Supplemental Data

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Data Article TCF7L2 is a master regulator of insulin production and processing ZHOU, Yuedan, et al. Abstract Genome-wide association studies have revealed >60 loci associated with type 2 diabetes (T2D), but the underlying causal variants and functional mechanisms remain largely elusive. Although variants in TCF7L2 confer the strongest risk of T2D among common variants by presumed effects on islet function, the molecular mechanisms are not yet well understood. Using RNA-sequencing, we have identified a TCF7L2-regulated transcriptional network responsible for its effect on insulin secretion in rodent and human pancreatic islets. ISL1 is a primary target of TCF7L2 and regulates proinsulin production and processing via MAFA, PDX1, NKX6.1, PCSK1, PCSK2 and SLC30A8, thereby providing evidence for a coordinated regulation of insulin production and processing. The risk T-allele of rs7903146 was associated with increased TCF7L2 expression, and decreased insulin content and secretion. Using gene expression profiles of 66 human pancreatic islets donors', we also show that the identified TCF7L2-ISL1 transcriptional network is regulated in a genotype-dependent manner. Taken together, these results demonstrate that not only synthesis of [...] Reference ZHOU, Yuedan, et al. TCF7L2 is a master regulator of insulin production and processing. Human Molecular Genetics, 2014, vol. 23, no. 24, p. 6419-6431 DOI : 10.1093/hmg/ddu359 PMID : 25015099 Available at: http://archive-ouverte.unige.ch/unige:45177 Disclaimer: layout of this document may differ from the published version. 1 / 1 Gene ID log(Fold Change) logCPM Likelihood ratio P Value value FDR correc Description Gene Symbol ENSRNOG00000020811 -1.884144098 6.14183448 182.8588856 1.15E-41 1.74E-37 Interleukin-6 receptor subunit alpha [Source:UniProtKB/Swiss-Prot;Acc:P22273] Il6r ENSRNOG00000017619 3.250568327 6.22643 180.5260665 3.72E-41 2.80E-37 Retinal dehydrogenase 1 [Source:UniProtKB/Swiss-Prot;Acc:P51647] Aldh1a1 ENSRNOG00000003251 -2.048987017 4.87056849 136.3135032 1.70E-31 6.42E-28 beta-1,3-galactosyltransferase 2 [Source:RefSeq peptide;Acc:NP_001102962] B3galt2 ENSRNOG00000007512 1.692218023 6.29829189 136.3430377 1.68E-31 6.42E-28 signal recognition particle 14 kDa protein [Source:RefSeq peptide;Acc:NP_001099967] Srp14 ENSRNOG00000033531 -2.18464309 7.80561272 133.5052453 7.01E-31 2.08E-27 Voltage-dependent calcium channel subunit alpha-2/delta-1Voltage-dependent calcium channel subunit alpha-2-1Voltage- Cacna2d1 dependent calcium channel subunit delta-1 [Source:UniProtKB/Swiss-Prot;Acc:P54290] ENSRNOG00000018082 -1.833829027 6.88465482 133.1739369 8.28E-31 2.08E-27 Sulfate transporter [Source:UniProtKB/Swiss-Prot;Acc:O70531] Slc26a2 ENSRNOG00000014550 -1.427981119 6.21976673 124.1122101 7.96E-29 1.72E-25 RGD1308448 ENSRNOG00000020578 -1.468888395 6.16475527 118.3789909 1.43E-27 2.70E-24 Carcinoembryonic antigen-related cell adhesion molecule 1 [Source:UniProtKB/Swiss-Prot;Acc:P16573] Ceacam1 ENSRNOG00000014740 -3.095060926 3.57761596 116.0514741 4.63E-27 7.76E-24 GastrinBig gastrinGastrin [Source:UniProtKB/Swiss-Prot;Acc:P04563] Gast ENSRNOG00000011352 1.472419643 7.49273232 115.8194425 5.21E-27 7.85E-24 Furin [Source:UniProtKB/Swiss-Prot;Acc:P23377] Furin ENSRNOG00000007583 1.189980628 7.6066405 108.6959566 1.89E-25 2.59E-22 Glycogen phosphorylase, brain form [Source:UniProtKB/Swiss-Prot;Acc:P53534] Pygb ENSRNOG00000031163 2.731920784 3.69878814 101.875131 5.91E-24 7.43E-21 NF-kappa-B inhibitor zeta [Source:RefSeq peptide;Acc:NP_001100565] Nfkbiz ENSRNOG00000003492 -1.680421521 5.9035401 98.91506493 2.64E-23 3.06E-20 Growth arrest-specific protein 7 [Source:UniProtKB/Swiss-Prot;Acc:O55148] Gas7 ENSRNOG00000034025 -1.685605215 5.49014494 98.20133676 3.78E-23 4.07E-20 protein tyrosine phosphatase, receptor type, J [Source:MGI Symbol;Acc:MGI:104574] Ptprj ENSRNOG00000010915 -1.365012655 7.59666154 96.93629925 7.16E-23 7.20E-20 Endoplasmic reticulum metallopeptidase 1 [Source:UniProtKB/Swiss-Prot;Acc:Q6UPR8] Ermp1 ENSRNOG00000007808 -2.423152566 4.22623711 95.66550493 1.36E-22 1.28E-19 Nucleosome assembly protein 1-like 5 [Source:UniProtKB/Swiss-Prot;Acc:Q5PPG6] Nap1l5 ENSRNOG00000025406 -2.126140228 4.61588368 92.80178369 5.78E-22 5.13E-19 Protein LOC100360623 [Source:UniProtKB/TrEMBL;Acc:F1LW74] Iqgap2 ENSRNOG00000008401 1.661230655 5.10569967 92.52384436 6.65E-22 5.57E-19 caspase recruitment domain-containing protein 10 [Source:RefSeq peptide;Acc:NP_001124026] Card10 ENSRNOG00000008088 -1.41145632 5.61211989 89.99888389 2.38E-21 1.89E-18 BTB/POZ domain-containing protein 3 [Source:RefSeq peptide;Acc:NP_001101252] Btbd3 ENSRNOG00000012181 -1.935960177 5.93691867 89.04029023 3.87E-21 2.92E-18 Lipoprotein lipase [Source:UniProtKB/Swiss-Prot;Acc:Q06000] Lpl ENSRNOG00000011439 -1.830216615 4.57317382 88.19842921 5.92E-21 4.25E-18 G protein-coupled receptor kinase 5 [Source:UniProtKB/Swiss-Prot;Acc:Q62833] Grk5 ENSRNOG00000012235 -2.895221597 2.85748939 87.89385692 6.91E-21 4.73E-18 G-substrate [Source:RefSeq peptide;Acc:NP_703197] Gsbs ENSRNOG00000011522 -2.391710509 3.22495114 81.45312465 1.79E-19 1.18E-16 Potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 1 [Source:UniProtKB/Swiss- Hcn1 ENSRNOG00000036661 -1.46704422 5.15806374 81.13873348 2.10E-19 1.32E-16 ras-related protein Rab-40B [Source:RefSeq peptide;Acc:NP_001100546] Rab40b ENSRNOG00000006523 -1.145210832 7.27846694 80.86454888 2.42E-19 1.46E-16 ADP-ribosylation factor-like 6 interacting protein 2 [Source:RefSeq peptide;Acc:NP_001094141] Atl2 ENSRNOG00000002654 2.102935356 3.62821177 80.31720584 3.19E-19 1.85E-16 Protein FAM120C [Source:UniProtKB/TrEMBL;Acc:D3ZNI4] FAM120C ENSRNOG00000026277 1.93434437 4.31273257 79.71236406 4.33E-19 2.42E-16 zinc finger CCCH domain-containing protein 6 [Source:RefSeq peptide;Acc:NP_001101242] Zc3h6 ENSRNOG00000024346 -2.637854506 2.86869803 79.4445456 4.96E-19 2.58E-16 Placenta-expressed transcript 1 protein [Source:UniProtKB/Swiss-Prot;Acc:Q5HZW7] LOC363060 ENSRNOG00000028169 1.345026004 7.40201982 79.45114326 4.94E-19 2.58E-16 protein phosphatase 1L [Source:RefSeq peptide;Acc:NP_001101151] Ppm1l ENSRNOG00000010086 1.611745287 5.42399559 79.2901701 5.36E-19 2.65E-16 pleiomorphic adenoma gene-like 2 [Source:RefSeq peptide;Acc:NP_001099998] Plagl2 ENSRNOG00000031997 -1.532688187 6.12050701 79.26019727 5.44E-19 2.65E-16 tubulin tyrosine ligase-like family, member 7 [Source:MGI Symbol;Acc:MGI:1918142] Ttll7 ENSRNOG00000016210 -1.650728107 4.42459748 77.41710161 1.38E-18 6.52E-16 MICAL C-terminal-like protein [Source:UniProtKB/Swiss-Prot;Acc:Q4G091] Micalcl ENSRNOG00000007427 1.334437858 5.2330087 76.78368015 1.91E-18 8.72E-16 Ectonucleoside triphosphate diphosphohydrolase 6 [Source:UniProtKB/Swiss-Prot;Acc:Q9ER31] Entpd6 ENSRNOG00000004747 -1.460640406 8.5304176 76.3103252 2.42E-18 1.08E-15 Zinc transporter 8 [Source:UniProtKB/Swiss-Prot;Acc:P0CE46] Slc30a8 ENSRNOG00000019559 -1.24220285 6.41624815 75.5102877 3.64E-18 1.57E-15 ELL-associated factor 1 [Source:RefSeq peptide;Acc:NP_001100763] Eaf1 ENSRNOG00000018911 -1.077842242 6.19674891 75.27551277 4.09E-18 1.71E-15 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 36-phosphofructo-2-kinaseFructose-2,6-bisphosphatase Pfkfb3 [Source:UniProtKB/Swiss-Prot;Acc:O35552] ENSRNOG00000016704 -1.322711989 7.17712665 74.95251575 4.82E-18 1.97E-15 Prenylcysteine oxidase [Source:UniProtKB/Swiss-Prot;Acc:Q99ML5] Pcyox1 ENSRNOG00000008055 -1.13286363 6.45256662 73.33881001 1.09E-17 4.33E-15 G1/S-specific cyclin-E2 [Source:RefSeq peptide;Acc:NP_001102126] Ccne2 ENSRNOG00000007989 1.573907077 4.95414373 72.25411701 1.89E-17 7.32E-15 Carbohydrate sulfotransferase 1 [Source:UniProtKB/Swiss-Prot;Acc:Q5RJQ0] Chst1 ENSRNOG00000010389 1.825619605 4.45619599 72.16015284 1.98E-17 7.48E-15 Protein NDRG2 [Source:UniProtKB/Swiss-Prot;Acc:Q8VBU2] Ndrg2 ENSRNOG00000003873 -1.258524309 8.27317166 69.9225615 6.17E-17 2.27E-14 Carboxypeptidase D [Source:UniProtKB/Swiss-Prot;Acc:Q9JHW1] Cpd ENSRNOG00000020151 -1.082052889 9.38456979 69.86331384 6.36E-17 2.28E-14 Cadherin-1E-Cad/CTF1E-Cad/CTF2E-Cad/CTF3 [Source:UniProtKB/Swiss-Prot;Acc:Q9R0T4] Cdh1 ENSRNOG00000021182 1.152885053 7.85036331 69.12500674 9.24E-17 3.24E-14 Synaptic vesicle glycoprotein 2A [Source:UniProtKB/Swiss-Prot;Acc:Q02563] Sv2a ENSRNOG00000027230 -1.621175318 5.39870771 68.17640157 1.50E-16 5.12E-14 formin homology 2 domain containing 3 (Fhod3), mRNA [Source:RefSeq mRNA;Acc:NM_001271332] Fhod3 ENSRNOG00000017983 1.064445121 6.12881425 67.90526741 1.72E-16 5.75E-14 Ubiquitin-associated domain-containing protein 1 [Source:UniProtKB/Swiss-Prot;Acc:Q5XIR9] Ubac1 ENSRNOG00000042246 -1.06814083 8.2185739 67.84660453 1.77E-16 5.79E-14 RCG54937Uncharacterized protein [Source:UniProtKB/TrEMBL;Acc:D3ZI94] LOC690918 ENSRNOG00000027514 0.985567875 7.0962325 67.55145813 2.05E-16 6.59E-14 NEDD8-conjugating enzyme Ubc12 [Source:RefSeq peptide;Acc:NP_001101941] Ube2m ENSRNOG00000013774 0.948789382 7.13294968 67.2392396 2.40E-16 7.56E-14 Lamin-B1 [Source:UniProtKB/Swiss-Prot;Acc:P70615] Lmnb1 ENSRNOG00000009029 -1.212945481 6.59228518 67.13631457 2.53E-16 7.80E-14 Neuroplastin [Source:UniProtKB/Swiss-Prot;Acc:P97546] Nptn ENSRNOG00000002922 -1.451922294 4.29423384 66.6764751 3.20E-16 9.65E-14 Adenosine receptor A2b [Source:UniProtKB/Swiss-Prot;Acc:P29276] Adora2b ENSRNOG00000004828 -2.269841398 3.35863648 66.58233116 3.36E-16 9.73E-14 Activin receptor type-1C [Source:UniProtKB/Swiss-Prot;Acc:P70539] Acvr1c ENSRNOG00000005609 -1.193132577 6.80065742 66.61173486 3.31E-16 9.73E-14 Neurogenic differentiation
Recommended publications
  • Are Zinc Transporter Type 8 Antibodies a Marker of Autoimmune Thyroiditis in Non-Obese Adults with New-Onset Diabetes?
    A Rogowicz-Frontczak and ZnT8A are associated with ATPO 170:4 651–658 Clinical Study others in diabetes Are zinc transporter type 8 antibodies a marker of autoimmune thyroiditis in non-obese adults with new-onset diabetes? Anita Rogowicz-Frontczak1, Dorota Zozulin´ ska-Zio´ łkiewicz1, Monika Litwinowicz2, Paweł Niedz´wiecki1, Krystyna Wyka3 and Bogna Wierusz-Wysocka1 Correspondence should be addressed to Departments of 1Internal Medicine and Diabetology and 2Internal Diseases, Metabolic Disorders and Dietetics, A Rogowicz-Frontczak Poznan University of Medical Sciences, Mickiewicza 2, 60-834 Poznan, Poland and 3Medical University of Lodz, Email Lodz, Poland [email protected] Abstract Objective: The diagnosis of autoimmune diabetes in non-obese adults is based on the detection of glutamic acid decarboxylase autoantibodies (GADA), islet cell antibodies (ICA) and antibodies to tyrosine phosphatase (IA-2A). Zinc transporter 8 (ZnT8) has been identified as a new autoantigen in patients with type 1 diabetes mellitus. The coincidence of autoimmune thyroiditis (AITD) with diabetes is common; therefore, screening of TSH and thyroid peroxidase antibodies (ATPO) is recommended during the diagnosis of diabetes. In this study, we determined whether the occurrence of islet autoantibodies is associated with a positive titre of ATPO in newly diagnosed adult-onset autoimmune diabetic patients. Design and methods: The study involved 80 non-obese adults aged 44 (interquartile range (IQR): 37–51) years with a BMI of 24.0 (IQR: 22.2–26.0) kg/m2 and new-onset diabetes. The markers of autoimmune diabetes (GADA, ICA, IA-2A and ZnT8A), TSH and thyroid peroxidase antibodies (ATPO) were evaluated. Results: In the study population, 70% (nZ56) of the subjects were positive for at least one of the four assessed markers of autoimmune diabetes (83.9% GADA, 62.5% ICA, 42.8% IA-2A and 33% ZnT8A) and 37.5% of the subjects were positive for ATPO.
    [Show full text]
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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 Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • Supplemental Figure 1. Vimentin
    Double mutant specific genes Transcript gene_assignment Gene Symbol RefSeq FDR Fold- FDR Fold- FDR Fold- ID (single vs. Change (double Change (double Change wt) (single vs. wt) (double vs. single) (double vs. wt) vs. wt) vs. single) 10485013 BC085239 // 1110051M20Rik // RIKEN cDNA 1110051M20 gene // 2 E1 // 228356 /// NM 1110051M20Ri BC085239 0.164013 -1.38517 0.0345128 -2.24228 0.154535 -1.61877 k 10358717 NM_197990 // 1700025G04Rik // RIKEN cDNA 1700025G04 gene // 1 G2 // 69399 /// BC 1700025G04Rik NM_197990 0.142593 -1.37878 0.0212926 -3.13385 0.093068 -2.27291 10358713 NM_197990 // 1700025G04Rik // RIKEN cDNA 1700025G04 gene // 1 G2 // 69399 1700025G04Rik NM_197990 0.0655213 -1.71563 0.0222468 -2.32498 0.166843 -1.35517 10481312 NM_027283 // 1700026L06Rik // RIKEN cDNA 1700026L06 gene // 2 A3 // 69987 /// EN 1700026L06Rik NM_027283 0.0503754 -1.46385 0.0140999 -2.19537 0.0825609 -1.49972 10351465 BC150846 // 1700084C01Rik // RIKEN cDNA 1700084C01 gene // 1 H3 // 78465 /// NM_ 1700084C01Rik BC150846 0.107391 -1.5916 0.0385418 -2.05801 0.295457 -1.29305 10569654 AK007416 // 1810010D01Rik // RIKEN cDNA 1810010D01 gene // 7 F5 // 381935 /// XR 1810010D01Rik AK007416 0.145576 1.69432 0.0476957 2.51662 0.288571 1.48533 10508883 NM_001083916 // 1810019J16Rik // RIKEN cDNA 1810019J16 gene // 4 D2.3 // 69073 / 1810019J16Rik NM_001083916 0.0533206 1.57139 0.0145433 2.56417 0.0836674 1.63179 10585282 ENSMUST00000050829 // 2010007H06Rik // RIKEN cDNA 2010007H06 gene // --- // 6984 2010007H06Rik ENSMUST00000050829 0.129914 -1.71998 0.0434862 -2.51672
    [Show full text]
  • Upregulation of Peroxisome Proliferator-Activated Receptor-Α And
    Upregulation of peroxisome proliferator-activated receptor-α and the lipid metabolism pathway promotes carcinogenesis of ampullary cancer Chih-Yang Wang, Ying-Jui Chao, Yi-Ling Chen, Tzu-Wen Wang, Nam Nhut Phan, Hui-Ping Hsu, Yan-Shen Shan, Ming-Derg Lai 1 Supplementary Table 1. Demographics and clinical outcomes of five patients with ampullary cancer Time of Tumor Time to Age Differentia survival/ Sex Staging size Morphology Recurrence recurrence Condition (years) tion expired (cm) (months) (months) T2N0, 51 F 211 Polypoid Unknown No -- Survived 193 stage Ib T2N0, 2.41.5 58 F Mixed Good Yes 14 Expired 17 stage Ib 0.6 T3N0, 4.53.5 68 M Polypoid Good No -- Survived 162 stage IIA 1.2 T3N0, 66 M 110.8 Ulcerative Good Yes 64 Expired 227 stage IIA T3N0, 60 M 21.81 Mixed Moderate Yes 5.6 Expired 16.7 stage IIA 2 Supplementary Table 2. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of an ampullary cancer microarray using the Database for Annotation, Visualization and Integrated Discovery (DAVID). This table contains only pathways with p values that ranged 0.0001~0.05. KEGG Pathway p value Genes Pentose and 1.50E-04 UGT1A6, CRYL1, UGT1A8, AKR1B1, UGT2B11, UGT2A3, glucuronate UGT2B10, UGT2B7, XYLB interconversions Drug metabolism 1.63E-04 CYP3A4, XDH, UGT1A6, CYP3A5, CES2, CYP3A7, UGT1A8, NAT2, UGT2B11, DPYD, UGT2A3, UGT2B10, UGT2B7 Maturity-onset 2.43E-04 HNF1A, HNF4A, SLC2A2, PKLR, NEUROD1, HNF4G, diabetes of the PDX1, NR5A2, NKX2-2 young Starch and sucrose 6.03E-04 GBA3, UGT1A6, G6PC, UGT1A8, ENPP3, MGAM, SI, metabolism
    [Show full text]
  • Propranolol-Mediated Attenuation of MMP-9 Excretion in Infants with Hemangiomas
    Supplementary Online Content Thaivalappil S, Bauman N, Saieg A, Movius E, Brown KJ, Preciado D. Propranolol-mediated attenuation of MMP-9 excretion in infants with hemangiomas. JAMA Otolaryngol Head Neck Surg. doi:10.1001/jamaoto.2013.4773 eTable. List of All of the Proteins Identified by Proteomics This supplementary material has been provided by the authors to give readers additional information about their work. © 2013 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 10/01/2021 eTable. List of All of the Proteins Identified by Proteomics Protein Name Prop 12 mo/4 Pred 12 mo/4 Δ Prop to Pred mo mo Myeloperoxidase OS=Homo sapiens GN=MPO 26.00 143.00 ‐117.00 Lactotransferrin OS=Homo sapiens GN=LTF 114.00 205.50 ‐91.50 Matrix metalloproteinase‐9 OS=Homo sapiens GN=MMP9 5.00 36.00 ‐31.00 Neutrophil elastase OS=Homo sapiens GN=ELANE 24.00 48.00 ‐24.00 Bleomycin hydrolase OS=Homo sapiens GN=BLMH 3.00 25.00 ‐22.00 CAP7_HUMAN Azurocidin OS=Homo sapiens GN=AZU1 PE=1 SV=3 4.00 26.00 ‐22.00 S10A8_HUMAN Protein S100‐A8 OS=Homo sapiens GN=S100A8 PE=1 14.67 30.50 ‐15.83 SV=1 IL1F9_HUMAN Interleukin‐1 family member 9 OS=Homo sapiens 1.00 15.00 ‐14.00 GN=IL1F9 PE=1 SV=1 MUC5B_HUMAN Mucin‐5B OS=Homo sapiens GN=MUC5B PE=1 SV=3 2.00 14.00 ‐12.00 MUC4_HUMAN Mucin‐4 OS=Homo sapiens GN=MUC4 PE=1 SV=3 1.00 12.00 ‐11.00 HRG_HUMAN Histidine‐rich glycoprotein OS=Homo sapiens GN=HRG 1.00 12.00 ‐11.00 PE=1 SV=1 TKT_HUMAN Transketolase OS=Homo sapiens GN=TKT PE=1 SV=3 17.00 28.00 ‐11.00 CATG_HUMAN Cathepsin G OS=Homo
    [Show full text]
  • Figure S1. DMD Module Network. the Network Is Formed by 260 Genes from Disgenet and 1101 Interactions from STRING. Red Nodes Are the Five Seed Candidate Genes
    Figure S1. DMD module network. The network is formed by 260 genes from DisGeNET and 1101 interactions from STRING. Red nodes are the five seed candidate genes. Figure S2. DMD module network is more connected than a random module of the same size. It is shown the distribution of the largest connected component of 10.000 random modules of the same size of the DMD module network. The green line (x=260) represents the DMD largest connected component, obtaining a z-score=8.9. Figure S3. Shared genes between BMD and DMD signature. A) A meta-analysis of three microarray datasets (GSE3307, GSE13608 and GSE109178) was performed for the identification of differentially expressed genes (DEGs) in BMD muscle biopsies as compared to normal muscle biopsies. Briefly, the GSE13608 dataset included 6 samples of skeletal muscle biopsy from healthy people and 5 samples from BMD patients. Biopsies were taken from either biceps brachii, triceps brachii or deltoid. The GSE3307 dataset included 17 samples of skeletal muscle biopsy from healthy people and 10 samples from BMD patients. The GSE109178 dataset included 14 samples of controls and 11 samples from BMD patients. For both GSE3307 and GSE10917 datasets, biopsies were taken at the time of diagnosis and from the vastus lateralis. For the meta-analysis of GSE13608, GSE3307 and GSE109178, a random effects model of effect size measure was used to integrate gene expression patterns from the two datasets. Genes with an adjusted p value (FDR) < 0.05 and an │effect size│>2 were identified as DEGs and selected for further analysis. A significant number of DEGs (p<0.001) were in common with the DMD signature genes (blue nodes), as determined by a hypergeometric test assessing the significance of the overlap between the BMD DEGs and the number of DMD signature genes B) MCODE analysis of the overlapping genes between BMD DEGs and DMD signature genes.
    [Show full text]
  • A Consensus Report from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)
    Diabetologia https://doi.org/10.1007/s00125-020-05181-w CONSENSUS REPORT Precision medicine in diabetes: a Consensus Report from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) Wendy K. Chung1,2 & Karel Erion3 & Jose C. Florez4,5,6,7,8 & Andrew T. Hattersley9 & Marie-France Hivert5,10 & Christine G. Lee11 & Mark I. McCarthy12,13,14 & John J. Nolan15 & Jill M. Norris16 & Ewan R. Pearson17 & Louis Philipson 18,19 & Allison T. McElvaine20 & William T. Cefalu11 & Stephen S. Rich21,22 & Paul W. Franks23,24 # European Association for the Study of Diabetes and American Diabetes Association 2020 Abstract The convergence of advances in medical science, human biology, data science and technology has enabled the generation of new insights into the phenotype known as ‘diabetes’. Increased knowledge of this condition has emerged from popu- lations around the world, illuminating the differences in how diabetes presents, its variable prevalence and how best practice in treatment varies between populations. In parallel, focus has been placed on the development of tools for the application of precision medicine to numerous conditions. This Consensus Report presents the American Diabetes Association (ADA) Precision Medicine in Diabetes Initiative in partnership with the European Association for the Study of Diabetes (EASD), including its mission, the current state of the field and prospects for the future. Expert opinions are presented on areas of precision diagnostics and precision therapeutics (including prevention and treatment) and key barriers to and opportunities for implementation of precision diabetes medicine, with better care and outcomes around the globe, are highlighted. Cases where precision diagnosis is already feasible and effective (i.e.
    [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]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • Breast Cancer Antiestrogen Resistance-3 Expression Regulates Breast Cancer Cell Migration Through Promotion of P130cas Membrane Localization Andmembrane Ruffling
    Research Article Breast Cancer Antiestrogen Resistance-3 Expression Regulates Breast Cancer Cell Migration through Promotion of p130Cas Membrane Localization andMembrane Ruffling Randy S. Schrecengost,1 Rebecca B. Riggins,2 Keena S. Thomas,1 Michael S. Guerrero,1 and Amy H. Bouton1 1Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia and 2Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia Abstract antiestrogen resistance-1 (BCAR1; also known as p130Cas; refs. 2, 3). Antiestrogens such as tamoxifen are widely used in the clinic A second protein, BCAR3 (the human homologue of the murine to treat estrogen receptor–positive breast tumors. Resistance protein AND-34), was identified in a genetic screen along with to tamoxifen can occur either de novo or develop over time in BCAR1 as a gene product whose overexpression conferred tamoxifen resistance in vitro (4). BCAR3 is a member of the novel a large proportion of these tumors. Additionally, resistance is S p associated with enhanced motility and invasiveness in vitro. rc homology 2 (SH2)–containing rotein (NSP) family that One molecule that has been implicated in tamoxifen resis- includes two other members, Chat/SHEP1 and NSP1. These tance, breast cancer antiestrogen resistance-3 (BCAR3), has proteins share a common domain structure consisting of an also been shown to regulate migration of fibroblasts. In this amino-terminal SH2 domain and a carboxyl-terminal domain with study, we investigated the role of BCAR3 in breast cancer cell sequence homology to the Cdc25-family of guanine nucleotide exchange factors (GEF). Several studies have shown that BCAR3 migration and invasion.
    [Show full text]
  • 9-Azido Analogs of Three Sialic Acid Forms for Metabolic Remodeling Of
    Supporting Information 9-Azido Analogs of Three Sialic Acid Forms for Metabolic Remodeling of Cell-Surface Sialoglycans Bo Cheng,†,‡ Lu Dong,†,§ Yuntao Zhu,†,‡ Rongbing Huang,†,‡ Yuting Sun,†,‖ Qiancheng You,†,‡ Qitao Song,†,§ James C. Paton, ∇ Adrienne W. Paton,∇ and Xing Chen*,†,‡,§,⊥,# †College of Chemistry and Molecular Engineering, ‡Beijing National Laboratory for Molecular Sciences, §Peking−Tsinghua Center for Life Sciences,‖Academy for Advanced Interdisciplinary Studies, ⊥Synthetic and Functional Biomolecules Center, and #Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China ∇Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide SA 5005, Australia Page S1 Table of Contents: Scheme S1.……………………………………………………….........……………. S3 Figure S1……………………………………………………..………..……………. S3 Figure S2……………………………………………………..………..…………… S4 Figure S3……………………………………………………..………..…………… S4 Figure S4……………………………………………………..………..…………… S5 Figure S5……………………………………………………..………..…………… S6 Figure S6……………………………………………………..………..…………….S7 Figure S7……………………………………………………..………..…………….S8 Figure S8……………………………………………………..………..…………….S9 Experimental Procedures……………………………….…........…………....S10-S27 Table S1………………………………………………..………..…………….S28-S48 Supporting Reference……………………………………………….......………...S49 Page S2 Scheme S1. Synthesis of 9AzNeu5Gc Figure S1: a, b, c, d) Representative scatter plots (FSC vs. SSC) and histograms of flow cytometry analysis
    [Show full text]
  • Whole Proteome Analysis of Human Tankyrase Knockout Cells Reveals Targets of Tankyrase- Mediated Degradation
    ARTICLE DOI: 10.1038/s41467-017-02363-w OPEN Whole proteome analysis of human tankyrase knockout cells reveals targets of tankyrase- mediated degradation Amit Bhardwaj1, Yanling Yang2, Beatrix Ueberheide2 & Susan Smith1 Tankyrase 1 and 2 are poly(ADP-ribose) polymerases that function in pathways critical to cancer cell growth. Tankyrase-mediated PARylation marks protein targets for proteasomal 1234567890 degradation. Here, we generate human knockout cell lines to examine cell function and interrogate the proteome. We show that either tankyrase 1 or 2 is sufficient to maintain telomere length, but both are required to resolve telomere cohesion and maintain mitotic spindle integrity. Quantitative analysis of the proteome of tankyrase double knockout cells using isobaric tandem mass tags reveals targets of degradation, including antagonists of the Wnt/β-catenin signaling pathway (NKD1, NKD2, and HectD1) and three (Notch 1, 2, and 3) of the four Notch receptors. We show that tankyrases are required for Notch2 to exit the plasma membrane and enter the nucleus to activate transcription. Considering that Notch signaling is commonly activated in cancer, tankyrase inhibitors may have therapeutic potential in targeting this pathway. 1 Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016, USA. 2 Proteomics Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA. Correspondence and requests for materials should be addressed to S.S. (email: [email protected]) NATURE COMMUNICATIONS | 8: 2214 | DOI: 10.1038/s41467-017-02363-w | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02363-w ankyrases function in cellular pathways that are critical to function in human cells will provide insights into the clinical cancer cell growth including telomere cohesion and length utility of tankyrase inhibitors.
    [Show full text]