), Metabolic Unit, Great Ormond Street Hospital and Institute of Child Health, University College London, London SE1 9RT, UK; [email protected] * Correspondence: [email protected]; Tel.: +48-606-415-888 Abstract: Most plasma proteins, cell membrane proteins and other proteins are glycoproteins with sugar chains attached to the polypeptide-glycans. Glycosylation is the main element of the post- translational transformation of most human proteins. Since glycosylation processes are necessary for many different biological processes, patients present a diverse spectrum of phenotypes and severity of symptoms. The most frequently observed neurological symptoms in congenital disorders of glycosylation (CDG) are: epilepsy, intellectual disability, myopathies, neuropathies and stroke-like episodes. Epilepsy is seen in many CDG subtypes and particularly present in the case of mutations
    [Show full text]
  • GM2 Gangliosidoses: Clinical Features, Pathophysiological Aspects, and Current Therapies
    International Journal of Molecular Sciences Review GM2 Gangliosidoses: Clinical Features, Pathophysiological Aspects, and Current Therapies Andrés Felipe Leal 1 , Eliana Benincore-Flórez 1, Daniela Solano-Galarza 1, Rafael Guillermo Garzón Jaramillo 1 , Olga Yaneth Echeverri-Peña 1, Diego A. Suarez 1,2, Carlos Javier Alméciga-Díaz 1,* and Angela Johana Espejo-Mojica 1,* 1 Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; [email protected] (A.F.L.); [email protected] (E.B.-F.); [email protected] (D.S.-G.); [email protected] (R.G.G.J.); [email protected] (O.Y.E.-P.); [email protected] (D.A.S.) 2 Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 110231, Colombia * Correspondence: [email protected] (C.J.A.-D.); [email protected] (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.) Received: 6 July 2020; Accepted: 7 August 2020; Published: 27 August 2020 Abstract: GM2 gangliosidoses are a group of pathologies characterized by GM2 ganglioside accumulation into the lysosome due to mutations on the genes encoding for the β-hexosaminidases subunits or the GM2 activator protein. Three GM2 gangliosidoses have been described: Tay–Sachs disease, Sandhoff disease, and the AB variant. Central nervous system dysfunction is the main characteristic of GM2 gangliosidoses patients that include neurodevelopment alterations, neuroinflammation, and neuronal apoptosis. Currently, there is not approved therapy for GM2 gangliosidoses, but different therapeutic strategies have been studied including hematopoietic stem cell transplantation, enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, and gene therapy.
    [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]
  • Salmonella Degrades the Host Glycocalyx Leading to Altered Infection and Glycan Remodeling
    UC Davis UC Davis Previously Published Works Title Salmonella Degrades the Host Glycocalyx Leading to Altered Infection and Glycan Remodeling. Permalink https://escholarship.org/uc/item/0nk8n7xb Journal Scientific reports, 6(1) ISSN 2045-2322 Authors Arabyan, Narine Park, Dayoung Foutouhi, Soraya et al. Publication Date 2016-07-08 DOI 10.1038/srep29525 Peer reviewed eScholarship.org Powered by the California Digital Library University of California www.nature.com/scientificreports OPEN Salmonella Degrades the Host Glycocalyx Leading to Altered Infection and Glycan Remodeling Received: 09 February 2016 Narine Arabyan1, Dayoung Park2, Soraya Foutouhi1, Allison M. Weis1, Bihua C. Huang1, Accepted: 17 June 2016 Cynthia C. Williams2, Prerak Desai1,†, Jigna Shah1,‡, Richard Jeannotte1,3,§, Nguyet Kong1, Published: 08 July 2016 Carlito B. Lebrilla2,4 & Bart C. Weimer1 Complex glycans cover the gut epithelial surface to protect the cell from the environment. Invasive pathogens must breach the glycan layer before initiating infection. While glycan degradation is crucial for infection, this process is inadequately understood. Salmonella contains 47 glycosyl hydrolases (GHs) that may degrade the glycan. We hypothesized that keystone genes from the entire GH complement of Salmonella are required to degrade glycans to change infection. This study determined that GHs recognize the terminal monosaccharides (N-acetylneuraminic acid (Neu5Ac), galactose, mannose, and fucose) and significantly (p < 0.05) alter infection. During infection, Salmonella used its two GHs sialidase nanH and amylase malS for internalization by targeting different glycan structures. The host glycans were altered during Salmonella association via the induction of N-glycan biosynthesis pathways leading to modification of host glycans by increasing fucosylation and mannose content, while decreasing sialylation.
    [Show full text]