Supplementary Information

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Information SUPPLEMENTARY INFORMATION 1. SUPPLEMENTARY FIGURE LEGENDS Supplementary Figure 1. Long-term exposure to sorafenib increases the expression of progenitor cell-like features. A) mRNA expression levels of PROM-1 (CD133), THY-1 (CD90), EpCAM, KRT19, and VIM assessed by quantitative real-time PCR. Data represent the mean expression value for a gene in each phenotypic type of cells, displayed as fold-changes normalized to 1 (expression value of its corresponding parental non-treated cell line). Expression level is relative to the GAPDH gene. Bars indicate standard deviation. Significant statistical differences are set at p<0.05. B) Immunocitochemical staining of CD90 and vimentin in Hep3B sorafenib resistant cell line and its parental cell line. C) Western blot analysis comparing protein levels in resistant Hu6 and Hep3B cells vs their corresponding parental cells lines. Supplementary Figure 2. Efficacy of gene silencing of IGF1R and FGFR1 and evaluation of MAPK14 signaling activation. IGF1R and FGFR1 knockdown expression 48h after transient transfection with siRNAs (50 nM), in non-treated parental cells and sorafenib-acquired resistant tumor derived cells was assessed by quantitative RT-PCR (A) and western blot (B). C) Activation status of MAPK14 signaling was evaluated by western blot analysis in vivo, in tumors with acquired resistance to sorafenib in comparison to non-treated tumors (right panel), as well as in in vitro, in sorafenib resistant cell lines vs parental non-treated. Supplementary Figure 3. Gene expression levels of several pro-angiogenic factors. mRNA expression levels of FGF1, FGF2, VEGFA, IL8, ANGPT2, KDR, FGFR3, FGFR4 assessed by quantitative real-time PCR in tumors harvested from mice. Plotted data represent the mean expression value for a gene in each experimental group, displayed as fold-changes normalized to 1 (mean expression value in non- treated animals). Expression level is relative to the GAPDH. Bars indicate standard deviation. Significant statistical differences are set at p<0.05. Supplementary Figure 4. Increased expression and activation of FGFR1 and IGF1R in acquired resistance tumors. A) Immunohistochemical analysis of FGFR1 and IGF1R protein levels in sorafenib-sensitive and sorafenib-acquired resistant tumors. B) Immunostaining showing activation (phosphorylation) of FGFR1 and IGF1R in viable clusters of cells close to necrotic areas in sorafenib-resistant tumors. 2. SUPPLEMENTARY MATERIAL & METHODS Sphere formation assay To obtain single cell suspension, subcutaneous Huh7-derived tumors were excised and mechanically dissociated by gentle pipetting in Ca/Mg-free PBS on ice, and filtered through a 40 µm cell strainer (BD Falcon). Then, cells were exposed to red cell lysis buffer (Sigma-Aldrich) to remove red blood cells. No enzymatic digestion was used to isolate cells. Cell viability was quantified in a Neubauer cytometer using the trypan blue (1%; Sigma Aldrich) exclusion method. After tumor harvest and generation of single cell suspensions tumor cells where directly cultured into non-adherent conditions to allow the formation of spheroids as previously described(1). Briefly, cells were grown in serum free epithelial growth medium (MEGM, BioWhittaker) supplemented with B27 (Invitrogen), 20 ng/mL EGF and 20 ng/mL bFGF (BD Biosciences), and 4 μg/mL insulin (Sigma). Live cells were then plated in 96-well ultra-low attachment plates (Corning) at limiting dilution (10, 102, 103 and 104 cells) and sphere formation was assessed microscopically after 10 days of growth. Cell culture, reagents and drugs Parental Huh7, Hep3B and H6 cell lines were cultured in DMEN with 10% fetal bovine serum (Gibco, Grand Island, NY). The Hep3B-sorafenib resistant cell line(2) and H6- sorafenib resistant cells line were kindly provided Dr. Desbois-Mouthon (INSERM, Paris) and grown in the presence of sorafenib at a concentration of 4 µM and 3 µM, respectively. Tumor-derived spheres were grown under non-adherent culture conditions in serum free epithelial growth medium (MEGM, BioWhittaker) supplemented with B27 (Invitrogen), 20 ng/mL EGF and 20 ng/mL bFGF (BD Biosciences), and 4 μg/mL insulin (Sigma). Cells from sorafenib-acquired resistant tumors were grown in the presence of 4 µM of sorafenib. Sorafenib was purchased from LC Laboratories (Woburn, MA). Linsitinib (IGF1R inhibitor) and BGJ398 (pan inhibitor of FGFR1, -2, -3) were purchased from BioVision (Milpitas, CA). Brivanib (FGFR inhibitor) was provided by Bristol-Myers Squibb (BMS, New York, NY). In vitro and the in vivo doses of the compounds were chosen after revision of published preclinical studies, as well as, by following the manufacturer’s recommendations(3-5). For in vivo oral administration, sorafenib was dissolved in ethanol 95%/cremophor EL/sterile water (12.5:12.5:75%) at 30 mg/kg/day dosage. Brivanib was dissolved following BMS recommendations and orally dosed at 100 mg/kg/day. For MTT assay dissociated spheres were plated at a density of 5,000 cells/well in 96-well ultra-low attachment. Huh7, Hep3B, Hep3B-sora, Huh6 and Huh6-sora cells were seeded at 2,500-3,000 cells/well density under adherent conditions. Then cell viability was evaluated after 48-72 hours in the presence of sorafenib or/and other inhibitors (0.1-10 M). Each experimental condition was measured in triplicate in 3 independent experiments. siRNA transfection Cells were plated on 96-well plates or 6-well plates and transiently transfected with Silencer® Select siRNAs from Ambion (Life technologies, Inc.) (cat. #4390824 ID: #s7211 for IGF1R siRNA, ID: #s5164 for FGFR1 siRNA, cat. # 4390649 for control GAPDH siRNA, cat. 4390646 negative control #2) at 50 nM using Lipofectamine® 3000 tranfection reagent (Life technologies, Life Technologies) following manufacturer’s intructions. 4 hours post transfection sorafenib at 3 µM was added to the medium and cells were allowed to grown for additional 48 hours. Protein extraction and Western blot analysis Cells were plated into 15 cm culture dishes and grown to subconfluency. Total protein was quantified using Bradford Reagent (Sigma, Saint Louis, MO). To isolate cytoplasmic proteins cells were lysed with RIPA buffer plus protease and phosphatase inhibitors (50 mM Tris pH=7.4, 150 mM NaCl, 1% Trithon X-100, 0.1% SDS, 0.25 mM EDTA, 1% Sodium deoxycholate). A modified lysis buffer was used to better preserve IGF1R and FGFR1 in whole lysates (Hepes pH=7.5 50 mM, NaCl 150 mM, MgCl2 1.5 mM, EDTA 1 mM, 10% Glycerol, 1% Trithon X-100). Both buffers contained a mixture of protease and phosphatase inhibitors; Cocktail Tablets (Roche, Mannheim, Germany), NaF 1 mM, NaPiP 1 mM and Na3VO4 0.2 mM. For western blot analysis equal amounts of total protein were resolved in polyacrylamide gels and transferred to nitrocellulose membranes (Pierce, Rockford, IL). Membranes were BSA-blocked and hybridized overnight at 4ºC with the primary antibodies; IGF1R, phospho-IGF1R, FGFR1, phospho-FGFR1, ERK, phospho-ERK, Akt, phospho-Akt, MAPK14, phospho-MAPK14, EpCAM, vimentin, CK19 (Cell Signaling) and later at room temperature with HRP-conjugated secondary antibodies. To evaluate the activation status of FGFR1, immunoprecipitation with total anti-FGFR1 and western blot analysis using the anti-phosphotyrosine clone PY20 HRP (Santa Cruz) was performed as previously described(5). Signals were visualized with Amersham ECL Prime (GE Healthcare, Buckinghamshire, UK) using a LAS-4000 imaging system (Fujifilm, Tokyo, Japan). Immunohistochemical analysis For immunohistochemistry, 5-m paraffin-embedded sections of mice xenograft tumors were baked at 55 C for 1 hour, deparaffinized in xylene, and rehydrated in a graded series of ethanol solutions. Antigens were unmasked by microwave heating the samples in 10 mM sodium citrate buffer (pH 6.0) for 15 minutes (5 minutes, 3 times), and the reaction was quenched using hydrogen peroxide 3%. After washing with PBS, samples were incubated with the following primary antibody concentrations: p-FGFR1 (Tyr766) 1:50, FGFR1 1:100 (both from Abcam plc, Cambridge, UK), anti-p-RPS6 (phosphoSer240/244) 1:200, anti-pERK (phosphoThr202/Tyr204) 1:100, anti-ERK 1:100 (all from Cell Signaling, Danvers, MA), and anti-p-IGF-1R (phosphoTyr1316)(6) 1:100, at 4ºC overnight. DAB (3,3'-diaminobenzidine) was used as a detection system (EnVision+ System-HRP, Dako). Samples were contrasted with Gill’s hematoxilyn (Panreac, Castellar del Valles, Spain), dehydrated and mounted with DPX (Richmond, IL). Immunoreactivity was graded by a liver pathologist (MS). The variables measured were immunostaining intensity and staining pattern. For immunocytochemistry cells were seeded on poly-L lysine treated coverslips on 24- well plates for 16h, fixed, permeabilized and incubated O/N at 4ºC with primary antibodies for CK19 (Dako), EpCAM, CD133, CD90 (Abcam), and vimentin (Cell Signaling). Gene expression analysis profiling Briefly, 1 g of total RNA was hybridized to the Human Gene 1.0 ST platform. Differential expression among experimental groups was analyzed using Array File Maker 4.0 (AFM) software. Data was normalized by RMA method and analyzed with GenePattern software (www.broadinstitute.org/genepattern). The dataset is deposited at GEO Omnibus (http://www.ncbi.nlm.nih.gov/geo) under accession number GEO (GSE73571). For relative mRNA quantification TaqMan® Gene Expression Assays were used following manufacturer’s instructions (Applied Biosystems, Foster City, CA). GAPDH and ribosomal RNA (18S) were chosen as endogenous reference genes. TaqMan® probes are listed in Supplementary Table 2. For the purpose of the study, we used data from the transcriptomic analysis of 442 HCCs performed by the HCC Genomic Consortium in previous studies (7-11). Computation of sphere- and tumor-initiating cell frequencies To compute the frequency of sphere- and tumor-initiating cells, the maximum-likelihood estimation method was used(4, 12). Similar results were obtained using a second method(13), and internal consistency was validated by chi-squared analysis (data not shown). Bioinformatic and statistical analysis Gene expression profiling among experimental groups was analyzed with the module Comparative Marker Selection as implemented in Gene Pattern, and Array File Maker 4.0 (AFM) tools.
Recommended publications
  • Cytoplasmic Activation-Induced Cytidine Deaminase (AID) Exists in Stoichiometric Complex with Translation Elongation Factor 1Α (Eef1a)
    Cytoplasmic activation-induced cytidine deaminase (AID) exists in stoichiometric complex with translation elongation factor 1α (eEF1A) Julien Häsler, Cristina Rada, and Michael S. Neuberger1 Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom Edited by Frederick W. Alt, Howard Hughes Medical Institute, Harvard Medical School, Children’s Hospital Immune Disease Institute, Boston, MA, and approved October 12, 2011 (received for review April 27, 2011) Activation-induced cytidine deaminase (AID) is a B lymphocyte- results reveal that endogenous cytoplasmic AID partakes in a specific DNA deaminase that acts on the Ig loci to trigger antibody complex containing stoichiometric quantities of translation elon- gene diversification. Most AID, however, is retained in the cyto- gation factor 1α (eEF1A), with this association likely implicated in plasm and its nuclear abundance is carefully regulated because the regulation of AID’s intracellular trafficking. off-target action of AID leads to cancer. The nature of the cytosolic AID complex and the mechanisms regulating its release from the Results cytoplasm and import into the nucleus remain unknown. Here, we Flag-Tagging the Endogenous AID Locus in DT40 Cells. We generated show that cytosolic AID in DT40 B cells is part of an 11S complex derivatives of the DT40 B-cell line in which the endogenous AID and, using an endogenously tagged AID protein to avoid overex- locus was modified so as to incorporate a single Flag tag at the pression artifacts, that it is bound in good stoichiometry to the AID N terminus. To allow targeting of both alleles, one targeting translation elongation factor 1 alpha (eEF1A).
    [Show full text]
  • Peking University-Juntendo University Joint Symposium on Cancer Research and Treatment ADAM28 (A Disintegrin and Metalloproteinase 28) in Cancer Cell Proliferation and Progression
    Whatʼs New from Juntendo University, Tokyo Juntendo Medical Journal 2017. 63(5), 322-325 Peking University - Juntendo University Joint Symposium on Cancer Research and Treatment ADAM28 (a Disintegrin and Metalloproteinase 28) in Cancer Cell Proliferation and Progression YASUNORI OKADA* *Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan A disintegrinandmetalloproteinase 28 (ADAM28) is overexpressedpredominantlyby carcinoma cells in more than 70% of the non-small cell lung carcinomas, showing positive correlations with carcinoma cell proliferation and metastasis. ADAM28 cleaves insulin-like growth factor binding protein-3 (IGFBP-3) in the IGF-I/IGFBP-3 complex, leading to stimulation of cell proliferation by intact IGF-I released from the complex. ADAM28 also degrades von Willebrand factor (VWF), which induces apoptosis in human carcinoma cell lines with negligible ADAM28 expression, andthe VWF digestionby ADAM28-expressing carcinoma cells facilitates them to escape from VWF-induced apoptosis, resulting in promotion of metastasis. We have developed human antibodies against ADAM28 andshown that one of them significantly inhibits tumor growth andmetastasis using lung adenocarcinoma cells. Our data suggest that ADAM28 may be a new molecular target for therapy of the patients with ADAM28-expressing non-small cell lung carcinoma. Key words: a disintegrin and metalloproteinase 28 (ADAM28), cell proliferation, invasion, metastasis, human antibody inhibitor Introduction human cancers 2). However, development of the synthetic inhibitors of MMPs andtheir application Cancer cell proliferation andprogression are for treatment of the cancer patients failed 3). modulated by proteolytic cleavage of tissue micro- On the other hand, members of the ADAM (a environmental factors such as extracellular matrix disintegrin and metalloproteinase) gene family, (ECM), growth factors andcytokines, receptors another family belonging to the metzincin gene andcell adhesionmolecules.
    [Show full text]
  • Identification of Potential Autoantigens in Anti-CCP-Positive and Anti-CCP-Negative Rheumatoid Arthritis Using Citrulline-Specific Protein Arrays
    www.nature.com/scientificreports OPEN Identifcation of potential autoantigens in anti‑CCP‑positive and anti‑CCP‑negative rheumatoid arthritis using citrulline‑specifc protein arrays Thomas B. G. Poulsen 1,2, Dres Damgaard 3, Malene M. Jørgensen 4,5, Ladislav Senolt6, Jonathan M. Blackburn 7, Claus H. Nielsen 3,8 & Allan Stensballe 1,8* The presence or absence of autoantibodies against citrullinated proteins (ACPAs) distinguishes two main groups of rheumatoid arthritis (RA) patients with diferent etiologies, prognoses, disease severities, and, presumably, disease pathogenesis. The heterogeneous responses of RA patients to various biologics, even among ACPA‑positive patients, emphasize the need for further stratifcation of the patients. We used high‑density protein array technology for fngerprinting of ACPA reactivity. Identifcation of the proteome recognized by ACPAs may be a step to stratify RA patients according to immune reactivity. Pooled plasma samples from 10 anti‑CCP‑negative and 15 anti‑CCP‑positive RA patients were assessed for ACPA content using a modifed protein microarray containing 1631 diferent natively folded proteins citrullinated in situ by protein arginine deiminases (PADs) 2 and PAD4. IgG antibodies from anti‑CCP‑positive RA plasma showed high‑intensity binding to 87 proteins citrullinated by PAD2 and 99 proteins citrullinated by PAD4 without binding signifcantly to the corresponding native proteins. Curiously, the binding of IgG antibodies in anti‑CCP‑negative plasma was also enhanced by PAD2‑ and PAD4‑mediated citrullination of 29 and 26 proteins, respectively. For only four proteins, signifcantly more ACPA binding occurred after citrullination with PAD2 compared to citrullination with PAD4, while the opposite was true for one protein.
    [Show full text]
  • The Protein Arginine Deiminases
    The Protein Arginine Deiminases: Inhibitors and Characteristics By Elizabeth Joanne Curiel Tejeda A thesis submitted in conformity with the requirements for the degree of Master of Science Pharmaceutical Sciences University of Toronto © Copyright by Elizabeth Joanne Curiel Tejeda (2015) Abstract The Protein Arginine Deiminases: Inhibitors and Characteristics MSc Pharmaceutical Sciences Convocation year of 2015 Elizabeth Joanne Curiel Tejeda Department of Pharmaceutical Sciences University of Toronto Protein arginine deiminases (PADs) are Ca2+ dependent enzymes involved in the post- translational modification of proteins through citrullination. Overexpression of PAD2 and PAD4 has been observed in a number of neurodegenerative diseases. In this thesis, two broad investigations were undertaken to look at the expression patterns of endogenous PADs and how they provide means to the development of disease modifying treatments. First, a BODIPY-based fluorescent probe was designed to assess the expression levels in vitro and in vivo of endogenous PAD. Secondly, medicinal chemistry approaches were adopted to design a library of novel non-covalent compounds to establish a structure activity relationship on PADs and identify a potential hit. Results indicated that a BODIPY-based biomarker was a feasible approach to monitor PAD enzymes in vivo. Through structure-activity relationship investigations, it was established that small heterocycles on an amino acid side chain were contributing to the inhibitory activities towards PADs. Declaration of Work: All synthetic experiments, synthesis of library of compounds, and confocal microscopy studies were performed by Elizabeth J. Curiel Tejeda. Biological assays and evaluations were performed by Ms. Ewa Wasilewski. ii Acknowledgements I would like thank Dr. Lakshmi Kotra for giving me the opportunity to work in his laboratory and expand my knowledge in the drug development research field.
    [Show full text]
  • Acetyl-Coa Synthetase 3 Promotes Bladder Cancer Cell Growth Under Metabolic Stress Jianhao Zhang1, Hongjian Duan1, Zhipeng Feng1,Xinweihan1 and Chaohui Gu2
    Zhang et al. Oncogenesis (2020) 9:46 https://doi.org/10.1038/s41389-020-0230-3 Oncogenesis ARTICLE Open Access Acetyl-CoA synthetase 3 promotes bladder cancer cell growth under metabolic stress Jianhao Zhang1, Hongjian Duan1, Zhipeng Feng1,XinweiHan1 and Chaohui Gu2 Abstract Cancer cells adapt to nutrient-deprived tumor microenvironment during progression via regulating the level and function of metabolic enzymes. Acetyl-coenzyme A (AcCoA) is a key metabolic intermediate that is crucial for cancer cell metabolism, especially under metabolic stress. It is of special significance to decipher the role acetyl-CoA synthetase short chain family (ACSS) in cancer cells confronting metabolic stress. Here we analyzed the generation of lipogenic AcCoA in bladder cancer cells under metabolic stress and found that in bladder urothelial carcinoma (BLCA) cells, the proportion of lipogenic AcCoA generated from glucose were largely reduced under metabolic stress. Our results revealed that ACSS3 was responsible for lipogenic AcCoA synthesis in BLCA cells under metabolic stress. Interestingly, we found that ACSS3 was required for acetate utilization and histone acetylation. Moreover, our data illustrated that ACSS3 promoted BLCA cell growth. In addition, through analyzing clinical samples, we found that both mRNA and protein levels of ACSS3 were dramatically upregulated in BLCA samples in comparison with adjacent controls and BLCA patients with lower ACSS3 expression were entitled with longer overall survival. Our data revealed an oncogenic role of ACSS3 via regulating AcCoA generation in BLCA and provided a promising target in metabolic pathway for BLCA treatment. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Introduction acetyl-CoA synthetase short chain family (ACSS), which In cancer cells, considerable number of metabolic ligates acetate and CoA6.
    [Show full text]
  • Epithelial Delamination Is Protective During Pharmaceutical-Induced Enteropathy
    Epithelial delamination is protective during pharmaceutical-induced enteropathy Scott T. Espenschieda, Mark R. Cronana, Molly A. Mattya, Olaf Muellera, Matthew R. Redinbob,c,d, David M. Tobina,e,f, and John F. Rawlsa,e,1 aDepartment of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710; bDepartment of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; cDepartment of Biochemistry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; dDepartment of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; eDepartment of Medicine, Duke University School of Medicine, Durham, NC 27710; and fDepartment of Immunology, Duke University School of Medicine, Durham, NC 27710 Edited by Dennis L. Kasper, Harvard Medical School, Boston, MA, and approved July 15, 2019 (received for review February 12, 2019) Intestinal epithelial cell (IEC) shedding is a fundamental response to in mediating intestinal responses to injury remains poorly un- intestinal damage, yet underlying mechanisms and functions have derstood for most xenobiotics. been difficult to define. Here we model chronic intestinal damage in Gastrointestinal pathology is common in people using phar- zebrafish larvae using the nonsteroidal antiinflammatory drug maceuticals, including nonsteroidal antiinflammatory drugs (NSAID) Glafenine. Glafenine induced the unfolded protein response (NSAIDs) (11). While gastric ulceration has historically been a (UPR) and inflammatory pathways in IECs, leading to delamination. defining clinical presentation of NSAID-induced enteropathy, Glafenine-induced inflammation was augmented by microbial colo- small intestinal pathology has also been observed, although the nizationandassociatedwithchanges in intestinal and environmental incidence may be underreported due to diagnostic limitations microbiotas.
    [Show full text]
  • Global Analysis of Protein Folding Thermodynamics for Disease State Characterization
    Global Analysis of Protein Folding Thermodynamics for Disease State Characterization and Biomarker Discovery by Jagat Adhikari Department of Biochemistry Duke University Date:_______________________ Approved: ___________________________ Michael C. Fitzgerald, Supervisor ___________________________ Kenneth Kreuzer ___________________________ Terrence G. Oas ___________________________ Jiyong Hong ___________________________ Seok-Yong Lee Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2015 ABSTRACT Global Analysis of Protein Folding Thermodynamics for Disease State Characterization and Biomarker Discovery by Jagat Adhikari Department of Biochemistry Duke University Date:_______________________ Approved: ___________________________ Michael C. Fitzgerald, Supervisor ___________________________ Kenneth Kreuzer ___________________________ Terrence G. Oas ___________________________ Jiyong Hong ___________________________ Seok-Yong Lee An abstract of a dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2015 Copyright by Jagat Adhikari 2015 Abstract Protein biomarkers can facilitate the diagnosis of many diseases such as cancer and they can be important for the development of effective therapeutic interventions. Current large-scale biomarker discovery and disease state characterization
    [Show full text]
  • Comparative Transcriptome Analysis of Embryo Invasion in the Mink Uterus
    Placenta 75 (2019) 16–22 Contents lists available at ScienceDirect Placenta journal homepage: www.elsevier.com/locate/placenta Comparative transcriptome analysis of embryo invasion in the mink uterus T ∗ Xinyan Caoa,b, , Chao Xua,b, Yufei Zhanga,b, Haijun Weia,b, Yong Liuc, Junguo Caoa,b, Weigang Zhaoa,b, Kun Baoa,b, Qiong Wua,b a Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China b State Key Laboratory for Molecular Biology of Special Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, China c Key Laboratory of Embryo Development and Reproductive Regulation of Anhui Province, College of Biological and Food Engineering, Fuyang Teachers College, Fuyang, China ABSTRACT Introduction: In mink, as many as 65% of embryos die during gestation. The causes and the mechanisms of embryonic mortality remain unclear. The purpose of our study was to examine global gene expression changes during embryo invasion in mink, and thereby to identify potential signaling pathways involved in implantation failure and early pregnancy loss. Methods: Illumina's next-generation sequencing technology (RNA-Seq) was used to analyze the differentially expressed genes (DEGs) in implantation (IMs) and inter- implantation sites (inter-IMs) of uterine tissue. Results: We identified a total of 606 DEGs, including 420 up- and 186 down-regulated genes in IMs compared to inter-IMs. Gene annotation analysis indicated multiple biological pathways to be significantly enriched for DEGs, including immune response, ECM complex, cytokine activity, chemokine activity andprotein binding. The KEGG pathway including cytokine-cytokine receptor interaction, Jak-STAT, TNF and the chemokine signaling pathway were the most enriched.
    [Show full text]
  • Supplemental Table 1. Complete Gene Lists and GO Terms from Figure 3C
    Supplemental Table 1. Complete gene lists and GO terms from Figure 3C. Path 1 Genes: RP11-34P13.15, RP4-758J18.10, VWA1, CHD5, AZIN2, FOXO6, RP11-403I13.8, ARHGAP30, RGS4, LRRN2, RASSF5, SERTAD4, GJC2, RHOU, REEP1, FOXI3, SH3RF3, COL4A4, ZDHHC23, FGFR3, PPP2R2C, CTD-2031P19.4, RNF182, GRM4, PRR15, DGKI, CHMP4C, CALB1, SPAG1, KLF4, ENG, RET, GDF10, ADAMTS14, SPOCK2, MBL1P, ADAM8, LRP4-AS1, CARNS1, DGAT2, CRYAB, AP000783.1, OPCML, PLEKHG6, GDF3, EMP1, RASSF9, FAM101A, STON2, GREM1, ACTC1, CORO2B, FURIN, WFIKKN1, BAIAP3, TMC5, HS3ST4, ZFHX3, NLRP1, RASD1, CACNG4, EMILIN2, L3MBTL4, KLHL14, HMSD, RP11-849I19.1, SALL3, GADD45B, KANK3, CTC- 526N19.1, ZNF888, MMP9, BMP7, PIK3IP1, MCHR1, SYTL5, CAMK2N1, PINK1, ID3, PTPRU, MANEAL, MCOLN3, LRRC8C, NTNG1, KCNC4, RP11, 430C7.5, C1orf95, ID2-AS1, ID2, GDF7, KCNG3, RGPD8, PSD4, CCDC74B, BMPR2, KAT2B, LINC00693, ZNF654, FILIP1L, SH3TC1, CPEB2, NPFFR2, TRPC3, RP11-752L20.3, FAM198B, TLL1, CDH9, PDZD2, CHSY3, GALNT10, FOXQ1, ATXN1, ID4, COL11A2, CNR1, GTF2IP4, FZD1, PAX5, RP11-35N6.1, UNC5B, NKX1-2, FAM196A, EBF3, PRRG4, LRP4, SYT7, PLBD1, GRASP, ALX1, HIP1R, LPAR6, SLITRK6, C16orf89, RP11-491F9.1, MMP2, B3GNT9, NXPH3, TNRC6C-AS1, LDLRAD4, NOL4, SMAD7, HCN2, PDE4A, KANK2, SAMD1, EXOC3L2, IL11, EMILIN3, KCNB1, DOK5, EEF1A2, A4GALT, ADGRG2, ELF4, ABCD1 Term Count % PValue Genes regulation of pathway-restricted GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, SMAD protein phosphorylation 9 6.34 1.31E-08 ENG pathway-restricted SMAD protein GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, phosphorylation
    [Show full text]
  • ASPA Gene Aspartoacylase
    ASPA gene aspartoacylase Normal Function The ASPA gene provides instructions for making an enzyme called aspartoacylase. In the brain, this enzyme breaks down a compound called N-acetyl-L-aspartic acid (NAA) into aspartic acid (an amino acid that is a building block of many proteins) and another molecule called acetic acid. The production and breakdown of NAA appears to be critical for maintaining the brain's white matter, which consists of nerve fibers surrounded by a myelin sheath. The myelin sheath is the covering that protects nerve fibers and promotes the efficient transmission of nerve impulses. The precise function of NAA is unclear. Researchers had suspected that it played a role in the production of the myelin sheath, but recent studies suggest that NAA does not have this function. The enzyme may instead be involved in the transport of water molecules out of nerve cells (neurons). Health Conditions Related to Genetic Changes Canavan disease More than 80 mutations in the ASPA gene are known to cause Canavan disease, which is a rare inherited disorder that affects brain development. Researchers have described two major forms of this condition: neonatal/infantile Canavan disease, which is the most common and most severe form, and mild/juvenile Canavan disease. The ASPA gene mutations that cause the neonatal/infantile form severely impair the activity of aspartoacylase, preventing the breakdown of NAA and allowing this substance to build up to high levels in the brain. The mutations that cause the mild/juvenile form have milder effects on the enzyme's activity, leading to less accumulation of NAA.
    [Show full text]
  • Functional Genomics Atlas of Synovial Fibroblasts Defining Rheumatoid Arthritis
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.16.20248230; this version posted December 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Functional genomics atlas of synovial fibroblasts defining rheumatoid arthritis heritability Xiangyu Ge1*, Mojca Frank-Bertoncelj2*, Kerstin Klein2, Amanda Mcgovern1, Tadeja Kuret2,3, Miranda Houtman2, Blaž Burja2,3, Raphael Micheroli2, Miriam Marks4, Andrew Filer5,6, Christopher D. Buckley5,6,7, Gisela Orozco1, Oliver Distler2, Andrew P Morris1, Paul Martin1, Stephen Eyre1* & Caroline Ospelt2*,# 1Versus Arthritis Centre for Genetics and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK 2Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland 3Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia 4Schulthess Klinik, Zurich, Switzerland 5Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK 6NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK 7Kennedy Institute of Rheumatology, University of Oxford Roosevelt Drive Headington Oxford UK *These authors contributed equally #corresponding author: [email protected] NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. 1 medRxiv preprint doi: https://doi.org/10.1101/2020.12.16.20248230; this version posted December 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
    [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]