1
|
Chadha S, Kumar A, Srivastava SA, Behl T, Ranjan R. Inulin as a Delivery Vehicle for Targeting Colon-Specific Cancer. Curr Drug Deliv 2021; 17:651-674. [PMID: 32459607 DOI: 10.2174/1567201817666200527133719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/11/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
Natural polysaccharides, as well as biopolymers, are now days widely developed for targeting colon cancer using various drug delivery systems. Currently, healing conformations are being explored that can efficiently play a multipurpose role. Owing to the capability of extravagance colonic diseases with the least adverse effects, biopolymers for site specific colon delivery have developed an increased curiosity over the past decades. Inulin (INU) was explored for its probable application as an entrapment material concerning its degradation by enzymes in the colonic microflora and its drug release behavior in a sustained and controlled manner. INU is a polysaccharide and it consists of 2 to 1 linkage having an extensive array of beneficial uses such as a carrier for delivery of therapeutic agents as an indicative/investigative utensil or as a dietary fiber with added well-being aids. In the main, limited research, as well as information, is available on the delivery of therapeutic agents using inulin specifically for colon cancer because of its capability to subsist in the stomach's acidic medium. This exceptional steadiness and robustness properties are exploited in numerous patterns to target drugs securely for the management of colonic cancer, where they effectively act and kills colonic tumor cells easily. In this review article, recent efforts and inulin-based nano-technological approaches for colon cancer targeting are presented and discussed.
Collapse
Affiliation(s)
- Swati Chadha
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Tapan Behl
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rishu Ranjan
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
2
|
de Man SMA, Zwanenburg G, van der Wal T, Hink MA, van Amerongen R. Quantitative live-cell imaging and computational modeling shed new light on endogenous WNT/CTNNB1 signaling dynamics. eLife 2021; 10:e66440. [PMID: 34190040 PMCID: PMC8341982 DOI: 10.7554/elife.66440] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
WNT/CTNNB1 signaling regulates tissue development and homeostasis in all multicellular animals, but the underlying molecular mechanism remains incompletely understood. Specifically, quantitative insight into endogenous protein behavior is missing. Here, we combine CRISPR/Cas9-mediated genome editing and quantitative live-cell microscopy to measure the dynamics, diffusion characteristics and absolute concentrations of fluorescently tagged, endogenous CTNNB1 in human cells under both physiological and oncogenic conditions. State-of-the-art imaging reveals that a substantial fraction of CTNNB1 resides in slow-diffusing cytoplasmic complexes, irrespective of the activation status of the pathway. This cytoplasmic CTNNB1 complex undergoes a major reduction in size when WNT/CTNNB1 is (hyper)activated. Based on our biophysical measurements, we build a computational model of WNT/CTNNB1 signaling. Our integrated experimental and computational approach reveals that WNT pathway activation regulates the dynamic distribution of free and complexed CTNNB1 across different subcellular compartments through three regulatory nodes: the destruction complex, nucleocytoplasmic shuttling, and nuclear retention.
Collapse
Affiliation(s)
- Saskia MA de Man
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Gooitzen Zwanenburg
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Tanne van der Wal
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Mark A Hink
- Molecular Cytology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
- van Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
3
|
Westphal D, Garzarolli M, Sergon M, Horak P, Hutter B, Becker JC, Wiegel M, Maczey E, Blum S, Grosche-Schlee S, Rütten A, Ugurel S, Stenzinger A, Glimm H, Aust D, Baretton G, Beissert S, Fröhling S, Redler S, Surowy H, Meier F. High tumour mutational burden and EGFR/MAPK pathway activation are therapeutic targets in metastatic porocarcinoma. Br J Dermatol 2021; 185:1186-1199. [PMID: 34185311 DOI: 10.1111/bjd.20604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Eccrine porocarcinoma (EPC) is a rare skin cancer arising from the eccrine sweat glands. Due to the lack of effective therapies, metastasis is associated with a high mortality rate. OBJECTIVES To investigate the drivers of EPC progression. METHODS We carried out genomic and transcriptomic profiling of metastatic EPC (mEPC), validation of the observed alterations in an EPC patient-derived cell line, confirmation of relevant observations in a large patient cohort of 30 tumour tissues, and successful treatment of a patient with mEPC under the identified treatment regimens. RESULTS mEPC was characterized by a high tumour mutational burden (TMB) with an ultraviolet signature, widespread copy number alterations and gene expression changes that affected cancer-relevant cellular processes such as cell cycle regulation and proliferation, including a pathogenic TP53 (tumour protein 53) mutation, a copy number deletion in the CDKN2A (cyclin dependent kinase inhibitor 2A) region and a CTNND1/PAK1 [catenin delta 1/p21 (RAC1) activated kinase 1] gene fusion. The overexpression of EGFR (epidermal growth factor receptor), PAK1 and MAP2K1 (mitogen-activated protein kinase kinase 1; also known as MEK1) genes translated into strong protein expression and respective pathway activation in the tumour tissue. Furthermore, a patient-derived cell line was sensitive to EGFR and MEK inhibition, confirming the functional relevance of the pathway activation. Immunohistochemistry analyses in a large patient cohort showed the relevance of the observed changes to the pathogenesis of EPC. Our results indicate that mEPC should respond to immune or kinase inhibitor therapy. Indeed, the advanced disease of our index patient was controlled by EGFR-directed therapy and immune checkpoint inhibition for more than 2 years. CONCLUSIONS Molecular profiling demonstrated high TMB and EGFR/MAPK pathway activation to be novel therapeutic targets in mEPC.
Collapse
Affiliation(s)
- D Westphal
- Department of Dermatology, University Hospital Carl Gustav Carus at Technische Universität (TU) Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - M Garzarolli
- Department of Dermatology, University Hospital Carl Gustav Carus at Technische Universität (TU) Dresden, Dresden, Germany
| | - M Sergon
- Institute of Pathology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - P Horak
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany
| | - B Hutter
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Computational Oncology, Molecular Diagnostics Program, NCT Heidelberg and DKFZ, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany
| | - J C Becker
- Department of Dermatology, University Hospital Essen, Essen, Germany.,Translational Skin Cancer Research, DKTK, Partner Site Essen, Essen, Germany
| | - M Wiegel
- Department of Dermatology, University Hospital Carl Gustav Carus at Technische Universität (TU) Dresden, Dresden, Germany
| | - E Maczey
- Department of Dermatology, University Medical Center Tübingen, Tübingen, Germany
| | - S Blum
- Institute and Policlinic of Diagnostic and Interventional Radiology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - S Grosche-Schlee
- Clinic and Policlinic of Nuclear Medicine, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - A Rütten
- Dermatopathology Friedrichshafen, Friedrichshafen, Germany
| | - S Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - A Stenzinger
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - H Glimm
- Translational Functional Cancer Genomics, NCT Heidelberg and DKFZ, Heidelberg, Germany.,Department of Translational Medical Oncology NCT Dresden and DKFZ, Dresden, Germany.,Center for Personalized Oncology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany.,DKTK, Dresden, Germany
| | - D Aust
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Institute of Pathology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany.,Tumor and Normal Tissue Bank of the UCC/NCT Site Dresden, NCT Dresden and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - G Baretton
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Institute of Pathology, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany.,Tumor and Normal Tissue Bank of the UCC/NCT Site Dresden, NCT Dresden and University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| | - S Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus at Technische Universität (TU) Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - S Fröhling
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany
| | - S Redler
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - H Surowy
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - F Meier
- Department of Dermatology, University Hospital Carl Gustav Carus at Technische Universität (TU) Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Skin Cancer Center at the University Cancer Center Dresden, University Hospital Carl Gustav Carus at TU Dresden, Dresden, Germany
| |
Collapse
|
4
|
Yamulla RJ, Nalubola S, Flesken-Nikitin A, Nikitin AY, Schimenti JC. Most Commonly Mutated Genes in High-Grade Serous Ovarian Carcinoma Are Nonessential for Ovarian Surface Epithelial Stem Cell Transformation. Cell Rep 2021; 32:108086. [PMID: 32877668 DOI: 10.1016/j.celrep.2020.108086] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/07/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the fifth leading cause of cancer-related deaths of women in the United States. Disease-associated mutations have been identified by the Cancer Genome Atlas Research Network. However, aside from mutations in TP53 or the RB1 pathway that are common in HGSOC, the contributions of mutation combinations are unclear. Here, we report CRISPR mutagenesis of 20 putative HGSOC driver genes to identify combinatorial disruptions of genes that transform either ovarian surface epithelium stem cells (OSE-SCs) or non-stem cells (OSE-NSs). Our results support the OSE-SC theory of HGSOC initiation and suggest that most commonly mutated genes in HGSOC have no effect on OSE-SC transformation initiation. Our results indicate that disruption of TP53 and PTEN, combined with RB1 disruption, constitutes a core set of mutations driving efficient transformation in vitro. The combined data may contribute to more accurate modeling of HGSOC development.
Collapse
Affiliation(s)
- Robert Joseph Yamulla
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Shreya Nalubola
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA; New York Medical College, Valhalla, NY 10595, USA
| | - Andrea Flesken-Nikitin
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - John C Schimenti
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA.
| |
Collapse
|
5
|
de Man SMA, van Amerongen R. Zooming in on the WNT/CTNNB1 Destruction Complex: Functional Mechanistic Details with Implications for Therapeutic Targeting. Handb Exp Pharmacol 2021; 269:137-173. [PMID: 34486095 DOI: 10.1007/164_2021_522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
WNT/CTNNB1 signaling is crucial for balancing cell proliferation and differentiation in all multicellular animals. CTNNB1 accumulation is the hallmark of WNT/CTNNB1 pathway activation and the key downstream event in both a physiological and an oncogenic context. In the absence of WNT stimulation, the cytoplasmic and nuclear levels of CTNNB1 are kept low because of its sequestration and phosphorylation by the so-called destruction complex, which targets CTNNB1 for proteasomal degradation. In the presence of WNT proteins, or as a result of oncogenic mutations, this process is impaired and CTNNB1 levels become elevated.Here we discuss recent advances in our understanding of destruction complex activity and inactivation, focusing on the individual components and interactions that ultimately control CTNNB1 turnover (in the "WNT off" situation) and stabilization (in the "WNT on" situation). We especially highlight the insights gleaned from recent quantitative, image-based studies, which paint an unprecedentedly detailed picture of the dynamic events that control destruction protein complex composition and function. We argue that these mechanistic details may reveal new opportunities for therapeutic intervention and could result in the destruction complex re-emerging as a target for therapy in cancer.
Collapse
Affiliation(s)
- Saskia Madelon Ada de Man
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Aghabozorgi AS, Ebrahimi R, Bahiraee A, Tehrani SS, Nabizadeh F, Setayesh L, Jafarzadeh-Esfehani R, Ferns GA, Avan A, Rashidi Z. The genetic factors associated with Wnt signaling pathway in colorectal cancer. Life Sci 2020; 256:118006. [PMID: 32593708 DOI: 10.1016/j.lfs.2020.118006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is a common cancer with poor prognosis and high mortality. There is growing information about the factors involved in the pathogenesis of CRC. However, the knowledge of the predisposing factors is limited. The development of CRC is strongly associated with the Wingless/Integrated (Wnt) signaling pathway. This pathway comprises several major target proteins, including LRP5/6, GSK3β, adenomatous polyposis coli (APC), axis inhibition protein (Axin), and β-catenin. Genetic variations in these components of the Wnt signaling pathway may lead to the activation of β-catenin, potentially increasing the proliferation of colorectal cells. Because of the potentially important role of the Wnt signaling pathway in CRC, we aimed to review the involvement of different mutations in the main downstream proteins of this pathway, including LRP5/6, APC, GSK3β, Axin, and β-catenin. Determination of the genetic risk factors involved in the progression of CRC may lead to novel approaches for the early diagnosis of CRC and the identification of potential therapeutic targets in the treatment of CRC.
Collapse
Affiliation(s)
- Amirsaeed Sabeti Aghabozorgi
- Medical Genetics Research Center, Basic Medical Sciences Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabizadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Zahra Rashidi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Vodicka P, Krskova L, Odintsov I, Krizova L, Sedlackova E, Schutzner J, Zamecnik J. Expression of molecules of the Wnt pathway and of E-cadherin in the etiopathogenesis of human thymomas. Oncol Lett 2020; 19:2413-2421. [PMID: 32194741 PMCID: PMC7039126 DOI: 10.3892/ol.2020.11343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/10/2019] [Indexed: 01/22/2023] Open
Abstract
The molecular pathogenesis of thymoma remains largely unknown. It has been recently demonstrated, that activation of Wnt signaling pathway leads to increased incidence of thymoma in murine models. The present study investigated the activation of molecules of the Wnt signaling pathway in human thymoma. A total of 112 thymoma cases with complete clinical and follow-up data and 8 controls were included in the present study. Patients with thymoma and controls were examined immunohistochemically for β-catenin and E-cadherin. The mRNA expression levels of CTNNB1, CCND1, MYC, AXIN2 and CDH1 were analyzed by reverse transcription-quantitative PCR. Immunohistochemically, β-catenin and E-cadherin were overexpressed in neoplastic cells of all thymomas. In type A, B1 and non-invasive type B2 thymoma, both molecules were located in the cytoplasm, in contrast to invasive type B2 and B3 thymoma, where membranous immunopositivities were observed. mRNA expression levels of genes involved in the Wnt pathway and of E-cadherin were significantly increased in both type A and B thymoma compared with controls; increasing gradually from type B1 to B3, and with higher stage of disease. In recurrent type B thymoma, the mRNA expression of the molecules was significantly higher. Despite the activation of Wnt pathway in indolent type A thymoma, the negative feedback of the pathway was preserved by overexpression of inhibitory molecule axin2, which was not overexpressed in type B thymoma. In summary, the Wnt pathway was activated in human thymoma and may contribute to oncogenesis. Detection of molecules of the Wnt pathway may be of diagnostic and prognostic value.
Collapse
Affiliation(s)
- Prokop Vodicka
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic.,First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague 128 08, Czech Republic
| | - Lenka Krskova
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Igor Odintsov
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Ludmila Krizova
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital, Prague 128 08, Czech Republic
| | - Eva Sedlackova
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital, Prague 128 08, Czech Republic
| | - Jan Schutzner
- Third Department of Surgery, First Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| | - Josef Zamecnik
- Department of Pathology and Molecular Medicine, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czech Republic
| |
Collapse
|
8
|
Hsa_circ_0009361 acts as the sponge of miR-582 to suppress colorectal cancer progression by regulating APC2 expression. Clin Sci (Lond) 2019; 133:1197-1213. [PMID: 31109967 DOI: 10.1042/cs20190286] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/27/2022]
Abstract
Circular RNA (circRNA) plays an important role in the development of human malignant tumors. Recently, an increasing number of circRNAs have been identified and investigated in various tumors. However, the expression pattern and biological function of circRNAs in colorectal cancer (CRC) still remain largely unexplored. In the present study, hsa_circ_0009361 was significantly down-regulated in CRC tissues and cells. Low expression level of hsa_circ_0009361 promoted the proliferation, epithelial-mesenchymal transition (EMT), migration, and invasion of CRC cells. Hsa_circ_0009361 was identified as the sponge of miR-582 by fluorescence in situ hybridization (FISH), RNA immunoprecipitation (RIP), and luciferase reporter assays. Overexpression of hsa_circ_0009361 up-regulated the expression of adenomatous polyposis coli 2 (APC2) and inhibited the activity of the Wnt/β-catenin pathway by competitively combining with miR-582. Exogenous miR-582 and APC2 interventions could reverse the multiple biological functions mediated by hsa_circ_0009361 in CRC cells. In vivo experiments also confirmed that hsa_circ_0009361 inhibited the growth and metastasis of CRC. Hsa_circ_0009361 acted as a tumor suppressive sponge of miR-582, which could up-regulate the expression of APC2, inhibit the Wnt/β-catenin signaling, and suppress the growth and metastasis of CRC. Collectively, the hsa_circ_0009361/miR-582/APC2 network could be employed as a potential therapeutic target for CRC patients.
Collapse
|
9
|
Schaefer KN, Peifer M. Wnt/Beta-Catenin Signaling Regulation and a Role for Biomolecular Condensates. Dev Cell 2019; 48:429-444. [PMID: 30782412 PMCID: PMC6386181 DOI: 10.1016/j.devcel.2019.01.025] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/06/2018] [Accepted: 01/29/2019] [Indexed: 12/19/2022]
Abstract
Wnt/β-Catenin signaling plays key roles in tissue homeostasis and cell fate decisions in embryonic and post-embryonic development across the animal kingdom. As a result, pathway mutations are associated with developmental disorders and many human cancers. The multiprotein destruction complex keeps signaling off in the absence of Wnt ligands and needs to be downregulated for pathway activation. We discuss new insights into destruction complex activity and regulation, highlighting parallels to the control of other cell biological processes by biomolecular condensates that form by phase separation to suggest that the destruction complex acts as a biomolecular condensate in Wnt pathway regulation.
Collapse
Affiliation(s)
- Kristina N Schaefer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
10
|
Ji L, Lu B, Wang Z, Yang Z, Reece-Hoyes J, Russ C, Xu W, Cong F. Identification of ICAT as an APC Inhibitor, Revealing Wnt-Dependent Inhibition of APC-Axin Interaction. Mol Cell 2018; 72:37-47.e4. [DOI: 10.1016/j.molcel.2018.07.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 06/18/2018] [Accepted: 07/27/2018] [Indexed: 12/13/2022]
|
11
|
Mao X, Tong J, Wang Y, Zhu Z, Yin Y, Wang Y. Triptolide exhibits antitumor effects by reversing hypermethylation of WIF‑1 in lung cancer cells. Mol Med Rep 2018; 18:3041-3049. [PMID: 30015908 DOI: 10.3892/mmr.2018.9263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 03/21/2018] [Indexed: 11/05/2022] Open
Abstract
Triptolide (TP) exhibits numerous biological activities, including immunosuppressive, anti‑inflammatory and antitumor effects. The aim of the present study was to investigate the role of TP as a potent therapeutic drug for the treatment of lung cancer and to investigate the underlying therapeutic mechanisms. Western blot analyses and reverse transcription‑quantitative polymerase chain reaction (PCR) were performed to investigate the expression of genes at transcriptional and translational levels, respectively. Methylation‑specific PCR assays were conducted to investigate whether TP affects the Wnt inhibitory factor‑1 (WIF‑1) methylation status and subsequently affects apoptosis, migration or the invasion of lung cancer cells. The results of the present study revealed that the methylation status of WIF‑1 in lung cancer cell lines A549 and H460 was significantly enhanced compared with the human normal bronchial epithelial cell line HBE, whereas treatment with TP was revealed to induce the demethylation of WIF‑1. The present study aimed to investigate whether the biological activities of TP are regulated by inhibiting the Wnt signaling pathway via an increase in WIF‑1 expression levels. The results of the present study revealed that Wnt signaling was suppressed in cells following treatment with TP, which was concluded by the downregulation of Axin 2 and β‑catenin expression. Further investigation demonstrated that the silencing of WIF‑1 expression with small interfering RNA reversed the TP‑induced upregulation of WIF‑1 expression, upregulated Axin 2 and β‑catenin expression and enhanced the activation of Wnt signaling. Notably, an upregulation of cellular tumor antigen p53 expression, and downregulation of matrix metalloproteinase‑9 (MMP‑9) and phosphorylated‑nuclear factor‑κB (NF‑κB) P65 (p‑P65) levels was observed following TP treatment. These results suggest that the Wnt, p53 and NF‑κB signaling pathways mediate the potent antitumor effects of TP. Notably, the silencing of WIF‑1 did not completely recover the levels of p53, MMP‑9 and p‑P65 in cells treated with TP compared with the control cells, thus suggesting that TP exhibits further functions in addition to the targeting of WIF‑1.
Collapse
Affiliation(s)
- Xiaoliang Mao
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Jichun Tong
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yong Wang
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zheng Zhu
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yajun Yin
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yeming Wang
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
12
|
Schaefer KN, Bonello TT, Zhang S, Williams CE, Roberts DM, McKay DJ, Peifer M. Supramolecular assembly of the beta-catenin destruction complex and the effect of Wnt signaling on its localization, molecular size, and activity in vivo. PLoS Genet 2018; 14:e1007339. [PMID: 29641560 PMCID: PMC5912785 DOI: 10.1371/journal.pgen.1007339] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 04/23/2018] [Accepted: 03/28/2018] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling provides a paradigm for cell-cell signals that regulate embryonic development and stem cell homeostasis and are inappropriately activated in cancers. The tumor suppressors APC and Axin form the core of the multiprotein destruction complex, which targets the Wnt-effector beta-catenin for phosphorylation, ubiquitination and destruction. Based on earlier work, we hypothesize that the destruction complex is a supramolecular entity that self-assembles by Axin and APC polymerization, and that regulating assembly and stability of the destruction complex underlie its function. We tested this hypothesis in Drosophila embryos, a premier model of Wnt signaling. Combining biochemistry, genetic tools to manipulate Axin and APC2 levels, advanced imaging and molecule counting, we defined destruction complex assembly, stoichiometry, and localization in vivo, and its downregulation in response to Wnt signaling. Our findings challenge and revise current models of destruction complex function. Endogenous Axin and APC2 proteins and their antagonist Dishevelled accumulate at roughly similar levels, suggesting competition for binding may be critical. By expressing Axin:GFP at near endogenous levels we found that in the absence of Wnt signals, Axin and APC2 co-assemble into large cytoplasmic complexes containing tens to hundreds of Axin proteins. Wnt signals trigger recruitment of these to the membrane, while cytoplasmic Axin levels increase, suggesting altered assembly/disassembly. Glycogen synthase kinase3 regulates destruction complex recruitment to the membrane and release of Armadillo/beta-catenin from the destruction complex. Manipulating Axin or APC2 levels had no effect on destruction complex activity when Wnt signals were absent, but, surprisingly, had opposite effects on the destruction complex when Wnt signals were present. Elevating Axin made the complex more resistant to inactivation, while elevating APC2 levels enhanced inactivation. Our data suggest both absolute levels and the ratio of these two core components affect destruction complex function, supporting models in which competition among Axin partners determines destruction complex activity.
Collapse
Affiliation(s)
- Kristina N. Schaefer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Teresa T. Bonello
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Shiping Zhang
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Clara E. Williams
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - David M. Roberts
- Department of Biology, Franklin and Marshall College, Lancaster, PA, United States of America
| | - Daniel J. McKay
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- * E-mail:
| |
Collapse
|
13
|
Murray PG, Stevens A, De Leonibus C, Koledova E, Chatelain P, Clayton PE. Transcriptomics and machine learning predict diagnosis and severity of growth hormone deficiency. JCI Insight 2018; 3:93247. [PMID: 29618660 DOI: 10.1172/jci.insight.93247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/28/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The effect of gene expression data on diagnosis remains limited. Here, we show how diagnosis and classification of growth hormone deficiency (GHD) can be achieved from a single blood sample using a combination of transcriptomics and random forest analysis. METHODS Prepubertal treatment-naive children with GHD (n = 98) were enrolled from the PREDICT study, and controls (n = 26) were acquired from online data sets. Whole blood gene expression was correlated with peak growth hormone (GH) using rank regression and a random forest algorithm tested for prediction of the presence of GHD and in classification of GHD as severe (peak GH <4 μg/l) and nonsevere (peak ≥4 μg/l). Performance was assessed using area under the receiver operating characteristic curve (AUC-ROC). RESULTS Rank regression identified 347 probe sets in which gene expression correlated with peak GH concentrations (r = ± 0.28, P < 0.01). These 347 probe sets yielded an AUC-ROC of 0.95 for prediction of GHD status versus controls and an AUC-ROC of 0.93 for prediction of GHD severity. CONCLUSION This study demonstrates highly accurate diagnosis and disease classification for GHD using a combination of transcriptomics and random forest analysis. TRIAL REGISTRATION NCT00256126 and NCT00699855. FUNDING Merck and the National Institute for Health Research (CL-2012-06-005).
Collapse
Affiliation(s)
- Philip G Murray
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Adam Stevens
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Chiara De Leonibus
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Ekaterina Koledova
- Global Medical Affairs Endocrinology, Global Medical, Safety & CMO Office, Merck KGaA, Darmstadt, Germany
| | - Pierre Chatelain
- Department Pediatrie, Hôpital Mère-Enfant - Université Claude Bernard, Lyon, France
| | - Peter E Clayton
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester, United Kingdom.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
14
|
Wei LH, Lin JM, Chu JF, Chen HW, Li QY, Peng J. Scutellaria barbata D. Don inhibits colorectal cancer growth via suppression of Wnt/β-catenin signaling pathway. Chin J Integr Med 2017; 23:858-863. [PMID: 29080197 DOI: 10.1007/s11655-017-2775-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To investigate the effect of the ethanol extract of Scutellaria barbata D. Don (EESB) on colorectal cancer (CRC) growth and Wnt/β-catenin signaling pathway in vivo and in vitro. METHODS In vivo experiment, CRC xenograft mouse model was constructed with injection of HT-29 cells. Following xenograft implantation, twenty mice were randomly divided into EESB-treated group (n=10) and control group (n=10) by a random number table, and were given with intra-gastric administration of 2 g/kg EESB or saline, 5 days a week for 16 days, respectively. At the end of experiment, tumors were removed and weighed by electronic scales. The proliferation biomarker Ki-67 of tumor was evaluated by immunohistochemistry (IHC) assay. In vitro study, HT-29 cells were treated with 0, 0.5, 1.5, 2.5 mg/mL EESB for 24 h. At the end of the treatment, the viability and survival of HT-29 cells were determined by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and colony formation assay, respectively. The mRNA expression of c-Myc, Survivin and adenomatous polyposis coli (APC) was examined by reverse transcription-polymerase chain reaction (RT-PCR) both in tumor tissues of CRC xenograft mice and HT-29 cells. Protein expression of c-Myc, Survivin, APC, and β-catenin as well as β-catenin phosphorylation level were evaluated by IHC assay or Western blotting. RESULTS EESB significantly reduced tumor weight in CRC xenografts mice, compared with the control group (P<0.05). IHC assay showed that EESB significantly inhibited protein expression of Ki-67 in tumor tissues (P<0.05). MTT assay showed that EESB significantly reduced HT-29 cell viability in a dose-dependent manner (P<0.05). Colony formation assay showed that EESB dose-dependently decreased the survival of HT-29 cells (P<0.05). In addition, RT-PCR assay showed that EESB decreased the mRNA expression of c-Myc and Survivin and increased APC expression, both in tumor tissues of CRC xenograft mice and HT-29 cells (P<0.05). IHC assay or Western blotting showed that EESB decreased protein expression of β-catenin, c-Myc and Survivin, as well as increased APC expression and β-catenin phosphorylation in tumor tissues or HT-29 cells (P<0.05). CONCLUSIONS EESB significantly reduced tumor growth in CRC xenografts mice, and inhibited the viability and survival of HT-29 cells. EESB could suppress the activation of the Wnt/β-catenin pathway, which might be one of the mechanisms whereby Scutellaria barbata D. Don exerts its anticancer activity.
Collapse
Affiliation(s)
- Li-Hui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jiu-Mao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jian-Feng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Hong-Wei Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Qing-Yu Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
15
|
Bourroul GM, Fragoso HJ, Gomes JWF, Bourroul VSO, Oshima CTF, Gomes TS, Saba GT, Palma RT, Waisberg J. The destruction complex of beta-catenin in colorectal carcinoma and colonic adenoma. EINSTEIN-SAO PAULO 2017; 14:135-42. [PMID: 27462886 PMCID: PMC4943346 DOI: 10.1590/s1679-45082016ao3678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/03/2016] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE To evaluate the destruction complex of beta-catenin by the expression of the proteins beta-catetenin, adenomatous polyposis coli, GSK3β, axin and ubiquitin in colorectal carcinoma and colonic adenoma. METHODS Tissue samples from 64 patients with colorectal carcinoma and 53 patients with colonic adenoma were analyzed. Tissue microarray blocks and slides were prepared and subjected to immunohistochemistry with polyclonal antibodies in carcinoma, adjacent non-neoplastic mucosa, and adenoma tissues. The immunoreactivity was evaluated by the percentage of positive stained cells and by the intensity assessed through of the stained grade of proteins in the cytoplasm and nucleus of cells. In the statistical analysis, the Spearman correlation coefficient, Student's t, χ2, Mann-Whitney, and McNemar tests, and univariate logistic regression analysis were used. RESULTS In colorectal carcinoma, the expressions of beta-catenin and adenomatous polyposis coli proteins were significantly higher than in colonic adenomas (p<0.001 and p<0.0001, respectively). The immunoreactivity of GSK3β, axin 1 and ubiquitin proteins was significantly higher (p=0.03, p=0.039 and p=0.03, respectively) in colorectal carcinoma than in the colonic adenoma and adjacent non-neoplastic mucosa. The immunohistochemistry staining of these proteins did not show significant differences with the clinical and pathological characteristics of colorectal cancer and colonic adenoma. CONCLUSIONS These results suggest that, in adenomas, the lower expression of the beta-catenin, axin 1 and GSK3β proteins indicated that the destruction complex of beta-catenin was maintained, while in colorectal carcinoma, the increased expression of beta-catenin, GSK3β, axin 1, and ubiquitin proteins indicated that the destruction complex of beta-catenin was disrupted. OBJETIVO Avaliar o complexo de destruição da betacatenina no carcinoma colorretal e no adenoma do colo pela expressão das proteínas betacatenina, adenomatous polyposis coli, GSK3β, axina e ubiquitina. MÉTODOS Amostras de tecidos de 64 doentes com carcinoma colorretal e de 53 pacientes com adenoma do colo foram analisadas. Blocos de tecidos foram submetidos ao estudo imuno-histoquímico com anticorpos policlonais nos tecidos do carcinoma, mucosa não neoplásica adjacente e adenoma. A imunorreatividade foi avaliada pela porcentagem de positividade de células coradas e pela intensidade do grau de coloração das proteínas no citoplasma e no núcleo das células. Na análise estatística, foram utilizados o coeficiente de correlação de Spearman, os testes t de Student, χ2, Mann-Whitney e de McNemar, e a análise de regressão logística univariada. RESULTADOS No carcinoma colorretal, as expressões da betacatenina e da adenomatous polyposis coli foram significativamente maiores do que em adenomas do colo (p<0,001 e p<0,0001, respectivamente). A imunorreatividade das proteínas GSK3β, axina 1 e ubiquitina foi significativamente maior (p=0,03, p=0,039 e p=0,03, respectivamente) no carcinoma colorretal do que no adenoma e na mucosa não neoplásica adjacente. A coloração imuno-histoquímica dessas proteínas não apresentou diferenças significantes em relação às características clinicopatológicas do câncer colorretal e do adenoma. CONCLUSÕES Em adenomas, as menores expressões de betacatenina, axina 1 e GSK3β indicaram que o complexo de destruição da betacatenina estava conservado, enquanto que, no carcinoma colorretal, o aumento das expressões da betacatenina, GSK3β, 1 axina, e ubiquitina indicaram que o complexo de destruição de betacatenina estava alterado.
Collapse
Affiliation(s)
| | - Hélio José Fragoso
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| | | | | | | | - Thiago Simão Gomes
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Rogério Tadeu Palma
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| | - Jaques Waisberg
- Hospital do Servidor Público Estadual "Francisco Morato de Oliveira", São Paulo, SP, Brazil
| |
Collapse
|
16
|
Aristizabal-Pachon AF, Castillo WO. Role of GSK3β in breast cancer susceptibility. Cancer Biomark 2017; 18:169-175. [DOI: 10.3233/cbm-160120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrés Felipe Aristizabal-Pachon
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Cell-Based Therapy, National Institute of Science and Technology in Stem Cell and Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
| | - Willian Orlando Castillo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
17
|
Abstract
Background Germline mutations in the coding sequence of the tumour suppressor APC gene give rise to familial adenomatous polyposis (which leads to colorectal cancer) and are associated with many other oncopathologies. The loss of APC function because of deletion of putative promoter 1A or 1B also results in the development of colorectal cancer. Since the regions of promoters 1A and 1B contain many single nucleotide polymorphisms (SNPs), the aim of this study was to perform functional analysis of some of these SNPs by means of an electrophoretic mobility shift assay (EMSA) and a luciferase reporter assay. Results First, it was shown that both putative promoters of APC (1A and 1B) drive transcription in an in vitro reporter experiment. From eleven randomly selected SNPs of promoter 1A and four SNPs of promoter 1B, nine and two respectively showed differential patterns of binding of nuclear proteins to oligonucleotide probes corresponding to alternative alleles. The luciferase reporter assay showed that among the six SNPs tested, the rs75612255 C allele and rs113017087 C allele in promoter 1A as well as the rs138386816 T allele and rs115658307 T allele in promoter 1B significantly increased luciferase activity in the human erythromyeloblastoid leukaemia cell line K562. In human colorectal cancer HCT-116 cells, none of the substitutions under study had any effect, with the exception of minor allele G of rs79896135 in promoter 1B. This allele significantly decreased the luciferase reporter’s activity Conclusion Our results indicate that many SNPs in APC promoters 1A and 1B are functionally relevant and that allele G of rs79896135 may be associated with the predisposition to colorectal cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0460-8) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Pronobis MI, Deuitch N, Posham V, Mimori-Kiyosue Y, Peifer M. Reconstituting regulation of the canonical Wnt pathway by engineering a minimal β-catenin destruction machine. Mol Biol Cell 2016; 28:41-53. [PMID: 27852897 PMCID: PMC5221518 DOI: 10.1091/mbc.e16-07-0557] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/01/2016] [Accepted: 11/07/2016] [Indexed: 02/05/2023] Open
Abstract
APC and Axin are key negative regulators of Wnt signaling in development and oncogenesis. They form a multiprotein complex targeting the key Wnt effector β-catenin for destruction. Essential components of APC and Axin required for their cooperative function are identified, and the data are used to design a minimal β-catenin–destruction machine. Negatively regulating key signaling pathways is critical to development and altered in cancer. Wnt signaling is kept off by the destruction complex, which is assembled around the tumor suppressors APC and Axin and targets β-catenin for destruction. Axin and APC are large proteins with many domains and motifs that bind other partners. We hypothesized that if we identified the essential regions required for APC:Axin cooperative function and used these data to design a minimal β-catenin-destruction machine, we would gain new insights into the core mechanisms of destruction complex function. We identified five key domains/motifs in APC or Axin that are essential for their function in reconstituting Wnt regulation. Strikingly, however, certain APC and Axin mutants that are nonfunctional on their own can complement one another in reducing β-catenin, revealing that the APC:Axin complex is a highly robust machine. We used these insights to design a minimal β-catenin-destruction machine, revealing that a minimized chimeric protein covalently linking the five essential regions of APC and Axin reconstitutes destruction complex internal structure, size, and dynamics, restoring efficient β-catenin destruction in colorectal tumor cells. On the basis of our data, we propose a new model of the mechanistic function of the destruction complex as an integrated machine.
Collapse
Affiliation(s)
- Mira I Pronobis
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natalie Deuitch
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Vinya Posham
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yuko Mimori-Kiyosue
- Cellular Dynamics Analysis Unit, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 .,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
19
|
Lui C, Mok MTS, Henderson BR. Characterization of Adenomatous Polyposis Coli Protein Dynamics and Localization at the Centrosome. Cancers (Basel) 2016; 8:cancers8050047. [PMID: 27144584 PMCID: PMC4880864 DOI: 10.3390/cancers8050047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/20/2016] [Accepted: 04/26/2016] [Indexed: 12/11/2022] Open
Abstract
The adenomatous polyposis coli (APC) tumor suppressor is a multifunctional regulator of Wnt signaling and acts as a mobile scaffold at different cellular sites. APC was recently found to stimulate microtubule (MT) growth at the interphase centrosome; however, little is known about its dynamics and localization at this site. To address this, we analysed APC dynamics in fixed and live cells by fluorescence microscopy. In detergent-extracted cells, we discovered that APC was only weakly retained at the centrosome during interphase suggesting a rapid rate of exchange. This was confirmed in living cells by fluorescence recovery after photobleaching (FRAP), which identified two pools of green fluorescent protein (GFP)-APC: a major rapidly exchanging pool (~86%) and minor retained pool (~14%). The dynamic exchange rate of APC was unaffected by C-terminal truncations implicating a targeting role for the N-terminus. Indeed, we mapped centrosome localization to N-terminal armadillo repeat (ARM) domain amino acids 334–625. Interestingly, the rate of APC movement to the centrosome was stimulated by intact MTs, and APC dynamics slowed when MTs were disrupted by nocodazole treatment or knockdown of γ-tubulin. Thus, the rate of APC recycling at the centrosome is enhanced by MT growth, suggesting a positive feedback to stimulate its role in MT growth.
Collapse
Affiliation(s)
- Christina Lui
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.
| | - Myth T S Mok
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Beric R Henderson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.
| |
Collapse
|
20
|
Croy HE, Fuller CN, Giannotti J, Robinson P, Foley AVA, Yamulla RJ, Cosgriff S, Greaves BD, von Kleeck RA, An HH, Powers CM, Tran JK, Tocker AM, Jacob KD, Davis BK, Roberts DM. The Poly(ADP-ribose) Polymerase Enzyme Tankyrase Antagonizes Activity of the β-Catenin Destruction Complex through ADP-ribosylation of Axin and APC2. J Biol Chem 2016; 291:12747-12760. [PMID: 27068743 DOI: 10.1074/jbc.m115.705442] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 12/18/2022] Open
Abstract
Most colon cancer cases are initiated by truncating mutations in the tumor suppressor, adenomatous polyposis coli (APC). APC is a critical negative regulator of the Wnt signaling pathway that participates in a multi-protein "destruction complex" to target the key effector protein β-catenin for ubiquitin-mediated proteolysis. Prior work has established that the poly(ADP-ribose) polymerase (PARP) enzyme Tankyrase (TNKS) antagonizes destruction complex activity by promoting degradation of the scaffold protein Axin, and recent work suggests that TNKS inhibition is a promising cancer therapy. We performed a yeast two-hybrid (Y2H) screen and uncovered TNKS as a putative binding partner of Drosophila APC2, suggesting that TNKS may play multiple roles in destruction complex regulation. We find that TNKS binds a C-terminal RPQPSG motif in Drosophila APC2, and that this motif is conserved in human APC2, but not human APC1. In addition, we find that APC2 can recruit TNKS into the β-catenin destruction complex, placing the APC2/TNKS interaction at the correct intracellular location to regulate β-catenin proteolysis. We further show that TNKS directly PARylates both Drosophila Axin and APC2, but that PARylation does not globally regulate APC2 protein levels as it does for Axin. Moreover, TNKS inhibition in colon cancer cells decreases β-catenin signaling, which we find cannot be explained solely through Axin stabilization. Instead, our findings suggest that TNKS regulates destruction complex activity at the level of both Axin and APC2, providing further mechanistic insight into TNKS inhibition as a potential Wnt pathway cancer therapy.
Collapse
Affiliation(s)
- Heather E Croy
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Caitlyn N Fuller
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Jemma Giannotti
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Paige Robinson
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Andrew V A Foley
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Robert J Yamulla
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Sean Cosgriff
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Bradford D Greaves
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Ryan A von Kleeck
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Hyun Hyung An
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Catherine M Powers
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Julie K Tran
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Aaron M Tocker
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Kimberly D Jacob
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - Beckley K Davis
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604
| | - David M Roberts
- From the Department of Biology, Franklin & Marshall College, Lancaster, Pennsylvania 17604.
| |
Collapse
|
21
|
Lui C, Ashton C, Sharma M, Brocardo MG, Henderson BR. APC functions at the centrosome to stimulate microtubule growth. Int J Biochem Cell Biol 2015; 70:39-47. [PMID: 26556314 DOI: 10.1016/j.biocel.2015.10.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 11/26/2022]
Abstract
The adenomatous polyposis coli (APC) tumor suppressor is multi-functional. APC is known to localize at the centrosome, and in mitotic cells contributes to formation of the mitotic spindle. To test whether APC contributes to nascent microtubule (MT) growth at interphase centrosomes, we employed MT regrowth assays in U2OS cells to measure MT assembly before and after nocodazole treatment and release. We showed that siRNA knockdown of full-length APC delayed both initial MT aster formation and MT elongation/regrowth. In contrast, APC-mutant SW480 cancer cells displayed a defect in MT regrowth that was unaffected by APC knockdown, but which was rescued by reconstitution of full-length APC. Our findings identify APC as a positive regulator of centrosome MT initial assembly and suggest that this process is disrupted by cancer mutations. We confirmed that full-length APC associates with the MT-nucleation factor γ-tubulin, and found that the APC cancer-truncated form (1-1309) also bound to γ-tubulin through APC amino acids 1-453. While binding to γ-tubulin may help target APC to the site of MT nucleation complexes, additional C-terminal sequences of APC are required to stimulate and stabilize MT growth.
Collapse
Affiliation(s)
- Christina Lui
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Cahora Ashton
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Manisha Sharma
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Mariana G Brocardo
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Beric R Henderson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.
| |
Collapse
|
22
|
Kunttas-Tatli E, Von Kleeck RA, Greaves BD, Vinson D, Roberts DM, McCartney BM. The two SAMP repeats and their phosphorylation state in Drosophila Adenomatous polyposis coli-2 play mechanistically distinct roles in negatively regulating Wnt signaling. Mol Biol Cell 2015; 26:4503-18. [PMID: 26446838 PMCID: PMC4666143 DOI: 10.1091/mbc.e15-07-0515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/28/2015] [Indexed: 11/30/2022] Open
Abstract
The colon cancer tumor suppressor Adenomatous polyposis coli (APC) negatively regulates Wnt signaling destruction complex by binding to β-catenin and facilitating its phosphorylation and degradation. The two SAMP repeats and their phosphorylation state in Drosophila APC2 play distinct roles in negatively regulating Wnt signaling. The tumor suppressor Adenomatous polyposis coli (APC) plays a key role in regulating the canonical Wnt signaling pathway as an essential component of the β-catenin destruction complex. C-terminal truncations of APC are strongly implicated in both sporadic and familial forms of colorectal cancer. However, many questions remain as to how these mutations interfere with APC’s tumor suppressor activity. One set of motifs frequently lost in these cancer-associated truncations is the SAMP repeats that mediate interactions between APC and Axin. APC proteins in both vertebrates and Drosophila contain multiple SAMP repeats that lack high sequence conservation outside of the Axin-binding motif. In this study, we tested the functional redundancy between different SAMPs and how these domains are regulated, using Drosophila APC2 and its two SAMP repeats as our model. Consistent with sequence conservation–based predictions, we show that SAMP2 has stronger binding activity to Axin in vitro, but SAMP1 also plays an essential role in the Wnt destruction complex in vivo. In addition, we demonstrate that the phosphorylation of SAMP repeats is a potential mechanism to regulate their activity. Overall our findings support a model in which each SAMP repeat plays a mechanistically distinct role but they cooperate for maximal destruction complex function.
Collapse
Affiliation(s)
- Ezgi Kunttas-Tatli
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Ryan A Von Kleeck
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Bradford D Greaves
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - David Vinson
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - David M Roberts
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
23
|
Abstract
Colorectal cancer (CRC) is a complex disease that develops as a consequence of both genetic and environmental risk factors. A small proportion (3-5%) of cases arise from hereditary syndromes predisposing to early onset CRC as a result of mutations in over a dozen well defined genes. In contrast, CRC is predominantly a late onset 'sporadic' disease, developing in individuals with no obvious hereditary syndrome. In recent years, genome wide association studies have discovered that over 40 genetic regions are associated with weak effects on sporadic CRC, and it has been estimated that increasingly large genome wide scans will identify many additional novel genetic regions. Subsequent experimental validations have identified the causally related variant(s) in a limited number of these genetic regions. Further biological insight could be obtained through ethnically diverse study populations, larger genetic sequencing studies and development of higher throughput functional experiments. Along with inherited variation, integration of the tumour genome may shed light on the carcinogenic processes in CRC. In addition to summarising the genetic architecture of CRC, this review discusses genetic factors that modify environmental predictors of CRC, as well as examples of how genetic insight has improved clinical surveillance, prevention and treatment strategies. In summary, substantial progress has been made in uncovering the genetic architecture of CRC, and continued research efforts are expected to identify additional genetic risk factors that further our biological understanding of this disease. Subsequently these new insights will lead to improved treatment and prevention of colorectal cancer.
Collapse
Affiliation(s)
- Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Stephanie Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Niha Zubair
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
24
|
Pronobis MI, Rusan NM, Peifer M. A novel GSK3-regulated APC:Axin interaction regulates Wnt signaling by driving a catalytic cycle of efficient βcatenin destruction. eLife 2015; 4:e08022. [PMID: 26393419 PMCID: PMC4568445 DOI: 10.7554/elife.08022] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/20/2015] [Indexed: 01/11/2023] Open
Abstract
APC, a key negative regulator of Wnt signaling in development and oncogenesis, acts in the destruction complex with the scaffold Axin and the kinases GSK3 and CK1 to target βcatenin for destruction. Despite 20 years of research, APC's mechanistic function remains mysterious. We used FRAP, super-resolution microscopy, functional tests in mammalian cells and flies, and other approaches to define APC's mechanistic role in the active destruction complex when Wnt signaling is off. Our data suggest APC plays two roles: (1) APC promotes efficient Axin multimerization through one known and one novel APC:Axin interaction site, and (2) GSK3 acts through APC motifs R2 and B to regulate APC:Axin interactions, promoting high-throughput of βcatenin to destruction. We propose a new dynamic model of how the destruction complex regulates Wnt signaling and how this goes wrong in cancer, providing insights into how this multiprotein signaling complex is assembled and functions via multivalent interactions. DOI:http://dx.doi.org/10.7554/eLife.08022.001 An embryo starts off as a small ball of stem cells, each of which has the potential to become any type of cell in the body. Adult organs and tissues also contain small numbers of stem cells that can replace old or damaged cells. In both of these processes, stem cells need to ‘decide’ when they should start to change into a more specialized cell type, and which cell fate to choose (e.g., liver cell vs kidney cell). A signaling pathway involving Wnt proteins helps to direct many of these decisions. But if the ‘Wnt signaling pathway’ becomes activated at the wrong time, it can lead to cancer. For example, the first step in development of colon cancer is the inappropriate activation of Wnt signaling, and is most often caused by mutations in the gene that encodes a protein called APC. The APC protein is a tumor suppressor and normally inhibits Wnt signaling. However, even after over 20 years of effort, it remains largely mysterious how APC does this. APC is known to work with another protein called Axin as part of a large protein machine. This protein complex performs one of the first steps in a process that ultimately marks a key component of the Wnt signaling pathway for destruction. Pronobis et al. have now used a range of techniques to define APC's role in this so-called ‘destruction complex’. This analysis revealed the internal structure of a complex made from APC and Axin, and showed that cable- and sheet-like assemblies of Axin were intertwined with APC cables. Further experiments then revealed how APC and Axin proteins are added into or leave these complexes, and showed that this is critical for this protein machine to work. Pronobis et al.'s data also suggest that APC plays two roles, which make the destruction complex more efficient. Firstly, it can interact with Axin via two separate interaction sites that help to assemble the destruction complex. Secondly, specific features in APC allow it to interact with a third protein (called GSK3), which can then regulate how APC interacts with Axin. One of the next challenges will be to uncover how APC helps to transfer the components of Wnt signaling to the next step of their destruction, and to clear up the role played by GSK3. DOI:http://dx.doi.org/10.7554/eLife.08022.002
Collapse
Affiliation(s)
- Mira I Pronobis
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, Bethesda, United States
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
25
|
Lu YY, Wang JJ, Zhang XK, Li WB, Guo XL. 1118-20, an indazole diarylurea compound, inhibits hepatocellular carcinoma HepG2 proliferation and tumour angiogenesis involving Wnt/β-catenin pathway and receptor tyrosine kinases. J Pharm Pharmacol 2015; 67:1393-405. [DOI: 10.1111/jphp.12440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/12/2015] [Indexed: 12/17/2022]
Abstract
Abstract
Objectives
Sorafenib is a first multi-kinase inhibitor and one of the most widely used small-molecule oral-targeted drugs. It has been widely used for the treatment of patients with advanced renal cell carcinoma and hepatocellular carcinoma. However, some common adverse effects of sorafenib may impact quality of life. In this study, we evaluated the inhibitory effect on the growth of hepatocellular carcinoma cell line (HepG2) and suppression on angiogenesis of 1118-20, a newly synthesized indazole diarylurea compound.
Methods
We evaluated the activity of 1118-20 against HepG2 cells growth and tumour angiogenesis of human umbilical vascular endothelial cell line (HUVECs) with sorafenib as a positive control.
Key findings
The cytotoxic efficacy of 1118-20 was higher in HepG2 cells than human normal liver cell line (HL-7702). 1118-20 significantly suppressed the proliferation of HepG2 cells by apoptosis induction via Bcl-2 family-mediated mitochondria pathway and inhibition on Wnt/β-catenin signalling pathway. 1118-20 effectively blunt the motility and migration, and inhibited the formation of capillary tube of HUVECs through suppression of angiogenic factors expression. Moreover, the results indicated that 1118-20 exerted higher efficacy than sorafenib on tumour cell proliferation and angiogenesis.
Conclusions
Compared with its parent drug sorafenib, we found that 1118-20 possessed more potential on inhibition of angiogenesis and cancer cells growth. Inhibitory effect of 1118-20 on non-tumour liver cell HL-7702 was lower than that on hepatoma carcinoma cell HepG2. These results suggest that 1118-20 is a promising candidate compound that could be developed to a potent anticancer agent.
Collapse
Affiliation(s)
- Yu-Yin Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jing-Jing Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Xin-Ke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wen-Bao Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiu-Li Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
26
|
Ding SL, Yang ZW, Wang J, Zhang XL, Chen XM, Lu FM. Integrative analysis of aberrant Wnt signaling in hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol 2015; 21:6317-6328. [PMID: 26034368 PMCID: PMC4445110 DOI: 10.3748/wjg.v21.i20.6317] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 01/22/2015] [Accepted: 03/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To comprehensively understand the underlying molecular events accounting for aberrant Wnt signaling activation in hepatocellular carcinoma (HCC).
METHODS: This study was retrospective. The HCC tissue specimens used in this research were obtained from patients who underwent liver surgery. The Catalogue of Somatic Mutations in Cancer (COSMIC) database was searched for the mutation statuses of CTNNB1, TP53, and protein degradation regulator genes of CTNNB1. Dual-luciferase reporter assay was performed with TOP/FOP reporters to detect whether TP53 gain-of-function (GOF) mutations could enhance the transcriptional activity of Wnt signaling. Methylation sensitive restriction enzyme-quantitative PCR was used to explore the methylation status of CpG islands located in the promoters of APC, SFRP1, and SFRP5 in HCCs with different risk factors. Finally, nested-reverse transcription PCR was performed to examine the integration of HBx in front of LINE1 element and the existence of HBx-LINE1 chimeric transcript in Hepatitis B virus-related HCC. All results in this article were analyzed with the software SPSS version 19.0 for Windows, and different groups were compared by χ2 test as appropriate.
RESULTS: Based on the data from COSMIC database, compared with other solid tumors, mutation frequency of CTNNB1 was significantly higher in HCC (P < 0.01). The rate of CTNNB1 mutation was significantly less frequent in Hepatitis B virus-related HCC than in other etiologies (P < 0.01). Dual-luciferase reporter system and TOP/FOP reporter assays confirmed that TP53 GOF mutants were able to enhance the transcriptional ability of Wnt signaling. An exclusive relationship between the status of TP53 and CTNNB1 mutations was observed. However, according to the COSMIC database, TP53 GOF mutation is rare in HCC, which indicates that TP53 GOF mutation is not a reason for the aberrant activation of Wnt signaling in HCC. APC and AXIN1 were mutated in HCC. By using methylation sensitive restriction enzyme-quantitative PCR, hypermethylation of APC was detected in HCC with different risk factors, whereas SFRP1 and SFRP5 were not hypermethylated in any of the HCC etiologies, which indicates that the mutation of APC and AXIN1, together with the methylation of APC could take part in the overactivation of Wnt signaling. Nested-reverse transcription PCR failed to detect the integration of HBx before the LINE1 element, or the existence of an HBx-LINE1 chimeric transcript, suggesting that integration could not play a role in the aberrant activation of Wnt signaling in HCC.
CONCLUSION: In HCC, genetic/epigenetic aberration of CTNNB1 and its protein degradation regulators are the major cause of Wnt signaling overactivation.
Collapse
MESH Headings
- Animals
- Binding Sites
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Transformation, Viral
- Chi-Square Distribution
- CpG Islands
- DNA Methylation
- Databases, Genetic
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- HEK293 Cells
- Hepatitis B, Chronic/complications
- Hepatitis B, Chronic/diagnosis
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/virology
- Mutation
- Phenotype
- Promoter Regions, Genetic
- Proteolysis
- Retrospective Studies
- Systems Biology
- Systems Integration
- Transcriptional Activation
- Transfection
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Wnt Signaling Pathway/genetics
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
|
27
|
MiR-939 promotes the proliferation of human ovarian cancer cells by repressing APC2 expression. Biomed Pharmacother 2015; 71:64-9. [DOI: 10.1016/j.biopha.2015.02.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 02/15/2015] [Indexed: 12/30/2022] Open
|
28
|
Sinkovics JG. The cell survival pathways of the primordial RNA-DNA complex remain conserved in the extant genomes and may function as proto-oncogenes. Eur J Microbiol Immunol (Bp) 2015; 5:25-43. [PMID: 25883792 PMCID: PMC4397846 DOI: 10.1556/eujmi-d-14-00034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/22/2014] [Indexed: 01/09/2023] Open
Abstract
Malignantly transformed (cancer) cells of multicellular hosts, including human cells, operate activated biochemical pathways that recognizably derived from unicellular ancestors. The descendant heat shock proteins of thermophile archaea now chaperon oncoproteins. The ABC cassettes of toxin-producer zooxantella Symbiodinia algae pump out the cytoplasmic toxin molecules; malignantly transformed cells utilize the derivatives of these cassettes to get rid of chemotherapeuticals. High mobility group helix-loop-helix proteins, protein arginine methyltransferases, proliferating cell nuclear antigens, and Ki-67 nuclear proteins, that protect and repair DNA in unicellular life forms, support oncogenes in transformed cells. The cell survival pathways of Wnt-β-catenin, Hedgehog, PI3K, MAPK-ERK, STAT, Ets, JAK, Pak, Myb, achaete scute, circadian rhythms, Bruton kinase and others, which are physiological in uni- and early multicellular eukaryotic life forms, are constitutively encoded in complex oncogenic pathways in selected single cells of advanced multicellular eukaryotic hosts. Oncogenes and oncoproteins in advanced multicellular hosts recreate selected independently living and immortalized unicellular life forms, which are similar to extinct and extant protists. These unicellular life forms are recognized at the clinics as autologous "cancer cells".
Collapse
Affiliation(s)
- J G Sinkovics
- St. Joseph's Hospital Cancer Institute Affiliated with the H. L. Moffitt Comprehensive Cancer Center, Morsani College of Medicine, Department of Molecular Medicine, The University of South Florida Tampa, FL USA
| |
Collapse
|
29
|
Kunttas-Tatli E, Roberts DM, McCartney BM. Self-association of the APC tumor suppressor is required for the assembly, stability, and activity of the Wnt signaling destruction complex. Mol Biol Cell 2014; 25:3424-36. [PMID: 25208568 PMCID: PMC4214788 DOI: 10.1091/mbc.e14-04-0885] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The tumor suppressor adenomatous polyposis coli (APC) is an essential negative regulator of Wnt signaling through its activity in the destruction complex with Axin, GSK3β, and CK1 that targets β-catenin/Armadillo (β-cat/Arm) for proteosomal degradation. The destruction complex forms macromolecular particles we termed the destructosome. Whereas APC functions in the complex through its ability to bind both β-cat and Axin, we hypothesize that APC proteins play an additional role in destructosome assembly through self-association. Here we show that a novel N-terminal coil, the APC self-association domain (ASAD), found in vertebrate and invertebrate APCs, directly mediates self-association of Drosophila APC2 and plays an essential role in the assembly and stability of the destructosome that regulates β-cat degradation in Drosophila and human cells. Consistent with this, removal of the ASAD from the Drosophila embryo results in β-cat/Arm accumulation and aberrant Wnt pathway activation. These results suggest that APC proteins are required not only for the activity of the destructosome, but also for the assembly and stability of this macromolecular machine.
Collapse
Affiliation(s)
- Ezgi Kunttas-Tatli
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - David M Roberts
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17604
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|