Analysis of Phosphatases in ER-Negative Breast Cancers Identifies DUSP4 As a Critical Regulator of Growth and Invasion

Total Page:16

File Type:pdf, Size:1020Kb

Analysis of Phosphatases in ER-Negative Breast Cancers Identifies DUSP4 As a Critical Regulator of Growth and Invasion Breast Cancer Res Treat DOI 10.1007/s10549-016-3892-y PRECLINICAL STUDY Analysis of phosphatases in ER-negative breast cancers identifies DUSP4 as a critical regulator of growth and invasion 1 1 1 2 Abhijit Mazumdar • Graham M. Poage • Jonathan Shepherd • Anna Tsimelzon • 1 1 1 1 Zachary C. Hartman • Petra Den Hollander • Jamal Hill • Yun Zhang • 3 2 2 2 Jenny Chang • Susan G. Hilsenbeck • Suzanne Fuqua • C. Kent Osborne • 4 1 Gordon B. Mills • Powel H. Brown Received: 28 January 2016 / Accepted: 27 June 2016 Ó The Author(s) 2016. This article is published with open access at Springerlink.com Abstract Estrogen receptor (ER)-negative cancers have a phosphoproteins. We identified 48 phosphatase genes are poor prognosis, and few targeted therapies are available for significantly differentially expressed in ER-negative com- their treatment. Our previous analyses have identified pared to those in ER-positive breast tumors. We discovered potential kinase targets critical for the growth of ER-neg- that 31 phosphatases were more highly expressed, while 11 ative, progesterone receptor (PR)-negative and HER2- were underexpressed specifically in ER-negative breast negative, or ‘‘triple-negative’’ breast cancer (TNBC). cancers. The DUSP4 gene is underexpressed in ER-nega- Because phosphatases regulate the function of kinase sig- tive breast cancer and is deleted in approximately 50 % of naling pathways, in this study, we investigated whether breast cancers. Induced DUSP4 expression suppresses both phosphatases are also differentially expressed in ER-neg- in vitro and in vivo growths of breast cancer cells. Our ative compared to those in ER-positive breast cancers. We studies show that induced DUSP4 expression blocks the compared RNA expression in 98 human breast cancers (56 cell cycle at the G1/S checkpoint; inhibits ERK1/2, p38, ER-positive and 42 ER-negative) to identify phosphatases JNK1, RB, and NFkB p65 phosphorylation; and inhibits differentially expressed in ER-negative compared to those invasiveness of TNBC cells. These results suggest that that in ER-positive breast cancers. We then examined the DUSP4 is a critical regulator of the growth and invasion of effects of one selected phosphatase, dual specificity phos- triple-negative breast cancer cells. phatase 4 (DUSP4), on proliferation, cell growth, migration and invasion, and on signaling pathways using protein Keywords TNBC Á Phosphatase Á Mouse xenograft Á microarray analyses of 172 proteins, including MAPK pathways Electronic supplementary material The online version of this Introduction article (doi:10.1007/s10549-016-3892-y) contains supplementary material, which is available to authorized users. Breast cancer is the most frequently diagnosed cancer in & Powel H. Brown women and the second highest cause of cancer-related [email protected] death in United States women in 2016 [1]. Selective 1 estrogen receptor modulators (SERMs) and aromatase Department of Clinical Cancer Prevention, The University of inhibitors are currently used to treat ER-positive breast Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA cancers [2–6], while anti-HER2 drugs are used to treat ER- negative HER2-positive breast cancers [7–9]. However, 2 Department of Medicine and the Dan L. Duncan Cancer Center, Lester and Sue Smith Breast Center, Baylor College there are few treatment options available for breast cancers of Medicine, Houston, USA that do not express ER, progesterone receptor (PR), or the 3 Methodist Cancer Center, The Methodist Hospital Research HER2 protein, otherwise known as ‘‘triple-negative’’ breast Institute, Houston, USA cancers (TNBCs). As no targeted therapy is available for 4 Department of Systems Biology, The University of Texas most TNBC tumors, cytotoxic chemotherapy is the current M.D. Anderson Cancer Center, Houston, USA treatment strategy. 123 Breast Cancer Res Treat We previously conducted cDNA microarray studies to genes. Statistical analysis was done using Partek software identify potential therapeutic targets of ER-negative breast (http://partek.com). The following criteria were used to cancers [10]. These studies identified several kinases crit- find genes differentially expressed between ER-negative ical for the growth of TNBC. In this study, we sought to and ER-positive tumors: Benjamini–Hochberg false dis- identify phosphatases differentially expressed in ER-neg- covery rate (FDR) p value B 0.1, fold change C1.2 ative tumors that also regulate breast cancer growth. We or B0.8. identified 11 underexpressed and 31 overexpressed phos- phatases in ER-negative compared to those in ER-positive Cell lines and cell culture breast cancers. We initially focused on those 11 phos- phatases underexpressed exhibiting decreased expression The MCF-7 (HTB-22), MDA-MB-231 (HTB-26), and in ER-negative tumors and found that of these 11 under- MDA-MB-468 (HTB-132) cell lines (American Type expressed phosphatases, DUSP4 is the most frequently Culture Collection (ATCC), Manassas, VA, USA) were deleted phosphatase in ER-negative breast cancer. cultured in DMEM (Cellgro by Mediatech, Inc., Manassas, DUSP4 specifically regulates extracellular regulated VA, USA) supplemented with 10 % fetal bovine serum, kinase (ERK), and the phosphatase activity of DUSP4 is 100 U/ml penicillin, and 100 mg/ml streptomycin. These enhanced upon interaction with ERK and p38 [11–13]. were grown and maintained as described in Supplementary DUSP4 expression has been shown to play an important Methods. STR profiles were compared to (1) known ATCC role in senescence [14] and emerging evidence suggests fingerprints (ATCC.org); (2) the Cell Line Integrated DUSP4 is involved in the growth and progression of cancer Molecular Authentication database (CLIMA) version [15, 16]. Our results demonstrate that DUSP4 is frequently 0.1.200808 (http://bioinformatics.istge.it/clima/)[17]; and deleted in breast cancer and underexpressed in TNBCs, and (3) the MD Anderson fingerprint database. that DUSP4 overexpression halts TNBC tumor growth and invasion. Our results also demonstrate that all three MAPK Reagents and antibodies proteins (ERK1/2, p38, and JNK1) are negatively regulated by DUSP4. Moreover, re-expression of DUSP4 also inhi- The DUSP4 antibody was purchased from BD Transduc- bits other growth inducing pathways, including NFjB and tion Laboratories (Lexington, KY, USA). Phospho-ERK1/2 Rb. Overall, this study demonstrates that DUSP4 is fre- (#4370) and total ERK1/2 (#4372) were purchased from quently deleted in breast cancers and is a critical regulator Cell Signaling Technology, Inc. (Danvers, MA, USA). of growth and invasion of TNBCs, suggesting DUSP4 is an Antiactin antibody was purchased from Sigma-Aldrich important tumor suppressor gene in these aggressive breast Corp. (St. Louis, MO, USA). Antimouse (#NA931 V) and cancers. antirabbit (NA934 V) secondary antibodies were obtained from GE Healthcare Bio-sciences Corp. (Piscataway, NJ, USA). Materials and methods Viral vectors and modification of cell lines Breast tumor and microarray datasets The DUSP4 ORF clone was obtained from Open Biosystems All tumors were collected by Dr. Jenny Chang and (Huntsville, AL, USA) and cloned into tetracycline (Tet)- approved by the institutional review board of Baylor Col- inducible vector (pTIPZ) through a gateway LR clonase lege of Medicine. This tumor set, including tumor stage, reaction. Final constructs were verified through restriction size, and patient race and menopausal status, has been digestion and sequencing. Lentiviral vectors were prepared previously described [10]. In this study, four samples were as described previously [18]. Stable cell lines expressing removed from the analysis. These four samples had an inducible cDNAs were generated by lentiviral infection unconfirmed ER status or appeared as outliers on the using a pTIPZ lentiviral expression system in the presence of principal component analysis (PCA) plot and thus removed 4 lg/ml polybrene, followed by puromycin selection at 48 h from the dataset. Therefore, 98 samples, 56 ER-positive post infection. All pTIPZ stable cell lines were maintained in and 42 ER-negative invasive breast cancers, were used for media with Tet-safe serum (Clontech Laboratories Inc., this analysis. Gene expression was estimated using the Mountain View, CA, USA). Robust Multi-array Average (RMA) procedure. We limited our data and clustering analysis to 262 genes (454 probe- Quantitative RT-PCR (qRT-PCR) assessments sets on Affymetrix U133A chip), which encode known phosphatases and proteins with phosphatase in their name. Quantitative PCR assays of reverse-transcribed transcripts We used the most variable probeset for each of the 262 (4 replicates per transcript) were carried out using an ABI 123 Breast Cancer Res Treat PRISM 7900 Sequence Detection System (Life Technolo- Reverse phase protein array (RPPA) gies (formerly Applied Biosystems Inc., Foster City, CA, USA), as previously described [10]. RPPA assays were performed in the MD Anderson Core facility. Experimental details are provided in Supplemen- Cell growth assays tary Materials. Cellular proliferation and anchorage-independent growth Mouse experiments were measured as described previously [19]. Experiments using nude mice (The Jackson Laboratory, Cell cycle assays Bar Harbor, ME, USA) were performed in accordance with M.D. Anderson Institutional Animal Care and Use Com- To measure cell cycle distribution, Tet-inducible cells were mittee (IACUC)-approved protocols. Experimental details treated for 4 days with or without doxycycline
Recommended publications
  • Myotubularin-Related Protein (MTMR) 9 Determines the Enzymatic Activity, Substrate Specificity, and Role in Autophagy of MTMR8
    Myotubularin-related protein (MTMR) 9 determines the enzymatic activity, substrate specificity, and role in autophagy of MTMR8 Jun Zoua,1, Chunfen Zhangb,1,2, Jasna Marjanovicc, Marina V. Kisselevab, Philip W. Majerusb,d,2, and Monita P. Wilsonb,2 aDepartment of Pathology and Immunology, bDivision of Hematology, Department of Internal Medicine, and dDepartment of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110; and cDivision of Basic and Pharmaceutical Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110 Contributed by Philip W. Majerus, May 1, 2012 (sent for review February 24, 2012) The myotubularins are a large family of inositol polyphosphate myotubularin proteins (16–21). One mechanism that regulates 3-phosphatases that, despite having common substrates, subsume the myotubularins is the formation of heterodimers between unique functions in cells that are disparate. The myotubularin catalytically active and inactive proteins. The interaction between family consists of 16 different proteins, 9 members of which different myotubularin proteins has a significant effect on en- possess catalytic activity, dephosphorylating phosphatidylinositol zymatic activity. For example, the association of myotubularin 3-phosphate [PtdIns(3)P] and phosphatidylinositol 3,5-bisphos- (MTM1) with MTMR12 results in a threefold increase in the 3- phate [PtdIns(3,5)P2] at the D-3 position. Seven members are in- phosphatase activity of MTM1, alters the subcellular localiza- active because they lack the conserved cysteine residue in the tion of MTM1 from the plasma membrane to the cytosol, and CX5R motif required for activity. We studied a subfamily of homol- attenuates the filopodia formation seen with MTM1 overex- ogous myotubularins, including myotubularin-related protein 6 pression (21, 22).
    [Show full text]
  • Myotubularin-Related Phosphatase 5 Is a Critical Determinant of Autophagy in Neurons
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.20.453106; this version posted July 20, 2021. 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. Myotubularin-related phosphatase 5 is a critical determinant of autophagy in neurons Jason P. Chua*1,8, Karan Bedi2,3,4, Michelle T. Paulsen2,4, Mats Ljungman2,4, Elizabeth M. H. Tank1, Erin S. Kim1, Jennifer M. Colón-Mercado7, Michael E. Ward7, Lois S. Weisman5,6, and Sami J. Barmada*1,8 1Department of Neurology, University of Michigan, Ann Arbor, MI, USA 2Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA 3Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA 4Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA 5Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA 6Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA 7National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA 8Lead contact *Correspondence: [email protected] (J.P.C.), [email protected] (S.J.B.) 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.07.20.453106; this version posted July 20, 2021. 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 Autophagy is a conserved, multi-step process of capturing proteolytic cargo in autophagosomes for lysosome degradation. The capacity to remove toxic proteins that accumulate in neurodegenerative disorders attests to the disease-modifying potential of the autophagy pathway.
    [Show full text]
  • Genes Retina/RPE Choroid Sclera
    Supplementary Materials: Genes Retina/RPE Choroid Sclera Fold Change p-value Fold Change p-value Fold Change p-value PPFIA2 NS NS 2.35 1.3X10-3 1.5 1.6X10-3 PTPRF 1.24 2.65X10-5 6.42 7X10-4 1.11 1X10-4 1.19 2.65X10-5 NS NS 1.11 3.3X10-3 PTPRR 1.44 2.65X10-5 3.04 4.7X10-3 NS NS Supplementary Table S1. Genes Differentially Expressed Related to Candidate Genes from Association. Genes selected for follow up validation by real time quantitative PCR. Multiple values for each gene indicate multiple probes within the same gene. NS indicates the fold change was not statistically significant. Gene/SNP Assay ID rs4764971 C__30866249_10 rs7134216 C__30023434_10 rs17306116 C__33218892_10 rs3803036 C__25749934_20 rs824311 C___8342112_10 PPFIA2 Hs00170308_m1 PTPRF Hs00160858_m1 PTPRR Hs00373136_m1 18S Hs03003631_g1 GAPDH Hs02758991_g1 Supplementary Table S2. Taqman® Genotyping and Gene Expression Assay Identification Numbers. SNP Chimp Orangutan Rhesus Marmoset Mouse Rat Cow Pig Guinea Pig Dog Elephant Opossum Chicken rs3803036 X X X X X X X X X X X X X rs1520562 X X X X X X rs1358228 X X X X X X X X X X X rs17306116 X X X X X X rs790436 X X X X X X X rs1558726 X X X X X X X X rs741525 X X X X X X X X rs7134216 X X X X X X rs4764971 X X X X X X X Supplementary Table S3. Conservation of Top SNPs from Association. X indicates SNP is conserved.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Distinct Transcriptional Programming Drive Response to MAPK Inhibition in BRAF -Mutant Melanoma Patient-Derived Xenografts
    Published OnlineFirst September 16, 2019; DOI: 10.1158/1535-7163.MCT-19-0028 Cancer Biology and Translational Studies Molecular Cancer Therapeutics Distinct Transcriptional Programming Drive Response to MAPK Inhibition in BRAFV600-Mutant Melanoma Patient-Derived Xenografts Tianshu Feng, Javad Golji, Ailing Li, Xiamei Zhang, David A. Ruddy, Daniel P. Rakiec, Felipe C. Geyer, Jane Gu, Hui Gao, Juliet A.Williams, Darrin D. Stuart, and Matthew J. Meyer Abstract Inhibitors targeting BRAF and its downstream kinase MEK are preferentially sensitive to MAPK inhibition. These gene- produce robust response in patients with advanced BRAFV600- expression programs are somewhat similar to the MITF-high mutant melanoma. However, the duration and depth of and -low phenotypes described in cancer cell lines, but response vary significantly between patients; therefore, pre- demonstrate an inverse relationship with drug response. dicting response a priori remains a significant challenge. Here, This suggests a discrepancy between in vitro and in vivo we utilized the Novartis collection of patient-derived xeno- experimental systems that warrants future investigations. grafts to characterize transcriptional alterations elicited by Finally, BRAFV600-mutant melanoma relies on either MAPK BRAF and MEK inhibitors in vivo, in an effort to identify or alternative pathways for survival under BRAF and MEK mechanisms governing differential response to MAPK inhibi- inhibition in vivo, which in turn predicts their response to tion. We show that the expression of an MITF-high, "epithelial- further pathway suppression using a combination of BRAF, like" transcriptional program is associated with reduced MEK, and ERK inhibitors. Our findings highlight the inter- sensitivity and adaptive response to BRAF and MEK inhibitor tumor heterogeneity in BRAFV600-mutant melanoma, and treatment.
    [Show full text]
  • Molecular and Genetic Medicine
    Bertazzi et al., J Mol Genet Med 2015, 8:2 Molecular and Genetic Medicine http://dx.doi.org/10.4172/1747-0862.1000116 Review Article Open Access Myotubularin MTM1 Involved in Centronuclear Myopathy and its Roles in Human and Yeast Cells Dimitri L. Bertazzi#, Johan-Owen De Craene# and Sylvie Friant* Department of Molecular and Cellular Genetics, UMR7156, Université de Strasbourg and CNRS, France #Authors contributed equally to this work. *Corresponding author: Friant S, Department of Molecular and Cellular Genetics, UMR7156, Université de Strasbourg and CNRS, 67084 Strasbourg, France, E-mail: [email protected] Received date: April 17, 2014; Accepted date: July 21, 2014; Published date: July 28, 2014 Copyright: © 2014 Bertazzi DL, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Abstract Mutations in the MTM1 gene, encoding the phosphoinositide phosphatase myotubularin, are responsible for the X-linked centronuclear myopathy (XLCNM) or X-linked myotubular myopathy (XLMTM). The MTM1 gene was first identified in 1996 and its function as a PtdIns3P and PtdIns(,5)P2 phosphatase was discovered in 2000. In recent years, very important progress has been made to set up good models to study MTM1 and the XLCNM disease such as knockout or knockin mice, the Labrador Retriever dog, the zebrafish and the yeast Saccharomyces cerevisiae. These helped to better understand the cellular function of MTM1 and of its four conserved domains: PH-GRAM (Pleckstrin Homology-Glucosyltransferase, Rab-like GTPase Activator and Myotubularin), RID (Rac1-Induced recruitment Domain), PTP/DSP (Protein Tyrosine Phosphatase/Dual-Specificity Phosphatase) and SID (SET-protein Interaction Domain).
    [Show full text]
  • Bioinformatics-Based Screening of Key Genes for Transformation of Liver
    Jiang et al. J Transl Med (2020) 18:40 https://doi.org/10.1186/s12967-020-02229-8 Journal of Translational Medicine RESEARCH Open Access Bioinformatics-based screening of key genes for transformation of liver cirrhosis to hepatocellular carcinoma Chen Hao Jiang1,2, Xin Yuan1,2, Jiang Fen Li1,2, Yu Fang Xie1,2, An Zhi Zhang1,2, Xue Li Wang1,2, Lan Yang1,2, Chun Xia Liu1,2, Wei Hua Liang1,2, Li Juan Pang1,2, Hong Zou1,2, Xiao Bin Cui1,2, Xi Hua Shen1,2, Yan Qi1,2, Jin Fang Jiang1,2, Wen Yi Gu4, Feng Li1,2,3 and Jian Ming Hu1,2* Abstract Background: Hepatocellular carcinoma (HCC) is the most common type of liver tumour, and is closely related to liver cirrhosis. Previous studies have focussed on the pathogenesis of liver cirrhosis developing into HCC, but the molecular mechanism remains unclear. The aims of the present study were to identify key genes related to the transformation of cirrhosis into HCC, and explore the associated molecular mechanisms. Methods: GSE89377, GSE17548, GSE63898 and GSE54236 mRNA microarray datasets from Gene Expression Omni- bus (GEO) were analysed to obtain diferentially expressed genes (DEGs) between HCC and liver cirrhosis tissues, and network analysis of protein–protein interactions (PPIs) was carried out. String and Cytoscape were used to analyse modules and identify hub genes, Kaplan–Meier Plotter and Oncomine databases were used to explore relationships between hub genes and disease occurrence, development and prognosis of HCC, and the molecular mechanism of the main hub gene was probed using Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis.
    [Show full text]
  • The Regulatory Roles of Phosphatases in Cancer
    Oncogene (2014) 33, 939–953 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW The regulatory roles of phosphatases in cancer J Stebbing1, LC Lit1, H Zhang, RS Darrington, O Melaiu, B Rudraraju and G Giamas The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3--kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies. Oncogene (2014) 33, 939–953; doi:10.1038/onc.2013.80; published online 18 March 2013 Keywords: cancer; phosphatases; solid tumours GASTROINTESTINAL MALIGNANCIES abs in sera were significantly associated with poor survival in Oesophageal cancer advanced ESCC, suggesting that they may have a clinical utility in Loss of PTEN (phosphatase and tensin homologue deleted on ESCC screening and diagnosis.5 chromosome 10) expression in oesophageal cancer is frequent, Cao et al.6 investigated the role of protein tyrosine phosphatase, among other gene alterations characterizing this disease. Zhou non-receptor type 12 (PTPN12) in ESCC and showed that PTPN12 et al.1 found that overexpression of PTEN suppresses growth and protein expression is higher in normal para-cancerous tissues than induces apoptosis in oesophageal cancer cell lines, through in 20 ESCC tissues.
    [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]
  • Targeting Protein Tyrosine Phosphatases in Cancer Lakshmi Reddy Bollu, Abhijit Mazumdar, Michelle I
    Published OnlineFirst January 13, 2017; DOI: 10.1158/1078-0432.CCR-16-0934 Molecular Pathways Clinical Cancer Research Molecular Pathways: Targeting Protein Tyrosine Phosphatases in Cancer Lakshmi Reddy Bollu, Abhijit Mazumdar, Michelle I. Savage, and Powel H. Brown Abstract The aberrant activation of oncogenic signaling pathways is a act as tumor suppressor genes by terminating signal responses universal phenomenon in cancer and drives tumorigenesis and through the dephosphorylation of oncogenic kinases. More malignant transformation. This abnormal activation of signal- recently, it has become clear that several PTPs overexpressed ing pathways in cancer is due to the altered expression of in human cancers do not suppress tumor growth; instead, they protein kinases and phosphatases. In response to extracellular positively regulate signaling pathways and promote tumor signals, protein kinases activate downstream signaling path- development and progression. In this review, we discuss both ways through a series of protein phosphorylation events, ulti- types of PTPs: those that have tumor suppressor activities as mately producing a signal response. Protein tyrosine phospha- well as those that act as oncogenes. We also discuss the tases (PTP) are a family of enzymes that hydrolytically remove potential of PTP inhibitors for cancer therapy. Clin Cancer Res; phosphate groups from proteins. Initially, PTPs were shown to 23(9); 1–7. Ó2017 AACR. Background in cancer and discuss the current status of PTP inhibitors for cancer therapy. Signal transduction is a complex process that transmits extra- PTPs belong to a superfamily of enzymes that hydrolytically cellular signals effectively through a cascade of events involving remove phosphate groups from proteins (2).
    [Show full text]
  • Bortezomib Inhibits Lung Fibrosis and Fibroblast Activation Without Proteasome
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.26.433086; this version posted February 26, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Bortezomib inhibits lung fibrosis and fibroblast activation without proteasome 2 inhibition 3 4 Loka Raghu Kumar Penke, Jennifer Speth, Scott Wettlaufer, Christina Draijer and Marc 5 Peters-Golden1* 6 7 Affiliations 8 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, 9 University of Michigan, Ann Arbor, Michigan, USA. 10 1 Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA. 11 12 Correspondence: 13 Marc Peters-Golden, M.D. 14 6301 MSRBIII, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-5642 15 Tel: 734-936-5047 16 Fax: 734-764-4556 17 Email: [email protected] 18 Author Contributions: L.R.K.P. planned and performed experiments, analyzed the 19 data, organized data for presentation, and wrote the manuscript. J.M.S, S.H.W and C.D 20 performed experiments. M.P.-G. planned experiments, analyzed data, and wrote the 21 manuscript. 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.26.433086; this version posted February 26, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.
    [Show full text]
  • A Peripheral Blood Gene Expression Signature to Diagnose Subclinical Acute Rejection
    CLINICAL RESEARCH www.jasn.org A Peripheral Blood Gene Expression Signature to Diagnose Subclinical Acute Rejection Weijia Zhang,1 Zhengzi Yi,1 Karen L. Keung,2 Huimin Shang,3 Chengguo Wei,1 Paolo Cravedi,1 Zeguo Sun,1 Caixia Xi,1 Christopher Woytovich,1 Samira Farouk,1 Weiqing Huang,1 Khadija Banu,1 Lorenzo Gallon,4 Ciara N. Magee,5 Nader Najafian,5 Milagros Samaniego,6 Arjang Djamali ,7 Stephen I. Alexander,2 Ivy A. Rosales,8 Rex Neal Smith,8 Jenny Xiang,3 Evelyne Lerut,9 Dirk Kuypers,10,11 Maarten Naesens ,10,11 Philip J. O’Connell,2 Robert Colvin,8 Madhav C. Menon,1 and Barbara Murphy1 Due to the number of contributing authors, the affiliations are listed at the end of this article. ABSTRACT Background In kidney transplant recipients, surveillance biopsies can reveal, despite stable graft function, histologic features of acute rejection and borderline changes that are associated with undesirable graft outcomes. Noninvasive biomarkers of subclinical acute rejection are needed to avoid the risks and costs associated with repeated biopsies. Methods We examined subclinical histologic and functional changes in kidney transplant recipients from the prospective Genomics of Chronic Allograft Rejection (GoCAR) study who underwent surveillance biopsies over 2 years, identifying those with subclinical or borderline acute cellular rejection (ACR) at 3 months (ACR-3) post-transplant. We performed RNA sequencing on whole blood collected from 88 indi- viduals at the time of 3-month surveillance biopsy to identify transcripts associated with ACR-3, developed a novel sequencing-based targeted expression assay, and validated this gene signature in an independent cohort.
    [Show full text]