The Transcription Factor TCF-1 Initiates the Differentiation of TFH Cells During Acute Viral Infection

Total Page:16

File Type:pdf, Size:1020Kb

The Transcription Factor TCF-1 Initiates the Differentiation of TFH Cells During Acute Viral Infection ARTICLES The transcription factor TCF-1 initiates the differentiation of TFH cells during acute viral infection Lifan Xu1,4, Yi Cao1,4, Zhunyi Xie1,4, Qizhao Huang1, Qiang Bai1, Xia Yang1, Ran He1, Yaxing Hao1, Haoqiang Wang1, Tingting Zhao1, Zhonglei Fan2,3, Aijian Qin2,3, Jianqiang Ye2,3, Xinyuan Zhou1,5, Lilin Ye1,5 & Yuzhang Wu1,5 Induction of the transcriptional repressor Bcl-6 in CD4+ T cells is critical for the differentiation of follicular helper T cells (TFH cells), which are essential for B cell–mediated immunity. In contrast, the transcription factor Blimp1 (encoded by Prdm1) inhibits TFH differentiation by antagonizing Bcl-6. Here we found that the transcription factor TCF-1 was essential for both the initiation of TFH differentiation and the effector function of differentiated TFH cells during acute viral infection. Mechanistically, TCF-1 bound directly to the Bcl6 promoter and Prdm1 5′ regulatory regions, which promoted Bcl-6 expression but repressed Blimp1 expression. TCF-1-null TFH cells upregulated genes associated with non-TFH cell lineages. Thus, TCF-1 functions as an important hub upstream of the Bcl-6–Blimp1 axis to initiate and secure the differentiation of TFH cells during acute viral infection. Most antimicrobial vaccines licensed for human use elicit their effects been reported to bind to the Bcl6 locus and activate its expression18. by inducing the production of memory B cells and long-lived plasma The transcription factor Blimp1 (encoded by Prdm1) binds to the Bcl6 cells1, both of which are generally differentiated from germinal center promoter and strongly represses Bcl6 transcription14. Furthermore, (GC) B cells in secondary lymphoid tissues2. Follicular helper T cells Bcl-6 inhibits its own transcription14. Despite such advances in under- + (TFH cells), a unique subset of CD4 helper T cells, are specialized to standing, whether and how other factors are involved in the regulation facilitate the establishment of a GC reaction and the selection of GC of Bcl-6 expression in activated CD4+ T cells has remained unclear. Nature America, Inc. All rights reserved. Inc. Nature America, B cells bearing high-affinity antigen receptors for final differentiation The transcription factor TCF-1 (encoded by Tcf7) is a downstream 5 3 into memory B cells and long-lived plasma cells . TFH cells express effector of the canonical Wnt signaling pathway and is critical for abundant chemokine receptor CXCR5 to enable homing to B cell T cell development19,20. The role of TCF-1 in T cell–mediated © 201 follicles4–6, where they engage cognate B cells and provide essential immune responses is also emerging. In a Listeria infection model, help by secreting the cytokines interleukin 21 (IL-21) and IL-4 and TCF-1 has been shown to favor the formation of memory CD8+ T cells expressing CD40L (the ligand for the costimulatory receptor CD40) by inducing expression of the transcription factor Eomes21,22. TCF-1 7,8 and the costimulatory receptor ICOS . promotes commitment to the T helper type 2 (TH2) fate but inhibits 8 TFH differentiation is a multistage, multifactorial process . The T helper type 1 (TH1) differentiation by inducing expression of the transcriptional repressor Bcl-6, identified as a ‘master regulator’ transcription factor GATA-3 while repressing interferon-γ (IFN-γ) 9–11 23 of TFH differentiation , emerges on early TFH-like cells together production . In addition, TCF-1 reportedly reduces the inflamma- 12 with CXCR5 at day 2 or 3 after infection or immunization . Next, tory effects of the TH17 subset of helper T cells by dampening IL-17A + + 24 CXCR5 Bcl-6 TFH-like cells accumulate at the T cell–B cell border, expression . Despite these profound effects of TCF-1 on the regula- where they engage activated B cells. Subsequently, they migrate into tion of various T cell responses, its role in TFH differentiation has not B cell follicles and initiate GC reactions13, during which reinforced been determined. Bcl-6 expression drives the differentiation of fully functional TFH The transcriptional signatures of TFH cells overlap considerably 14 + 12 cells . Given the central role of Bcl-6 in TFH differentiation, it is with those of CD8 memory precursor cells . Indeed, both subsets critical to understand how Bcl-6 expression is regulated in activated have high expression of TCF-1 mRNA during acute infection with CD4+ T cells. Several axes of cytokines and members of the STAT lymphocytic choriomeningitis virus (LCMV)12. TCF-1 regulates family of transcription factors, including IL-6–STAT1, IL-12–STAT4 the formation of CD8+ memory cells mainly by inducing Eomes and IL-21–STAT3 (refs. 15–17), have been linked to the induction expression, which leads to upregulation of expression of the IL-2Rβ of Bcl-6 expression. In addition, the transcription factor Batf has cytokine receptor chain21. However, both Eomes and IL-2Rβ seem to 1Institute of Immunology, Third Military Medical University, Chongqing, China. 2Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, China. 3Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China. 4These authors contributed equally to this work. 5These authors jointly directed this work. Correspondence should be addressed to L.Y. ([email protected]), X.Z. ([email protected]) or Y.W. ([email protected]). Received 6 May; accepted 15 June; published online 27 July 2015; doi:10.1038/ni.3229 NATURE IMMUNOLOGY ADVANCE ONLINE PUBLICATION 1 ARTICLES 25,26 be unnecessary for TFH differentiation . Thus, it is unclear whether Role of TCF-1 in TFH differentiation high TCF-1 expression in the TFH cell subset serves a function or To investigate whether a greater abundance of TCF-1 is essential for merely represents a nonfunctional marker of the TFH-differentiation TFH differentiation during acute viral infection, we crossed mice with pathway. In this study, we determined that TCF-1 directly bound loxP-flanked Tcf7 alleles (Tcf7fl/fl) to mice with transgenic expression to the Bcl6 promoter and Prdm1 5′ regulatory regions, which acti- of Cre recombinase from the T cell–specific Cd4 promoter (Cd4- vated Bcl-6 expression but repressed Blimp1 expression. In addition, Cre) to generate mice with conditional deletion of Tcf7 in CD4+ TCF-1 formed a complex with Bcl-6 and increased Bcl6 transcrip- T cells (Tcf7fl/flCd4-Cre mice; called ‘Tcf7−/− mice’ here). CD4+ −/− tion. Thus, our findings suggest that TCF-1 promotes TFH differ- T cells from Tcf7 mice had largely abrogated TCF-1 expression entiation through multiple regulatory mechanisms upstream of the (Supplementary Fig. 2a). At day 8 after infection with LCMV, we Bcl-6–Blimp1 axis. observed a similar frequency and number of gp66 tetramer–positive CD4+ T cells in Tcf7−/− mice and control mice (Tcf7fl/fl mice with- RESULTS out Cd4-Cre) (Supplementary Fig. 2b), indicative of normal clonal + −/− + Higher TCF-1 expression in TFH cells than in naive CD4 T cells expansion of virus-specific Tcf7 CD4 T cells. However, very few + lo + −/− Upon acute viral infection, virus-specific naive CD4 T cells generally tetramer-positive SLAM CXCR5 TFH cells were present in Tcf7 27,28 + differentiate into TH1 cells or TFH cells . To investigate the expres- mice, whereas approximately 35% of the tetramer-positive CD4 sion of TCF-1 during this bifurcated differentiation, we transferred T cells in the control mice were TFH cells (Fig. 2a). This phenotype naive SMARTA cells (which have transgenic expression of a T cell anti- was recapitulated for other TFH cell markers, including ICOS, Bcl-6 gen receptor specific for LCMV epitope of glycoprotein amino acids and PD-1 (Supplementary Fig. 2c). The number of TFH cells was 66–77 (gp66) presented by I-Ab) into wild-type C57BL/6J recipients, also much lower in Tcf7−/− mice than in control mice (Fig. 2b). We which we subsequently infected with the Armstrong strain of LCMV. observed a similar phenotype for bulk activated Foxp3− CD4+ T cells At day 2 after infection, we observed that TCF-1 expression was nearly in Tcf7−/− mice (Supplementary Fig. 2d). Consistent with their lower hi − −/− tenfold lower in TH1 cells (Tim3 CXCR5 ) but twofold higher in TFH abundance of TFH cells, Tcf7 mice had a much lower frequency and cells (Tim3loCXCR5+) relative to its expression in naive cells (Fig. 1a). number of GC B cells, as well as a much lower titer of LCMV-specific We also observed the higher expression of TCF-1 in TFH cells than in immunoglobulin G (IgG), relative to that of their control counterparts −/− TH1 cells at day 8 after infection (Fig. 1a). Furthermore, at day 2 after (data not shown). The reduction in the TFH cell population in Tcf7 infection, we observed substantial nuclear enrichment for TCF-1 in mice was probably not due to enhanced differentiation of follicular TFH cells, but this was minimal in TH1 cells (Fig. 1b). regulatory T cells (Supplementary Fig. 2e) or altered apoptosis or Next we investigated the correlation between TCF-1 expression and proliferation (Supplementary Fig. 2f,g). Indeed, TCF-1 deficiency TFH differentiation. At day 2 after infection, TCF-1 expression was influenced the programming of TFH differentiation, as indicated by −/− positively correlated with expression of the key TFH-differentiation lower expression of Bcl-6, ICOS and CXCR5 in Tcf7 mice than in molecules CXCR5 and Bcl-6 and was negatively correlated with control mice (Fig. 2c). −/− + expression of the TH1 cell–associated molecules CD25, Tim3 and In Tcf7 mice, CD8 T cells are also deficient in TCF-1 expression, granzyme B (Fig. 1c). We observed a similar phenomenon at day 8 which may affect antigen loads and then influence the differentia- 21 after infection (Supplementary Fig. 1).
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Open Full Page
    Research Article Distinct Genomic Profiles in Hereditary Breast Tumors Identified by Array-Based Comparative Genomic Hybridization Go¨ran Jo¨nsson,1 Tara L. Naylor,5 Johan Vallon-Christersson,1 Johan Staaf,1 Jia Huang,5 M. Renee Ward,5 Joel D. Greshock,5 Lena Luts,4 Ha˚kan Olsson,1 Nazneen Rahman,6 Michael Stratton,6 Markus Ringne´r,3 A˚ke Borg,1,2 and Barbara L. Weber5 1Department of Oncology, University Hospital; 2Lund Strategic Research Center for Stem Cell Biology and Cell Therapy and 3Department of Theoretical Physics, Lund University; and 4Department of Pathology, University Hospital, Lund, Sweden; 5Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania; and 6Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, United Kingdom Abstract of additional predisposing genes, although technical limitations Mutations in BRCA1 and BRCA2 account for a significant and the complexity of BRCA gene regulation and mutation proportion of hereditary breast cancers. Earlier studies have spectrum can probably explain why some disease-causing muta- shown that inherited and sporadic tumors progress along tions are missed (2). BRCA1 and BRCA2 function as classic tumor different somatic genetic pathways and that global gene suppressor genes with frequent loss of the wild-type allele in expression profiles distinguish between these groups. To tumors of mutation carriers. The BRCA1 protein has been determine whether genomic profiles similarly discriminate implicated in a broad range of cellular functions, including repair among BRCA1, BRCA2, and sporadic tumors, we established of double-strand breaks by homologous recombination, cell cycle DNA copy number profiles using comparative genomic checkpoint control, chromatin remodeling, and transcriptional hybridization to BAC-clone microarrays providing <1 Mb regulation.
    [Show full text]
  • Primary Driver Mutations in GTF2I Specific to the Development Of
    cancers Article Primary Driver Mutations in GTF2I Specific to the Development of Thymomas Rumi Higuchi 1, Taichiro Goto 1,* , Yosuke Hirotsu 2 , Yujiro Yokoyama 1, Takahiro Nakagomi 1, Sotaro Otake 1, Kenji Amemiya 2,3, Toshio Oyama 3, Hitoshi Mochizuki 2 and Masao Omata 2,4 1 Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Yamanashi 400-8506, Japan; [email protected] (R.H.); [email protected] (Y.Y.); [email protected] (T.N.); [email protected] (S.O.) 2 Genome Analysis Center, Yamanashi Central Hospital, Yamanashi 400-8506, Japan; [email protected] (Y.H.); [email protected] (K.A.); [email protected] (H.M.); [email protected] (M.O.) 3 Department of Pathology, Yamanashi Central Hospital, Yamanashi 400-8506, Japan; [email protected] 4 Department of Gastroenterology, The University of Tokyo Hospital, Tokyo 113-8655, Japan * Correspondence: [email protected]; Tel.: +81-55-253-7111 Received: 16 June 2020; Accepted: 22 July 2020; Published: 24 July 2020 Abstract: Thymomas are rare mediastinal tumors that are difficult to treat and pose a major public health concern. Identifying mutations in target genes is vital for the development of novel therapeutic strategies. Type A thymomas possess a missense mutation in GTF2I (chromosome 7 c.74146970T>A) with high frequency. However, the molecular pathways underlying the tumorigenesis of other thymomas remain to be elucidated. We aimed to detect this missense mutation in GTF2I in other thymoma subtypes (types B). This study involved 22 patients who underwent surgery for thymomas between January 2014 and August 2019.
    [Show full text]
  • Plenary and Platform Abstracts
    American Society of Human Genetics 68th Annual Meeting PLENARY AND PLATFORM ABSTRACTS Abstract #'s Tuesday, October 16, 5:30-6:50 pm: 4. Featured Plenary Abstract Session I Hall C #1-#4 Wednesday, October 17, 9:00-10:00 am, Concurrent Platform Session A: 6. Variant Insights from Large Population Datasets Ballroom 20A #5-#8 7. GWAS in Combined Cancer Phenotypes Ballroom 20BC #9-#12 8. Genome-wide Epigenomics and Non-coding Variants Ballroom 20D #13-#16 9. Clonal Mosaicism in Cancer, Alzheimer's Disease, and Healthy Room 6A #17-#20 Tissue 10. Genetics of Behavioral Traits and Diseases Room 6B #21-#24 11. New Frontiers in Computational Genomics Room 6C #25-#28 12. Bone and Muscle: Identifying Causal Genes Room 6D #29-#32 13. Precision Medicine Initiatives: Outcomes and Lessons Learned Room 6E #33-#36 14. Environmental Exposures in Human Traits Room 6F #37-#40 Wednesday, October 17, 4:15-5:45 pm, Concurrent Platform Session B: 24. Variant Interpretation Practices and Resources Ballroom 20A #41-#46 25. Integrated Variant Analysis in Cancer Genomics Ballroom 20BC #47-#52 26. Gene Discovery and Functional Models of Neurological Disorders Ballroom 20D #53-#58 27. Whole Exome and Whole Genome Associations Room 6A #59-#64 28. Sequencing-based Diagnostics for Newborns and Infants Room 6B #65-#70 29. Omics Studies in Alzheimer's Disease Room 6C #71-#76 30. Cardiac, Valvular, and Vascular Disorders Room 6D #77-#82 31. Natural Selection and Human Phenotypes Room 6E #83-#88 32. Genetics of Cardiometabolic Traits Room 6F #89-#94 Wednesday, October 17, 6:00-7:00 pm, Concurrent Platform Session C: 33.
    [Show full text]
  • Whole Genome Comparative Genomic Hybridization of Ewing Sarcoma Indicates Cytoskeleton, Migration and Protein Trafficking †
    Proceedings Whole Genome Comparative Genomic Hybridization of Ewing Sarcoma Indicates Cytoskeleton, Migration and Protein Trafficking † Burçin Baran 1, Safiye Aktaş 1,*, Hülya Tosun 1,2, Gülden Diniz 1,2, Yasemin Çakır 1,2, Tekincan Çağrı Aktaş 1, Zekiye Altun 1 and Nur Olgun 1 1 Institute of Oncology, Dokuz Eylul University, Izmir 35340, Turkey; [email protected] (B.B.); [email protected] (H.T.); [email protected] (G.D.); [email protected] (Y.Ç.); [email protected] (T.Ç.A.); [email protected] (Z.A.); [email protected] (N.O.) 2 Dr.Behcet Uz Children’s Research Hospital, Izmir 35210, Turkey * Correspondence: [email protected] † Presented at the 2nd International Cell Death Research Congress, Izmir, Turkey, 1–4 November 2018. Published: 5 December 2018 Abstract: Ewing sarcoma is a bone and soft tissue tumor either neuroectodermal or mesenchymal originated and affecting children and adolescents. In the present study, we aimed to find out prognostic and predictive biomarkers for Ewing sarcoma. Hence, we examined the copy number alterations (and related possible genes) among ten Ewing sarcoma patient samples and possible associations with the clinical outcome. DNA extraction from formalin fixed paraffin embedded archive tissues was performed. Whole genome Comparative Genomic Hybridization (CGH) was performed by NimbleGen and recorded as single Panel Rainbow through chromosomes 1–22, X and Y. Data was interpreted by SignalMap software and genetic regions matching the deletion or amplification loci were recorded. The mean age of the patients was 8.6 years. Three of the cases were male, while seven were female. According to CGH analysis, the most common DNA copy number alterations were found in SLIT-ROBO Rho GTPase activating protein (srGAP2), RANBP2 like GRIP domain (RGPD5), nephrocystin 1 (NPHP1), GTF2I repeat domain containing 2 (GTF2IRD2), pyridoxal dependent decarboxylase domain containing 1 (PXDC1), which were found down- regulated among 7 of 10 patients.
    [Show full text]
  • Supplementary Data
    SUPPLEMENTARY DATA A cyclin D1-dependent transcriptional program predicts clinical outcome in mantle cell lymphoma Santiago Demajo et al. 1 SUPPLEMENTARY DATA INDEX Supplementary Methods p. 3 Supplementary References p. 8 Supplementary Tables (S1 to S5) p. 9 Supplementary Figures (S1 to S15) p. 17 2 SUPPLEMENTARY METHODS Western blot, immunoprecipitation, and qRT-PCR Western blot (WB) analysis was performed as previously described (1), using cyclin D1 (Santa Cruz Biotechnology, sc-753, RRID:AB_2070433) and tubulin (Sigma-Aldrich, T5168, RRID:AB_477579) antibodies. Co-immunoprecipitation assays were performed as described before (2), using cyclin D1 antibody (Santa Cruz Biotechnology, sc-8396, RRID:AB_627344) or control IgG (Santa Cruz Biotechnology, sc-2025, RRID:AB_737182) followed by protein G- magnetic beads (Invitrogen) incubation and elution with Glycine 100mM pH=2.5. Co-IP experiments were performed within five weeks after cell thawing. Cyclin D1 (Santa Cruz Biotechnology, sc-753), E2F4 (Bethyl, A302-134A, RRID:AB_1720353), FOXM1 (Santa Cruz Biotechnology, sc-502, RRID:AB_631523), and CBP (Santa Cruz Biotechnology, sc-7300, RRID:AB_626817) antibodies were used for WB detection. In figure 1A and supplementary figure S2A, the same blot was probed with cyclin D1 and tubulin antibodies by cutting the membrane. In figure 2H, cyclin D1 and CBP blots correspond to the same membrane while E2F4 and FOXM1 blots correspond to an independent membrane. Image acquisition was performed with ImageQuant LAS 4000 mini (GE Healthcare). Image processing and quantification were performed with Multi Gauge software (Fujifilm). For qRT-PCR analysis, cDNA was generated from 1 µg RNA with qScript cDNA Synthesis kit (Quantabio). qRT–PCR reaction was performed using SYBR green (Roche).
    [Show full text]
  • The Nuclear Localization Pattern and Interaction Partners of GTF2IRD1 Demonstrate a Role in Chromatin Regulation
    Hum Genet DOI 10.1007/s00439-015-1591-0 ORIGINAL INVESTIGATION The nuclear localization pattern and interaction partners of GTF2IRD1 demonstrate a role in chromatin regulation Paulina Carmona‑Mora1 · Jocelyn Widagdo2 · Florence Tomasetig1 · Cesar P. Canales1 · Yeojoon Cha1 · Wei Lee1 · Abdullah Alshawaf3 · Mirella Dottori3 · Renee M. Whan4 · Edna C. Hardeman1 · Stephen J. Palmer1 Received: 11 February 2015 / Accepted: 4 August 2015 © Springer-Verlag Berlin Heidelberg 2015 Abstract GTF2IRD1 is one of the three members of the mostly involved in chromatin modification and transcrip- GTF2I gene family, clustered on chromosome 7 within a tional regulation, whilst others indicate an unexpected role 1.8 Mb region that is prone to duplications and deletions in connection with the primary cilium. Mapping of the sites in humans. Hemizygous deletions cause Williams–Beuren of protein interaction also indicates key features regarding syndrome (WBS) and duplications cause WBS duplica- the evolution of the GTF2IRD1 protein. These data provide tion syndrome. These copy number variations disturb a a visual and molecular basis for GTF2IRD1 nuclear func- variety of developmental systems and neurological func- tion that will lead to an understanding of its role in brain, tions. Human mapping data and analyses of knockout mice behaviour and human disease. show that GTF2IRD1 and GTF2I underpin the craniofacial abnormalities, mental retardation, visuospatial deficits and Abbreviations hypersociability of WBS. However, the cellular role of the hESC Human embryonic stem cells GTF2IRD1 protein is poorly understood due to its very PLA Proximity ligation assay low abundance and a paucity of reagents. Here, for the first STED Stimulated emission depletion time, we show that endogenous GTF2IRD1 has a punctate WBS Williams–Beuren syndrome pattern in the nuclei of cultured human cell lines and neu- Y2H Yeast two-hybrid rons.
    [Show full text]
  • Monoclonal Antibodies
    NOTE Clinical Pathology Establishment of rat anti-canine DEP domain containing 1B (DEPDC1B) monoclonal antibodies Masaya IGASE1), Yuki MORINAGA1), Masahiro KATO2), Toshihiro TSUKUI2), Yusuke SAKAI3), Masaru OKUDA4) and Takuya MIZUNO1)* 1)Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 2)Nippon Zenyaku Kogyo Co., Ltd., 1-1 Tairanoue, Sasagawa, Asaka-machi, Koriyama, Fukushima 963-0196, Japan 3)Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 4)Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan ABSTRACT. DEP domain-containing 1B (DEPDC1B) is involved in the regulation of cell de- J. Vet. Med. Sci. adhesion and actin cytoskeleton activity during the G2/M transition of the cell cycle, and its 82(4): 483–487, 2020 overexpression has been proven to be associated with cancer progression in several human cancers. Canine DEPDC1B was identified as a gene that was overexpressed in canine lymphoma doi: 10.1292/jvms.19-0667 tissues in our previous study. However, in dogs, the protein expression of DEPDC1B remains to be determined due to the lack of a specific monoclonal antibody. Here, we developed rat monoclonal antibodies against canine DEPDC1B and characterized their applicability for immunodetection Received: 12 December 2019 assays. Our findings demonstrated that these antibodies are functional and can be important tools Accepted: 20 February 2020 to investigate the precise role of DEPDC1B in canine tumors. Advanced Epub: 6 March 2020 KEY WORDS: antibody, cancer, DEP domain-containing 1B, dog The DEP domain-containing 1B (DEPDC1B) gene was recently identified and localized at human chromosome 5 (5q12.1).
    [Show full text]
  • The Tumor Suppressor Notch Inhibits Head and Neck Squamous Cell
    The Texas Medical Center Library DigitalCommons@TMC The University of Texas MD Anderson Cancer Center UTHealth Graduate School of The University of Texas MD Anderson Cancer Biomedical Sciences Dissertations and Theses Center UTHealth Graduate School of (Open Access) Biomedical Sciences 12-2015 THE TUMOR SUPPRESSOR NOTCH INHIBITS HEAD AND NECK SQUAMOUS CELL CARCINOMA (HNSCC) TUMOR GROWTH AND PROGRESSION BY MODULATING PROTO-ONCOGENES AXL AND CTNNAL1 (α-CATULIN) Shhyam Moorthy Shhyam Moorthy Follow this and additional works at: https://digitalcommons.library.tmc.edu/utgsbs_dissertations Part of the Biochemistry, Biophysics, and Structural Biology Commons, Cancer Biology Commons, Cell Biology Commons, and the Medicine and Health Sciences Commons Recommended Citation Moorthy, Shhyam and Moorthy, Shhyam, "THE TUMOR SUPPRESSOR NOTCH INHIBITS HEAD AND NECK SQUAMOUS CELL CARCINOMA (HNSCC) TUMOR GROWTH AND PROGRESSION BY MODULATING PROTO-ONCOGENES AXL AND CTNNAL1 (α-CATULIN)" (2015). The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access). 638. https://digitalcommons.library.tmc.edu/utgsbs_dissertations/638 This Dissertation (PhD) is brought to you for free and open access by the The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at DigitalCommons@TMC. It has been accepted for inclusion in The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access) by an authorized administrator of DigitalCommons@TMC. For more information, please contact [email protected]. THE TUMOR SUPPRESSOR NOTCH INHIBITS HEAD AND NECK SQUAMOUS CELL CARCINOMA (HNSCC) TUMOR GROWTH AND PROGRESSION BY MODULATING PROTO-ONCOGENES AXL AND CTNNAL1 (α-CATULIN) by Shhyam Moorthy, B.S.
    [Show full text]
  • (ENU) Mutagenized Mouse Model for Autosomal Dominant Non-Syndromic Kyphoscoliosis Due to Vertebral Fusion
    This is a repository copy of An N -ethyl- N -nitrosourea (ENU) mutagenized mouse model for autosomal dominant non-syndromic kyphoscoliosis due to vertebral fusion. White Rose Research Online URL for this paper: http://eprints.whiterose.ac.uk/133327/ Version: Published Version Article: Esapa, C.T., Piret, S.E., Nesbit, M.A. et al. (13 more authors) (2018) An N -ethyl- N -nitrosourea (ENU) mutagenized mouse model for autosomal dominant non-syndromic kyphoscoliosis due to vertebral fusion. JBMR Plus, 2 (3). pp. 154-163. ISSN 2473-4039 https://doi.org/10.1002/jbm4.10033 Reuse This article is distributed under the terms of the Creative Commons Attribution (CC BY) licence. This licence allows you to distribute, remix, tweak, and build upon the work, even commercially, as long as you credit the authors for the original work. More information and the full terms of the licence here: https://creativecommons.org/licenses/ Takedown If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request. [email protected] https://eprints.whiterose.ac.uk/ ORIGINAL ARTICLE An N-Ethyl-N-Nitrosourea (ENU) Mutagenized Mouse Model for Autosomal Dominant Nonsyndromic Kyphoscoliosis Due to Vertebral Fusion Christopher T Esapa,1,2Ã Sian E Piret,1Ã M Andrew Nesbit,1,3 Gethin P Thomas,4,5 Leslie A Coulton,6 Orla M Gallagher,6 Michelle M Simon,2 Saumya Kumar,2,7 Ann-Marie Mallon,2 Ilaria Bellantuono,6 Matthew A Brown,4 Peter
    [Show full text]
  • Curcumin Alters Gene Expression-Associated DNA Damage, Cell Cycle, Cell Survival and Cell Migration and Invasion in NCI-H460 Human Lung Cancer Cells in Vitro
    ONCOLOGY REPORTS 34: 1853-1874, 2015 Curcumin alters gene expression-associated DNA damage, cell cycle, cell survival and cell migration and invasion in NCI-H460 human lung cancer cells in vitro I-TSANG CHIANG1,2, WEI-SHU WANG3, HSIN-CHUNG LIU4, SU-TSO YANG5, NOU-YING TANG6 and JING-GUNG CHUNG4,7 1Department of Radiation Oncology, National Yang‑Ming University Hospital, Yilan 260; 2Department of Radiological Technology, Central Taiwan University of Science and Technology, Taichung 40601; 3Department of Internal Medicine, National Yang‑Ming University Hospital, Yilan 260; 4Department of Biological Science and Technology, China Medical University, Taichung 404; 5Department of Radiology, China Medical University Hospital, Taichung 404; 6Graduate Institute of Chinese Medicine, China Medical University, Taichung 404; 7Department of Biotechnology, Asia University, Taichung 404, Taiwan, R.O.C. Received March 31, 2015; Accepted June 26, 2015 DOI: 10.3892/or.2015.4159 Abstract. Lung cancer is the most common cause of cancer CARD6, ID1 and ID2 genes, associated with cell survival and mortality and new cases are on the increase worldwide. the BRMS1L, associated with cell migration and invasion. However, the treatment of lung cancer remains unsatisfactory. Additionally, 59 downregulated genes exhibited a >4-fold Curcumin has been shown to induce cell death in many human change, including the DDIT3 gene, associated with DNA cancer cells, including human lung cancer cells. However, the damage; while 97 genes had a >3- to 4-fold change including the effects of curcumin on genetic mechanisms associated with DDIT4 gene, associated with DNA damage; the CCPG1 gene, these actions remain unclear. Curcumin (2 µM) was added associated with cell cycle and 321 genes with a >2- to 3-fold to NCI-H460 human lung cancer cells and the cells were including the GADD45A and CGREF1 genes, associated with incubated for 24 h.
    [Show full text]
  • Supplementary Figure Legends
    1 Supplementary Figure legends 2 Supplementary Figure 1. 3 Experimental workflow. 4 5 Supplementary Figure 2. 6 IRF9 binding to promoters. 7 a) Verification of mIRF9 antibody by site-directed ChIP. IFNβ-stimulated binding of IRF9 to 8 the ISRE sequences of Mx2 was analyzed using BMDMs of WT and Irf9−/− (IRF9-/-) mice. 9 Cells were treated with 250 IU/ml of IFNβ for 1.5h. Data represent mean and SEM values of 10 three independent experiments. P-values were calculated using the paired ratio t-test (*P ≤ 11 0.05; **P ≤ 0.01, ***P ≤ 0.001). 12 b) Browser tracks showing complexes assigned as STAT-IRF9 in IFNγ treated wild type 13 BMDMs. Input, STAT2, IRF9 (scale 0-200). STAT1 (scale 0-150). 14 15 Supplementary Figure 3. 16 Experimental system for BioID. 17 a) Kinetics of STAT1, STAT2 and IRF9 synthesis in Raw 264.7 macrophages and wild type 18 BMDMs treated with 250 IU/ml as indicated. Whole-cell extracts were tested in western blot 19 for STAT1 phosphorylation at Y701 and of STAT2 at Y689 as well as total STAT1, STAT2, 20 IRF9 and GAPDH levels. The blots are representative of three independent experiments. b) 21 Irf9-/- mouse embryonic fibroblasts (MEFs) were transiently transfected with the indicated 22 expression vectors, including constitutively active IRF7-M15. One day after transfection, 23 RNA was isolated and Mx2 expression determined by qPCR. c) Myc-BirA*-IRF9 transgenic 24 Raw 264.7 were treated with increasing amounts of doxycycline (dox) (0,2µg/ml, 0,4µg/ml, 25 0,6µg/ml, 0,8µg/ml, 1mg/ml) and 50µM biotin.
    [Show full text]