1
|
A new IRES-mediated truncated Cx32 isoform inhibits global mRNA translation to suppress glioblastoma. Biomed Pharmacother 2023; 161:114513. [PMID: 36931032 DOI: 10.1016/j.biopha.2023.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most lethal malignant primary brain tumor. Although multimodal therapy has been applied for GBM, the median survival time remains less than 16 months. Thus, better therapeutic targets in GBM are urgently needed. Herein, we first identified five new N-terminal-truncated Cx32 isoforms (GJB1-28k, GJB1-22k, GJB1-20k, GJB1-15k, and GJB1-13k) and further demonstrated that they were generated via cap-independent internal translation through internal ribosome entry sites (IRESs) in the coding sequence of GJB1 mRNA. Among these isoforms, GJB1-13k inhibited proliferation, promoted apoptosis, and limited cell cycle progression in GBM cells by inhibiting global mRNA translation. In vivo experiments further confirmed the antitumor activity of GJB1-13k against GBM cells. In addition, TSR3, a ribosomal maturation factor, was demonstrated to directly interact with GJB1-13k. Moreover, GBM cells with high TSR3 expression exhibited low sensitivity to GJB1-13k treatment, while GJB1-13k sensitivity was restored by TSR3 knockdown. Our work identifies a new IRES-mediated protein, GJB1-13k, and suggests that overexpression of GJB1-13k in GBM cells with low TSR3 expression or combined targeting of GJB1-13k and TSR3 in GBM cells with high TSR3 expression constitutes a potential therapeutic strategy for GBM.
Collapse
|
2
|
Engineered Zinc Finger Protein Targeting 2LTR Inhibits HIV Integration in Hematopoietic Stem and Progenitor Cell-Derived Macrophages: In Vitro Study. Int J Mol Sci 2022; 23:ijms23042331. [PMID: 35216446 PMCID: PMC8875109 DOI: 10.3390/ijms23042331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/22/2022] Open
Abstract
Human hematopoietic stem/progenitor cell (HSPC)-based gene therapy is a promising direction for curing HIV-1-infected individuals. The zinc finger protein (2LTRZFP) designed to target the 2-LTR-circle junction of HIV-1 cDNA was previously reported as an intracellular antiviral molecular scaffold that prevents HIV integration. Here, we elucidate the efficacy and safety of using 2LTRZFP in human CD34+ HSPCs. We transduced 2LTRZFP which has the mCherry tag (2LTRZFPmCherry) into human CD34+ HSPCs using a lentiviral vector. The 2LTRZFPmCherry-transduced HSPCs were subsequently differentiated into macrophages. The expression levels of pro-apoptotic proteins of the 2LTRZFPmCherry-transduced HSPCs showed no significant difference from those of the non-transduced control. Furthermore, the 2LTRZFPmCherry-transduced HSPCs were successfully differentiated into mature macrophages, which had normal phagocytic function. The cytokine secretion assay demonstrated that 2LTRZFPmCherry-transduced CD34+ derived macrophages promoted the polarization towards classically activated (M1) subtypes. More importantly, the 2LTRZFPmCherry transduced cells significantly exhibited resistance to HIV-1 integration in vitro. Our findings demonstrate that the 2LTRZFPmCherry-transduced macrophages were found to be functionally and phenotypically normal, with no adverse effects of the anti-HIV-1 scaffold. Our data suggest that the anti-HIV-1 integrase scaffold is a promising antiviral molecule that could be applied to human CD34+ HSPC-based gene therapy for AIDS patients.
Collapse
|
3
|
Saliakoura M, Sebastiano MR, Nikdima I, Pozzato C, Konstantinidou G. Restriction of extracellular lipids renders pancreatic cancer dependent on autophagy. J Exp Clin Cancer Res 2022; 41:16. [PMID: 34998392 PMCID: PMC8742413 DOI: 10.1186/s13046-021-02231-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND KRAS is the predominant oncogene mutated in pancreatic ductal adenocarcinoma (PDAC), the fourth cause of cancer-related deaths worldwide. Mutant KRAS-driven tumors are metabolically programmed to support their growth and survival, which can be used to identify metabolic vulnerabilities. In the present study, we aimed to understand the role of extracellularly derived fatty acids in KRAS-driven pancreatic cancer. METHODS To assess the dependence of PDAC cells on extracellular fatty acids we employed delipidated serum or RNAi-mediated suppression of ACSL3 (to inhibit the activation and cellular retention of extracellular fatty acids) followed by cell proliferation assays, qPCR, apoptosis assays, immunoblots and fluorescence microscopy experiments. To assess autophagy in vivo, we employed the KrasG12D/+;p53flox/flox;Pdx1-CreERT2 (KPC) mice crossed with Acsl3 knockout mice, and to assess the efficacy of the combination therapy of ACSL3 and autophagy inhibition we used xenografted human cancer cell-derived tumors in immunocompromised mice. RESULTS Here we show that depletion of extracellularly derived lipids either by serum lipid restriction or suppression of ACSL3, triggers autophagy, a process that protects PDAC cells from the reduction of bioenergetic intermediates. Combined extracellular lipid deprivation and autophagy inhibition exhibits anti-proliferative and pro-apoptotic effects against PDAC cell lines in vitro and promotes suppression of xenografted human pancreatic cancer cell-derived tumors in mice. Therefore, we propose lipid deprivation and autophagy blockade as a potential co-targeting strategy for PDAC treatment. CONCLUSIONS Our work unravels a central role of extracellular lipid supply in ensuring fatty acid provision in cancer cells, unmasking a previously unappreciated metabolic vulnerability of PDAC cells.
Collapse
Affiliation(s)
- Maria Saliakoura
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | | - Ioanna Nikdima
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | - Chiara Pozzato
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | |
Collapse
|
4
|
Wavelet-Vermuse C, Groux-Degroote S, Vicogne D, Cogez V, Venturi G, Trinchera M, Brysbaert G, Krzewinski-Recchi MA, Hadj Bachir E, Schulz C, Vincent A, Van Seuningen I, Harduin-Lepers A. Analysis of the proximal promoter of the human colon-specific B4GALNT2 (Sd a synthase) gene: B4GALNT2 is transcriptionally regulated by ETS1. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2021; 1864:194747. [PMID: 34500083 DOI: 10.1016/j.bbagrm.2021.194747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND The Sda antigen and corresponding biosynthetic enzyme B4GALNT2 are primarily expressed in normal colonic mucosa and are down-regulated to a variable degree in colon cancer tissues. Although their expression profile is well studied, little is known about the underlying regulatory mechanisms. METHODS To clarify the molecular basis of Sda expression in the human gastrointestinal tract, we investigated the transcriptional regulation of the human B4GALNT2 gene. The proximal promoter region was delineated using luciferase assays and essential trans-acting factors were identified through transient overexpression and silencing of several transcription factors. RESULTS A short cis-regulatory region restricted to the -72 to +12 area upstream of the B4GALNT2 short-type transcript variant contained the essential promoter activity that drives the expression of the human B4GALNT2 regardless of the cell type. We further showed that B4GALNT2 transcriptional activation mostly requires ETS1 and to a lesser extent SP1. CONCLUSIONS Results presented herein are expected to provide clues to better understand B4GALNT2 regulatory mechanisms.
Collapse
Affiliation(s)
- Cindy Wavelet-Vermuse
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Sophie Groux-Degroote
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Dorothée Vicogne
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Virginie Cogez
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Giulia Venturi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), General Pathology Building, University of Bologna, 40126 Bologna, Italy
| | - Marco Trinchera
- Department of Medicine and Surgery, University of Insubria, 21100, Varese, Italy
| | - Guillaume Brysbaert
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | | | - Elsa Hadj Bachir
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020 - U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Céline Schulz
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Audrey Vincent
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020 - U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020 - U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France.
| |
Collapse
|
5
|
April-Monn SL, Andreasi V, Schiavo Lena M, Sadowski MC, Kim-Fuchs C, Buri MC, Ketkar A, Maire R, Di Domenico A, Schrader J, Muffatti F, Doglioni C, Partelli S, Falconi M, Perren A, Marinoni I. EZH2 Inhibition as New Epigenetic Treatment Option for Pancreatic Neuroendocrine Neoplasms (PanNENs). Cancers (Basel) 2021; 13:cancers13195014. [PMID: 34638497 PMCID: PMC8508156 DOI: 10.3390/cancers13195014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms are epigenetically driven tumors, but therapies against underlying epigenetic drivers are currently not available in the clinical practice. We aimed to investigate EZH2 (Enhancer of Zest homolog) expression in PanNEN and the impact of EZH2 inhibition in three different PanNEN preclinical models. EZH2 expression in PanNEN patient samples (n = 172) was assessed by immunohistochemistry and correlated with clinico-pathological data. Viability of PanNEN cell lines treated with EZH2 inhibitor (GSK126) was determined in vitro. Lentiviral transduction of shRNA targeting EZH2 was performed in QGP1 cells, and cell proliferation was measured. Rip1TAG2 mice underwent GSK126 treatment for three weeks starting from week 10 of age. Primary cells isolated from PanNEN patients (n = 6) were cultivated in 3D as islet-like tumoroids and monitored for 10 consecutive days upon GSK126 treatment. Viability was measured continuously for the whole duration of the treatment. We found that high EZH2 expression correlated with higher tumor grade (p < 0.001), presence of distant metastases (p < 0.001), and shorter disease-free survival (p < 0.001) in PanNEN patients. Inhibition of EZH2 in vitro in PanNEN cell lines and in patient-derived islet-like tumoroids reduced cell viability and impaired cell proliferation, while inhibition of EZH2 in vivo in Rip1TAG2 mice reduced tumor burden. Our results show that EZH2 is highly expressed in high-grade PanNENs, and during disease progression it may contribute to aberrations in the epigenetic cellular landscape. Targeting EZH2 may represent a valuable epigenetic treatment option for patients with PanNEN.
Collapse
Affiliation(s)
- Simon Leonhard April-Monn
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Valentina Andreasi
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.M.); (S.P.); (M.F.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Marco Schiavo Lena
- Unit of Pathology, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Martin Carl Sadowski
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
| | - Corina Kim-Fuchs
- Department of Visceral Surgery and Medicine, University Hospital Bern, University of Bern, 3008 Bern, Switzerland;
| | - Michelle Claudine Buri
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
| | - Avanee Ketkar
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
| | - Renaud Maire
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
| | - Annunziata Di Domenico
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
| | - Jörg Schrader
- Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Francesca Muffatti
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.M.); (S.P.); (M.F.)
| | - Claudio Doglioni
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- Unit of Pathology, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.M.); (S.P.); (M.F.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.M.); (S.P.); (M.F.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Aurel Perren
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
- Bern Center for Precision Medicine, University & University Hospital of Bern, 3008 Bern, Switzerland
- Correspondence: (A.P.); (I.M.)
| | - Ilaria Marinoni
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (S.L.A.-M.); (V.A.); (M.C.S.); (M.C.B.); (A.K.); (R.M.); (A.D.D.)
- Correspondence: (A.P.); (I.M.)
| |
Collapse
|
6
|
Reducing FASN expression sensitizes acute myeloid leukemia cells to differentiation therapy. Cell Death Differ 2021; 28:2465-2481. [PMID: 33742137 PMCID: PMC8329134 DOI: 10.1038/s41418-021-00768-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 02/14/2021] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Fatty acid synthase (FASN) is the only human lipogenic enzyme available for de novo fatty acid synthesis and is often highly expressed in cancer cells. We found that FASN mRNA levels were significantly higher in acute myeloid leukemia (AML) patients than in healthy granulocytes or CD34+ hematopoietic progenitors. Accordingly, FASN levels decreased during all-trans retinoic acid (ATRA)-mediated granulocytic differentiation of acute promyelocytic leukemia (APL) cells, partially via autophagic degradation. Furthermore, our data suggest that inhibition of FASN expression levels using RNAi or (-)-epigallocatechin-3-gallate (EGCG) accelerated the differentiation of APL cell lines and significantly re-sensitized ATRA refractory non-APL AML cells. FASN reduction promoted translocation of transcription factor EB (TFEB) to the nucleus, paralleled by activation of CLEAR network genes and lysosomal biogenesis. Together, our data demonstrate that inhibition of FASN expression in combination with ATRA treatment facilitates granulocytic differentiation of APL cells and may extend differentiation therapy to non-APL AML cells.
Collapse
|
7
|
Li J, Li G, Qi Y, Lu Y, Wang H, Shi K, Li D, Shi J, Stovall DB, Sui G. SRSF5 regulates alternative splicing of DMTF1 pre-mRNA through modulating SF1 binding. RNA Biol 2021; 18:318-336. [PMID: 34291726 DOI: 10.1080/15476286.2021.1947644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
ABBREVIATIONS ARF: alternative reading frame, that is, p14ARF, or CDKN2A (cyclin-dependent kinase inhibitor 2A); β-gal: β-galactosidase; CLIP-seq: crosslinking and immunoprecipitation-sequencing; DMTF1: the cyclin D binding myb-like transcription factor 1; ESS/ESE: exonic splicing silencer/enhancer; Ex: exon; FBS: fetal bovine serum; Gluc: Gaussia luciferase; hnRNPs: heterogeneous nuclear ribonucleoproteins; In: intron; ISS/ISE: intronic splicing silencer/enhancer; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PSI: percent-splice-in; qPCR: quantitative real-time PCR; RIP: RNA immunoprecipitation; RNAseq: RNA sequencing; RT: reverse transcription; SF1: splicing factor 1; SR: serine/arginine-rich proteins; SRSF5: serine and arginine-rich splicing factor 5; TCGA: the cancer genome atlas; UCSC: University of California, Santa Cruz. WT: Wild type.
Collapse
Affiliation(s)
- Jialiang Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Guangyue Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Yige Qi
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Yao Lu
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Hao Wang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Ke Shi
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Dangdang Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Jinming Shi
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Daniel B Stovall
- College of Arts and Sciences, Winthrop University, Rock Hill, SC, USA
| | - Guangchao Sui
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
8
|
Tokarchuk I, Janser FA, Schläfli AM, Pinto MT, Humbert M, Niklaus NJ, Berezowska S, Langer R, Tschan MP. Increased LAMP2A levels correlate with a shorter disease-free survival of HER2 negative breast cancer patients and increased breast cancer cell viability. Biochem Biophys Res Commun 2021; 569:47-53. [PMID: 34229122 DOI: 10.1016/j.bbrc.2021.06.082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 06/23/2021] [Indexed: 02/05/2023]
Abstract
Chaperone Mediated Autophagy (CMA) is a selective autophagy pathway deregulated in many cancers. In this study, we were aiming at understanding the importance of CMA in breast cancer. To this end, we examined the expression of the CMA markers HSP8 and LAMP2A in different breast cancer cell lines and found a wide range of LAMP2A expression levels across the cell lines analyzed. Next, we applied a specific immunohistochemical staining protocol to a tissue microarray derived from a cohort of 365 breast cancer patients. Therefore, we were able to find a correlation of high LAMP2A but not HSPA8 (HSC70) with worse disease free survival in patients with HER2 negative tumors (p = 0.026) which was independent prognostic parameter from pT category, pN category and grading in a multivariate model (HR = 1.889; 95% CI = 1.039-3.421; p = 0.037). In line, low LAMP2A levels decrease proliferation of the breast cancer cell lines T47D and MCF-7 in vitro. Our data suggest that LAMP2A supports a more severe breast cancer cell phenotype.
Collapse
Affiliation(s)
- Igor Tokarchuk
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Félice A Janser
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Anna M Schläfli
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Marta Teixeira Pinto
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal
| | - Magali Humbert
- Institute of Pathology, University of Bern, Bern, Switzerland; TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action, CA15138, Spain
| | - Nicolas J Niklaus
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland; Institute of Pathology, Kepler University Hospital and Johannes Kepler University Linz, Linz, Austria.
| | - Mario P Tschan
- Institute of Pathology, University of Bern, Bern, Switzerland; TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action, CA15138, Spain.
| |
Collapse
|
9
|
Ercolano G, Gomez-Cadena A, Dumauthioz N, Vanoni G, Kreutzfeldt M, Wyss T, Michalik L, Loyon R, Ianaro A, Ho PC, Borg C, Kopf M, Merkler D, Krebs P, Romero P, Trabanelli S, Jandus C. PPARɣ drives IL-33-dependent ILC2 pro-tumoral functions. Nat Commun 2021; 12:2538. [PMID: 33953160 PMCID: PMC8100153 DOI: 10.1038/s41467-021-22764-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 03/25/2021] [Indexed: 01/27/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play a critical role in protection against helminths and in diverse inflammatory diseases by responding to soluble factors such as the alarmin IL-33, that is often overexpressed in cancer. Nonetheless, regulatory factors that dictate ILC2 functions remain poorly studied. Here, we show that peroxisome proliferator-activated receptor gamma (PPARγ) is selectively expressed in ILC2s in humans and in mice, acting as a central functional regulator. Pharmacologic inhibition or genetic deletion of PPARγ in ILC2s significantly impair IL-33-induced Type-2 cytokine production and mitochondrial fitness. Further, PPARγ blockade in ILC2s disrupts their pro-tumoral effect induced by IL-33-secreting cancer cells. Lastly, genetic ablation of PPARγ in ILC2s significantly suppresses tumor growth in vivo. Our findings highlight a crucial role for PPARγ in supporting the IL-33 dependent pro-tumorigenic role of ILC2s and suggest that PPARγ can be considered as a druggable pathway in ILC2s to inhibit their effector functions. Hence, PPARγ targeting might be exploited in cancer immunotherapy and in other ILC2-driven mediated disorders, such as asthma and allergy.
Collapse
Affiliation(s)
- Giuseppe Ercolano
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Alejandra Gomez-Cadena
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Nina Dumauthioz
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Giulia Vanoni
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Tania Wyss
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Romain Loyon
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Angela Ianaro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ping-Chih Ho
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Christophe Borg
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Pedro Romero
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland
| | - Sara Trabanelli
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. .,Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| |
Collapse
|
10
|
Schläfli AM, Tokarchuk I, Parejo S, Jutzi S, Berezowska S, Engedal N, Tschan MP. ALK inhibition activates LC3B-independent, protective autophagy in EML4-ALK positive lung cancer cells. Sci Rep 2021; 11:9011. [PMID: 33907223 PMCID: PMC8079437 DOI: 10.1038/s41598-021-87966-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 04/06/2021] [Indexed: 01/08/2023] Open
Abstract
ALK inhibitors effectively target EML4-ALK positive non-small cell lung cancer, but their effects are hampered by treatment resistance. In the present study, we asked whether ALK inhibition affects autophagy, and whether this may influence treatment response. Whereas the impact of targeted therapies on autophagic activity previously have been assessed by surrogate marker proteins such as LC3B, we here thoroughly examined effects on functional autophagic activity, i.e. on the sequestration and degradation of autophagic cargo, in addition to autophagic markers. Interestingly, the ALK inhibitor Ceritinib decreased mTOR activity and increased GFP-WIPI1 dot formation in H3122 and H2228 EML4-ALK+ lung cancer cells, suggesting autophagy activation. Moreover, an mCherry-EGFP-LC3B based assay indicated elevated LC3B carrier flux upon ALK inhibition. In accordance, autophagic cargo sequestration and long-lived protein degradation significantly increased upon ALK inhibition. Intriguingly, autophagic cargo flux was dependent on VPS34 and ULK1, but not LC3B. Co-treating H3122 cells with Ceritinib and a VPS34 inhibitor or Bafilomycin A1 resulted in reduced cell numbers. Moreover, VPS34 inhibition reduced clonogenic recovery of Ceritinib-treated cells. In summary, our results indicate that ALK inhibition triggers LC3B-independent macroautophagic flux in EML4-ALK+ cells to support cancer cell survival and clonogenic growth.
Collapse
Affiliation(s)
- Anna M Schläfli
- Institute of Pathology, University of Bern, Bern, Switzerland.
| | - Igor Tokarchuk
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sarah Parejo
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Susanne Jutzi
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Sabina Berezowska
- Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Mario P Tschan
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Li J, Shi K, Xu T, Hu J, Li T, Li G, Chen K, Li D, Inoue K, Sui G. Mechanisms regulating DMTF1β/γ expression and their functional interplay with DMTF1α. Int J Oncol 2020; 58:20-32. [PMID: 33367929 PMCID: PMC7721083 DOI: 10.3892/ijo.2020.5146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/25/2020] [Indexed: 11/24/2022] Open
Abstract
The cyclin D binding myb-like transcription factor 1 (DMTF1), a haplo-insufficient tumor suppressor gene, has 3 alternatively spliced mRNA isoforms encoding DMTF1α, β and γ proteins. Previous studies have indicated a tumor suppressive role of DMTF1α and the oncogenic activity of DMTF1β, while the function of DMTF1γ remains largely undetermined. In the present study, the mechanisms regulating DMTF1 isoform expression were investigated and the functional interplay of DMTF1β and γ with DMTF1α was characterized. It was found that specific regions of DMTF1β and γ transcripts can impair their mRNA integrity or stability, and thus reduce protein expression levels. Additionally, DMTF1β and γ proteins exhibited a reduced stability compared to DMTF1α and all 3 DMTF1 isoforms were localized in the nuclei. Two basic residues, K52 and R53, in the DMTF1 isoforms determined their nuclear localization. Importantly, both DMTF1β and γ could associate with DMTF1α and antagonize its transactivation of the ARF promoter. Consistently, the ratios of both DMTF1β/α and γ/α were significantly associated with a poor prognoses of breast cancer patients, suggesting oncogenic roles of DMTF1β and γ isoforms in breast cancer development.
Collapse
Affiliation(s)
- Jialiang Li
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Ke Shi
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Tianqi Xu
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Jingru Hu
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Tianxin Li
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Guangyue Li
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Kuida Chen
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Dangdang Li
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Kazushi Inoue
- Department of Pathology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston‑Salem, NC 27157, USA
| | - Guangchao Sui
- Key Laboratory of Saline‑Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
12
|
Zhang J, Shen Y, Xu X, Dai Y, Li J. Transcriptome Analysis of the Liver and Muscle Tissues of Black Carp (Mylopharyngodon piceus) of Different Growth Rates. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:706-716. [PMID: 32914204 DOI: 10.1007/s10126-020-09994-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/25/2020] [Indexed: 06/11/2023]
Abstract
In this study, we used RNA-seq to analyze the muscle and liver tissues of black carps (Mylopharyngodon piceus) of different growth rates from the same batch to evaluate their growth traits. We have two groups; they are the black carp group with fast-growth rate and the slow-growth rate. A total of 23,132 genes were enriched in the Gene Ontology analysis, and 285 related pathways were found in the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. The KEGG pathway analysis showed significant differences in the expression of some genes involved in growth- and development-related metabolic pathways such as the FoxO signaling pathway, p53 signaling pathway, PI3K-Akt signaling pathway, apoptosis, TGF-β signaling pathway, and insulin signaling pathway. The numbers of differentially expressed genes in muscle and liver are 1913 and 1775. Nine of the differently expressed genes involved in the different growth traits and accuracy of the transcriptome data were validated using quantitative real-time PCR. We found that the expression levels of some growth-related genes were significantly higher in the fast-growth rate black carps than in the slow-growth rate black carps. The large number of transcriptome sequences obtained in this study has enriched the black carp gene resources, and the obtained differentially expressed genes and related pathway analysis provide valuable information for understanding the growth traits of the black carp.
Collapse
Affiliation(s)
- Jiahua Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rual Affairs, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rual Affairs, Shanghai Ocean University, Shanghai, China.
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
- College of Aquaculture and Life science, Shanghai Ocean University, Shanghai, 201306, China.
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rual Affairs, Shanghai Ocean University, Shanghai, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yafan Dai
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rual Affairs, Shanghai Ocean University, Shanghai, China
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rual Affairs, Shanghai Ocean University, Shanghai, China.
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
13
|
Chaperone-Mediated Autophagy Markers LAMP2A and HSC70 Are Independent Adverse Prognostic Markers in Primary Resected Squamous Cell Carcinomas of the Lung. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8506572. [PMID: 33029283 PMCID: PMC7527932 DOI: 10.1155/2020/8506572] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/11/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
LAMP2A and HSC70 are crucial players in chaperone-mediated autophagy (CMA), a targeted, lysosome-dependent protein degradation pathway. Elevated LAMP2A levels, indicative of increased CMA activity, are observed in several malignancies, and CMA downregulation may be exploited therapeutically. We evaluated the impact of LAMP2A and HSC70 in pulmonary squamous cell carcinomas (pSQCC). Antibodies were validated by knockdown and overexpression experiments using three different cell lines. Expression levels in tissue were analyzed by immunohistochemistry in a cohort of 336 consecutive pSQCC using tissue microarrays. There was no significant correlation between the two markers among each other and no association with pathological parameters (TNM categories, grading). However, both high LAMP2A and HSC70 expression were associated with worse outcome, including overall survival (OS; p = 0.012 and p = 0.001) and disease free survival (DFS; p = 0.049 and p = 0.036). In multivariate analysis, both markers and a combination of them were independent adverse prognostic factors for OS (LAMP2Ahigh: HR = 2.059; p < 0.001; HSC70high: HR = 1.987; p < 0.001; LAMP2Ahigh/HSC70high: HR = 1.529; p < 0.001) and DFS (LAMP2Ahigh: HR = 1.709; p = 0.004; HSC70high: HR = 1.484; p = 0.027; LAMP2Ahigh/HSC70high: HR = 1.342, p < 0.001). The negative prognostic impact of high LAMP2A and HSC70 and their variable expression in pSQCC may justify the use of these proteins as potential biomarkers for future CMA-inhibiting therapies.
Collapse
|
14
|
Orfali N, Shan-Krauer D, O'Donovan TR, Mongan NP, Gudas LJ, Cahill MR, Tschan MP, McKenna SL. Inhibition of UBE2L6 attenuates ISGylation and impedes ATRA-induced differentiation of leukemic cells. Mol Oncol 2020; 14:1297-1309. [PMID: 31820845 PMCID: PMC7266268 DOI: 10.1002/1878-0261.12614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/29/2019] [Accepted: 12/06/2019] [Indexed: 01/18/2023] Open
Abstract
Ubiquitin/ISG15‐conjugating enzyme E2L6 (UBE2L6) is a critical enzyme in ISGylation, a post‐translational protein modification that conjugates the ubiquitin‐like modifier, interferon‐stimulated gene 15 (ISG15), to target substrates. Previous gene expression studies in acute promyelocytic leukemia (APL) cells showed that all‐trans‐retinoic acid (ATRA) altered the expression of many genes, including UBE2L6 (200‐fold) and other members of the ISGylation pathway. Through gene expression analyses in a cohort of 98 acute myeloid leukemia (AML) patient samples and in primary neutrophils from healthy donors, we found that UBE2L6 gene expression is reduced in primary AML cells compared with normal mature granulocytes. To assess whether UBE2L6 expression is important for leukemic cell differentiation—two cell line models were employed: the human APL cell line NB4 and its ATRA‐resistant NB4R counterpart, as well as the ATRA‐sensitive human AML HL60 cells along with their ATRA‐resistant subclone—HL60R. ATRA strongly induced UBE2L6 in NB4 APL cells and in ATRA‐sensitive HL60 AML cells, but not in the ATRA‐resistant NB4R and HL60R cells. Furthermore, short hairpin (sh)RNA‐mediated UBE2L6 depletion in NB4 cells impeded ATRA‐mediated differentiation, suggesting a functional role for UBE2L6 in leukemic cell differentiation. In addition, ATRA induced ISG15 gene expression in NB4 APL cells, leading to increased levels of both free ISG15 protein and ISG15 conjugates. UBE2L6 depletion attenuated ATRA‐induced ISG15 conjugation. Knockdown of ISG15 in NB4 APL cells inhibited ISGylation and also attenuated ATRA‐induced differentiation. In summary, we demonstrate the functional importance of UBE2L6 in ATRA‐induced neutrophil differentiation of APL cells and propose that this may be mediated by its catalytic role in ISGylation.
Collapse
Affiliation(s)
- Nina Orfali
- Cork Cancer Research Centre & Cancer Research at UCC, University College Cork, Ireland.,Department of Hematology, Cork University Hospital, Ireland.,Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Deborah Shan-Krauer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland
| | - Tracey R O'Donovan
- Cork Cancer Research Centre & Cancer Research at UCC, University College Cork, Ireland
| | - Nigel P Mongan
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.,Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Mary R Cahill
- Cork Cancer Research Centre & Cancer Research at UCC, University College Cork, Ireland.,Department of Hematology, Cork University Hospital, Ireland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland
| | - Sharon L McKenna
- Cork Cancer Research Centre & Cancer Research at UCC, University College Cork, Ireland
| |
Collapse
|
15
|
Inoue K, Fry EA. Tumor suppression by the EGR1, DMP1, ARF, p53, and PTEN Network. Cancer Invest 2018; 36:520-536. [PMID: 30396285 PMCID: PMC6500763 DOI: 10.1080/07357907.2018.1533965] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/25/2018] [Accepted: 10/05/2018] [Indexed: 01/08/2023]
Abstract
Recent studies have indicated that EGR1 is a direct regulator of tumor suppressors including TGFβ1, PTEN, and p53. The Myb-like transcription factor Dmp1 is a physiological regulator of the Arf-p53 pathway through transactivation of the Arf promoter and physical interaction of p53. The Dmp1 promoter has binding sites for Egr proteins, and Egr1 is a target for Dmp1. Crosstalks between p53 and PTEN have been reported. The Egr1-Dmp1-Arf-p53-Pten pathway displays multiple modes of interaction with each other, suggesting the existence of a functional network of tumor suppressors that maintain normal cell growth and prevent the emergence of incipient cancer cells.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences,
Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Elizabeth A. Fry
- The Department of Pathology, Wake Forest University Health Sciences,
Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
16
|
Her2-Targeted Therapy Induces Autophagy in Esophageal Adenocarcinoma Cells. Int J Mol Sci 2018; 19:ijms19103069. [PMID: 30297650 PMCID: PMC6213363 DOI: 10.3390/ijms19103069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/04/2018] [Accepted: 10/04/2018] [Indexed: 01/17/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a highly lethal cancer type with an overall poor survival rate. Twenty to thirty percent of EAC overexpress the human epidermal growth factor receptor 2 (Her2), a transmembrane receptor tyrosine kinase promoting cell growth and proliferation. Patients with Her2 overexpressing breast and gastroesophageal cancer may benefit from Her2 inhibitors. Therapy resistance, however, is well documented. Since autophagy, a lysosome-dependent catabolic process, is implicated in cancer resistance mechanisms, we tested whether autophagy modulation influences Her2 inhibitor sensitivity in EAC. Her2-positive OE19 EAC cells showed an induction in autophagic flux upon treatment with the small molecule Her2 inhibitor Lapatinib. Newly generated Lapatinib-resistant OE19 (OE19 LR) cells showed increased basal autophagic flux compared to parental OE19 (OE19 P) cells. Based on these results, we tested if combining Lapatinib with autophagy inhibitors might be beneficial. OE19 P showed significantly reduced cell viability upon double treatment, while OE19 LR were already sensitive to autophagy inhibition alone. Additionally, Her2 status and autophagy marker expression (LC3B and p62) were investigated in a treatment-naïve EAC patient cohort (n = 112) using immunohistochemistry. Here, no significant correlation between Her2 status and expression of LC3B and p62 was found. Our data show that resistance to Her2-directed therapy is associated with a higher basal autophagy level, which is not per se associated with Her2 status. Therefore, we propose that autophagy may contribute to acquired resistance to Her2-targeted therapy in EAC, and that combining Her2 and autophagy inhibition might be beneficial for EAC patients.
Collapse
|
17
|
Graule J, Uth K, Fischer E, Centeno I, Galván JA, Eichmann M, Rau TT, Langer R, Dawson H, Nitsche U, Traeger P, Berger MD, Schnüriger B, Hädrich M, Studer P, Inderbitzin D, Lugli A, Tschan MP, Zlobec I. CDX2 in colorectal cancer is an independent prognostic factor and regulated by promoter methylation and histone deacetylation in tumors of the serrated pathway. Clin Epigenetics 2018; 10:120. [PMID: 30257705 PMCID: PMC6158822 DOI: 10.1186/s13148-018-0548-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/27/2018] [Indexed: 01/01/2023] Open
Abstract
Background In colorectal cancer, CDX2 expression is lost in approximately 20% of cases and associated with poor outcome. Here, we aim to validate the clinical impact of CDX2 and investigate the role of promoter methylation and histone deacetylation in CDX2 repression and restoration. Methods CDX2 immunohistochemistry was performed on multi-punch tissue microarrays (n = 637 patients). Promoter methylation and protein expression investigated on 11 colorectal cancer cell lines identified two CDX2 low expressors (SW620, COLO205) for treatment with decitabine (DNA methyltransferase inhibitor), trichostatin A (TSA) (general HDAC inhibitor), and LMK-235 (specific HDAC4 and HDAC5 inhibitor). RNA and protein levels were assessed. HDAC5 recruitment to the CDX2 gene promoter region was tested by chromatin immunoprecipitation. Results Sixty percent of tumors showed focal CDX2 loss; 5% were negative. Reduced CDX2 was associated with lymph node metastasis (p = 0.0167), distant metastasis (p = 0.0123), and unfavorable survival (multivariate analysis: p = 0.0008; HR (95%CI) 0.922 (0.988–0.997)) as well as BRAFV600E, mismatch repair deficiency, and CpG island methylator phenotype. Decitabine treatment alone induced CDX2 RNA and protein with values from 2- to 25-fold. TSA treatment ± decitabine also led to successful restoration of RNA and/or protein. Treatment with LMK-235 alone had marked effects on RNA and protein levels, mainly in COLO205 cells that responded less to decitabine. Lastly, decitabine co-treatment was more effective than LMK-235 alone at restoring CDX2. Conclusion CDX2 loss is an adverse prognostic factor and linked to molecular features of the serrated pathway. RNA/protein expression is restored in CDX2 low-expressing CRC cell lines by demethylation and HDAC inhibition. Importantly, our data underline HDAC4 and HDAC5 as new epigenetic CDX2 regulators that warrant further investigation. Electronic supplementary material The online version of this article (10.1186/s13148-018-0548-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janina Graule
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Kristin Uth
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Elia Fischer
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Irene Centeno
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - José A Galván
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Micha Eichmann
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Tilman T Rau
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Rupert Langer
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Heather Dawson
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Ulrich Nitsche
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich, 81675, Germany
| | - Peter Traeger
- Careanesth AG, Nelkenstrasse 15, Zürich, 8006, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, University Hospital of Bern, 3010, Bern, Switzerland.,Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| | - Beat Schnüriger
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Marion Hädrich
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Peter Studer
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Daniel Inderbitzin
- University of Bern and Bürgerspital Solothurn, Schöngrünstrasse 42, 4500, Solothurn, Switzerland
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Mario P Tschan
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.
| |
Collapse
|
18
|
Niklaus NJ, Humbert M, Tschan MP. Cisplatin sensitivity in breast cancer cells is associated with particular DMTF1 splice variant expression. Biochem Biophys Res Commun 2018; 503:2800-2806. [PMID: 30100063 DOI: 10.1016/j.bbrc.2018.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/05/2018] [Indexed: 01/08/2023]
Abstract
The cyclin D binding myb-like transcription factor 1 (DMTF1) is a tumor suppressor gene that activates p14ARF transcription and thereby stabilizing the p53 tumor suppressor. The DMTF1 gene locus encodes for three different alternatively spliced isoforms, namely DMTF1α, β and γ. The oncogenic DMTF1β isoform negatively interferes with the transcriptional activity of DMTF1α. Increased DMTF1β is associated with increased cell proliferation in a variety of cancer cell types. In this study, we aimed at identifying the role of DMTF1 isoforms in response to cisplatin treatment in breast cancer cells. First, we used SKBR3 (cisplatin sensitive) and MCF7 (cisplatin resistant) breast cancer cell lines to quantify DMTF1 expression in response to cisplatin treatment. Total DMTF1 mRNA levels increased in a dose dependent manner in both cell lines upon cisplatin treatment. However, the mRNA levels of the isoforms revealed that the sensitive cell line, SKBR3, showed increased levels of both isoforms, whereas the resistant cell, MCF7, only showed increased levels of the oncogenic DMTF1β isoform. Silencing all DMTF1 isoforms led to increased cell survival upon cisplatin treatment. Furthermore, we found a significant increase in the percentage of quiescent cells in SKBR3 shDMTF1. Together, our data suggest that DMTF1 expression levels are associated with increased cisplatin resistance.
Collapse
Affiliation(s)
- Nicolas J Niklaus
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Magali Humbert
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland.
| |
Collapse
|
19
|
Adams O, Janser FA, Dislich B, Berezowska S, Humbert M, Seiler CA, Kroell D, Slotta-Huspenina J, Feith M, Ott K, Tschan MP, Langer R. A specific expression profile of LC3B and p62 is associated with nonresponse to neoadjuvant chemotherapy in esophageal adenocarcinomas. PLoS One 2018; 13:e0197610. [PMID: 29897944 PMCID: PMC5999293 DOI: 10.1371/journal.pone.0197610] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/04/2018] [Indexed: 12/11/2022] Open
Abstract
Paclitaxel is a powerful chemotherapeutic drug, used for the treatment of many cancer types, including esophageal adenocarcinomas (EAC). Autophagy is a lysosome-dependent degradation process maintaining cellular homeostasis. Defective autophagy has been implicated in cancer biology and therapy resistance. We aimed to assess the impact of autophagy on chemotherapy response in EAC, with a special focus on paclitaxel. Responsiveness of EAC cell lines, OE19, FLO-1, OE33 and SK-GT-4, to paclitaxel was assessed using Alamar Blue assays. Autophagic flux upon paclitaxel treatment in vitro was assessed by immunoblotting of LC3B-II and quantitative assessment of WIP1 mRNA. Immunohistochemistry for the autophagy markers LC3B and p62 was applied on tumor tissue from 149 EAC patients treated with neoadjuvant chemotherapy, including pre- and post-therapeutic samples (62 matched pairs). Tumor response was assessed by histology. For comparison, previously published data on 114 primary resected EAC cases were used. EAC cell lines displayed differing responsiveness to paclitaxel treatment; however this was not associated with differential autophagy regulation. High p62 cytoplasmic expression on its own (p ≤ 0.001), or in combination with low LC3B (p = 0.034), was associated with nonresponse to chemotherapy, regardless of whether or not the regiments contained paclitaxel, but there was no independent prognostic value of LC3B or p62 expression patterns for EAC after neoadjuvant treatment. p62 and related pathways, most likely other than autophagy, play a role in chemotherapeutic response in EAC in a clinical setting. Therefore p62 could be a novel therapeutic target to overcome chemoresistance in EAC.
Collapse
Affiliation(s)
- Olivia Adams
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Félice A. Janser
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Bastian Dislich
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Magali Humbert
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Christian A. Seiler
- Department of Visceral Surgery and Medicine, Inselspital University Hospital Bern and University of Bern, Bern, Switzerland
| | - Dino Kroell
- Department of Visceral Surgery and Medicine, Inselspital University Hospital Bern and University of Bern, Bern, Switzerland
| | | | - Marcus Feith
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, München, Germany
| | - Katja Ott
- Department of Surgery, RoMED Klinikum, Rosenheim, Germany
| | - Mario P. Tschan
- Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
20
|
Wiedmer T, Blank A, Pantasis S, Normand L, Bill R, Krebs P, Tschan MP, Marinoni I, Perren A. Autophagy Inhibition Improves Sunitinib Efficacy in Pancreatic Neuroendocrine Tumors via a Lysosome-dependent Mechanism. Mol Cancer Ther 2017; 16:2502-2515. [PMID: 28729403 DOI: 10.1158/1535-7163.mct-17-0136] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 06/08/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022]
Abstract
Increasing the efficacy of approved systemic treatments in metastasized pancreatic neuroendocrine tumors (PanNET) is an unmet medical need. The antiangiogenic tyrosine kinase inhibitor sunitinib is approved for PanNET treatment. In addition, sunitinib is a lysosomotropic drug and such drugs can induce lysosomal membrane permeabilization as well as autophagy. We investigated sunitinib-induced autophagy as a possible mechanism of PanNET therapy resistance. Sunitinib accumulated in lysosomes and induced autophagy in PanNET cell lines. Adding the autophagy inhibitor chloroquine reduced cell viability in cell lines and in primary cells isolated from PanNET patients. The same treatment combination reduced tumor burden in the Rip1Tag2 transgenic PanNET mouse model. The combination of sunitinib and chloroquine reduced recovery and induced apoptosis in vitro, whereas single treatments did not. Knockdown of key autophagy proteins in combination with sunitinib showed similar effect as chloroquine. Sunitinib also induced lysosomal membrane permeabilization, which further increased in the presence of chloroquine or knockdown of lysosome-associated membrane protein (LAMP2). Both combinations led to cell death. Our data indicate that chloroquine increases sunitinib efficacy in PanNET treatment via autophagy inhibition and lysosomal membrane permeabilization. We suggest that adding chloroquine to sunitinib treatment will increase efficacy of PanNET treatment and that such patients should be included in respective ongoing clinical trials. Mol Cancer Ther; 16(11); 2502-15. ©2017 AACR.
Collapse
Affiliation(s)
- Tabea Wiedmer
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Annika Blank
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Sophia Pantasis
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Lea Normand
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ruben Bill
- Department of Internal Medicine, Regional Hospital Emmental Burgdorf, Burgdorf, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mario P Tschan
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ilaria Marinoni
- Institute of Pathology, University of Bern, Bern, Switzerland.
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
21
|
Schläfli AM, Isakson P, Garattini E, Simonsen A, Tschan MP. The autophagy scaffold protein ALFY is critical for the granulocytic differentiation of AML cells. Sci Rep 2017; 7:12980. [PMID: 29021535 PMCID: PMC5636880 DOI: 10.1038/s41598-017-12734-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/13/2017] [Indexed: 01/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignancy of myeloid progenitor cells that are blocked in differentiation. Acute promyelocytic leukemia (APL) is a rare form of AML, which generally presents with a t(15;17) translocation causing expression of the fusion protein PML-RARA. Pharmacological doses of all-trans retinoic acid (ATRA) induce granulocytic differentiation of APL cells leading to cure rates of >80% if combined with conventional chemotherapy. Autophagy is a lysosomal degradation pathway for the removal of cytoplasmic content and recycling of macromolecules. ATRA induces autophagy in ATRA-sensitive AML and APL cells and autophagy inhibition attenuates ATRA-triggered differentiation. In this study, we aimed at identifying if the autophagy-linked FYVE-domain containing protein (ALFY/WDFY3) is involved in autophagic degradation of protein aggregates contributes to ATRA therapy-induced autophagy. We found that ALFY mRNA levels increase significantly during the course of ATRA-induced differentiation of APL and AML cell lines. Importantly ALFY depletion impairs ATRA-triggered granulocytic differentiation of these cells. In agreement with its function in aggrephagy, knockdown of ALFY results in reduced ATRA-induced proteolysis. Our data further suggest that PML-RARα is an autophagy substrate degraded with the help of ALFY. In summary, we present a crucial role for ALFY in retinoid triggered maturation of AML cells.
Collapse
Affiliation(s)
- Anna M Schläfli
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Pauline Isakson
- Clinical immunology & transfusion medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - E Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche 'Mario Negri', Milano, Italy
| | - Anne Simonsen
- Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland. .,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
22
|
Mager LF, Koelzer VH, Stuber R, Thoo L, Keller I, Koeck I, Langenegger M, Simillion C, Pfister SP, Faderl M, Genitsch V, Tcymbarevich I, Juillerat P, Li X, Xia Y, Karamitopoulou E, Lyck R, Zlobec I, Hapfelmeier S, Bruggmann R, McCoy KD, Macpherson AJ, Müller C, Beutler B, Krebs P. The ESRP1-GPR137 axis contributes to intestinal pathogenesis. eLife 2017; 6:28366. [PMID: 28975893 PMCID: PMC5665647 DOI: 10.7554/elife.28366] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
Aberrant alternative pre-mRNA splicing (AS) events have been associated with several disorders. However, it is unclear whether deregulated AS directly contributes to disease. Here, we reveal a critical role of the AS regulator epithelial splicing regulator protein 1 (ESRP1) for intestinal homeostasis and pathogenesis. In mice, reduced ESRP1 function leads to impaired intestinal barrier integrity, increased susceptibility to colitis and altered colorectal cancer (CRC) development. Mechanistically, these defects are produced in part by modified expression of ESRP1-specific Gpr137 isoforms differently activating the Wnt pathway. In humans, ESRP1 is downregulated in inflamed biopsies from inflammatory bowel disease patients. ESRP1 loss is an adverse prognostic factor in CRC. Furthermore, generation of ESRP1-dependent GPR137 isoforms is altered in CRC and expression of a specific GPR137 isoform predicts CRC patient survival. These findings indicate a central role of ESRP1-regulated AS for intestinal barrier integrity. Alterations in ESRP1 function or expression contribute to intestinal pathology.
Collapse
Affiliation(s)
- Lukas Franz Mager
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Regula Stuber
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Lester Thoo
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Irene Keller
- Department of BioMedical Research, University of Bern, Bern, Switzerland.,Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Ivonne Koeck
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Cedric Simillion
- Department of BioMedical Research, University of Bern, Bern, Switzerland.,Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Simona P Pfister
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Martin Faderl
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Vera Genitsch
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Irina Tcymbarevich
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pascal Juillerat
- Department of Gastroenterology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xiaohong Li
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yu Xia
- Department of Genetics, The Scripps Research Institute, La Jolla, United States
| | | | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Kathy D McCoy
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Andrew J Macpherson
- Department of BioMedical Research, University of Bern, Bern, Switzerland.,Department of Gastroenterology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Bruce Beutler
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, United States
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Bakula D, Müller AJ, Zuleger T, Takacs Z, Franz-Wachtel M, Thost AK, Brigger D, Tschan MP, Frickey T, Robenek H, Macek B, Proikas-Cezanne T. WIPI3 and WIPI4 β-propellers are scaffolds for LKB1-AMPK-TSC signalling circuits in the control of autophagy. Nat Commun 2017; 8:15637. [PMID: 28561066 PMCID: PMC5460038 DOI: 10.1038/ncomms15637] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 04/13/2017] [Indexed: 12/31/2022] Open
Abstract
Autophagy is controlled by AMPK and mTOR, both of which associate with ULK1 and control the production of phosphatidylinositol 3-phosphate (PtdIns3P), a prerequisite for autophagosome formation. Here we report that WIPI3 and WIPI4 scaffold the signal control of autophagy upstream of PtdIns3P production and have a role in the PtdIns3P effector function of WIPI1-WIPI2 at nascent autophagosomes. In response to LKB1-mediated AMPK stimulation, WIPI4-ATG2 is released from a WIPI4-ATG2/AMPK-ULK1 complex and translocates to nascent autophagosomes, controlling their size, to which WIPI3, in complex with FIP200, also contributes. Upstream, WIPI3 associates with AMPK-activated TSC complex at lysosomes, regulating mTOR. Our WIPI interactome analysis reveals the scaffold functions of WIPI proteins interconnecting autophagy signal control and autophagosome formation. Our functional kinase screen uncovers a novel regulatory link between LKB1-mediated AMPK stimulation that produces a direct signal via WIPI4, and we show that the AMPK-related kinases NUAK2 and BRSK2 regulate autophagy through WIPI4. During autophagy, AMPK and mTOR associate with ULK1 and regulate phosphatidylinositol 3-phosphate (PtdIns3P) production that mediates autophagosome formation via WIPI proteins. Here the authors show WIPI3 and WIPI4 have a scaffolding function upstream of PtdIns3P production and have a role in the PtdIns3P effector function of WIPI1-WIPI2 at nascent autophagosomes.
Collapse
Affiliation(s)
- Daniela Bakula
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany.,International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Amelie J Müller
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany.,International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Theresia Zuleger
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Zsuzsanna Takacs
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany.,International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tuebingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Ann-Katrin Thost
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Daniel Brigger
- Division of Experimental Pathology, Institute of Pathology, University of Bern, CH-3008 Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, CH-3008 Bern, Switzerland
| | - Tancred Frickey
- Department of Biology, Applied Bioinformatics, Konstanz University, D-78457 Konstanz, Germany
| | - Horst Robenek
- Institute of Experimental Musculoskeletal Medicine, University Hospital Muenster, D-48149 Muenster, Germany
| | - Boris Macek
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany.,Proteome Center Tuebingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| | - Tassula Proikas-Cezanne
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany.,International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, D-72076 Tuebingen, Germany
| |
Collapse
|
24
|
Kendig RD, Kai F, Fry EA, Inoue K. Stabilization of the p53-DNA Complex by the Nuclear Protein Dmp1α. Cancer Invest 2017; 35:301-312. [PMID: 28406729 PMCID: PMC6262109 DOI: 10.1080/07357907.2017.1303505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/14/2016] [Accepted: 03/03/2017] [Indexed: 01/19/2023]
Abstract
We recently reported the existence of a physical interaction between the Myb-like transcription factor Dmp1 (Dmtf1) and p53 in which Dmp1 antagonized polyubiquitination of p53 by Mdm2 and promoted its nuclear localization. Dmp1 significantly stabilized p53-DNA complexes on promoters that contained p53-consensus sequences, which were either supershifted or disrupted with antibodies to Dmp1. Lysates from mice injected with doxorubicin showed that Dmp1 bound to p21Cip1, Bbc3, and Thbs1 gene regulatory regions in a p53-dependent fashion. Our data suggest that acceleration of DNA-binding of p53 by Dmp1 is a critical process for Dmp1 to increase the p53 function in Arf-deficient cells.
Collapse
Affiliation(s)
- Robert D Kendig
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Fumitake Kai
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Elizabeth A Fry
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Kazushi Inoue
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| |
Collapse
|
25
|
Haimovici A, Humbert M, Federzoni EA, Shan-Krauer D, Brunner T, Frese S, Kaufmann T, Torbett BE, Tschan MP. PU.1 supports TRAIL-induced cell death by inhibiting NF-κB-mediated cell survival and inducing DR5 expression. Cell Death Differ 2017; 24:866-877. [PMID: 28362429 PMCID: PMC5423115 DOI: 10.1038/cdd.2017.40] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
The hematopoietic Ets-domain transcription factor PU.1/SPI1 orchestrates myeloid, B- and T-cell development, and also supports hematopoietic stem cell maintenance. Although PU.1 is a renowned tumor suppressor in acute myeloid leukemia (AML), a disease characterized by an accumulation of immature blast cells, comprehensive studies analyzing the role of PU.1 during cell death responses in AML treatment are missing. Modulating PU.1 expression in AML cells, we found that PU.1 supports tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis via two mechanisms: (a) by repressing NF-κB activity via a novel direct PU.1-RelA/p65 protein-protein interaction, and (b) by directly inducing TRAIL receptor DR5 expression. Thus, expression of NF-κB-regulated antiapoptotic genes was sustained in PU.1-depleted AML cells upon TRAIL treatment and DR5 levels were decreased. Last, PU.1 deficiency significantly increased AML cell resistance to anthracycline treatment. Altogether, these results reveal a new facet of PU.1's tumor suppressor function during antileukemic therapies.
Collapse
Affiliation(s)
- Aladin Haimovici
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, Bern, Switzerland
| | - Magali Humbert
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Elena A Federzoni
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Deborah Shan-Krauer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thomas Brunner
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Steffen Frese
- Department of Thoracic Surgery, ELK Berlin Chest Hospital, Berlin, Germany
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, Bern, Switzerland
| |
Collapse
|
26
|
Tian N, Li J, Shi J, Sui G. From General Aberrant Alternative Splicing in Cancers and Its Therapeutic Application to the Discovery of an Oncogenic DMTF1 Isoform. Int J Mol Sci 2017; 18:ijms18030191. [PMID: 28257090 PMCID: PMC5372486 DOI: 10.3390/ijms18030191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
Alternative pre-mRNA splicing is a crucial process that allows the generation of diversified RNA and protein products from a multi-exon gene. In tumor cells, this mechanism can facilitate cancer development and progression through both creating oncogenic isoforms and reducing the expression of normal or controllable protein species. We recently demonstrated that an alternative cyclin D-binding myb-like transcription factor 1 (DMTF1) pre-mRNA splicing isoform, DMTF1β, is increasingly expressed in breast cancer and promotes mammary tumorigenesis in a transgenic mouse model. Aberrant pre-mRNA splicing is a typical event occurring for many cancer-related functional proteins. In this review, we introduce general aberrant pre-mRNA splicing in cancers and discuss its therapeutic application using our recent discovery of the oncogenic DMTF1 isoform as an example. We also summarize new insights in designing novel targeting strategies of cancer therapies based on the understanding of deregulated pre-mRNA splicing mechanisms.
Collapse
Affiliation(s)
- Na Tian
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Jialiang Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Jinming Shi
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Guangchao Sui
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
27
|
Marinoni I, Wiederkeher A, Wiedmer T, Pantasis S, Di Domenico A, Frank R, Vassella E, Schmitt A, Perren A. Hypo-methylation mediates chromosomal instability in pancreatic NET. Endocr Relat Cancer 2017; 24:137-146. [PMID: 28115389 DOI: 10.1530/erc-16-0554] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022]
Abstract
DAXX and or ATRX loss occur in 40% of pancreatic neuroendocrine tumors (PanNETs). PanNETs negative for DAXX or ATRX show an increased risk of relapse. The tumor-associated pathways activated upon DAXX or ATRX loss and how this event may induce chromosomal instability (CIN) and alternative lengthening telomeres (ALT) are still unknown. Both DAXX and ATRX are involved in DNA methylation regulation. DNA methylation of heterochromatin and of non-coding sequences is extremely important for the maintenance of genomic stability. We analyzed the association of DAXX and/or ATRX loss and CIN with global DNA methylation in human PanNET samples and the effect of DAXX knock-down on methylation and cell proliferation. We assessed LINE1 as well as global DNA methylation in 167 PanNETs, and we found that DAXX and or ATRX-negative tumors and tumors with CIN were hypomethylated. DAXX knock-down in PanNET cell lines blocked cells in G1/G0 phase and seemed to increase CIN in QGP-1 cells. However, no direct changes in DNA methylation were observed after DAXX knock-down in vitro In conclusion, our data indicate that epigenetic changes are crucial steps in the progression of PanNETs loss and suggest that DNA methylation is the mechanism via which CIN is induced, allowing clonal expansion and selection.
Collapse
Affiliation(s)
- I Marinoni
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - A Wiederkeher
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - T Wiedmer
- Institute of PathologyUniversity of Bern, Bern, Switzerland
- GCB Graduate School BernBern, Switzerland
| | - S Pantasis
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - A Di Domenico
- Institute of PathologyUniversity of Bern, Bern, Switzerland
- GCB Graduate School BernBern, Switzerland
| | - R Frank
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - E Vassella
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - A Schmitt
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| | - A Perren
- Institute of PathologyUniversity of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Inoue K, Fry EA. Aberrant splicing of the DMP1-ARF-MDM2-p53 pathway in cancer. Int J Cancer 2016; 139:33-41. [PMID: 26802432 PMCID: PMC5047959 DOI: 10.1002/ijc.30003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/01/2016] [Indexed: 12/11/2022]
Abstract
Alternative splicing (AS) of mRNA precursors is a ubiquitous mechanism for generating numerous transcripts with different activities from one genomic locus in mammalian cells. The gene products from a single locus can thus have similar, dominant-negative or even opposing functions. Aberrant AS has been found in cancer to express proteins that promote cell growth, local invasion and metastasis. This review will focus on the aberrant splicing of tumor suppressor/oncogenes that belong to the DMP1-ARF-MDM2-p53 pathway. Our recent study shows that the DMP1 locus generates both tumor-suppressive DMP1α (p53-dependent) and oncogenic DMP1β (p53-independent) splice variants, and the DMP1β/α ratio increases with neoplastic transformation of breast epithelial cells. This process is associated with high DMP1β protein expression and shorter survival of breast cancer (BC) patients. Accumulating pieces of evidence show that ARF is frequently inactivated by aberrant splicing in human cancers, demonstrating its involvement in human malignancies. Splice variants from the MDM2 locus promote cell growth in culture and accelerate tumorigenesis in vivo. Human cancers expressing these splice variants are associated with advanced stage/metastasis, and thus have negative clinical impacts. Although they lack most of the p53-binding domain, their activities are mostly dependent on p53 since they bind to wild-type MDM2. The p53 locus produces splice isoforms that have either favorable (β/γ at the C-terminus) or negative impact (Δ40, Δ133 at the N-terminus) on patients' survival. As the oncogenic AS products from these loci are expressed only in cancer cells, they may eventually become targets for molecular therapies.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Elizabeth A. Fry
- The Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
29
|
Rothschild SI, Gautschi O, Batliner J, Gugger M, Fey MF, Tschan MP. MicroRNA-106a targets autophagy and enhances sensitivity of lung cancer cells to Src inhibitors. Lung Cancer 2016; 107:73-83. [PMID: 27372519 DOI: 10.1016/j.lungcan.2016.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/30/2016] [Accepted: 06/10/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Src tyrosine kinase inhibitors (TKIs) significantly inhibit cell migration and invasion in lung cancer cell lines with minor cytotoxic effects. In clinical trials, however, they show modest activity in combination with chemotherapeutic agents. Possible resistance mechanisms include the induction of cytoprotective autophagy upon Src inhibition. Autophagy is a cellular recycling process that allows cell survival in response to a variety of stress stimuli including responses to various treatments. MATERIAL AND METHODS We screened autophagic activity in A549, H460, and H1299 NSCLC cell lines treated with two different Src-TKIs (saracatinib, dasatinib) or shRNA targeting SRC. The autophagy response was determined by LC3B-I to -II conversion, increased ULK1 epxression and increased GFP-LC3B dot formation. Autophagy was inhibited by pharmacological (bafilomycin A, chloroquine) or genetic (ULK1 shRNA) means. Expression of miR-106a and miR-20b was analyzed by qPCR, and we used different lentivral vectors for ectopic expression of either miR-106a mimetics, anti-sense miR-106a or different miR-106a-363 cluster constructs. RESULTS In the current study we found that Src-TKIs induce autophagy in lung adenocarcinoma cell lines and that a combination of autophagy and Src tyrosine kinase inhibition results in cell death. Moreover, Src-TKI induced autophagy depends on the induction of the key autophagy kinase ULK1. This ULK1 upregulation is caused by downregulation of the ULK1-targeting microRNA-106a. An inverse correlation of miR-106a and ULK1 expression was seen in lung adenocarcinoma. Accordingly, ectopic expression of miR-106a in combination with Src-TKI treatment resulted in significant cell death as compared to control transduced cells. CONCLUSIONS Autophagy protects lung adenocarcinoma cells from Src-TKIs via a newly identified miR-106a-ULK1 signaling pathway. The combined inhibition of Src and ULK1/autophagy might represent a promising treatment option for future clinical trials. Lastly, our data might challenge the term "oncogenic" miR-106a as it can promote sensitivity to Src-TKIs thereby underlining the context-dependent function of miRNAs.
Collapse
Affiliation(s)
- Sacha I Rothschild
- Department of Medical Oncology, Inselspital, Bern University Hospital, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland; Department Internal Medicine, Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Oliver Gautschi
- Department of Medical Oncology, Inselspital, Bern University Hospital, Switzerland; Medical Oncology, Cantonal Hospital, Luzern, Switzerland
| | - Jasmin Batliner
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Mathias Gugger
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Martin F Fey
- Department of Medical Oncology, Inselspital, Bern University Hospital, Switzerland; Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
30
|
Wang WQ, Wang FH, Qin WX, Liu HY, Lu B, Chung C, Zhu J, Gu Q, Shi W, Wen C, Wu F, Zhang K, Sun XD. Joint Antiangiogenic Effect of ATN-161 and Anti-VEGF Antibody in a Rat Model of Early Wet Age-Related Macular Degeneration. Mol Pharm 2016; 13:2881-90. [PMID: 27089240 DOI: 10.1021/acs.molpharmaceut.6b00056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The wet form of age-related macular degeneration (AMD) is a leading cause of blindness among elderly Americans and is characterized by abnormal vessel growth, termed choroidal neovascularization (CNV). Integrin α5β1 is a transmembrane receptor that binds matrix macromolecules and proteinases to stimulate angiogenesis. We recently demonstrated that integrin α5β1 plays a critical role in the development of choroidal neovascularization. In this study, we determined the role and underlying mechanisms of integrin α5β1 in angiogenesis in human choroidal endothelial cells and evaluated the antiangiogenic effects of delivering a combination therapy of ATN-161, an integrin α5β1 inhibitor, and an anti-VEGF monoclonal antibody to rats with laser-induced CNV. Vascular endothelial growth factor (VEGF) is a signaling protein that stimulates vasculogenesis and angiogenesis through a pathway that is distinct from the integrin α5β1 signaling pathway. Our results indicate that fibronectin binds to integrin α5β1 and synergizes VEGF-induced angiogenesis via two independent signaling pathways, FN/integrin α5β1/FAK/ERK1/2 and FN/integrin α5β1/FAK/AKT. Integrin α5 knockdown by shRNA inhibits endothelial cell migration, tube formation, and proliferation, while ATN-161 only partially decreases integrin α5 function. Treatment with ATN-161 combined with anti-VEGF antibody showed joint effects in attenuating angiogenesis. In summary, our results provide the first evidence for the mechanisms by which integrin α5β1 is involved in ocular pathological neovascularization in vivo, suggesting that dual inhibition of integrin α5β1 and VEGF may be a promising novel therapeutic strategy for CNV in wet AMD.
Collapse
Affiliation(s)
- Wen-Qiu Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Feng-Hua Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Wen-Xin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, 200032, China
| | - Hai-Yun Liu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Bing Lu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Christopher Chung
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Jie Zhu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Qing Gu
- Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| | - William Shi
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Cindy Wen
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Frances Wu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Kang Zhang
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China.,Veterans Administration Healthcare System , San Diego, California 92161, United States
| | - Xiao-Dong Sun
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| |
Collapse
|
31
|
Inoue K, Fry EA. Novel Molecular Markers for Breast Cancer. BIOMARKERS IN CANCER 2016; 8:25-42. [PMID: 26997872 PMCID: PMC4790586 DOI: 10.4137/bic.s38394] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/16/2016] [Accepted: 02/14/2016] [Indexed: 01/15/2023]
Abstract
The use of molecular biomarkers assures that breast cancer (BC) patients receive optimal treatment. Established biomarkers, such as estrogen receptor, progesterone receptor, HER2, and Ki67, have been playing significant roles in the subcategorization of BC to predict the prognosis and decide the specific therapy to each patient. Antihormonal therapy using 4-hydroxytamoxifen or aromatase inhibitors have been employed in patients whose tumor cells express hormone receptors, while monoclonal antibody to HER2 has been administered to HER2-positive BCs. Although new therapeutic agents have been developed in the past few decades, many patients still die of the disease due to relapse; thus, novel molecular markers that predict therapeutic failure and those that can be targets for specific therapy are expected. We have chosen four of such molecules by reviewing recent publications, which are cyclin E, B-Myb, Twist, and DMP1β. The oncogenicity of these molecules has been demonstrated in vivo and/or in vitro through studies using transgenic mice or siRNAs, and their expressions have been shown to be associated with shortened overall or disease-free survival of BC patients. The former three molecules have been shown to accelerate epithelial-mesenchymal transition that is often associated with cancer stem cell-ness and metastasis; all these four can be novel therapeutic targets as well. Thus, large prospective studies employing immunohistochemistry will be needed to establish the predictive values of these molecules in patients with BC.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
32
|
Wampfler J, Federzoni EA, Torbett BE, Fey MF, Tschan MP. The RNA binding proteins RBM38 and DND1 are repressed in AML and have a novel function in APL differentiation. Leuk Res 2015; 41:96-102. [PMID: 26740055 DOI: 10.1016/j.leukres.2015.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/15/2015] [Indexed: 12/16/2022]
Abstract
The RNA binding proteins RBM binding motif protein 38 (RBM38) and DEAD END 1 (DND1) selectively stabilize mRNAs by attenuating RNAse activity or protecting them from micro(mi)RNA-mediated cleavage. Furthermore, both proteins can efficiently stabilize the mRNA of the cell cycle inhibitor p21(CIP1). Since acute myeloid leukemia (AML) differentiation requires cell cycle arrest and RBM38 as well as DND1 have antiproliferative functions, we hypothesized that decreased RBM38 and DND1 expression may contribute to the differentiation block seen in this disease. We first quantified RBM38 and DND1 mRNA expression in clinical AML patient samples and CD34(+) progenitor cells and mature granulocytes from healthy donors. We found significantly lower RBM38 and DND1 mRNA levels in AML blasts and CD34(+) progenitor cells as compared to mature neutrophils from healthy donors. Furthermore, the lowest expression of both RBM38 and DND1 mRNA correlated with t(8;21). In addition, neutrophil differentiation of CD34(+) cells in vitro with G-CSF (granulocyte colony stimulating factor) resulted in a significant increase of RBM38 and DND1 mRNA levels. Similarly, neutrophil differentiation of NB4 acute promyelocytic leukemia (APL) cells was associated with a significant induction of RBM38 and DND1 expression. To address the function of RBM38 and DND1 in neutrophil differentiation, we generated two independent NB4RBM38 as well as DND1 knockdown cell lines. Inhibition of both RBM38 and DND1 mRNA significantly attenuated NB4 differentiation and resulted in decreased p21(CIP1) mRNA expression. Our results clearly indicate that expression of the RNA binding proteins RBM38 and DND1 is repressed in primary AML patients, that neutrophil differentiation is dependent on increased expression of both proteins, and that these proteins have a critical role in regulating p21(CIP1) expression during APL differentiation.
Collapse
Affiliation(s)
- Julian Wampfler
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.
| | - Elena A Federzoni
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, United States.
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, United States.
| | - Martin F Fey
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland.
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
33
|
Abstract
Cyclin D1 binds and activates cyclin-dependent kinases 4/6 (Cdk4/6) to phosphorylate the retinoblastoma (RB) family proteins, relieving E2F/DPs from the negative restraint of RB proteins and histone deacetylases. The cyclin D-Cdk4/6 complexes activate cyclin E/Cdk2 through titration of the Cdk inhibitors p21Cip1/p27Kip1. Cyclin E/Cdk2 further phosphorylates RBs, thereby activating E2F/DPs, and cells enter the S phase of the cell cycle. Cyclin D-Cdk4/6 also phosphorylates MEP50 subunit of the protein arginine methyltransferase 5 (PRMT5), which cooperates with cyclin D1 to drive lymphomagenesis in vivo. Activated PRMPT5 causes arginine methylation of p53 to suppress expression of pro-apoptotic and anti-proliferative target genes, explaining the molecular mechanism for tumorigenesis. Cyclin D1 physically interacts with transcription factors such as estrogen receptor, androgen receptor, and Myb family proteins to regulate gene expression in Cdk-independent fashion. Dmp1 is a Myb-like protein that quenches the oncogenic signals from activated Ras or HER2 by inducing Arf/p53-dependent cell cycle arrest. Cyclin D1 binds to Dmp1α to activate both Arf and Ink4a promoters to induce cell cycle arrest or apoptosis in non-transformed cells to prevent them from neoplastic transformation. Dmp1-deficiency significantly accelerates mouse mammary tumorigenesis with reduced apoptosis and increased metastasis. Cyclin D1 interferes with ligand activation of PPARγ involved in cellular differentiation; it also physically interacts with histone deacetylases (HDACs) and p300 to repress gene expression. It has also been shown that cyclin D1 accelerates tumorigenesis through transcriptional activation of miR-17/20 and Dicer1 which, in turn, represses cyclin D1 expression. Identification of cyclin D1-binding proteins/promoters will be essential for further clarification of its biological activities.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Elizabeth A Fry
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
34
|
Tschan MP, Federzoni EA, Haimovici A, Britschgi C, Moser BA, Jin J, Reddy VA, Sheeter DA, Fischer KM, Sun P, Torbett BE. Human DMTF1β antagonizes DMTF1α regulation of the p14(ARF) tumor suppressor and promotes cellular proliferation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1198-208. [PMID: 26187004 DOI: 10.1016/j.bbagrm.2015.07.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022]
Abstract
The human DMTF1 (DMP1) transcription factor, a DNA binding protein that interacts with cyclin D, is a positive regulator of the p14ARF (ARF) tumor suppressor. Our earlier studies have shown that three differentially spliced human DMP1 mRNAs, α, β and γ, arise from the human gene. We now show that DMP1α, β and γ isoforms differentially regulate ARF expression and promote distinct cellular functions. In contrast to DMP1α, DMP1β and γ did not activate the ARF promoter, whereas only β resulted in a dose-dependent inhibition of DMP1α-induced transactivation of the ARF promoter. Ectopic expression of DMP1β reduced endogenous ARF mRNA levels in human fibroblasts. The DMP1β- and γ-isoforms share domains necessary for the inhibitory function of the β-isoform. That DMP1β may interact with DMP1α to antagonize its function was shown in DNA binding assays and in cells by the close proximity of DMP1α/β in the nucleus. Cells stably expressing DMP1β, as well as shRNA targeting all DMP1 isoforms, disrupted cellular growth arrest induced by serum deprivation or in PMA-derived macrophages in the presence or absence of cellular p53. DMP1 mRNA levels in acute myeloid leukemia samples, as compared to granulocytes, were reduced. Treatment of acute promyelocytic leukemia patient samples with all-trans retinoic acid promoted differentiation to granulocytes and restored DMP1 transcripts to normal granulocyte levels. Our findings imply that DMP1α- and β-ratios are tightly regulated in hematopoietic cells and DMP1β antagonizes DMP1α transcriptional regulation of ARF resulting in the alteration of cellular control with a gain in proliferation.
Collapse
Affiliation(s)
- Mario P Tschan
- Department of Molecular and Experimental Medicine, La Jolla, CA 92037, USA; Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern CH-3010, Switzerland
| | - Elena A Federzoni
- Department of Molecular and Experimental Medicine, La Jolla, CA 92037, USA
| | - Aladin Haimovici
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern CH-3010, Switzerland
| | | | - Bettina A Moser
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jing Jin
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern CH-3010, Switzerland
| | | | - Dennis A Sheeter
- Department of Molecular and Experimental Medicine, La Jolla, CA 92037, USA
| | | | - Peiqing Sun
- Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
35
|
Wampfler J, Federzoni EA, Torbett BE, Fey MF, Tschan MP. Low DICER1 expression is associated with attenuated neutrophil differentiation and autophagy of NB4 APL cells. J Leukoc Biol 2015; 98:357-63. [PMID: 25990244 DOI: 10.1189/jlb.1ab0514-258r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 04/21/2015] [Indexed: 12/21/2022] Open
Abstract
Successful myeloid differentiation depends on the expression of a series of miRNAs. Thus, it is hardly surprising that miRNAs are globally repressed in AML, a disease mainly characterized by a block in cellular myeloid differentiation. Studies investigating the mechanisms for low miRNA expression in AML has mostly focused on altered transcriptional regulation or deletions, whereas defective miRNA processing has received less attention. In this study, we report that the expression of the key miRNA processing enzyme DICER1 is down-regulated in primary AML patient samples and healthy CD34(+) progenitor cells as compared with granulocytes. In line with these findings, Dicer1 expression was induced significantly in AML cell lines upon neutrophil differentiation. The knocking down of DICER1 in AML cells significantly attenuated neutrophil differentiation, which was paralleled by decreased expression of miRNAs involved in this process. Moreover, we found that inhibiting DICER1 attenuated the activation of autophagy, a cellular recycling process that is needed for proper neutrophil differentiation of AML cells. Our results clearly indicate that DICER1 plays a novel role in neutrophil differentiation as well as in myeloid autophagy of AML cells.
Collapse
Affiliation(s)
- Julian Wampfler
- *Division of Experimental Pathology, Institute of Pathology, and Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA; and Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Elena A Federzoni
- *Division of Experimental Pathology, Institute of Pathology, and Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA; and Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Bruce E Torbett
- *Division of Experimental Pathology, Institute of Pathology, and Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA; and Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Martin F Fey
- *Division of Experimental Pathology, Institute of Pathology, and Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA; and Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mario P Tschan
- *Division of Experimental Pathology, Institute of Pathology, and Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA; and Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
36
|
Maglic D, Stovall DB, Cline JM, Fry EA, Mallakin A, Taneja P, Caudell DL, Willingham MC, Sui G, Inoue K. DMP1β, a splice isoform of the tumour suppressor DMP1 locus, induces proliferation and progression of breast cancer. J Pathol 2015; 236:90-102. [PMID: 25537728 DOI: 10.1002/path.4504] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 12/13/2014] [Accepted: 12/21/2014] [Indexed: 01/20/2023]
Abstract
Our recent work has indicated that the DMP1 locus on 7q21, encoding a haplo-insufficient tumour suppressor, is hemizygously deleted at a high frequency in breast cancer. The locus encodes DMP1α protein, an activator of the p53 pathway leading to cell cycle arrest and senescence, and two other functionally undefined isoforms, DMP1β and DMP1γ. In this study, we show that the DMP1 locus is alternatively spliced in ∼30% of breast cancer cases with relatively decreased DMP1α and increased DMP1β expression. RNA-seq analyses of a publicly available database showed significantly increased DMP1β mRNA in 43-55% of human breast cancers, dependent on histological subtypes. Similarly, DMP1β protein was found to be overexpressed in ∼60% of tumours relative to their surrounding normal tissue. Importantly, alteration of DMP1 splicing and DMP1β overexpression were associated with poor clinical outcomes of the breast cancer patients, indicating that DMP1β may have a biological function. Indeed, DMP1β increased proliferation of non-tumourigenic mammary epithelial cells and knockdown of endogenous DMP1 inhibited breast cancer cell growth. To determine DMP1β's role in vivo, we established MMTV-DMP1β transgenic mouse lines. DMP1β overexpression was sufficient to induce mammary gland hyperplasia and multifocal tumour lesions in mice at 7-18 months of age. The tumours formed were adenosquamous carcinomas with evidence of transdifferentiation and keratinized deposits. Overall, we identify alternative splicing as a mechanism utilized by cancer cells to modulate the DMP1 locus through diminishing DMP1α tumour suppressor expression, while simultaneously up-regulating the tumour-promoting DMP1β isoform.
Collapse
Affiliation(s)
- Dejan Maglic
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Pathology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Graduate Program in Molecular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Brigger D, Proikas-Cezanne T, Tschan MP. WIPI-dependent autophagy during neutrophil differentiation of NB4 acute promyelocytic leukemia cells. Cell Death Dis 2014; 5:e1315. [PMID: 24991767 PMCID: PMC4123064 DOI: 10.1038/cddis.2014.261] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/13/2014] [Accepted: 05/14/2014] [Indexed: 12/29/2022]
Abstract
Members of the WD-repeat protein interacting with phosphoinositides (WIPI) family are phosphatidylinositol 3-phosphate (PI3P) effectors that are essential for the formation of autophagosomes. Autophagosomes, unique double-membraned organelles, are characteristic for autophagy, a bulk degradation mechanism with cytoprotective and homeostatic function. Both, WIPI-1 and WIPI-2 are aberrantly expressed in several solid tumors, linking these genes to carcinogenesis. We now found that the expression of WIPI-1 was significantly reduced in a large cohort of 98 primary acute myeloid leukemia (AML) patient samples (complex karyotypes; t(8;21); t(15,17); inv(16)). In contrast, the expression of WIPI-2 was only reduced in acute promyelocytic leukemia (APL), a distinct subtype of AML (t(15,17)). As AML cells are blocked in their differentiation, we tested if the expression levels of WIPI-1 and WIPI-2 increase during all-trans retinoic acid (ATRA)-induced neutrophil differentiation of APL. According to the higher WIPI-1 expression in granulocytes compared with immature blast cells, WIPI-1 but not WIPI-2 expression was significantly induced during neutrophil differentiation of NB4 APL cells. Interestingly, the induction of WIPI-1 expression was dependent on the transcription factor PU.1, a master regulator of myelopoiesis, supporting our notion that WIPI-1 expression is reduced in AML patients lacking proper PU-1 activity. Further, knocking down WIPI-1 in NB4 cells markedly attenuated the autophagic flux and significantly reduced neutrophil differentiation. This result was also achieved by knocking down WIPI-2, suggesting that both WIPI-1 and WIPI-2 are functionally required and not redundant in mediating the PI3P signal at the onset of autophagy in NB4 cells. In line with these data, downregulation of PI3KC3 (hVPS34), which generates PI3P upstream of WIPIs, also inhibited neutrophil differentiation. In conclusion, we demonstrate that both WIPI-1 and WIPI-2 are required for the PI3P-dependent autophagic activity during neutrophil differentiation, and that PU.1-dependent WIPI-1 expression is significantly repressed in primary AML patient samples and that the induction of autophagic flux is associated with neutrophil differentiation of APL cells.
Collapse
Affiliation(s)
- D Brigger
- 1] Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland [2] Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - T Proikas-Cezanne
- Autophagy Laboratory, Interfaculty Institute for Cell Biology, Eberhard Karls University Tuebingen, Germany
| | - M P Tschan
- 1] Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland [2] Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland [3] Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
38
|
Haimovici A, Brigger D, Torbett BE, Fey MF, Tschan MP. Induction of the autophagy-associated gene MAP1S via PU.1 supports APL differentiation. Leuk Res 2014; 38:1041-7. [PMID: 25043887 DOI: 10.1016/j.leukres.2014.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
Abstract
The PU.1 transcription factor is essential for myeloid development. We investigated if the microtubule-associated protein 1S (MAP1S) is a novel PU.1 target with a link to autophagy, a cellular recycling pathway. Comparable to PU.1, MAP1S expression was significantly repressed in primary AML blasts as compared to mature neutrophils. Accordingly, MAP1S expression was induced during neutrophil differentiation of CD34(+) progenitor and APL cells. Moreover, PU.1 bound to the MAP1S promoter and induced MAP1S expression during APL differentiation. Inhibiting MAP1S resulted in aberrant neutrophil differentiation and autophagy. Taken together, our findings implicate the PU.1-regulated MAP1S gene in neutrophil differentiation and autophagy control.
Collapse
Affiliation(s)
- Aladin Haimovici
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Daniel Brigger
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Martin F Fey
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland; Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
39
|
Stutz E, Gautschi O, Fey MF, Gugger M, Tschan MP, Rothschild SI. Crizotinib inhibits migration and expression of ID1 in MET-positive lung cancer cells: implications for MET targeting in oncology. Future Oncol 2014; 10:211-7. [DOI: 10.2217/fon.13.179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Aims: ID1 is an important component of the MET–SRC signaling pathway, which is a regulator of cell migration and invasion. We hypothesized that the ALK/MET inhibitor crizotinib inhibits migration via MET–SRC–ID1, rather than ALK. Materials & methods: We used ALK fusion-positive and -negative lung cancer cell lines; crizotinib, PHA-665752, and saracatinib, and stable transfection with shMET. We performed western blotting for p-ALK, ALK, p-MET, MET, p-SRC, SRC and ID1, and quantitative real-time PCR for ID1. Results: Crizotinib decreased p-MET, p-SRC and ID1 levels in ALK- and or MET-positive cell lines and inhibited cell migration. Knockdown of MET was comparable with the effect of crizotinib. Conclusion: The effects of crizotinib on ID1 expression and cancer cell migration were associated with the presence of activated MET, rather than ALK fusion.
Collapse
Affiliation(s)
- Emanuel Stutz
- Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
| | - Oliver Gautschi
- Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
- Medical Oncology, Cantonal Hospital, 6000 Luzern, Switzerland
| | - Martin F Fey
- Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Mathias Gugger
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland
| | - Mario P Tschan
- Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
40
|
Fry EA, Taneja P, Maglic D, Zhu S, Sui G, Inoue K. Dmp1α inhibits HER2/neu-induced mammary tumorigenesis. PLoS One 2013; 8:e77870. [PMID: 24205004 PMCID: PMC3812138 DOI: 10.1371/journal.pone.0077870] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 08/29/2013] [Indexed: 01/09/2023] Open
Abstract
Our recent study shows a pivotal role of Dmp1 in quenching hyperproliferative signals from HER2 to the Arf-p53 pathway as a safety mechanism to prevent breast carcinogenesis. To directly demonstrate the role of Dmp1 in preventing HER2/neu-driven oncogenic transformation, we established Flag-Dmp1α transgenic mice (MDTG) under the control of the mouse mammary tumor virus (MMTV) promoter. The mice were viable but exhibited poorly developed mammary glands with markedly reduced milk production; thus more than half of parous females were unable to support the lives of new born pups. The mammary glands of the MDTG mice had very low Ki-67 expression but high levels of Arf, Ink4a, p53, and p21Cip1, markers of senescence and accelerated aging. In all strains of generated MDTG;neu mice, tumor development was significantly delayed with decreased tumor weight. Tumors from MDTG;neu mice expressed Flag-Dmp1α and Ki-67 in a mutually exclusive fashion indicating that transgenic Dmp1α prevented tumor growth in vivo. Genomic DNA analyses showed that the Dmp1α transgene was partially lost in half of the MDTG;neu tumors, and Western blot analyses showed Dmp1α protein downregulation in 80% of the cases. Our data demonstrate critical roles of Dmp1 in preventing mammary tumorigenesis and raise the possibility of treating breast cancer by restoring Dmp1α expression.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Extracellular Matrix Proteins/physiology
- Female
- Gene Dosage
- Humans
- Immunoenzyme Techniques
- Mammary Neoplasms, Experimental/etiology
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
Collapse
Affiliation(s)
- Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Pankaj Taneja
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Dejan Maglic
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Sinan Zhu
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
| | - Guangchao Sui
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- * E-mail: (GS); (KI)
| | - Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- Graduate Program in Molecular Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States of America
- * E-mail: (GS); (KI)
| |
Collapse
|
41
|
Humbert M, Federzoni EA, Britschgi A, Schläfli AM, Valk PJM, Kaufmann T, Haferlach T, Behre G, Simon HU, Torbett BE, Fey MF, Tschan MP. The tumor suppressor gene DAPK2 is induced by the myeloid transcription factors PU.1 and C/EBPα during granulocytic differentiation but repressed by PML-RARα in APL. J Leukoc Biol 2013; 95:83-93. [PMID: 24038216 DOI: 10.1189/jlb.1112608] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
DAPK2 is a proapoptotic protein that is mostly expressed in the hematopoietic tissue. A detailed DAPK2 expression analysis in two large AML patient cohorts revealed particularly low DAPK2 mRNA levels in APL. DAPK2 levels were restored in APL patients undergoing ATRA therapy. PML-RARA is the predominant lesion in APL causing transcriptional repression of genes important for neutrophil differentiation. We found binding of PML-RARA and PU.1, a myeloid master regulator, to RARA and PU.1 binding sites in the DAPK2 promoter. Ectopic expression of PML-RARA in non-APL, as well as knocking down PU.1 in APL cells, resulted in a significant reduction of DAPK2 expression. Restoring DAPK2 expression in PU.1 knockdown APL cells partially rescued neutrophil differentiation, thereby identifying DAPK2 as a relevant PU.1 downstream effector. Moreover, low DAPK2 expression is also associated with C/EBPα-mutated AML patients, and we found C/EBPα-dependent regulation of DAPK2 during APL differentiation. In conclusion, we identified first inhibitory mechanisms responsible for the low DAPK2 expression in particular AML subtypes, and the regulation of DAPK2 by two myeloid transcription factors underlines its importance in neutrophil development.
Collapse
Affiliation(s)
- Magali Humbert
- 1.Division of Experimental Pathology, TP2, University of Bern, Murtenstrasse 31, P.O. Box 62, CH-3010 Bern, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Brigger D, Torbett BE, Chen J, Fey MF, Tschan MP. Inhibition of GATE-16 attenuates ATRA-induced neutrophil differentiation of APL cells and interferes with autophagosome formation. Biochem Biophys Res Commun 2013; 438:283-8. [PMID: 23891751 PMCID: PMC4225710 DOI: 10.1016/j.bbrc.2013.07.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 07/16/2013] [Indexed: 12/11/2022]
Abstract
Autophagy is an intracellular bulk degradation process involved in cell survival upon stress induction, but also with a newly identified function in myeloid differentiation. The autophagy-related (ATG)8 protein family, including the GABARAP and LC3 subfamilies, is crucial for autophagosome biogenesis. In order to evaluate the significance of the GABARAPs in the pathogenesis of acute myeloid leukemia (AML), we compared their expression in primary AML patient samples, CD34(+) progenitor cells and in granulocytes from healthy donors. GABARAPL1 and GABARAPL2/GATE-16, but not GABARAP, were significantly downregulated in particular AML subtypes compared to normal granulocytes. Moreover, the expression of GABARAPL1 and GATE-16 was significantly induced during ATRA-induced neutrophil differentiation of acute promyelocytic leukemia cells (APL). Lastly, knocking down GABARAPL2/GATE-16 in APL cells attenuated neutrophil differentiation and decreased autophagic flux. In conclusion, low GABARAPL2/GATE-16 expression is associated with an immature myeloid leukemic phenotype and these proteins are necessary for neutrophil differentiation of APL cells.
Collapse
Affiliation(s)
- Daniel Brigger
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Bruce E. Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Joy Chen
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Martin F. Fey
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mario P. Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
43
|
Pardo LM, Rizzu P, Francescatto M, Vitezic M, Leday GGR, Sanchez JS, Khamis A, Takahashi H, van de Berg WDJ, Medvedeva YA, van de Wiel MA, Daub CO, Carninci P, Heutink P. Regional differences in gene expression and promoter usage in aged human brains. Neurobiol Aging 2013; 34:1825-36. [PMID: 23428183 DOI: 10.1016/j.neurobiolaging.2013.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/29/2012] [Accepted: 01/07/2013] [Indexed: 10/27/2022]
Abstract
To characterize the promoterome of caudate and putamen regions (striatum), frontal and temporal cortices, and hippocampi from aged human brains, we used high-throughput cap analysis of gene expression to profile the transcription start sites and to quantify the differences in gene expression across the 5 brain regions. We also analyzed the extent to which methylation influenced the observed expression profiles. We sequenced more than 71 million cap analysis of gene expression tags corresponding to 70,202 promoter regions and 16,888 genes. More than 7000 transcripts were differentially expressed, mainly because of differential alternative promoter usage. Unexpectedly, 7% of differentially expressed genes were neurodevelopmental transcription factors. Functional pathway analysis on the differentially expressed genes revealed an overrepresentation of several signaling pathways (e.g., fibroblast growth factor and wnt signaling) in hippocampus and striatum. We also found that although 73% of methylation signals mapped within genes, the influence of methylation on the expression profile was small. Our study underscores alternative promoter usage as an important mechanism for determining the regional differences in gene expression at old age.
Collapse
Affiliation(s)
- Luba M Pardo
- Section Medical Genomics, Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wojtalla A, Fischer B, Kotelevets N, Mauri FA, Sobek J, Rehrauer H, Wotzkow C, Tschan MP, Seckl MJ, Zangemeister-Wittke U, Arcaro A. Targeting the phosphoinositide 3-kinase p110-α isoform impairs cell proliferation, survival, and tumor growth in small cell lung cancer. Clin Cancer Res 2012; 19:96-105. [PMID: 23172887 DOI: 10.1158/1078-0432.ccr-12-1138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The phosphoinositide 3-kinase (PI3K) pathway is fundamental for cell proliferation and survival and is frequently altered and activated in neoplasia, including carcinomas of the lung. In this study, we investigated the potential of targeting the catalytic class I(A) PI3K isoforms in small cell lung cancer (SCLC), which is the most aggressive of all lung cancer types. EXPERIMENTAL DESIGN The expression of PI3K isoforms in patient specimens was analyzed. The effects on SCLC cell survival and downstream signaling were determined following PI3K isoform inhibition by selective inhibitors or downregulation by siRNA. RESULTS Overexpression of the PI3K isoforms p110-α and p110-β and the antiapoptotic protein Bcl-2 was shown by immunohistochemistry in primary SCLC tissue samples. Targeting the PI3K p110-α with RNA interference or selective pharmacologic inhibitors resulted in strongly affected cell proliferation of SCLC cells in vitro and in vivo, whereas targeting p110-β was less effective. Inhibition of p110-α also resulted in increased apoptosis and autophagy, which was accompanied by decreased phosphorylation of Akt and components of the mTOR pathway, such as the ribosomal S6 protein, and the eukaryotic translation initiation factor 4E-binding protein 1. A DNA microarray analysis revealed that p110-α inhibition profoundly affected the balance of pro- and antiapoptotic Bcl-2 family proteins. Finally, p110-α inhibition led to impaired SCLC tumor formation and vascularization in vivo. CONCLUSION Together our data show the key involvement of the PI3K isoform p110-α in the regulation of multiple tumor-promoting processes in SCLC.
Collapse
Affiliation(s)
- Anna Wojtalla
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
INTRODUCTION MicroRNAs are small, noncoding RNAs that suppress gene expression by binding to the 3' untranslated region (UTR) and thereby repress translation or decrease messenger RNA stability. Inhibitor of differentiation 1 (ID1) is a putative stem-cell gene involved in invasion and angiogenesis. We previously showed that ID1 is regulated by Src kinases, overexpressed in human lung adenocarcinoma, and targeted by Src-dependent microRNAs. The current study focused on the association between miR-381 and ID1 in lung adenocarcinoma. METHODS An ID1 3'UTR-luciferase reporter assay was used to determine whether miR-381 directly targets ID1. Human lung cancer cell lines were stably transduced with a precursor of miR-381 to evaluate its role on ID1 expression and to investigate changes in cell migration and invasion. The Src tyrosine kinase inhibitors saracatinib and dasatinib were used to repress ID1 expression. MiR-381 expression was measured in 18 human lung adenocarcinomas and corresponding normal lung tissue by quantitative reverse-transcription polymerase chain reaction. RESULTS ID1 is a direct target of miR-381 as shown by 3'UTR luciferase reporter assays. MiR-381 expression was negatively correlated with ID1 expression in lung cancer cell lines. Ectopic expression of miR-381 reduced ID1 mRNA and protein levels, and significantly decreased cell migration and invasion. Furthermore, miR-381 was significantly downregulated in human lung adenocarcinomas, and low miR-381 expression levels correlated with poor prognosis. CONCLUSION These results suggest that downregulation of miR-381 and thus induction of its target ID1 may contribute to the metastatic potential of lung adenocarcinomas. Further studies to explore potential therapeutic strategies, including Src inhibitors, are ongoing.
Collapse
|
46
|
Maglic D, Zhu S, Fry EA, Taneja P, Kai F, Kendig RD, Sugiyama T, Miller LD, Willingham MC, Inoue K. Prognostic value of the hDMP1-ARF-Hdm2-p53 pathway in breast cancer. Oncogene 2012; 32:4120-9. [PMID: 23045280 DOI: 10.1038/onc.2012.423] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 06/22/2012] [Accepted: 08/02/2012] [Indexed: 12/18/2022]
Abstract
Our recent study showed critical roles of Dmp1 as a sensor of oncogenic Ras, HER2/neu signaling and activation of the Arf-p53 pathway. To elucidate the role of human DMP1 (hDMP1) in breast cancer, one hundred and ten pairs of human breast cancer specimen were studied for the alterations of the hDMP1-ARF-Hdm2-p53 pathway with follow up of clinical outcomes. Loss of heterozygosity (LOH) of the hDMP1 locus was found in 42% of human breast carcinomas, while that of INK4a/ARF and p53 were found in 20 and 34%, respectively. Hdm2 amplification was found in 13% of the same sample, which was found independently of LOH for hDMP1. Conversely, LOH for hDMP1 was found in mutually exclusive fashion with that of INK4a/ARF and p53, and was associated with low Ki67 index and diploid karyotype. Consistently, LOH for hDMP1 was associated with luminal A category and longer relapse-free survival, while that of p53 was associated with non-luminal A and shorter survival. Thus, loss of hDMP1 could define a new disease category associated with prognosis of breast cancer patients. Human breast epithelial cells/cancer cells with wild-type p53 were sensitive to growth inhibition by activated Dmp1:ER while those that delete p14(ARF) or p53, and/or Hdm2 amplification showed partial or nearly complete resistance, indicating that p53 is a critical target for hDMP1 to exhibit its biological activity.
Collapse
Affiliation(s)
- D Maglic
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Humbert M, Mueller C, Fey MF, Tschan MP. Inhibition of damage-regulated autophagy modulator-1 (DRAM-1) impairs neutrophil differentiation of NB4 APL cells. Leuk Res 2012; 36:1552-6. [PMID: 22981223 DOI: 10.1016/j.leukres.2012.08.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 08/17/2012] [Accepted: 08/21/2012] [Indexed: 11/16/2022]
Abstract
The damage-regulator autophagy modulator 1 (DRAM-1) is a lysosomal protein that positively regulates autophagy in a p53-dependent manner. We aimed at analyzing the role of DRAM-1 in granulocytic differentiation of APL cells. We observed a significant increase of DRAM-1 expression during all-trans retinoic acid (ATRA)-induced neutrophil differentiation of NB4 APL cells but not in ATRA-resistant NB4-R2 cells. Next, knocking down DRAM-1 in NB4 APL cells was sufficient to impair neutrophil differentiation. Given that DRAM-1 is a transcriptional target of p53, we tested if DRAM-1 is regulated by the p53 relative p73. Indeed, inhibiting p73 prevented neutrophil differentiation and DRAM-1 induction of NB4 cells. In conclusion, we show for the first time that p73-regulated DRAM-1 is functionally involved in neutrophil differentiation of APL cells.
Collapse
Affiliation(s)
- Magali Humbert
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | |
Collapse
|
48
|
Federzoni EA, Valk PJM, Torbett BE, Haferlach T, Löwenberg B, Fey MF, Tschan MP. PU.1 is linking the glycolytic enzyme HK3 in neutrophil differentiation and survival of APL cells. Blood 2012; 119:4963-70. [PMID: 22498738 PMCID: PMC3367898 DOI: 10.1182/blood-2011-09-378117] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 02/08/2012] [Indexed: 11/20/2022] Open
Abstract
The transcription factor PU.1 is a master regulator of myeloid differentiation and function. On the other hand, only scarce information is available on PU.1-regulated genes involved in cell survival. We now identified the glycolytic enzyme hexokinase 3 (HK3), a gene with cytoprotective functions, as transcriptional target of PU.1. Interestingly, HK3 expression is highly associated with the myeloid lineage and was significantly decreased in acute myeloid leukemia patients compared with normal granulocytes. Moreover, HK3 expression was significantly lower in acute promyelocytic leukemia (APL) compared with non-APL patient samples. In line with the observations in primary APL patient samples, we observed significantly higher HK3 expression during neutrophil differentiation of APL cell lines. Moreover, knocking down PU.1 impaired HK3 induction during neutrophil differentiation. In vivo binding of PU.1 and PML-RARA to the HK3 promoter was found, and PML-RARA attenuated PU.1 activation of the HK3 promoter. Next, inhibiting HK3 in APL cell lines resulted in significantly reduced neutrophil differentiation and viability compared with control cells. Our findings strongly suggest that HK3 is: (1) directly activated by PU.1, (2) repressed by PML-RARA, and (3) functionally involved in neutrophil differentiation and cell viability of APL cells.
Collapse
MESH Headings
- Anthracyclines/pharmacology
- Anthracyclines/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Survival/drug effects
- Cell Survival/genetics
- Cells, Cultured
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/physiology
- Glycolysis/genetics
- Hexokinase/genetics
- Hexokinase/metabolism
- Hexokinase/physiology
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Neutrophils/drug effects
- Neutrophils/metabolism
- Neutrophils/pathology
- Neutrophils/physiology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/physiology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Trans-Activators/physiology
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
Collapse
Affiliation(s)
- Elena A Federzoni
- Experimental Oncology/Hematology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
49
|
Laemmle A, Lechleiter A, Roh V, Schwarz C, Portmann S, Furer C, Keogh A, Tschan MP, Candinas D, Vorburger SA, Stroka D. Inhibition of SIRT1 impairs the accumulation and transcriptional activity of HIF-1α protein under hypoxic conditions. PLoS One 2012; 7:e33433. [PMID: 22479397 PMCID: PMC3316573 DOI: 10.1371/journal.pone.0033433] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/09/2012] [Indexed: 12/28/2022] Open
Abstract
Sirtuins and hypoxia-inducible transcription factors (HIF) have well-established roles in regulating cellular responses to metabolic and oxidative stress. Recent reports have linked these two protein families by demonstrating that sirtuins can regulate the activity of HIF-1 and HIF-2. Here we investigated the role of SIRT1, a NAD+-dependent deacetylase, in the regulation of HIF-1 activity in hypoxic conditions. Our results show that in hepatocellular carcinoma (HCC) cell lines, hypoxia did not alter SIRT1 mRNA or protein expression, whereas it predictably led to the accumulation of HIF-1α and the up-regulation of its target genes. In hypoxic models in vitro and in in vivo models of systemic hypoxia and xenograft tumor growth, knockdown of SIRT1 protein with shRNA or inhibition of its activity with small molecule inhibitors impaired the accumulation of HIF-1α protein and the transcriptional increase of its target genes. In addition, endogenous SIRT1 and HIF-1α proteins co-immunoprecipitated and loss of SIRT1 activity led to a hyperacetylation of HIF-1α. Taken together, our data suggest that HIF-1α and SIRT1 proteins interact in HCC cells and that HIF-1α is a target of SIRT1 deacetylase activity. Moreover, SIRT1 is necessary for HIF-1α protein accumulation and activation of HIF-1 target genes under hypoxic conditions.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Blotting, Western
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Hypoxia
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Hep G2 Cells
- Humans
- Hypoxia
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Knockout
- Mice, Nude
- Naphthalenes/pharmacology
- Naphthols/pharmacology
- Protein Binding
- Pyrimidinones/pharmacology
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- Sirtuin 1/antagonists & inhibitors
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Transcriptional Activation
- Transplantation, Heterologous
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Alexander Laemmle
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Antje Lechleiter
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Vincent Roh
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Christa Schwarz
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Simone Portmann
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Cynthia Furer
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Adrian Keogh
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Mario P. Tschan
- Medical Oncology/Hematology, Department of Clinical Research, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Daniel Candinas
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Stephan A. Vorburger
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Deborah Stroka
- Clinic of Visceral Surgery and Medicine, Visceral and Transplantation Surgery, University Hospital Bern and University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Batliner J, Buehrer E, Fey MF, Tschan MP. Inhibition of the miR-143/145 cluster attenuated neutrophil differentiation of APL cells. Leuk Res 2012; 36:237-40. [DOI: 10.1016/j.leukres.2011.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 10/06/2011] [Accepted: 10/08/2011] [Indexed: 10/15/2022]
|