Rnai-Mediated HOXD3 Knockdown Inhibits Growth in Human RKO Cells

Total Page:16

File Type:pdf, Size:1020Kb

Rnai-Mediated HOXD3 Knockdown Inhibits Growth in Human RKO Cells ONCOLOGY REPORTS 36: 1793-1798, 2016 RNAi-mediated HOXD3 knockdown inhibits growth in human RKO cells FANGJUN CHEN1,2, GUOPING SUN1 and JUN PENG3 1Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022; 2Department of Clinical Medicine, Anqing Medical College, Anqing, Anhui 246052; 3Department of Pathology, The Affiliated Anqing Municipal Hospital of Anhui Medical University, Anqing, Anhui 246003, P.R. China Received January 25, 2016; Accepted March 2, 2016 DOI: 10.3892/or.2016.4993 Abstract. Numerous studies have shown that the multifunc- regulators of cell differentiation, morphogenesis, and tional Homeobox-containing (HOX) D3 gene is involved in organogenesis during development (5,6). In humans and other various physiological and pathological processes. To elucidate mammals, 39 HOX genes are clustered in four complexes the role and mechanism of HOXD3 in colorectal cancer called HOXA, B, C, and D. These are further subdivided into (CRC), we measured its expression in five CRC cell lines. 13 paralogous groups based on their sequence similarities and After determining that HOXD3 was highly expressed in the relative positions along the clusters (7-11). Mutational analyses human RKO cancer cell line, we used lentiviral-mediated demonstrated that HOXD genes play a pivotal role in deter- small interfering RNAs (siRNAs) to knock down HOXD3 mining the regional specificity of cells (12,13). For example, expression and assessed proliferation, cell cycle distribution, the HOXD13 gene is involved in the normal morphogenesis of apoptosis and colony formation using cell proliferation, flow limbs (12) and of the anal sphincter (13). HOXD3 is a member cytometric, and colony formation assays. The expression of of the third paralogous group of the HOXD gene family and is HOXD3 was strongly suppressed in the RKO cells infected involved in embryonic development (14,15). with the lentiviruse expressing an HOXD3 short hairpin RNA Studies have shown that the human HOXD3 gene plays (shRNA). The downregulation of HOXD3 expression in RKO a multifunctional role in cancer. For example, HOXD3 cells significantly decreased proliferation and colony forma- overexpression was found to regulate cell adhesion in tion, and increased apoptosis in vitro, compared to the cells human erythroleukemia HEL cells (16), induce coordinate infected with the mock control (p<0.01). Moreover, specific metastasis-related gene expression, and enhance the motility downregulation of HOXD3 led to the accumulation of cells at and invasiveness of human lung cancer A549 (17-19) and the G2 phase of the cell cycle. Our findings revealed that the melanoma cells (20). Additionally, studies have shown that HOXD3 gene promotes CRC cell growth and plays a pivotal HOXD3 induces angiogenesis by increasing pro-angiogenic role in the development and survival of malignant human molecules (21-25). While these studies clearly demonstrated colorectal cancer cells. that HOXD3 is involved in the development and growth of various types of cancers, the functional role of HOXD3 in Introduction human CRC has not yet been determined. In the present study, we demonstrated that HOXD3 is highly Worldwide, colorectal carcinoma (CRC) is one of the most expressed in the human CRC RKO cell line. Consequently, common malignant cancers and is the second most frequent we used a lentiviral vector to deliver small interfering RNA cause of cancer-related deaths (1-3). Despite marked advances (siRNA) to knock down HOXD3 expression in the RKO cells. in diagnostic and therapeutic approaches, the prognosis for Finally, we assessed the effects of HOXD3 knockdown on CRC patients remains poor (4). Consequently, there is an human CRC cell growth and survival in vitro. urgent need to develop new CRC treatment methods. Since homeobox-containing (HOX) genes encode Materials and methods DNA-binding proteins, they are master transcriptional Cell lines. DLD-1, HCT-116, SW620, HT-29, and RKO human colon carcinoma cell lines were purchased from the Shanghai Cell Bank of the Chinese Academy of Science (Shanghai, Correspondence to: Professor Guoping Sun, Department of China). Cell lines were maintained in Dulbecco's modi- Oncology, The First Affiliated Hospital of Anhui Medical University, fied Eagle's medium (DMEM; Corning, Shanghai, China) 218 Jixi Road, Hefei, Anhui 230022, P.R. China supplemented with 10% fetal bovine serum (FBS; Ausbian, E-mail: [email protected] Australia) at 37˚C in a 5% CO2 incubator. Key words: colorectal cancer, HOXD3, RKO cells, proliferation, colony formation, apoptosis Quantitative RT-PCR. Total RNA was extracted from the cells in each group (DLD-1, HCT-116, SW620, HT-29, and RKO) 1794 CHEN et al: HOXD3 knockdown inhibits RKO cell growth using TRIzol reagent (Invitrogen, Carlsbad, CA, USA), post-infection, the cells were incubated with 3-(4,5-dimethyl- according to the manufacturer's protocol. Two micrograms of thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT, 5 mg/ total RNA from each sample was reverse transcribed to single- ml; Promega, Shanghai, China) at a final concentration of stranded cDNA. One microgram of cDNA was used as a 0.5 mg/ml for 4 h. After discarding the supernatants, 150 µl template for the quantitative real-time PCR. The primers used of dimethyl sulfoxide (DMSO; Sigma-Aldrich Co., LLC, St. were as follows: HOXD3 forward, 5'-CGG CAA CTT CGT Louis, MO, USA) was added to each well. The plates were read CGA GTC C-3' and reverse, 5'-ATG AGG GTC GCA AGG at 490 nm using an ELISA reader (Tecan Infinite, Männedorf, TCC A-3'; and GAPDH forward, 5'-TGA CTT CAA CAG Switzerland). All experiments were performed in triplicate. CGA CAC CCA-3' and reverse, 5'-CAC CCT GTT GCT GTA GCC AAA-3'. Cycling conditions for quantitative RT-PCR Cell cycle distribution analysis. Flow cytometry (FCM) were as follows: 95˚C for 30 sec, then 45 cycles of 95˚C for was used to determine cell cycle distribution as previously 5 sec and 60˚C for 30 sec. The PCR products of HOXD3 and described (27). Briefly, RKO cells were infected with shCtrl GAPDH were 145 and 121 bp, respectively. The data were or shHOXD3 vector and incubated at 37˚C for 1, 2, 3, 4, or quantified using the 2 -ΔΔCt method. All analyses were performed 5 days. At the indicated time-point, the cells were collected in triplicate. and washed with ice-cold phosphate-buffered saline (PBS). Cells were centrifuged at 1,000 x g for 10 min and then fixed Recombinant lentiviral vector production and cell infection. in ice-cold 70% ethanol for 30 min at 4˚C. The cells were To create the RNAi target site, the complementary DNA washed with PBS and then resuspended and incubated in PBS sequence (CCA AAT CAC AGC CCA ATA T) of HOXD3 was containing 50 µg/ml propidium iodide (PI; Sigma-Aldrich) designed by Shanghai GeneChem Co., Ltd. (Shanghai, China) and 100 µg/ml RNase A (Fermentas, Shanghai, China) in the using the full-length human sequence (GenBank no. dark at 4˚C for 30 min. Cell cycle phase was analyzed using a NM_006898). The HOXD3 hairpin oligonucleotides were BD FACSCalibur flow cytometer (BD Biosciences, San Diego, synthesized and inserted into the pGV115-GFP (GeneChem CA, USA). All studies were performed in triplicate. Co. Ltd.) lentiviral vector. Lentivirus particles were prepared as previously described (26). Analysis of apoptosis. FCM was used to measure apoptosis For lentiviral infection, RKO cells were cultured in 6-well and was performed as previously described (28). At 48 h plates. The HOXD3-siRNA lentivirus (shHOXD3) or negative prior to transfection, the RKO cells were seeded in 6-well control lentivirus (shCtrl) was added according to a multiplicity plates. At 72 h post-transfection, the cells were collected and of infection (MOI 1:5). At 3 days post-infection, the cells were washed twice with ice-cold PBS. Cell densities were adjusted observed for presence of the GFP marker with a fluorescence to 1x106/ml using 1X staining buffer. Cell suspension (100 µl) microscope (MicroPublisher 3.3RTV; Olympus, Tokyo, and 5 µl Annexin V-APC (eBioscience, San Diego, CA, USA) Japan). At 5 days post-infection, the cells were harvested and were thoroughly mixed and incubated in darkness at room knockdown efficiency was analyzed by quantitative RT-PCR temperature for 15 min. All rates of apoptosis were measured and western blot analysis. by FCM within 1 h. Each experiment was performed in triplicate. Western blot analysis. While on ice, cell lysates were incu- bated for 10-15 min in ice-cold lysis buffer (100 mM Tris, Cell colony formation assay. At 72 h post-transfection, the pH 6.8, 2% β-mercaptoethanol, 20% glycerol, 4% SDS). The RKO cells were reseeded at 600 cells/well in 6-well plates lysates were centrifuged at 12,000 x g for 15 min at 4˚C, and and cultured at 37˚C for 10 days. The cells were collected and the supernatants were collected. Protein concentration was washed with ice-cold PBS. When a majority of single colonies determined using a BCA protein assay kit (Beyotime, Beijing, contained more than 50 cells, the samples were fixed using China). An equal amount of total protein from each sample 1 ml/well of 4% paraformaldehyde (Sinopharm, Shanghai, was partially separated in a 10% SDS-PAGE gel and blotted China) for 30 min at 37˚C. According to instructions, the onto PVDF membranes. Membranes were incubated with GFP samples were stained with 500 µl of Giemsa stain (Dingguo or GAPDH primary antibodies (Santa Cruz Biotechnology, Bio, Shanghai, China) at room temperature for 10 min and Santa Cruz, CA, USA) at 4˚C overnight, followed by horse- images were acquired. All experiments were performed in radish peroxidase (HRP)-conjugated goat anti-mouse IgG triplicate.
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • SUPPLEMENTARY MATERIAL Bone Morphogenetic Protein 4 Promotes
    www.intjdevbiol.com doi: 10.1387/ijdb.160040mk SUPPLEMENTARY MATERIAL corresponding to: Bone morphogenetic protein 4 promotes craniofacial neural crest induction from human pluripotent stem cells SUMIYO MIMURA, MIKA SUGA, KAORI OKADA, MASAKI KINEHARA, HIROKI NIKAWA and MIHO K. FURUE* *Address correspondence to: Miho Kusuda Furue. Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567-0085, Japan. Tel: 81-72-641-9819. Fax: 81-72-641-9812. E-mail: [email protected] Full text for this paper is available at: http://dx.doi.org/10.1387/ijdb.160040mk TABLE S1 PRIMER LIST FOR QRT-PCR Gene forward reverse AP2α AATTTCTCAACCGACAACATT ATCTGTTTTGTAGCCAGGAGC CDX2 CTGGAGCTGGAGAAGGAGTTTC ATTTTAACCTGCCTCTCAGAGAGC DLX1 AGTTTGCAGTTGCAGGCTTT CCCTGCTTCATCAGCTTCTT FOXD3 CAGCGGTTCGGCGGGAGG TGAGTGAGAGGTTGTGGCGGATG GAPDH CAAAGTTGTCATGGATGACC CCATGGAGAAGGCTGGGG MSX1 GGATCAGACTTCGGAGAGTGAACT GCCTTCCCTTTAACCCTCACA NANOG TGAACCTCAGCTACAAACAG TGGTGGTAGGAAGAGTAAAG OCT4 GACAGGGGGAGGGGAGGAGCTAGG CTTCCCTCCAACCAGTTGCCCCAAA PAX3 TTGCAATGGCCTCTCAC AGGGGAGAGCGCGTAATC PAX6 GTCCATCTTTGCTTGGGAAA TAGCCAGGTTGCGAAGAACT p75 TCATCCCTGTCTATTGCTCCA TGTTCTGCTTGCAGCTGTTC SOX9 AATGGAGCAGCGAAATCAAC CAGAGAGATTTAGCACACTGATC SOX10 GACCAGTACCCGCACCTG CGCTTGTCACTTTCGTTCAG Suppl. Fig. S1. Comparison of the gene expression profiles of the ES cells and the cells induced by NC and NC-B condition. Scatter plots compares the normalized expression of every gene on the array (refer to Table S3). The central line
    [Show full text]
  • Methylome and Transcriptome Maps of Human Visceral and Subcutaneous
    www.nature.com/scientificreports OPEN Methylome and transcriptome maps of human visceral and subcutaneous adipocytes reveal Received: 9 April 2019 Accepted: 11 June 2019 key epigenetic diferences at Published: xx xx xxxx developmental genes Stephen T. Bradford1,2,3, Shalima S. Nair1,3, Aaron L. Statham1, Susan J. van Dijk2, Timothy J. Peters 1,3,4, Firoz Anwar 2, Hugh J. French 1, Julius Z. H. von Martels1, Brodie Sutclife2, Madhavi P. Maddugoda1, Michelle Peranec1, Hilal Varinli1,2,5, Rosanna Arnoldy1, Michael Buckley1,4, Jason P. Ross2, Elena Zotenko1,3, Jenny Z. Song1, Clare Stirzaker1,3, Denis C. Bauer2, Wenjia Qu1, Michael M. Swarbrick6, Helen L. Lutgers1,7, Reginald V. Lord8, Katherine Samaras9,10, Peter L. Molloy 2 & Susan J. Clark 1,3 Adipocytes support key metabolic and endocrine functions of adipose tissue. Lipid is stored in two major classes of depots, namely visceral adipose (VA) and subcutaneous adipose (SA) depots. Increased visceral adiposity is associated with adverse health outcomes, whereas the impact of SA tissue is relatively metabolically benign. The precise molecular features associated with the functional diferences between the adipose depots are still not well understood. Here, we characterised transcriptomes and methylomes of isolated adipocytes from matched SA and VA tissues of individuals with normal BMI to identify epigenetic diferences and their contribution to cell type and depot-specifc function. We found that DNA methylomes were notably distinct between diferent adipocyte depots and were associated with diferential gene expression within pathways fundamental to adipocyte function. Most striking diferential methylation was found at transcription factor and developmental genes. Our fndings highlight the importance of developmental origins in the function of diferent fat depots.
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • Projeto De Tese De Doutorado
    2 3 Agradecimentos Todo este trabalho não teria sido realizado se não fosse pelo apoio e credibilidade de pessoas importantes que fazem parte da minha vida, com as quais eu pude contar incondicionalmente. Agradeço ao Professor Hernandes F. Carvalho, por me orientar desde meu treinamento técnico, por acreditar na minha capacidade profissional, aceitando-me como sua aluna no Mestrado em Biologia Celular e por sempre ter me dado todo apoio intelectual e estrutural para prosseguir neste caminho. A todos os integrantes do Laboratório de Matriz Extracelular: Adauto, Augusto, Fabiana, Fiama, Guilherme, Gustavo, Juliete, Renan, Rony, Sílvia, Umar, por sempre dividirem comigo conhecimentos teóricos e práticos, felicidades e angústias. Em especial ao ex e aos ainda integrantes do laboratório: Alexandre, por ter me ensinado muitas das coisas que hoje sei e me aceitado como colaboradora em seus trabalhos; à Ana Milena pela dedicação e colaboração essencial na escrita dos manuscritos; ao Danilo, pela paciência e ajuda integral nos tratamentos dos animais; à Taize, pela permanente disposição em ajudar nos experimentos e acolhimento desde a minha chegada à UNICAMP. Ao Ramon Vidal Oliveira, do LNBio pela assistência e realização da análise de bioinformática dos dados obtidos na técnica de microarray. À Maria Eugênia Ribeiro Camargo, pela disponibilidade e auxílio na utilização do Laboratório de Microarray (LNBio). À Professora Maria Julia Marques, por ter nos cedido seu equipamento para a revelação das memebranas de Western Blotting. Em especial às suas alunas Samara, Cíntia, Ana Paula, Letícia, Adriane, Adriele pela disposição e ajuda sempre que precisei. 4 À Professora Heide Dolder, por ter seu laboratório de portas abertas para utilização dos micrótomos.
    [Show full text]
  • WO 2015/048577 A2 April 2015 (02.04.2015) W P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2015/048577 A2 April 2015 (02.04.2015) W P O P C T (51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every A61K 48/00 (2006.01) kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, (21) International Application Number: BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, PCT/US20 14/057905 DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, (22) International Filing Date: HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, 26 September 2014 (26.09.2014) KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, (25) Filing Language: English PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, (26) Publication Language: English SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (30) Priority Data: 61/883,925 27 September 2013 (27.09.2013) US (84) Designated States (unless otherwise indicated, for every 61/898,043 31 October 2013 (3 1. 10.2013) US kind of regional protection available): ARIPO (BW, GH, GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, (71) Applicant: EDITAS MEDICINE, INC.
    [Show full text]
  • Xo PANEL DNA GENE LIST
    xO PANEL DNA GENE LIST ~1700 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes (at 400 -500x average coverage on tumor) Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11
    [Show full text]
  • BMC Biology Biomed Central
    BMC Biology BioMed Central Research article Open Access Classification and nomenclature of all human homeobox genes PeterWHHolland*†1, H Anne F Booth†1 and Elspeth A Bruford2 Address: 1Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK and 2HUGO Gene Nomenclature Committee, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK Email: Peter WH Holland* - [email protected]; H Anne F Booth - [email protected]; Elspeth A Bruford - [email protected] * Corresponding author †Equal contributors Published: 26 October 2007 Received: 30 March 2007 Accepted: 26 October 2007 BMC Biology 2007, 5:47 doi:10.1186/1741-7007-5-47 This article is available from: http://www.biomedcentral.com/1741-7007/5/47 © 2007 Holland et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: The homeobox genes are a large and diverse group of genes, many of which play important roles in the embryonic development of animals. Increasingly, homeobox genes are being compared between genomes in an attempt to understand the evolution of animal development. Despite their importance, the full diversity of human homeobox genes has not previously been described. Results: We have identified all homeobox genes and pseudogenes in the euchromatic regions of the human genome, finding many unannotated, incorrectly annotated, unnamed, misnamed or misclassified genes and pseudogenes.
    [Show full text]
  • Supplementary File Table S1
    Supplementary file Table S1 Grouping of Homeobox genes according to their main known function. Anatomical Structure Morphogenesis EN1, HOXC10, HOXC13, HOXD3, LBX1, SIX2, SIX4 Organ Morphogenesis CDX1, CDX2, HOXA11, HOXA13, ISL1, LHX1, PAX3, PDHX, PITX2, PITX3, PROX1, SIX6 Body Pattern Formation ALX3, EMX2, HHEX, HOXA11, HOXA2, HOXA4, HOXA5, HOXA6, HOXB1, HOXB5, HOXB6, HOXC5, HOXD10, HOXD8, LMX1B, PITX2 Ectoderm Development PROX1, VAX2 Endoderm Development HOXC11 Brain & Nervous System Development Brain Development ALX1, DLX2, EMX2 Nervous System Development: ARX, DLX5, DLX6, HOXD10, LBX1, LHX1, OTP, PAX3, PHOX2A, PHOX2B Skeletal Development: ALX3, ALX4, DLX3, DLX5, DLX6, EN1, HOXA11, HOXA13, HOXA2, HOXB6, HOXD10, HOXD13, MSX2 Muscle Development: BARX2, MKX, SIRT1, SIRT2, SIX1 Other Homeobox Genes Involved In BARX1, CDX4, CUX1, DLX1, EMX1, EN2, Multicellular Organismal HOXA1, HOXA7, HOXA9, HOXB13, HOXB2, Development: HOXB3, HOXB4, HOXB7, HOXB8, HOXB9, HOXC12, HOXC8, HOXC9, HOXD1, HOXD11, HOXD12, HOXD9, ISL2, LBX2, LMX1A, MEIS1, NKX3-1, OTX1, TLX1, VAX1, VSX1, VSX2 Homeobox Genes Involved In Cell ARX, EMX2, HHEX, HLX, HOPX, LBX1, LHX1, Differentiation: LMX1B, MIXL1, OTP, PHOX2A, SIRT1, VSX2 Other Genes: PHTF1, SIRT3, SIRT6, SIRT7, ZHX1, ZHX2 Homeobox genes include two subsets of genes coding for transcription factors involved in multiple functions. The clustered HOX genes are indicated in bold. Supplementary file Figure S2 5’ Spatial collinearity 3’ HOXA Chr. 7p15.3 HOXB Chr. 17q21.3 HOXC Chr. 12q13.3 HOXD Chr. 2q31 13 12 11 10 9 8 7 6 5 4 3 2 1 Paralogous HOX groups Distribution of the 39 human HOX genes in four clusters located in different chromosomal regions*. Blue indicates anterior HOX genes. Yellow, paralogy group 3 Hox genes, green and purple indicatete central HOX genes and Red the posterior HOX genes.
    [Show full text]
  • HOX Genes in High Grade Ovarian Cancer
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by UWL Repository cancers Review HOX Genes in High Grade Ovarian Cancer Praveena Idaikkadar 1, Richard Morgan 2 and Agnieszka Michael 1,* 1 Postgraduate Medical School, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7YS, UK 2 Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK * Correspondence: [email protected] Received: 3 July 2019; Accepted: 31 July 2019; Published: 3 August 2019 Abstract: HOX genes are highly conserved members of the homeobox superfamily that have a crucial role in determining cellular identity. High grade ovarian cancer is the most lethal gynaecological malignancy. Our understanding of the role of HOX genes in the oncogenesis of ovarian cancer is evolving, and here we review their dysregulated expression patterns, their function in cell survival and invasion, their potential uses as biomarkers, and ways in which HOX genes are being targeted with new and existing drugs. Keywords: HOX; ovarian cancer 1. Introduction HOX genes are a member of the homeobox superfamily, with “homeotic” or “HOX” genes coding for transcription factors that were originally recognized for their role in embryogenesis in the Drosophila melanogaster fruit fly [1]. There are now known to be 39 HOX genes in mammals, grouped into four paralogous clusters termed A to D on four different chromosomes (7, 17, 12 and 2 respectively). They are numbered 1 to 13 based on their 30 to 50 position [1]. HOX proteins can function as monomers, homodimers, heterodimers or heterotrimers, and the affinity and specificity of their binding to DNA can be modulated by binding proteins of the three amino acid loop extension (TALE) family of co-factors, especially Pre-B cell leukaemia transcription factor (PBX) and myeloid ectopic viral integration site (MEIS) [2].
    [Show full text]