1
|
Sobhani I, De Oliveira Alves N, Sadeghi M, Charpy C, Bergsten E, Amiot A, Barau C, Brunetti F, Vaysse A, Tournigand C, Chamaillard M, Khashayarsha K, Mestivier D. Poor prognosis in IBD-complicated colon cancer through gut dysbiosis-related immune response failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645177. [PMID: 40196693 PMCID: PMC11974894 DOI: 10.1101/2025.03.25.645177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Colorectal cancer (CRC) results from the accumulation of mutations and epigenetic changes in gut epithelial cells likely due to gut microbiota dysbiosis. However, limited research has been done to explore the link between host tumour dysbiosis and disease outcome. Methods The mechanisms influencing outcomes of 97 colorectal cancer (CRC) patients, including 13 with Lynch syndrome, 20 with inflammatory bowel disease (IBD), and 64 sporadic cases, were analyzed using a multiomics approach. These patients were categorized into two groups: "disease-free/stable disease" and "progression disease" survival outcomes. The analysis included tumor adherent microbiota composition (16S rRNA), somatic gene mutations (WES), gene expression (RNAseq), immune markers (RNAscope), and immune infiltrate cells (immunohistochemistry). Results IBD-CRC patients had worse outcomes than those with Lynch or sporadic CRC, regardless of TNM staging or treatment. Symbiotic bacteria like Lactococcus lactis were significantly reduced in IBD-CRC tissues. Patient outcomes were influenced by the abundance of virulent ( Escherichia coli ) relative to beneficial bacteria ( Lactococcus lactis ). Although no significant increase in deleterious somatic mutations was found in IBD-CRC. 16sRNA revealed increased virulent- and decreased anti-inflammatory symbiotic-bacteria correlating with the upregulation of oncogenes and downregulation of anti-oncogenes like PHLPP1. The multiplex in situ hybridization of CD8, IFNγ and PHLPP1 an anti-oncogene revealed significant decrease of immune cells with detectable PHLPP1 expression in IBD-CRC tumour tissues as compared to sporadic CRCs. Conclusion The poor outcomes in IBD-CRC patients are likely due to gut dysbiosis and immune cell alterations, possibly triggered by microbiota-related epigenetic pathways. What You Need to Know BACKGROUND AND CONTEXT: Colorectal cancer (CRC) is associated with gut microbiota dysbiosis. Inflammatory bowel disease-related CRC (IBD-CRC) is classified as an environment-related condition.NEW FINDINGS: In relation with patient outcomes, tumour tissues from three types of CRC (Sporadic-, IBD-, and Lynch syndrome-CRC) were analyzed using a multiomic approach. This included examining tissue adherent virulent bacteria, gene analyses, and quantifying immune cell infiltration in the mucosa. IBD-CRC patients had the worst outcomes, associated with the down regulation of PHLPP1 gene, virulent/symbiotic imbalance, and immune response failure.LIMITATIONS: Lack of animal experiments using FMT of fresh stool from IBD-CRC patients.CLINICAL AND TRANSLATIONAL RESEARCH RELEVANCE: Among the different types of CRC, IBD-CRC patients showed a greater imbalance between harmful and beneficial bacteria, along with immune response failure.Lay summary: This study compares the pathological and clinical characteristics of patients with colorectal cancer (CRC) across three distinct etiologies: sporadic CRC, inflammatory bowel disease (IBD)-associated CRC, and Lynch syndrome-associated CRC (LS-CRC). Distinct differences in tumor-adherent microbiota, gene expression and immune response profiles were observed. Notably, IBD-CRC patients demonstrated the poorest prognosis depending on microbe-host gene interaction highlighting potential biomarkers for disease prognosis and treatment strategies.
Collapse
|
2
|
Razmi M, Bayat AA, Mortazavi N, Kalantari E, Saeednejad Zanjani L, Saki S, Ghods R, Madjd Z. Clinical significance of "S" isoform of DCLK1 in different gastric cancer subtypes using newly produced monoclonal antibody. Cancer Biomark 2025; 42:18758592241301691. [PMID: 40109216 DOI: 10.1177/18758592241301691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundDoublecortin-like kinase 1 (DCLK1) isoforms play distinct roles in the progression of gastrointestinal cancers. For the first time ever, the current study aimed to generate DCLK1-S-specific monoclonal antibodies (mAbs) to evaluate the clinical value of DCLK1-S (short isoform) in gastric cancer (GC).Materials and methodsMice were immunized with a unique 7-mer synthetic peptide of DCLK1-S conjugated with keyhole limpet hemocyanin (KLH). Immunoreactivity of hybridomas and mAbs was determined by ELISA assays and immunohistochemistry (IHC). DCLK1-S expression in two GC cell lines was assessed by flow cytometry. After characterization, the expression pattern of DCLK1-S was investigated in different histological subtypes of GC (n=217) and adjacent normal tissues (n=28) using IHC on tissue microarrays. The association of clinical prognostic values with DCLK1-S expression was also investigated.ResultsELISA findings demonstrated that the generated monoclonal antibody (mAb) exhibited strong immunoreactivity towards the immunizing peptide. Positive control tissues, including GC and colorectal cancer, showed strong positive immunoreactivity with anti-DCLK1-S mAb whereas negative reagent control sections represented no staining, demonstrating the specificity of produced mAb. Flow cytometry analysis confirmed that the newly developed mAbs effectively recognized DCLK1-S on the cell surface. A mixture pattern of membranous, cytoplasmic, and nuclear DCLK1-S expression in the GC cells was observed. A significant and inverse association was identified between the expression DCLK1-S in the cell membrane and cytoplasm and PT stage, muscolarispropia, subserosa, and perineural invasion in intestinal subtype, respectively. In signet ring cell type, however, nuclear DCLK1-S expression was adversely associated with tumor size and PT stage. Furthermore, patients with low DCLK1-S expression had a shorter survival than patients with high expression, however, without a statistically significant association.ConclusionAn efficient and precise tool for detecting DCLK1-S in cancer tissues has been developed. Moreover, DCLK1-S overexpression might be considered a favorable clinical factor in GC patients.
Collapse
Affiliation(s)
- Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ali-Ahmad Bayat
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Nafiseh Mortazavi
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Pathology and Genomic Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sima Saki
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Karamikheirabad M, Zhang J, Ahn AR, Park HS, Park SH, Moon YJ, Kim KM, Jang KY. IL-13Rα2 Is Involved in Resistance to Doxorubicin and Survival of Osteosarcoma Patients. Pharmaceuticals (Basel) 2024; 17:1526. [PMID: 39598436 PMCID: PMC11597473 DOI: 10.3390/ph17111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Interleukin 13 receptor alpha 2 (IL-13Rα2) is a receptor with a high affinity for IL-13 and is involved in the progression of human cancers. However, studies on the role of IL-13Rα2 in osteosarcoma are limited. Therefore, this study aimed to investigate the expression and roles of IL-13Rα2 in the progression of osteosarcoma. METHODS This study evaluated the roles of IL-13Rα2 in osteosarcomas by evaluating tumor tissues from 37 human osteosarcomas and osteosarcoma cells. RESULTS Immunohistochemical positivity of IL-13Rα2 was an independent indicator of shorter overall survival and relapse-free survival of 37 osteosarcoma patients and 26 subpopulations of patients who received adjuvant chemotherapy with multivariate analysis. In U2OS and KHOS/NP osteosarcoma cells, overexpression of IL-13Rα2 significantly increased proliferation, migration, and invasion of cells, all of which decreased with knockdown of IL-13Rα2. Overexpression of IL-13Rα2 increased expression of TGF-β, snail, cyclin D1, and BCL2 but decreased BAX, and knockdown of IL-13Rα2 caused a decrease in expression of these molecules. In addition, both in vitro and in vivo, proliferation of osteosarcoma cells increased, and apoptosis decreased with overexpression of IL-13Rα2 under treatment with doxorubicin. Knockdown of IL-13Rα2 sensitized osteosarcoma cells to the cytotoxic effect of doxorubicin. CONCLUSIONS The results of this study suggest that the expression of IL13Rα2 might be used as a potential prognostic indicator in osteosarcoma patients. Furthermore, it is observed that IL13Rα2 influences the resistance to the chemotherapeutic agent doxorubicin. Therefore, a therapeutic trial targeting IL13Rα2 might be a new therapeutic strategy for osteosarcoma, especially those highly expressing IL13Rα2.
Collapse
Affiliation(s)
- Maryam Karamikheirabad
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
| | - Junyue Zhang
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
| | - Ae-Ri Ahn
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
| | - Ho Sung Park
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea;
| | - Young Jae Moon
- Department of Biochemistry and Molecular Biology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea;
- Department of Orthopedic Surgery, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Medical School, Jeonbuk National University, Jeonju 54896, Republic of Korea; (M.K.); (J.Z.); (A.-R.A.); (H.S.P.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
4
|
Ye L, Liu B, Huang J, Zhao X, Wang Y, Xu Y, Wang S. DCLK1 and its oncogenic functions: A promising therapeutic target for cancers. Life Sci 2024; 336:122294. [PMID: 38007147 DOI: 10.1016/j.lfs.2023.122294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
Doublecortin-like kinase 1 (DCLK1), a significant constituent of the protein kinase superfamily and the doublecortin family, has been recognized as a prooncogenic factor that exhibits a strong association with the malignant progression and clinical prognosis of various cancers. DCLK1 serves as a stem cell marker that governs tumorigenesis, tumor cell reprogramming, and epithelial-mesenchymal transition. Multiple studies have indicated the capable of DCLK1 in regulating the DNA damage response and facilitating DNA damage repair. Additionally, DCLK1 is involved in the regulation of the immune microenvironment and the promotion of tumor immune evasion. Recently, DCLK1 has emerged as a promising therapeutic target for a multitude of cancers. Several small-molecule inhibitors of DCLK1 have been identified. Nevertheless, the biological roles of DCLK1 are mainly ambiguous, particularly with the disparities between its α- and β-form transcripts in the malignant progression of cancers, which impedes the development of more precisely targeted drugs. This article focuses on tumor stem cells, tumor epithelial-mesenchymal transition, the DNA damage response, and the tumor microenvironment to provide a comprehensive overview of the association between DCLK1 and tumor malignant progression, address unsolved questions and current challenges, and project future directions for targeting DCLK1 for the diagnosis and treatment of cancers.
Collapse
Affiliation(s)
- Liu Ye
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Beibei Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jingling Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaolin Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yuan Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
| | - Yungen Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
5
|
Moore LL, Houchen CW. Epigenetic Landscape and Therapeutic Implication of Gene Isoforms of Doublecortin-Like Kinase 1 for Cancer Stem Cells. Int J Mol Sci 2023; 24:16407. [PMID: 38003596 PMCID: PMC10671580 DOI: 10.3390/ijms242216407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
While significant strides have been made in understanding cancer biology, the enhancement in patient survival is limited, underscoring the urgency for innovative strategies. Epigenetic modifications characterized by hereditary shifts in gene expression without changes to the DNA sequence play a critical role in producing alternative gene isoforms. When these processes go awry, they influence cancer onset, growth, spread, and cancer stemness. In this review, we delve into the epigenetic and isoform nuances of the protein kinase, doublecortin-like kinase 1 (DCLK1). Recognized as a hallmark of tumor stemness, DCLK1 plays a pivotal role in tumorigenesis, and DCLK1 isoforms, shaped by alternative promoter usage and splicing, can reveal potential therapeutic touchpoints. Our discussion centers on recent findings pertaining to the specific functions of DCLK1 isoforms and the prevailing understanding of its epigenetic regulation via its two distinct promoters. It is noteworthy that all DCLK1 isoforms retain their kinase domain, suggesting that their unique functionalities arise from non-kinase mechanisms. Consequently, our research has pivoted to drugs that specifically influence the epigenetic generation of these DCLK1 isoforms. We posit that a combined therapeutic approach, harnessing both the epigenetic regulators of specific DCLK1 isoforms and DCLK1-targeted drugs, may prove more effective than therapies that solely target DCLK1.
Collapse
Affiliation(s)
- Landon L. Moore
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Courtney W. Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Hardt R, Dehghani A, Schoor C, Gödderz M, Cengiz Winter N, Ahmadi S, Sharma R, Schork K, Eisenacher M, Gieselmann V, Winter D. Proteomic investigation of neural stem cell to oligodendrocyte precursor cell differentiation reveals phosphorylation-dependent Dclk1 processing. Cell Mol Life Sci 2023; 80:260. [PMID: 37594553 PMCID: PMC10439241 DOI: 10.1007/s00018-023-04892-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Oligodendrocytes are generated via a two-step mechanism from pluripotent neural stem cells (NSCs): after differentiation of NSCs to oligodendrocyte precursor/NG2 cells (OPCs), they further develop into mature oligodendrocytes. The first step of this differentiation process is only incompletely understood. In this study, we utilized the neurosphere assay to investigate NSC to OPC differentiation in a time course-dependent manner by mass spectrometry-based (phospho-) proteomics. We identify doublecortin-like kinase 1 (Dclk1) as one of the most prominently regulated proteins in both datasets, and show that it undergoes a gradual transition between its short/long isoform during NSC to OPC differentiation. This is regulated by phosphorylation of its SP-rich region, resulting in inhibition of proteolytic Dclk1 long cleavage, and therefore Dclk1 short generation. Through interactome analyses of different Dclk1 isoforms by proximity biotinylation, we characterize their individual putative interaction partners and substrates. All data are available via ProteomeXchange with identifier PXD040652.
Collapse
Affiliation(s)
- Robert Hardt
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Alireza Dehghani
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach, Germany
| | - Carmen Schoor
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Markus Gödderz
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Nur Cengiz Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
- Institute of Human Genetics, University Hospital Cologne, 50931, Cologne, Germany
| | - Shiva Ahmadi
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
- Bayer Pharmaceuticals, 42113, Wuppertal, Germany
| | - Ramesh Sharma
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Karin Schork
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Volkmar Gieselmann
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Nussallee 11, 53115, Bonn, Germany.
| |
Collapse
|
7
|
Carli ALE, Hardy JM, Hoblos H, Ernst M, Lucet IS, Buchert M. Structure-Guided Prediction of the Functional Impact of DCLK1 Mutations on Tumorigenesis. Biomedicines 2023; 11:biomedicines11030990. [PMID: 36979969 PMCID: PMC10046695 DOI: 10.3390/biomedicines11030990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Doublecortin-like kinase 1 (DCLK1) is a functional serine/threonine (S/T)-kinase and a member of the doublecortin family of proteins which are characterized by their ability to bind to microtubules (MTs). DCLK1 is a proposed cancer driver gene, and its upregulation is associated with poor overall survival in several solid cancer types. However, how DCLK1 associates with MTs and how its kinase function contributes to pro-tumorigenic processes is poorly understood. This review builds on structural models to propose not only the specific functions of the domains but also attempts to predict the impact of individual somatic missense mutations on DCLK1 functions. Somatic missense mutations in DCLK1 are most frequently located within the N-terminal MT binding region and likely impact on the ability of DCLK1 to bind to αβ-tubulin and to polymerize and stabilize MTs. Moreover, the MT binding affinity of DCLK1 is negatively regulated by its auto-phosphorylation, and therefore mutations that affect kinase activity are predicted to indirectly alter MT dynamics. The emerging picture portrays DCLK1 as an MT-associated protein whose interactions with tubulin heterodimers and MTs are tightly controlled processes which, when disrupted, may confer pro-tumorigenic properties.
Collapse
Affiliation(s)
- Annalisa L E Carli
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Joshua M Hardy
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Hanadi Hoblos
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Matthias Ernst
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Isabelle S Lucet
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael Buchert
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
8
|
Tanvir I, Hassan A, Albeladi F. DNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas. Cureus 2022; 14:e30743. [DOI: 10.7759/cureus.30743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/05/2022] Open
|
9
|
Chhetri D, Vengadassalapathy S, Venkadassalapathy S, Balachandran V, Umapathy VR, Veeraraghavan VP, Jayaraman S, Patil S, Iyaswamy A, Palaniyandi K, Gnanasampanthapandian D. Pleiotropic effects of DCLK1 in cancer and cancer stem cells. Front Mol Biosci 2022; 9:965730. [PMID: 36250024 PMCID: PMC9560780 DOI: 10.3389/fmolb.2022.965730] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Doublecortin-like kinase 1 (DCLK1), a protein molecule, has been identified as a tumor stem cell marker in the cancer cells of gastrointestinal, pancreas, and human colon. DCLK1 expression in cancers, such as breast carcinoma, lung carcinoma, hepatic cell carcinoma, tuft cells, and human cholangiocarcinoma, has shown a way to target the DCLK1 gene and downregulate its expression. Several studies have discussed the inhibition of tumor cell proliferation along with neoplastic cell arrest when the DCLK1 gene, which is expressed in both cancer and normal cells, was targeted successfully. In addition, previous studies have shown that DCLK1 plays a vital role in various cancer metastases. The correlation of DCLK1 with numerous stem cell receptors, signaling pathways, and genes suggests its direct or an indirect role in promoting tumorigenesis. Moreover, the impact of DCLK1 was found to be related to the functioning of an oncogene. The downregulation of DCLK1 expression by using targeted strategies, such as embracing the use of siRNA, miRNA, CRISPR/Cas9 technology, nanomolecules, specific monoclonal antibodies, and silencing the pathways regulated by DCLK1, has shown promising results in both in vitro and in vivo studies on gastrointestinal (GI) cancers. In this review, we will discuss about the present understanding of DCLK1 and its role in the progression of GI cancer and metastasis.
Collapse
Affiliation(s)
- Dibyashree Chhetri
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Srinivasan Vengadassalapathy
- Department of Pharmacology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | | | - Varadharaju Balachandran
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Sree Balaji Dental College and Hospital, Chennai, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| | - Ashok Iyaswamy
- Centre for Parkinsons Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| |
Collapse
|
10
|
Targeting Doublecortin-Like Kinase 1 (DCLK1)-Regulated SARS-CoV-2 Pathogenesis in COVID-19. J Virol 2022; 96:e0096722. [PMID: 35943255 PMCID: PMC9472619 DOI: 10.1128/jvi.00967-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Host factors play critical roles in SARS-CoV-2 infection-associated pathology and the severity of COVID-19. In this study, we systematically analyzed the roles of SARS-CoV-2-induced host factors, doublecortin-like kinase 1 (DCLK1), and S100A9 in viral pathogenesis. In autopsied subjects with COVID-19 and pre-existing chronic liver disease, we observed high levels of DCLK1 and S100A9 expression and immunosuppressive (DCLK1+S100A9+CD206+) M2-like macrophages and N2-like neutrophils in lungs and livers. DCLK1 and S100A9 expression were rarely observed in normal controls, COVID-19-negative subjects with chronic lung disease, or COVID-19 subjects without chronic liver disease. In hospitalized patients with COVID-19, we detected 2 to 3-fold increased levels of circulating DCLK1+S100A9+ mononuclear cells that correlated with disease severity. We validated the SARS-CoV-2-dependent generation of these double-positive immune cells in coculture. SARS-CoV-2-induced DCLK1 expression correlated with the activation of β-catenin, a known regulator of the DCLK1 promoter. Gain and loss of function studies showed that DCLK1 kinase amplified live virus production and promoted cytokine, chemokine, and growth factor secretion by peripheral blood mononuclear cells. Inhibition of DCLK1 kinase blocked pro-inflammatory caspase-1/interleukin-1β signaling in infected cells. Treatment of SARS-CoV-2-infected cells with inhibitors of DCLK1 kinase and S100A9 normalized cytokine/chemokine profiles and attenuated DCLK1 expression and β-catenin activation. In conclusion, we report previously unidentified roles of DCLK1 in augmenting SARS-CoV-2 viremia, inflammatory cytokine expression, and dysregulation of immune cells involved in innate immunity. DCLK1 could be a potential therapeutic target for COVID-19, especially in patients with underlying comorbid diseases associated with DCLK1 expression. IMPORTANCE High mortality in COVID-19 is associated with underlying comorbidities such as chronic liver diseases. Successful treatment of severe/critical COVID-19 remains challenging. Herein, we report a targetable host factor, DCLK1, that amplifies SARS-CoV-2 production, cytokine secretion, and inflammatory pathways via activation of β-catenin(p65)/DCLK1/S100A9/NF-κB signaling. Furthermore, we observed in the lung, liver, and blood an increased prevalence of immune cells coexpressing DCLK1 and S100A9, a myeloid-derived proinflammatory protein. These cells were associated with increased disease severity in COVID-19 patients. Finally, we used a novel small-molecule inhibitor of DCLK1 kinase (DCLK1-IN-1) and S100A9 inhibitor (tasquinimod) to decrease virus production in vitro and normalize hyperinflammatory responses known to contribute to disease severity in COVID-19.
Collapse
|
11
|
Kalantari E, Razmi M, Tajik F, Asadi-Lari M, Ghods R, Madjd Z. Oncogenic functions and clinical significances of DCLK1 isoforms in colorectal cancer: a systematic review and meta-analysis. Cancer Cell Int 2022; 22:217. [PMID: 35717205 PMCID: PMC9206744 DOI: 10.1186/s12935-022-02632-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background The oncogenic role of doublecortin-like kinase 1 (DCLK1) as a putative cancer stem cell (CSC) marker has been clarified in colorectal cancer (CRC). Isoform-specific functions of DCLK1 have shed new light on different functions of DCLK1 short (DCLK1-S) and DCLK1 long (DCLK1-L) isoforms in tumor initiation, growth, and metastasis. Therefore, the current systematic review and meta-analysis aimed to review the available in vitro, in vivo, and clinical evidence on the oncogenic roles and clinical significance of DCLK1 isoforms in colorectal cancer. Methods The literature databases of PubMed, Scopus, ISI Web of Science, and Embase were searched to identify eligible articles. The description characteristics of in vitro and pre-clinical studies were extracted from identified reports. In addition, hazard ratios (HRs) or odds ratios (ORs) with 95% confidence intervals (CIs) were recorded to determine the relationships between DCLK1-L and DCLK1-S expression and prognostic outcomes in patients with CRC. Results Both in vitro and in vivo evidence have emphasized the potential oncogenic functions of DCLK1 in tumor initiation, self-renewal ability, tumor invasion, epithelial-mesenchymal transition (EMT), and metastasis. However, the anti-DCLK1 antibodies generally utilized in these studies could detect sequence homology epitopes of both isoforms. Recent limited isoform-specific evidence has strongly supported the significant positive expression and rather oncogenic efficacy of DCLK1-S in tumorigenesis, EMT, and invasion compared with DCLK1-L in human CRC cell lines. Our meta-analysis findings of limited clinical studies indicated that only overexpression of DCLK1-S is associated with worse overall survival (OS) (HR = 7.930, 95% CI 2.252–27.924, p = 0.001). Increased expression of both DCLK1-S (HR = 1.610, 95% CI 1.020–2.541, p = 0.041) and DCLK1-L (HR = 5.890, 95% CI 1.219–28.453, p = 0.027) isoforms was closely associated with worse DSS/CSS in CRC patients. Furthermore, the high expression of DCLK1-S was found to be associated with poor DFS/RFS/PFS (HR = 1.913, 95% CI 1.230–2.973, p = 0.004). Conclusions The current findings strongly supported that the DCLK1-S isoform may play a crucial role in the invasion, aggressive tumor behavior, and worsened survival outcomes of CRC patients. However, further critical investigations related to the potential preclinical and clinical utilities of DCLK1-S as a specific CRC-CSC marker are warranted. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02632-9.
Collapse
Affiliation(s)
- Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohsen Asadi-Lari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
12
|
Wang J, Yokoyama Y, Hirose H, Shimomura Y, Bonkobara S, Itakura H, Kouda S, Morimoto Y, Minami K, Takahashi H, Shibata S, Kobayashi S, Uemura M, Tanaka S, Wu X, Tanaka S, Mori M, Yamamoto H. Functional assessment of miR‑1291 in colon cancer cells. Int J Oncol 2022; 60:13. [PMID: 34981812 PMCID: PMC8759348 DOI: 10.3892/ijo.2022.5303] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
miR‑1291 exerts an anti‑tumor effect in a subset of human carcinomas, including pancreatic cancer. However, its role in colorectal cancer (CRC) is largely unknown. In the present study, the expression and effect of miR‑1291 in CRC cells was investigated. It was identified that miR‑1291 significantly suppressed the proliferation, invasion, cell mobility and colony formation of CRC cells. Additionally, miR‑1291 induced cell apoptosis. A luciferase reporter assay revealed that miR‑1291 directly bound the 3'‑untranslated region sequence of doublecortin‑like kinase 1 (DCLK1). miR‑1291 also suppressed DCLK1 mRNA and protein expression in HCT116 cells that expressed DCLK1. Furthermore, miR‑1291 suppressed cancer stem cell markers BMI1 and CD133, and inhibited sphere formation. The inhibitory effects on sphere formation, invasion and mobility in HCT116 cells were also explored and verified using DCLK1 siRNAs. Furthermore, miR‑1291 induced CDK inhibitors p21WAF1/CIP1 and p27KIP1 in three CRC cell lines, and the overexpression of DCLK1 in HCT116 cells led to a decrease of p21WAF1/CIP1 and p27KIP1. Intravenous administration of miR‑1291 loaded on the super carbonate apatite delivery system significantly inhibited tumor growth in the DLD‑1 xenograft mouse model. Additionally, the resultant tumors exhibited significant upregulation of the p21WAF1/CIP1 and p27KIP1 protein with treatment of miR‑1291. Taken together, the results indicated that miR‑1291 served an anti‑tumor effect by modulating multiple functions, including cancer stemness and cell cycle regulation. The current data suggested that miR‑1291 may be a promising nucleic acid medicine against CRC.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuhki Yokoyama
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Haruka Hirose
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Shimomura
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Saki Bonkobara
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroaki Itakura
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shihori Kouda
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Morimoto
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazumasa Minami
- Department of Radiation Oncology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Satoshi Shibata
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shogo Kobayashi
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mamoru Uemura
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Susumu Tanaka
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Suita, Osaka 565-0871, Japan
| | - Xin Wu
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
- Department of Hepato-Billiary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Masaki Mori
- Tokai University, Graduate School of Medicine, Isehara, Kanagawa 259-1193, Japan
| | - Hirofumi Yamamoto
- Department of Molecular Pathology, Division of Health Sciences, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Surgery and Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
13
|
Cheng L, Yang Z, Guo W, Wu C, Liang S, Tong A, Cao Z, Thorne RF, Yang SY, Yu Y, Chen Q. DCLK1 autoinhibition and activation in tumorigenesis. Innovation (N Y) 2022; 3:100191. [PMID: 34977835 PMCID: PMC8686072 DOI: 10.1016/j.xinn.2021.100191] [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: 05/18/2021] [Accepted: 11/23/2021] [Indexed: 02/05/2023] Open
Abstract
Doublecortin-like kinase 1 (DCLK1) is upregulated in many tumors and is a marker for tumor stem cells. Accumulating evidence suggests DCLK1 constitutes a promising drug target for cancer therapy. However, the regulation of DCLK1 kinase activity is poorly understood, particularly the function of its autoinhibitory domain (AID), and, moreover, no physiological activators of DCLK1 have presently been reported. Here we determined the first DCLK1 kinase structure in the autoinhibited state and identified the neuronal calcium sensor HPCAL1 as an activator of DCLK1. The C-terminal AID functions to block the ATP-binding site and is competitive with ATP. HPCAL1 binds directly to the AID in a Ca2+-dependent manner, which releases the autoinhibition. We also analyzed cancer-associated mutations occurring in the AID and elucidate how these mutations disrupt DCLK1 autoinhibition to elicit kinase activity upregulation. Our results present a molecular mechanism for autoinhibition and activation of DCLK1 kinase activity and provide insights into DCLK1-associated tumorigenesis.
Collapse
Affiliation(s)
- Linna Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.,Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Zejing Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wenhao Guo
- Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450003, China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yamei Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| |
Collapse
|
14
|
Kalantari E, Ghods R, Zanjani LS, Rahimi M, Eini L, Razmi M, Asadi-Lari M, Madjd Z. Cytoplasmic expression of DCLK1-S, a novel DCLK1 isoform, is associated with tumor aggressiveness and worse disease-specific survival in colorectal cancer. Cancer Biomark 2021; 33:277-289. [DOI: 10.3233/cbm-210330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND: Isoform-specific function of doublecortin-like kinase 1 (DCLK1) has highlighted the key role of the DCLK1-S (short isoform) in the maintenance, progression, and invasion of the tumor. OBJECTIVE: This study was designed to produce an anti-DCLK1-S polyclonal antibody to evaluate DCLK1-S in human colorectal cancer (CRC) specifically. METHODS: The expression pattern and clinical significance of DCLK1-S were assessed in a well-defined tissue microarray (TMA) series of 348 CRC and 51 adjacent normal tissues during a follow-up period of 108 months. RESULTS: Expression of DCLK1-S was significantly higher in CRC samples compared to adjacent normal samples (P< 0.001). Cytoplasmic expression of DCLK1-S was significantly higher in the tumors at the advanced stage of cancer and with poorer differentiation (P< 0.001, P= 0.02). The patients with CRC whose tumors showed higher cytoplasmic expression of DCLK1-S had worse disease-specific survival (DSS) (log-rank test, P= 0.03) and 5-year DSS rates (P= 0.01). Additionally, an improved prognostic value was observed in the patients with CRC with high DCLK1-S expression vs. its moderate expression (HR: 2.70, 95% CI: 0.98–7.38; p= 0.04) by multivariate analysis. CONCLUSIONS: Our findings strongly supported that high cytoplasmic expression of DCLK1-S compared to its moderate expression could be considered an independent prognostic factor influencing DSS.
Collapse
Affiliation(s)
- Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Mandana Rahimi
- Hasheminejad Kidney Center, Pathology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leila Eini
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Division of Histology, Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohsen Asadi-Lari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
15
|
Vijai M, Baba M, Ramalingam S, Thiyagaraj A. DCLK1 and its interaction partners: An effective therapeutic target for colorectal cancer. Oncol Lett 2021; 22:850. [PMID: 34733368 PMCID: PMC8561619 DOI: 10.3892/ol.2021.13111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/02/2021] [Indexed: 12/23/2022] Open
Abstract
Doublecortin-like kinase protein 1 (DCLK1) is a microtubule-associated protein with a C-terminal serine/threonine kinase domain. Its expression was first reported in radial glial cells, where it serves an essential role in early neurogenesis, and since then, other functions of the DCLK1 protein have also been identified. Initially considered to be a marker of quiescent gastrointestinal and pancreatic stem cells, DCLK1 has recently been identified in the gastrointestinal tract as a marker of tuft cells. It has also been implicated in different types of cancer, where it regulates several vital pathways, such as Kras signaling. However, its underlying molecular mechanisms remain unclear. The present review discusses the different roles of DCLK1 and its interactions with other proteins that are homologically similar to DCLK1 to develop a novel therapeutic strategy to target cancer cells more accurately.
Collapse
Affiliation(s)
- Muthu Vijai
- Department of Genetic Engineering, SRM Institute of Science and Technology, Sri Ramaswamy Memorial (SRM) Nagar, Kattankulathur, Tamil Nadu 603203, India
| | - Mursaleen Baba
- Department of Genetic Engineering, SRM Institute of Science and Technology, Sri Ramaswamy Memorial (SRM) Nagar, Kattankulathur, Tamil Nadu 603203, India
| | - Satish Ramalingam
- Department of Genetic Engineering, SRM Institute of Science and Technology, Sri Ramaswamy Memorial (SRM) Nagar, Kattankulathur, Tamil Nadu 603203, India
| | - Anand Thiyagaraj
- Department of Genetic Engineering, SRM Institute of Science and Technology, Sri Ramaswamy Memorial (SRM) Nagar, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
16
|
Ge Y, Fan X, Huang X, Weygant N, Xiao Z, Yan R, Liu H, Liu J, An G, Yao J. DCLK1-Short Splice Variant Promotes Esophageal Squamous Cell Carcinoma Progression via the MAPK/ERK/MMP2 Pathway. Mol Cancer Res 2021; 19:1980-1991. [PMID: 34610960 DOI: 10.1158/1541-7786.mcr-21-0161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022]
Abstract
Cancer stem cell (CSC) marker doublecortin-like kinase 1 (DCLK1) contributes greatly to the malignancy of gastrointestinal cancers, and DCLK1-targeted agents have potential therapeutic value. However, the molecular pathways regulated by DCLK1-S (DCLK1 isoform 4), a shortened splice variant of DCLK1, still remain obscure. Here we found that the expression of DCLK1-S is significantly increased in human esophageal squamous cell carcinoma (ESCC) tissues and associated with malignant progression and poor prognosis. Functional studies indicated that silencing total of DCLK1 mediated by CRISPR/Cas9 inhibited ESCC cell proliferation, migration, and invasion. Conversely, these changes were largely reversed after DCLK1-S rescue or overexpression. More importantly, DCLK1-S significantly enhanced primary tumor formation and metastatic lung colonization in vivo. The Cancer Genome Atlas database and molecular analysis showed that DCLK1-S was closely related to the epithelial-mesenchymal transition (EMT) process in patients with ESCC. Further RNA sequencing and Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that MAPK signaling pathway was significantly enriched. Our in vitro study proclaimed that DCLK1-S induced MMP2 expression in ESCC cells via MAPK/ERK signaling, leading to the activation of EMT. In addition, administration of ERK1/2 blocker SCH772984 attenuated the proliferative and migratory phenotype induced by DCLK1-S. In conclusion, these findings suggest that DCLK1-S may be a key molecule in MAPK/ERK/MMP2 pathway-mediated progression of ESCC, and that it has potential as a biomarker or therapeutic target to improve outcomes in patients with ESCC. IMPLICATIONS: : DCLK1-S induces ESCC progression by activating the MAPK/ERK/MMP2 axis and may serve as a prognostic biomarker or therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Xiaona Fan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Xuying Huang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, P.R. China.,Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, P.R. China
| | - Zeru Xiao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Rui Yan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Heshu Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China.,Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China.
| | - Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China.
| |
Collapse
|
17
|
Structural basis for small molecule targeting of Doublecortin Like Kinase 1 with DCLK1-IN-1. Commun Biol 2021; 4:1105. [PMID: 34545159 PMCID: PMC8452690 DOI: 10.1038/s42003-021-02631-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/01/2021] [Indexed: 02/03/2023] Open
Abstract
Doublecortin-like kinase 1 (DCLK1) is an understudied bi-functional kinase with a proven role in tumour growth and development. However, the presence of tissue-specific spliced DCLK1 isoforms with distinct biological functions have challenged the development of effective strategies to understand the role of DCLK1 in oncogenesis. Recently, DCLK1-IN-1 was reported as a highly selective DCLK1 inhibitor, a powerful tool to dissect DCLK1 biological functions. Here, we report the crystal structures of DCLK1 kinase domain in complex with DCLK1-IN-1 and its precursors. Combined, our data rationalises the structure-activity relationship that informed the development of DCLK1-IN-1 and provides the basis for the high selectivity of DCLK1-IN-1, with DCLK1-IN-1 inducing a drastic conformational change of the ATP binding site. We demonstrate that DCLK1-IN-1 binds DCLK1 long isoforms but does not prevent DCLK1's Microtubule-Associated Protein (MAP) function. Together, our work provides an invaluable structural platform to further the design of isoform-specific DCLK1 modulators for therapeutic intervention.
Collapse
|
18
|
Jang DM, Lim HJ, Hahn H, Lee Y, Kim HK, Kim HS. Structural Basis of Inhibition of DCLK1 by Ruxolitinib. Int J Mol Sci 2021; 22:ijms22168488. [PMID: 34445192 PMCID: PMC8395186 DOI: 10.3390/ijms22168488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Given the functional attributes of Doublecortin-like kinase 1 (DCLK1) in tumor growth, invasion, metastasis, cell motility, and tumor stemness, it is emerging as a therapeutic target in gastrointestinal cancers. Although a series of specific or nonspecific ATP-competitive inhibitors were identified against DCLK1, different types of scaffolds that can be utilized for the development of highly selective inhibitors or structural understanding of binding specificities of the compounds remain limited. Here, we present our work to repurpose a Janus kinase 1 inhibitor, ruxolitinib as a DCLK1 inhibitor, showing micromolar binding affinity and inhibitory activity. Furthermore, to gain an insight into its interaction mode with DCLK1, a crystal structure of the ruxolitinib-complexed DCLK1 has been determined and analyzed. Ruxolitinib as a nonspecific DCLK1 inhibitor characterized in this work is anticipated to provide a starting point for the structure-guided discovery of selective DCLK1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Hark Kyun Kim
- Correspondence: (H.K.K.); (H.S.K.); Tel.: +82-31-920-2275 (H.S.K.)
| | - Hyoun Sook Kim
- Correspondence: (H.K.K.); (H.S.K.); Tel.: +82-31-920-2275 (H.S.K.)
| |
Collapse
|
19
|
Broner EC, Trujillo JA, Korzinkin M, Subbannayya T, Agrawal N, Ozerov IV, Zhavoronkov A, Rooper L, Kotlov N, Shen L, Pearson AT, Rosenberg AJ, Savage PA, Mishra V, Chatterjee A, Sidransky D, Izumchenko E. Doublecortin-Like Kinase 1 (DCLK1) Is a Novel NOTCH Pathway Signaling Regulator in Head and Neck Squamous Cell Carcinoma. Front Oncol 2021; 11:677051. [PMID: 34336664 PMCID: PMC8323482 DOI: 10.3389/fonc.2021.677051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/29/2021] [Indexed: 12/30/2022] Open
Abstract
Despite recent advancements, the 5 year survival of head and neck squamous cell carcinoma (HNSCC) hovers at 60%. DCLK1 has been shown to regulate epithelial-to-mesenchymal transition as well as serving as a cancer stem cell marker in colon, pancreatic and renal cancer. Although it was reported that DCLK1 is associated with poor prognosis in oropharyngeal cancers, very little is known about the molecular characterization of DCLK1 in HNSCC. In this study, we performed a comprehensive transcriptome-based computational analysis on hundreds of HNSCC patients from TCGA and GEO databases, and found that DCLK1 expression positively correlates with NOTCH signaling pathway activation. Since NOTCH signaling has a recognized role in HNSCC tumorigenesis, we next performed a series of in vitro experiments in a collection of HNSCC cell lines to investigate the role of DCLK1 in NOTCH pathway regulation. Our analyses revealed that DCLK1 inhibition, using either a pharmacological inhibitor or siRNA, resulted in substantially decreased proliferation, invasion, migration, and colony formation. Furthermore, these effects paralleled downregulation of active NOTCH1, and its downstream effectors, HEY1, HES1 and HES5, whereas overexpression of DCLK1 in normal keratinocytes, lead to an upregulation of NOTCH signaling associated with increased proliferation. Analysis of 233 primary and 40 recurrent HNSCC cancer biopsies revealed that high DCLK1 expression was associated with poor prognosis and showed a trend towards higher active NOTCH1 expression in tumors with elevated DCLK1. Our results demonstrate the novel role of DCLK1 as a regulator of NOTCH signaling network and suggest its potential as a therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Esther C. Broner
- Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Jonathan A. Trujillo
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| | | | | | - Nishant Agrawal
- Section of Otolaryngology-Head and Neck Surgery, University of Chicago, Chicago, IL, United States
| | - Ivan V. Ozerov
- InSilico Medicine Hong Kong Ltd., Pak Shek Kok, Hong Kong
| | | | - Lisa Rooper
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Nikita Kotlov
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Le Shen
- Department of Pathology, The University of Chicago Medicine, Chicago, IL, United States
| | - Alexander T. Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| | - Ari J. Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| | - Peter A. Savage
- Department of Pathology, The University of Chicago Medicine, Chicago, IL, United States
| | - Vasudha Mishra
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| | - Aditi Chatterjee
- Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - David Sidransky
- Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
20
|
Cheng L, Huang S, Chen L, Dong X, Zhang L, Wu C, Ye K, Shao F, Zhu Z, Thorne RF. Research Progress of DCLK1 Inhibitors as Cancer Therapeutics. Curr Med Chem 2021; 29:2261-2273. [PMID: 34254905 DOI: 10.2174/0929867328666210709110721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/29/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1) has emerged over the last decade as a unique stem cell marker within gastrointestinal tissues. Evidence from mouse models shows that high Dclk1 expression denotes a population of cells that promote tissue regeneration and serve as potential cancer stem cells. Moreover, since specific DCLK1 isoforms are overexpressed in many cancers and not normal cells, targeting the expression or kinase activity of DCLK1 can inhibit cancer cell growth. Here we review the evidence for DCLK1 as a prospective cancer target, including its isoform-specific expression and mutational status in human cancers. We further discuss the challenges and current progress in the development of small-molecule inhibitors of DCLK1.
Collapse
Affiliation(s)
- Linna Cheng
- Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Lijuan Chen
- Department of Medical Imaging, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Xiaoyan Dong
- Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Lei Zhang
- Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Chengye Wu
- Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Kaihong Ye
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, No.7, WeiWu Road, Zhengzhou, 450003, Henan, China
| | - Fengmin Shao
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, No.7, WeiWu Road, Zhengzhou, 450003, Henan, China
| | - Zunmin Zhu
- Institute of Hematology, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, No.7, WeiWu Road, Zhengzhou, 450003, Henan, China
| |
Collapse
|
21
|
DCLK1 isoforms and aberrant Notch signaling in the regulation of human and murine colitis. Cell Death Discov 2021; 7:169. [PMID: 34226497 PMCID: PMC8257684 DOI: 10.1038/s41420-021-00526-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 01/19/2023] Open
Abstract
Alternative promoter usage generates long and short isoforms (DCLK1-L and DCLK1-S) of doublecortin-like kinase-1 (DCLK1). Tight control of Notch signaling is important to prevent and restitute inflammation in the intestine. Our aim was to investigate whether Notch1–DCLK1 axis regulates the mucosal immune responses to infection and whether this is phenocopied in human models of colitis. In the FFPE (formalin-fixed paraffin-embedded) sections prepared from the colons of ulcerative colitis (UC) and immune-mediated colitis (IRAEC) patients, expression of DCLK1 isoforms correlated positively with Notch1 and negatively with a transcriptional repressor, FoxD3 (Forkhead Box D3). DCLK1 protein staining in these sections was predominantly sub-epithelial (stromal) wherein DCLK1 co-localized with NICD, CD68, CD11c, and neutrophil elastase (NE). NE also co-stained with Citrullinated-H3 indicating the presence of neutrophil extracellular traps. In human neutrophils, elevated levels of DCLK1-S, CXCL-10, Ly6G, MPO, NE, and Notch1/2 in LPS-treated cells were inhibited when LPS was added in conjunction with Notch blocker dibenzazepine (DBZ; LPS + DBZ group). In CR-infected Rag1−/− mice, higher levels of DCLK1 in the colonic crypts were inhibited when mice received DBZ for 10 days coincident with significant dysbiosis, barrier disruption, and colitis. Concurrently, DCLK1 immunoreactivity shifted toward the stroma in CR + DBZ mice with predominance of DCLK1-S that coincided with higher Notch1 levels. Upon antibiotic treatment, partial restoration of crypt DCLK1, reduction in MPO activity, and increased survival followed. When intestinal epithelial cell-specific Dclk1-knockout (Dclk1ΔIEC) or Dclk1ΔIEC;Rag1−/− double knockout (DKO) mice were infected with CR and given a single dose of DBZ, they developed barrier defect and severe colitis with higher levels of stromal DCLK1-S, Ly6G, NE, and Notch1. We therefore propose that, by regulating the mucosal immune responses, the Notch–DCLK1 axis may be integral to the development of murine or human colitis.
Collapse
|
22
|
Le Boiteux E, Court F, Guichet PO, Vaurs-Barrière C, Vaillant I, Chautard E, Verrelle P, Costa BM, Karayan-Tapon L, Fogli A, Arnaud P. Widespread overexpression from the four DNA hypermethylated HOX clusters in aggressive (IDHwt) glioma is associated with H3K27me3 depletion and alternative promoter usage. Mol Oncol 2021; 15:1995-2010. [PMID: 33720519 PMCID: PMC8334257 DOI: 10.1002/1878-0261.12944] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/17/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
In human, the 39 coding HOX genes and 18 referenced noncoding antisense transcripts are arranged in four genomic clusters named HOXA, B, C, and D. This highly conserved family belongs to the homeobox class of genes that encode transcription factors required for normal development. Therefore, HOX gene deregulation might contribute to the development of many cancer types. Here, we study HOX gene deregulation in adult glioma, a common type of primary brain tumor. We performed extensive molecular analysis of tumor samples, classified according to their isocitrate dehydrogenase (IDH1) gene mutation status, and of glioma stem cells. We found widespread expression of sense and antisense HOX transcripts only in aggressive (IDHwt) glioma samples, although the four HOX clusters displayed DNA hypermethylation. Integrative analysis of expression, DNA methylation, and histone modification signatures along the clusters revealed that HOX gene upregulation relies on canonical and alternative bivalent CpG island promoters that escape hypermethylation. H3K27me3 loss at these promoters emerges as the main cause of widespread HOX gene upregulation in IDHwt glioma cell lines and tumors. Our study provides the first comprehensive description of the epigenetic changes at HOX clusters and their contribution to the transcriptional changes observed in adult glioma. It also identified putative 'master' HOX proteins that might contribute to the tumorigenic potential of glioma stem cells.
Collapse
Affiliation(s)
- Elisa Le Boiteux
- CNRS, Inserm, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Franck Court
- CNRS, Inserm, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Pierre-Olivier Guichet
- INSERM-U1084, Poitiers, France.,Poitiers University, France.,Department of Cancer Biology, Poitiers Hospital, France
| | | | - Isabelle Vaillant
- CNRS, Inserm, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Pathology Department, Jean Perrin Center, Clermont-Ferrand, France.,INSERM, U1240 IMoST, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Pierre Verrelle
- CIMB, INSERM U1196 CNRS UMR9187, Curie Institute, Orsay, France.,Radiotherapy Department, Curie Institute, Paris, France.,Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucie Karayan-Tapon
- INSERM-U1084, Poitiers, France.,Poitiers University, France.,Department of Cancer Biology, Poitiers Hospital, France
| | - Anne Fogli
- CNRS, Inserm, GReD, Université Clermont Auvergne, Clermont-Ferrand, France.,Biochemistry and Molecular Biology Department, Clermont-Ferrand Hospital, France
| | - Philippe Arnaud
- CNRS, Inserm, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
23
|
Chakrapani B, Khan MIK, Kadumuri RV, Gupta S, Verma M, Awasthi S, Govindaraju G, Mahesh A, Rajavelu A, Chavali S, Dhayalan A. The uncharacterized protein FAM47E interacts with PRMT5 and regulates its functions. Life Sci Alliance 2021; 4:e202000699. [PMID: 33376131 PMCID: PMC7772775 DOI: 10.26508/lsa.202000699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) symmetrically dimethylates arginine residues in various proteins affecting diverse cellular processes such as transcriptional regulation, splicing, DNA repair, differentiation, and cell cycle. Elevated levels of PRMT5 are observed in several types of cancers and are associated with poor clinical outcomes, making PRMT5 an important diagnostic marker and/or therapeutic target for cancers. Here, using yeast two-hybrid screening, followed by immunoprecipitation and pull-down assays, we identify a previously uncharacterized protein, FAM47E, as an interaction partner of PRMT5. We report that FAM47E regulates steady-state levels of PRMT5 by affecting its stability through inhibition of its proteasomal degradation. Importantly, FAM47E enhances the chromatin association and histone methylation activity of PRMT5. The PRMT5-FAM47E interaction affects the regulation of PRMT5 target genes expression and colony-forming capacity of the cells. Taken together, we identify FAM47E as a protein regulator of PRMT5, which promotes the functions of this versatile enzyme. These findings imply that disruption of PRMT5-FAM47E interaction by small molecules might be an alternative strategy to attenuate the oncogenic function(s) of PRMT5.
Collapse
Affiliation(s)
- Baskar Chakrapani
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Mohd Imran K Khan
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Somlee Gupta
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Mamta Verma
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Sharad Awasthi
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Gayathri Govindaraju
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Arun Mahesh
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Arumugam Rajavelu
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | | |
Collapse
|
24
|
Kang XL, He LR, Chen YL, Wang SB. Role of doublecortin-like kinase 1 and leucine-rich repeat-containing G-protein-coupled receptor 5 in patients with stage II/III colorectal cancer: Cancer progression and prognosis. World J Gastroenterol 2020; 26:6853-6866. [PMID: 33268966 PMCID: PMC7684452 DOI: 10.3748/wjg.v26.i43.6853] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/29/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a subpopulation of cancer cells with the potential of self-renewal and differentiation. CSCs play critical roles in tumorigenesis, recurrence, metastasis, radiation tolerance and chemoresistance.
AIM To assess the expression patterns and clinical potential of doublecortin-like kinase 1 (DCLK1) and leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5), as prognostic CSC markers of colorectal cancer (CRC).
METHODS The expression of DCLK1 and Lgr5 in CRC tissue sections from 92 patients was determined by immunohistochemistry. Each case was evaluated using a combined scoring method based on signal intensity staining (scored 0-3) and the proportion of positively stained cancer cells (scored 0-3). The final staining score was calculated as the intensity score multiplied by the proportion score. Low expression of DCLK1 and Lgr5 was defined as a score of 0-3; high expression of DCLK1 and Lgr5 was defined as a score of ≥ 4. Specimens were categorized as either high or low expression, and the correlation between the expression of DCLK1 or Lgr5 and clinicopathological factors was investigated.
RESULTS DCLK1 and Lgr5 expression levels were significantly positively correlated. CRC patients with high DCLK1, Lgr5 and DCLK1/Lgr5 expressions had poorer progression-free survival and overall survival. Moreover, high expression of DCLK1 was an independent prognostic factor for recurrence and overall survival in patients with CRC by multivariate analysis (P = 0.026 and P = 0.049, respectively).
CONCLUSION DCLK1 may be a potential CSC marker for the recurrence and survival of CRC patients.
Collapse
Affiliation(s)
- Xue-Ling Kang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong Province, China
| | - Li-Rui He
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Yao-Li Chen
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Shu-Bin Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, China Cancer Institute of Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong Province, China
| |
Collapse
|
25
|
Sobhani I, Rotkopf H, Khazaie K. Bacteria-related changes in host DNA methylation and the risk for CRC. Gut Microbes 2020; 12:1800898. [PMID: 32931352 PMCID: PMC7575230 DOI: 10.1080/19490976.2020.1800898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer deaths in men and women combined. Colon-tumor growth is multistage and the result of the accumulation of spontaneous mutations and epigenetic events that silence tumor-suppressor genes and activate oncogenes. Environmental factors are primary contributors to these somatic gene alterations, which account for the increase in incidence of CRC in western countries. In recent decades, gut microbiota and their metabolites have been recognized as essential contributing factors to CRC, and now serve as biomarkers for the diagnosis and prognosis of CRC. In the present review, we highlight holistic approaches to understanding how gut microbiota contributes to CRC. We particularly focus herein on bacteria-related changes in host DNA methylation and the risk for CRC.
Collapse
Affiliation(s)
- Iradj Sobhani
- Head of the Department of Gastroenterology, Consultant in GI Oncology, Hopital Henri Mondor, APHP. Créteil-France; Head of the Research Team EC2M3, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Hugo Rotkopf
- Department of Gastroenterology Hospital Henri Mondor, APHP. Créteil-France; Member of Research Team EC2M3, Université Paris-Est Créteil (UPEC). Créteil, France
| | | |
Collapse
|
26
|
Razi S, Sadeghi A, Asadi-Lari Z, Tam KJ, Kalantari E, Madjd Z. DCLK1, a promising colorectal cancer stem cell marker, regulates tumor progression and invasion through miR-137 and miR-15a dependent manner. Clin Exp Med 2020; 21:139-147. [PMID: 32965580 DOI: 10.1007/s10238-020-00665-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022]
Abstract
Cancer stem cells (CSCs) are thought to be a major player in tumor initiation, progression, and metastasis. Targeting CSCs for elimination presents a promising therapeutic strategy; however, this approach will require a stronger understanding of CSC biology and identification of CSC-specific markers. The present study was conducted to examine the correlation between DCLK1 and miR-137 and miR-15a levels in colorectal cancer. A total of 222 samples, including 181 colorectal cancer specimens, 24 adenomatosis, and 17 non-adenomatosis colonic polyps, were stained for DCLK1 expression using immunohistochemistry. Also, expression of miR-137 and miR-15a was assessed in colorectal cancer with high and low DCLK1 expression levels. Most colorectal cancer specimens (76%) showed strong expression of DCLK1, whereas only 21% of adenomatous and none of non-adenomatous colonic polyps showed strong DCLK1 expression. A significant difference in DCLK1 expression was found between colorectal cancer, adenomatous, and non-adenomatous colonic polyps (P < 0.001). Higher expression of DCLK1 was more frequently detected in colorectal cases with larger tumor size (P = 0.03), poor differentiation (P = 0.03), and lymph node involvement (P = 0.04). Comparison of miR-137 and miR-15a in colorectal cancer cases revealed a significant inverse correlation with DCLK1 expression (P = 0.03 and P = 0.04, respectively). DCLK1 may act as a candidate marker for colorectal cancer stem cells. The critical role of DCLK1 in colorectal cancer suggests that it may represent an early diagnostic marker and therapeutic target; however, further investigation is warranted.
Collapse
Affiliation(s)
- Sepideh Razi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asieh Sadeghi
- Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | | | - Kevin J Tam
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran. .,Department of Molecular Medicine, Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Fan M, Yan H, Gou M, Qian N, Dai G. Divergent expression of DCLK1 in gastrointestinal neuroendocrine tumors and primary hepatic, gallbladder, and pancreatic neuroendocrine tumors. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2249-2258. [PMID: 33042329 PMCID: PMC7539880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/31/2020] [Indexed: 06/11/2023]
Abstract
Doublecortin-like kinase 1 (DCLK1) is reported to be a negative prognostic marker in colorectal cancer and is involved in tumorigenesis and progression through several miRNA pathways. In this study, We analyzed its expression in neuroendocrine tumor (NET) and explored its relation with survival outcome. 122 patients were enrolled in the study, including 60 cases of GI-NETs, 24 cases of primary hepatic NETs (PHNETs), 16 cases of gallbladder NETs (GBNETs) and 22 cases of pancreatic NETs (pNETs). IHC was performed for DCLK1 on tumor tissue. All patients underwent a baseline visit, histologic determination, and a follow-up for survival. In the 60 cases of GI-NETs, DCLK1 showed diffuse cytoplasmic expression. The positive rates of DCLK1, Syn and CgA were 100% (60/60), 100% (60/60) and 36.7% (22/60), respectively. However, DCLK1 showed negative staining in all of the 62 cases of PHNETs, GBNETs, and pNETs. The mean score of DCLK1, Syn, and CgA were (5.77±2.012), (5.13±2.078) and (2.68±2.797), respectively. DCLK1 was correlated with primary site (P<0.001) and Syn expression (P = 0.045). Additionally, in GI-NETs, we found that DCLK1 expression was associated with worse OS (log-rank = 5.212, P = 0.022). The divergent expression of DCLK1 in NETs suggests different functional roles of DCLK1 in different locations of NET within the digestive system. However, with the limited number of tumor samples, its outcome prediciton still needs further investigation. DCLK1 expression may aid in the diagnosis and prognosis of GI-NETs.
Collapse
Affiliation(s)
- Mengjiao Fan
- Oncology Department, Chinese PLA General Hospital Beijing, China
| | - Huan Yan
- Oncology Department, Chinese PLA General Hospital Beijing, China
| | - Miaomiao Gou
- Oncology Department, Chinese PLA General Hospital Beijing, China
| | - Niansong Qian
- Oncology Department, Chinese PLA General Hospital Beijing, China
| | - Guanghai Dai
- Oncology Department, Chinese PLA General Hospital Beijing, China
| |
Collapse
|
28
|
AP-TSS: A New Method for the Analysis of RNA Expression from Particular and Challenging Transcription Start Sites. Biomolecules 2020; 10:biom10060827. [PMID: 32481529 PMCID: PMC7355800 DOI: 10.3390/biom10060827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/12/2020] [Accepted: 05/21/2020] [Indexed: 11/25/2022] Open
Abstract
Alternative promoter usage involved in the regulation of transcription, splicing, and translation contributes to proteome diversity and is involved in a large number of diseases, in particular, cancer. Epigenetic mechanisms and cis regulatory elements are involved in alternative promoter activity. Multiple transcript isoforms can be produced from a gene, due to the initiation of transcription at different transcription start sites (TSS). These transcripts may not have regions that allow discrimination during RT-qPCR, making quantification technically challenging. This study presents a general method for the relative quantification of a transcript synthesized from a particular TSS that we called AP-TSS (analysis of particular TSS). AP-TSS is based on the specific elongation of the cDNA of interest, followed by its quantification by qPCR. As proof of principle, AP-TSS was applied to two non-coding RNA: telomeric repeat-containing RNAs (TERRA) from a particular subtelomeric TSS, and Alu transcripts. The treatment of cells with a DNA methylation inhibitor was associated with a global increase of the total TERRA level, but the TERRA expression from the TSS of interest did not change in HT1080 cells, and only modestly increased in HeLa cells. This result suggests that TERRA upregulation induced by global demethylation of the genome is mainly due to activation from sites other than this particular TSS. For Alu RNA, the signal obtained by AP-TSS is specific for the RNA Polymerase III-dependent Alu transcript. In summary, our method provides a tool to study regulation of gene expression from a given transcription start site, in different conditions that could be applied to many genes. In particular, AP-TSS can be used to investigate the epigenetic regulation of alternative TSS usage that is of importance for the development of epigenetic-targeted therapies.
Collapse
|
29
|
Subramaniam D, Angulo P, Ponnurangam S, Dandawate P, Ramamoorthy P, Srinivasan P, Iwakuma T, Weir SJ, Chastain K, Anant S. Suppressing STAT5 signaling affects osteosarcoma growth and stemness. Cell Death Dis 2020; 11:149. [PMID: 32094348 PMCID: PMC7039889 DOI: 10.1038/s41419-020-2335-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/26/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone tumor that primarily affects children and adolescents. Studies suggested that dysregulation JAK/STAT signaling promotes the development of OS. Cells treated with pimozide, a STAT5 inhibitor suppressed proliferation and colony formation and induced sub G0/G1 cell cycle arrest and apoptosis. There was a reduction in cyclin D1 and CDK2 expression and Rb phosphorylation, and activation of Caspase-3 and PARP cleavage. In addition, pimozide suppressed the formation of 3-dimensional osteospheres and growth of the cells in the Tumor in a Dish lung organoid system. Furthermore, there was a reduction in expression of cancer stem cell marker proteins DCLK1, CD44, CD133, Oct-4, and ABCG2. More importantly, it was the short form of DCLK1 that was upregulated in osteospheres, which was suppressed in response to pimozide. We further confirmed by flow cytometry a reduction in DCLK1+ cells. Moreover, pimozide inhibits the phosphorylation of STAT5, STAT3, and ERK in OS cells. Molecular docking studies suggest that pimozide interacts with STAT5A and STAT5B with binding energies of −8.4 and −6.4 Kcal/mol, respectively. Binding was confirmed by cellular thermal shift assay. To further understand the role of STAT5, we knocked down the two isoforms using specific siRNAs. While knockdown of the proteins did not affect the cells, knockdown of STAT5B reduced pimozide-induced necrosis and further enhanced late apoptosis. To determine the effect of pimozide on tumor growth in vivo, we administered pimozide intraperitoneally at a dose of 10 mg/kg BW every day for 21 days in mice carrying KHOS/NP tumor xenografts. Pimozide treatment significantly suppressed xenograft growth. Western blot and immunohistochemistry analyses also demonstrated significant inhibition of stem cell marker proteins. Together, these data suggest that pimozide treatment suppresses OS growth by targeting both proliferating cells and stem cells at least in part by inhibiting the STAT5 signaling pathway.
Collapse
Affiliation(s)
- Dharmalingam Subramaniam
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Pablo Angulo
- Division of Hematology and Oncology, Children's Mercy Hospital, Kansas City, MO, 64108, USA.,Banner Health, 1432S. Dobson Rd. Ste. 107, Mesa, AZ, 85202, USA
| | - Sivapriya Ponnurangam
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Prasad Dandawate
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Prabhu Ramamoorthy
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Pugazhendhi Srinivasan
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA.,Division of Hematology and Oncology, Children's Mercy Hospital, Kansas City, MO, 64108, USA
| | - Scott J Weir
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Katherine Chastain
- Division of Hematology and Oncology, Children's Mercy Hospital, Kansas City, MO, 64108, USA.,Janssen Inc, 1000 U.S. Route 202 South, Raritan, NJ, 08869, USA
| | - Shrikant Anant
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
30
|
Makino S, Takahashi H, Okuzaki D, Miyoshi N, Haraguchi N, Hata T, Matsuda C, Yamamoto H, Mizushima T, Mori M, Doki Y. DCLK1 integrates induction of TRIB3, EMT, drug resistance and poor prognosis in colorectal cancer. Carcinogenesis 2019; 41:303-312. [DOI: 10.1093/carcin/bgz157] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/20/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
Doublecortin-like kinase 1 (DCLK1) promotes tumour proliferation in human colorectal cancer (CRC). To elucidate the mechanism and clinical relevance of this association, we performed expression analysis using commercially available colon carcinoma cell lines (SW480, HCT116, CaCO2, SW48 and SKCO1) and immunohistochemical analysis of 200 resected CRC samples for correlation with clinical features. DCLK1 showed a high level of expression, especially in SW480 and HCT116 cells. Silencing DCLK1 expression using short hairpin DCLK1 (shDCLK1) RNA inhibited the growth and invasion capacities of these cell lines, which showed signs of entering into the mesenchymal–epithelial transition (MET). We found evidence of a strong correlation of DCLK1 expression with that of Tribbles homolog 3 (TRIB3), and silencing TRIB3 also led to the MET phenotype in these cells. In the clinical samples, compared with samples showing low expression of DCLK1, high expression was associated with poor prognosis in terms of overall and recurrence-free survival (P < 0.0001). The results of univariate and multivariate analysis suggested that high expression of DCLK1 in clinical colon cancer samples was tied to poor prognosis, cancer invasion depth and lymph node metastasis. DCLK1 expression correlates with malignant grade of colon cancer and offers a potential treatment target.
Collapse
Affiliation(s)
- Shunichiro Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Norikatsu Miyoshi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Naotsugu Haraguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Taishi Hata
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Chu Matsuda
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
- Department of Surgery and Science, Kyushu University Graduate School of Medicine, Maidashi, Higashi-ku, Fukuoka Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
31
|
Shafiei S, Kalantari E, Saeednejad Zanjani L, Abolhasani M, Asadi Lari MH, Madjd Z. Increased expression of DCLK1, a novel putative CSC maker, is associated with tumor aggressiveness and worse disease-specific survival in patients with bladder carcinomas. Exp Mol Pathol 2019; 108:164-172. [PMID: 31028726 DOI: 10.1016/j.yexmp.2019.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/13/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
Doublecortin-like kinase 1 (DCLK1) has been characterized as a novel potential cancer stem cell (CSC) marker in several types of cancer. It is considered as one of the most specific markers for distinguishing colorectal CSCs from normal stem cells. Yet, there are limited reports on the role of DCLK1 as a putative CSC marker in bladder cancer. Using immunohistochemistry, DCLK1 expression was examined in a well-defined tissue microarray series of 472 bladder cancer tissues. The association between DCLK1 protein expression and clinicopathological features, as well as survival outcomes, was assessed. Our findings showed strong, moderate, and weak DCLK1 expression in 123 (26.1%), 230 (48.7%), and 119 (25.2%) of the bladder cancer specimens, respectively. Higher expression of DCLK1 was significantly associated with increase in histological grade (P ≤ .001), pT stage (P = .014), lamina propria (P = .006), and lamina propria/muscularis (L/M) involvement (P = .014). On multivariate analysis, pT stage (P < .001), histological grade (P = .021), and lamina propria involvement (P = .001) were independent prognostic factors in DCLK1 expression. Moreover, the expression of DCLK1 was found to be an independent marker of poor prognosis for disease- specific survival (DSS) (P = .048) in bladder carcinomas. Our observations showed that DCLK1 expression was associated with more aggressive tumor behavior, more advanced disease, and poorer DSS in patients with bladder carcinomas. However, any potential clinical applications of DCLK1 as a novel target molecule in bladder cancer patients would require further investigations.
Collapse
Affiliation(s)
- Somayeh Shafiei
- Dep of Pathology, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| | - Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Maryam Abolhasani
- Dep of Pathology, Iran University of Medical Sciences, (IUMS), Tehran, Iran; Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Hasheminejad Kidney, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| | | | - Zahra Madjd
- Dep of Pathology, Iran University of Medical Sciences, (IUMS), Tehran, Iran; Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada..
| |
Collapse
|
32
|
Roy BC, Ahmed I, Ramalingam S, Jala V, Haribabu B, Ramamoorthy P, Ashcraft J, Valentino J, Anant S, Sampath V, Umar S. Co-localization of autophagy-related protein p62 with cancer stem cell marker dclk1 may hamper dclk1's elimination during colon cancer development and progression. Oncotarget 2019; 10:2340-2354. [PMID: 31040926 PMCID: PMC6481322 DOI: 10.18632/oncotarget.26684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/01/2019] [Indexed: 12/16/2022] Open
Abstract
Autophagy may play a critical role in colon cancer stem cells (CCSCs)-related cancer development. Here, we investigate whether accumulation of infection/injury-induced CCSCs due to impaired autophagy influences colon cancer development and progression. When Apc++ mice were infected with Citrobacter rodentium (CR; 109CFUs), we discovered presence of autophagosomes with increases in Beclin-1, LC3B and p62 staining during crypt hyperplasia. Apc1638N/+ mice when infected with CR or subjected to CR+AOM treatment, exhibited increased colon tumorigenesis with elevated levels of Ki-67, β-catenin, EZH2 and CCSC marker Dclk1, respectively. AOM/DSS treatment of Apc1638N/+ mice phenocopied CR+AOM treatment as colonic tumors exhibited pronounced changes in Ki-67, EZH2 and Dclk1 accompanied by infiltration of F4/80+ macrophages, CD3+ lymphocytes and CD3/β-catenin co-localization. Intestinal and colonic tumors also stained positive for migrating CSC markers CD110 and CDCP1 wherein, colonic tumors additionally exhibited stromal positivity. In tumors from CR-infected, CR+AOM or AOM/DSS-treated Apc1638N/+ mice and surgically-resected colon tumor/metastatic liver samples, significant accumulation of p62 and it's co-localization with LC3B and Dclk1 was evident. ApcMin/+ mice when infected with CR and BLT1−/−;ApcMin/+ mice, exhibited similar co-localization of p62 with LC3B and Dclk1 within the tumors. Studies in HCT116 and SW480 cells further confirmed p62/Dclk1 co-localization and Chloroquin/LPS-induced increases in Dclk1 promoter activity. Thus, co-localization of p62 with Dclk1 may hamper Dclk1's elimination to impact colon cancer development and progression.
Collapse
Affiliation(s)
- Badal Chandra Roy
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ishfaq Ahmed
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram, Tamil Nadu, India
| | - Venkatakrishna Jala
- James Graham Brown Cancer Center and Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Bodduluri Haribabu
- James Graham Brown Cancer Center and Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Prabhu Ramamoorthy
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - John Ashcraft
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joseph Valentino
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shrikant Anant
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Venkatesh Sampath
- Division of Neonatology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Shahid Umar
- Departments of Surgery and Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
33
|
Kim S, Trudo SP, Gallaher DD. Apiaceous and Cruciferous Vegetables Fed During the Post-Initiation Stage Reduce Colon Cancer Risk Markers in Rats. J Nutr 2019; 149:249-257. [PMID: 30649390 DOI: 10.1093/jn/nxy257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/20/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Vegetable consumption reduces colon cancer risk when fed in the initiation stage of carcinogenesis; however, the effect of vegetable consumption during the post-initiation stage has rarely been examined. OBJECTIVE We investigated the chemopreventive effects of feeding apiaceous and cruciferous vegetables on colon cancer risk in the post-initiation stage. METHODS Thirty male Wistar rats (∼5 wk, 92 g) were subcutaneously injected with 1,2-dimethylhydrazine 1 time/wk for 2 wk. One week after the last dose, rats were randomly assigned to 3 groups: the basal diet, an apiaceous vegetable-containing diet (API; 21% fresh wt/wt), or a cruciferous vegetable-containing diet (CRU; 21% fresh wt/wt). All diets contained ∼20% protein, 7% fat, and 63% digestible carbohydrate. Experimental diets were fed for 10 wk, after which colons were harvested. RESULTS CRU reduced aberrant crypt foci (ACF) number compared to the basal group (P = 0.014) and API (P = 0.013), whereas API decreased the proportion of dysplastic ACF relative to the basal group (P < 0.05). Both CRU and API reduced doublecortin-like kinase 1-positive marker expression relative to basal by 57.9% (P = 0.009) and 51.4% (P < 0.02). The numbers of CD44-positive ACF did not differ between the groups. We identified 14 differentially expressed microRNAs (miRNAs). Of these, expression of 6 miRNAs were greater or tended to be greater (P ≤ 0.10) in one or both vegetable-containing groups compared to the basal group. Bioinformatic analysis of these expression changes in miRNA predicted a change in WNT/β-catenin signaling, indicating downregulation of β-catenin in the vegetable-fed groups. Consistent with this bioinformatics analysis, β-catenin-accumulated ACF were decreased in CRU (93.1%, P = 0.012), but not in API (54.4%, P = 0.125), compared to the basal group. CONCLUSION Both apiaceous and cruciferous vegetables, fed post-initiation, reduce colonic preneoplastic lesions as well as cancer stem cell marker expression in rats, possibly by suppressing oncogenic signaling through changes in miRNA expression.
Collapse
Affiliation(s)
- Sangyub Kim
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Sabrina P Trudo
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN.,School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR
| | - Daniel D Gallaher
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| |
Collapse
|
34
|
Park SY, Kim JY, Choi JH, Kim JH, Lee CJ, Singh P, Sarkar S, Baek JH, Nam JS. Inhibition of LEF1-Mediated DCLK1 by Niclosamide Attenuates Colorectal Cancer Stemness. Clin Cancer Res 2018; 25:1415-1429. [PMID: 30446587 DOI: 10.1158/1078-0432.ccr-18-1232] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/10/2018] [Accepted: 11/13/2018] [Indexed: 01/31/2023]
Abstract
PURPOSE Niclosamide, an FDA-approved anthelmintic drug, has been characterized as a potent Wnt inhibitor that can suppress tumor growth and cancer stem-like cell (CSC) populations. However, the underlying molecular mechanisms remain poorly understood. This study aimed to examine how Wnt inhibition by niclosamide preferentially targets CSCs. EXPERIMENTAL DESIGN The mechanistic role of niclosamide in CSC inhibition was examined in public databases, human colorectal cancer cells, colorectal cancer xenografts, and azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colorectal cancer model. RESULTS Niclosamide suppresses CSC populations and their self-renewal activities in colorectal cancer cells, and this CSC-targeting effect leads to irreversible disruption of tumor-initiating potential in vivo. Mechanistically, niclosamide downregulates multiple signaling components of the Wnt pathway, specifically lymphoid enhancer-binding factor 1 (LEF1) expression, which is critical for regulating stemness. Subsequently, we identified that the doublecortin-like kinase 1 (DCLK1)-B is a target of LEF1 and upregulates cancer stemness in colorectal cancer cells. We first documented that niclosamide blocks the transcription of DCLK1-B by interrupting the binding of LEF1 to DCLK1-B promoter. DCLK1-B depletion impairs cancer stemness resulting in reduced survival potential and increased apoptosis, thus sensitizing colorectal cancer to chemoradiation. CONCLUSIONS Disruption of the LEF1/DCLK1-B axis by niclosamide eradicates cancer stemness and elicits therapeutic effects on colorectal cancer initiation, progression, and resistance. These findings provide a preclinical rationale to broaden the clinical evaluation of niclosamide for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- So-Yeon Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.,Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Ji-Young Kim
- Laboratory Animal Resource Center, Gwangju Institute of Science and Technology, Gwnagju, Republic of Korea
| | - Jang-Hyun Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jee-Heun Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Choong-Jae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Pomila Singh
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Shubhashish Sarkar
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Jeong-Heum Baek
- Division of Colon and Rectal Surgery, Department of Surgery, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Jeong-Seok Nam
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea. .,Cell Logistics Research Center, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.,Silver Health Bio Research Center, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
35
|
Dai T, Hu Y, Lv F, Ozawa T, Sun X, Huang J, Han X, Kishi H, Muraguchi A, Jin A. Analysis of the clinical significance of DCLK1 + colorectal cancer using novel monoclonal antibodies against DCLK1. Onco Targets Ther 2018; 11:5047-5057. [PMID: 30174443 PMCID: PMC6110630 DOI: 10.2147/ott.s169928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Introduction Doublecortin-like kinase 1 (DCLK1) is considered a putative tumor stem cell (TSC) marker and a promising therapeutic target, as DCLK1+ progeny cells exhibit high expression in tumors. However, the biological function of DCLK1+ cells in tumorigenesis and tumor progression remains unclear. Materials and methods We generated rabbit monoclonal antibodies (mAbs) against DCLK1, DCLK1-42, and DCLK1-87 mAbs, using a novel chip-based immunospot array assay on a chip system. First, the specificity of two mAbs to DCLK1 was confirmed by Western blot, which were bound to DCLK1-long in normal colon cells and to DCLK1-short in a cancer cell line as well as colorectal cancer (CRC) cells. Results Precise localization analysis using immunofluorescence revealed that both mAbs had cytoplasmic signal and exhibited a high degree of overlap with microtubules. Furthermore, bacterial display technology indicated that the antigenic epitope region of DCLK1-87 mAb was consistent with that of a commercial anti-DCLK1 polyclonal antibody. In addition, DCLK1-42 mAb has the common polyclonal antibody characteristic of binding to more than one site on DCLK1. By immunohistochemistry, it was found that DCLK1-87 mAb was more specific for DCLK1+ cell labeling than a commercial anti-DCLK1 polyclonal antibody. DCLK1 labeled with DCLK1-87 mAb might be a potential TSC marker because the tissue expression site covers the ALDH1 area in CRC tissues. Finally, we analyzed 100 pairs of cancer tissues and matching paracancerous tissue samples from patients with CRC who received 100 months of follow-up with the DCLK1-87 mAb. The results showed that patients with high DCLK1 expression exhibited a longer survival time than that of patients with low DCLK1 expression (P=0.0029). Discussion Our results indicated that we successfully generated an efficient tool for the precise detection of DCLK1+ cells in cancer tissues. Moreover, we found that high DCLK1 expression in CRC patients appears to play a protective role against tumor progression.
Collapse
Affiliation(s)
- Tianqi Dai
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| | - Yunlong Hu
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China, .,Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, People's Republic of China
| | - Fulian Lv
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| | - Tatsuhiko Ozawa
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Xin Sun
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| | - Jingjing Huang
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| | - Xiaojian Han
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Atsushi Muraguchi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Aishun Jin
- Department of Immunology, College of Basic Medical Sciences, Harbin Medical University, Harbin, People's Republic of China,
| |
Collapse
|
36
|
Liu W, Wang S, Sun Q, Yang Z, Liu M, Tang H. DCLK1 promotes epithelial-mesenchymal transition via the PI3K/Akt/NF-κB pathway in colorectal cancer. Int J Cancer 2018; 142:2068-2079. [PMID: 29277893 DOI: 10.1002/ijc.31232] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/06/2017] [Accepted: 12/18/2017] [Indexed: 12/29/2022]
Abstract
Double cortin-like kinase 1 (DCLK1) plays important roles during the epithelial-mesenchymal transition (EMT) process in human colorectal cancer (CRC). However, the role of DCLK1 in regulating the EMT of CRC is still poorly understood. In this study, we report evidence that DCLK1 acts as a potent oncogene to drive its extremely malignant character of EMT in an NF-κB-dependent manner in CRC cells. Mechanistic investigations showed that DCLK1 induced the NF-κBp65 subunit expression through the PI3K/Akt/Sp1 axis and activated NF-κBp65 through the PI3K/Akt/IκBα pathway during the EMT of CRC cells. Moreover, we found that silencing the expression of DCLK1 inhibited the invasion and metastasis of CRC cells in vivo. Collectively, our findings identify DCLK1 as a pivotal regulator of an EMT axis in CRC, thus implicating DCLK1 as a potential therapeutic target for CRC metastasis.
Collapse
Affiliation(s)
- Weiying Liu
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shixing Wang
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qi Sun
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhen Yang
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Min Liu
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Tang
- Tianjin Life Science Research Center and Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
Testa U, Pelosi E, Castelli G. Colorectal cancer: genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med Sci (Basel) 2018; 6:E31. [PMID: 29652830 PMCID: PMC6024750 DOI: 10.3390/medsci6020031] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/24/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
Colon cancer is the third most common cancer worldwide. Most colorectal cancer occurrences are sporadic, not related to genetic predisposition or family history; however, 20-30% of patients with colorectal cancer have a family history of colorectal cancer and 5% of these tumors arise in the setting of a Mendelian inheritance syndrome. In many patients, the development of a colorectal cancer is preceded by a benign neoplastic lesion: either an adenomatous polyp or a serrated polyp. Studies carried out in the last years have characterized the main molecular alterations occurring in colorectal cancers, showing that the tumor of each patient displays from two to eight driver mutations. The ensemble of molecular studies, including gene expression studies, has led to two proposed classifications of colorectal cancers, with the identification of four/five non-overlapping groups. The homeostasis of the rapidly renewing intestinal epithelium is ensured by few stem cells present at the level of the base of intestinal crypts. Various experimental evidence suggests that colorectal cancers may derive from the malignant transformation of intestinal stem cells or of intestinal cells that acquire stem cell properties following malignant transformation. Colon cancer stem cells seem to be involved in tumor chemoresistance, radioresistance and relapse.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
38
|
Kalantari E, Asadi Lari MH, Roudi R, Korourian A, Madjd Z. Lgr5High/DCLK1High phenotype is more common in early stage and intestinal subtypes of gastric carcinomas. Cancer Biomark 2017; 20:563-573. [DOI: 10.3233/cbm-170383] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Asadi Lari
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Korourian
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Kawamura D, Takemoto Y, Nishimoto A, Ueno K, Hosoyama T, Shirasawa B, Tanaka T, Kugimiya N, Harada E, Hamano K. Enhancement of cytotoxic effects of gemcitabine by Dclk1 inhibition through suppression of Chk1 phosphorylation in human pancreatic cancer cells. Oncol Rep 2017; 38:3238-3244. [PMID: 29048622 DOI: 10.3892/or.2017.5974] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Although gemcitabine (GEM) is frequently used in the treatment of pancreatic cancer, the effects are limited. To increase the inhibitory effect of GEM, the identification of a molecular target is needed. Recent studies have revealed that doublecortin-like kinase 1 (Dclk1) positively regulates tumor growth, invasion, metastasis, factors related to epithelial-mesenchymal transition (EMT), pluripotency, angiogenesis, and anti-apoptosis in pancreatic cancer cells. Therefore, Dclk1 is a potential therapeutic target for pancreatic cancer. However, the Dclk1-signaling pathway including its substrate proteins remains to be elucidated. To identify the candidate substrate proteins phosphorylated by Dclk1, we performed a cancer-related phosphorylated protein microarray using Dclk1-inhibited MIA Paca2 cells. Expression levels of phosphorylated cdc25A (p-cdc25A) and phosphorylated Chk1 (p-Chk1), belonging to the ATR pathway, were decreased by treatment with Dclk1 inhibitor LRRK2-IN-1 (LRRK), indicating Dclk1 involvement in the ATR pathway. Consistent with this finding, the GEM-induced p-Chk1 expression was significantly decreased by treatment with LRRK. Notably, combined treatment with GEM and LRRK allowed cell cycle progression without arresting at S phase, while individual treatment with GEM induced cell cycle arrest at S phase. In addition, combined treatment with GEM and LRRK increased the number of γ-H2AX-positive cells compared with that upon individual treatments. Moreover, LRRK alone, and combined treatment with GEM and LRRK, induced caspase-3 activation and PARP1 cleavage, in contrast to treatment with GEM alone. Finally, combined treatment with GEM and LRRK significantly reduced cell survival compared to individual treatment with GEM. These results indicate that Dclk1 inhibition in combination with GEM treatment offers a novel approach to treat pancreatic cancer cells.
Collapse
Affiliation(s)
- Daichi Kawamura
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Yoshihiro Takemoto
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Arata Nishimoto
- Department of Medical Education, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Tohru Hosoyama
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Bungo Shirasawa
- Department of Medical Education, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Toshiki Tanaka
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Naruji Kugimiya
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Eijiro Harada
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
40
|
Middelhoff M, Westphalen CB, Hayakawa Y, Yan KS, Gershon MD, Wang TC, Quante M. Dclk1-expressing tuft cells: critical modulators of the intestinal niche? Am J Physiol Gastrointest Liver Physiol 2017; 313:G285-G299. [PMID: 28684459 PMCID: PMC5668570 DOI: 10.1152/ajpgi.00073.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 01/31/2023]
Abstract
Dclk1-expressing tuft cells constitute a unique intestinal epithelial lineage that is distinct from enterocytes, Paneth cells, goblet cells, and enteroendocrine cells. Tuft cells express taste-related receptors and distinct transcription factors and interact closely with the enteric nervous system, suggesting a chemosensory cell lineage. In addition, recent work has shown that tuft cells interact closely with cells of the immune system, with a critical role in the cellular regulatory network governing responses to luminal parasites. Importantly, ablation of tuft cells severely impairs epithelial proliferation and tissue regeneration after injury, implicating tuft cells in the modulation of epithelial stem/progenitor function. Finally, tuft cells expand during chronic inflammation and in preneoplastic tissues, suggesting a possible early role in inflammation-associated tumorigenesis. Hence, we outline and discuss emerging evidence that strongly supports tuft cells as key regulatory cells in the complex network of the intestinal microenvironment.
Collapse
Affiliation(s)
- Moritz Middelhoff
- 1Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York; ,2II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany;
| | - C. Benedikt Westphalen
- 3Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany;
| | - Yoku Hayakawa
- 4Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan;
| | - Kelley S. Yan
- 1Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York; ,5Department of Genetics and Development, Columbia University Medical Center, New York, New York; and
| | - Michael D. Gershon
- 6Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Timothy C. Wang
- 1Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, New York;
| | - Michael Quante
- II. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany;
| |
Collapse
|
41
|
A novel antibody against cancer stem cell biomarker, DCLK1-S, is potentially useful for assessing colon cancer risk after screening colonoscopy. J Transl Med 2017; 97:1245-1261. [PMID: 28414327 PMCID: PMC5623180 DOI: 10.1038/labinvest.2017.40] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
DCLK1 expression is critically required for maintaining growth of human colon cancer cells (hCCCs). Human colorectal tumors (CRCs) and hCCCs express a novel short isoform of DCLK1 (DCLK1-S; isoform 2) from β-promoter of hDCLK1 gene, while normal colons express long isoform (DCLK1-L; isoform 1) from 5'(α)-promoter, suggesting that DCLK1-S, and not DCLK1-L, marks cancer stem cells (CSCs). Even though DCLK1-S differs from DCLK1-L by only six amino acids, we succeeded in generating a monospecific DCLK1-S-Antibody (PS41014), which does not cross-react with DCLK1-L, and specifically detects CSCs. Subcellular localization of S/L-isoforms was examined by immune-electron-microscopy (IEM). Surprisingly, besides plasma membrane and cytosolic fractions, S/L also localized to nuclear/mitochondrial fractions, with pronounced localization of S-isoform in the nuclei and mitochondria. Sporadic CRCs develop from adenomas. Screening colonoscopy is used for detection/resection of growths, and morphological/pathological criteria are used for risk assessment and recommendations for follow-up colonoscopy. But, these features are not precise and majority of the patients will never develop cancer. We hypothesized that antibody-based assay(s), which identify CSCs, will significantly improve prognostic value of morphological/pathological criteria. We conducted a pilot retrospective study with PS41014-Ab, by staining archived adenoma specimens from patients who developed (high-risk), or did not develop (low-risk) adenocarcinomas within 10-15 years. PS41014-Ab stained adenomas from initial and follow-up colonoscopies of high-risk patients, at significantly higher levels (three to fivefold) than adenomas from low-risk patients, suggesting that PS41014-Ab could be used as an additional tool for assessing CRC risk. CRC patients, with high DCLK1-S-expressing tumors (by qRT-PCR), were reported to have worse overall survival than low expressers. We now report that DCLK1-S-specific Ab may help to identify high-risk patients at the time of index/screening colonoscopy.
Collapse
|
42
|
Fan M, Qian N, Dai G. Expression and prognostic significance of doublecortin-like kinase 1 in patients with hepatocellular carcinoma. Oncol Lett 2017; 14:7529-7537. [PMID: 29344199 DOI: 10.3892/ol.2017.7082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/20/2017] [Indexed: 12/17/2022] Open
Abstract
Doublecortin-like kinase 1 (DCLK1), a putative cancer stem cell marker in intestinal and pancreatic tumors, is associated with tumor pathogenesis and progression, and poor survival outcomes in numerous types of cancer. However, DCLK1 expression and its prognostic value remain unclear in hepatocellular carcinoma (HCC). In the present study, the expression of DCLK1 was assessed using immunohistochemistry in 96 resected HCC and 68 adjacent tissue specimens. The staining intensity and the percentage of stained cells were scored on a scale of 0-3 and 0-4, respectively. Tissue was defined as positive for DCLK1 if the composite multiple score was >3. Cytoplasmic expression of DCLK1 was observed in HCC and adjacent tissue specimens with an expression rate of 81% (78/96) and 74% (50/68), respectively; the median score was 4.6 and 3.9, respectively, and no statistically significant difference was observed between HCC and adjacent tissues (P=0.087). DCLK1 expression was positively associated with intrahepatic metastasis (P=0.035). Furthermore, univariate analysis revealed that DCLK1 expression was significantly associated with poor disease-free survival (DFS) and overall survival (P=0.024 and 0.034). Multivariate analysis also demonstrated that DCLK1 expression was an independent prognostic factor for DFS in HCC (P=0.019; hazard ratio, 1.546; 95% confidence interval, 1.330-1.725). Stratified Kaplan-Meier survival curves revealed that DCLK1 expression predicted poorer DFS with respect to positivity for three characteristics: Portal venous metastasis, intrahepatic metastasis, and cirrhosis (P=0.020, P=0.007 and P=0.017, respectively). Collectively, the results of the present study suggested that DCLK1, functioning as a tumor promoter, is frequently overexpressed in HCC, and that DCLK1 expression is associated with poor DFS in patients with HCC. DCLK1 may represent a promising therapeutic target in HCC and requires further study.
Collapse
Affiliation(s)
- Mengjiao Fan
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Niansong Qian
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Guanghai Dai
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
43
|
Chao C, Widen SG, Wood TG, Zatarain JR, Johnson P, Gajjar A, Gomez G, Qiu S, Thompson J, Spratt H, Hellmich MR. Patient-derived Xenografts from Colorectal Carcinoma: A Temporal and Hierarchical Study of Murine Stromal Cell Replacement. Anticancer Res 2017; 37:3405-3412. [PMID: 28668828 DOI: 10.21873/anticanres.11707] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Patient-derived xenografting (PDX) of human colorectal cancer (CRC) is the preferred experimental model to study tumor response to therapeutic agents. Gradually, human stromal cells are replaced by mouse stromal cells; however, the exact timing of the replacement of human with murine stromal cells in human CRC xenograft has not been fully elucidated. We hypothesize that orthologous murine transcripts functionally substitutes for the loss due to replacement of human stromal genes. MATERIALS AND METHODS Human CRC were implanted in athymic nude mice in replicates and followed-up over time. Using next-generation sequencing, we determined the temporal kinetics of human stromal cell replacement with the orthologous murine transcripts. RESULTS CRC cell-induced re-organization of the normal, quiescent murine stromal cells into a protumorigenic phenotype supporting human CRC growth occurs at initial implantation. CONCLUSION Murine cell replacement occurs in a time- and size-dependent manner.
Collapse
Affiliation(s)
- Celia Chao
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A.
| | - Steve G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Thomas G Wood
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - John R Zatarain
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Paul Johnson
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Aakash Gajjar
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Guillermo Gomez
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Suimin Qiu
- Department of Surgical Pathology, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Jill Thompson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Heidi Spratt
- Department of Preventative Medicine and Community Health, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, U.S.A
| |
Collapse
|
44
|
Sarkar S, O'Connell MR, Okugawa Y, Lee BS, Toiyama Y, Kusunoki M, Daboval RD, Goel A, Singh P. FOXD3 Regulates CSC Marker, DCLK1-S, and Invasive Potential: Prognostic Implications in Colon Cancer. Mol Cancer Res 2017; 15:1678-1691. [PMID: 28851816 DOI: 10.1158/1541-7786.mcr-17-0287] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/26/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022]
Abstract
The 5' (α)-promoter of the human doublecortin-like kinase 1 (DCLK1) gene becomes epigenetically silenced during colon carcinogenesis, resulting in loss of expression of the canonical long(L)-isoform1 (DCLK1-L) in human colon adenocarcinomas (hCRCs). Instead, hCRCs express a short(S)-isoform2 (DCLK1-S) from an alternate (β)-promoter of DCLK1. The current study, examined if the transcriptional activity of the (β)-promoter is suppressed in normal versus cancerous cells. On the basis of in silico and molecular approaches, it was discovered that FOXD3 potently inhibits the transcriptional activity of the (β)-promoter. FOXD3 becomes methylated in human colon cancer cells (hCCC), with loss of FOXD3 expression, allowing expression of the DCLK1(S) variant in hCCCs/hCRCs. Relative levels of FOXD3/DCLK1(S/L) were measured in a cohort of CRC patient specimens (n = 92), in relation to overall survival (OS). Patients expressing high DCLK1(S), with or without low FOXD3, had significantly worse OS compared with patients expressing low DCLK1(S). The relative levels of DCLK1-L did not correlate with OS. In a pilot retrospective study, colon adenomas from high-risk patients (who developed CRCs in <15 years) demonstrated significantly higher staining for DCLK1(S) + significantly lower staining for FOXD3, compared with adenomas from low-risk patients (who remained free of CRCs). Latter results strongly suggest a prognostic value of measuring DCLK1(S)/FOXD3 in adenomas. Overexpression of DCLK1(S), but not DCLK1(L), caused a significant increase in the invasive potential of hCCCs, which may explain worse outcomes for patients with high DCLK1-S-expressing tumors. On the basis of these data, FOXD3 is a potent repressor of DCLK1-S expression in normal cells; loss of FOXD3 in hCCCs/hCRCs allows upregulation of DCLK1-S, imparting a potent invasive potential to the cells. Mol Cancer Res; 15(12); 1678-91. ©2017 AACR.
Collapse
Affiliation(s)
- Shubhashish Sarkar
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Malaney R O'Connell
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Yoshinaga Okugawa
- Gastrointestinal Cancer Research Laboratory, Division of Gastroenterology, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, Dallas, Texas.,Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie, Japan
| | - Brian S Lee
- Medical School, University of Texas Medical Branch, Galveston, Texas
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie, Japan
| | - Masato Kusunoki
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie, Japan
| | - Robert D Daboval
- Medical School, University of Texas Medical Branch, Galveston, Texas
| | - Ajay Goel
- Gastrointestinal Cancer Research Laboratory, Division of Gastroenterology, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, Dallas, Texas
| | - Pomila Singh
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
45
|
Increased DCLK1 correlates with the malignant status and poor outcome in malignant tumors: a meta-analysis. Oncotarget 2017; 8:100545-100557. [PMID: 29246000 PMCID: PMC5725042 DOI: 10.18632/oncotarget.20129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/30/2017] [Indexed: 01/04/2023] Open
Abstract
Doublecortin-like kinase 1 (DCLK1) has been found to be involved in malignant biological behavior of cancers and poor prognosis of cancer patients. The aim of this meta-analysis was to systematically clarify the relationships between expression level of DCLK1 and clinicopathological characteristics in tumors and assess its clinical value in cancer diagnosis and prognosis. 18 eligible studies with a total of 2660 patients were identified by searching the electronic bibliographic databases. Pooled results showed that DCLK1 was highly expressed in tissues from cancer patients compared to normal tissues (OR, 10.00), and overexpression of DCLK1 was significantly correlated with advanced clinical stage (OR, 2.48), positive lymph node metastasis (OR, 2.18), poorly differentiated cancers (OR, 1.83) and poor overall survival (HR, 2.15). The overall combined sensitivity and specificity for DCLK1 in distinguishing malignant tumors were 0.58 and 0.90, respectively. The mean diagnostic odds ratio was 12.70, and the corresponding area under the summary receiver operating characteristic curve was 0.78. In summary, our study indicated that DCLK1 could be a risk factor for development of malignant tumors and may serve as a promising diagnostic and prognostic biomarker for malignant tumors.
Collapse
|
46
|
Roles of Wnt Target Genes in the Journey of Cancer Stem Cells. Int J Mol Sci 2017; 18:ijms18081604. [PMID: 28757546 PMCID: PMC5577996 DOI: 10.3390/ijms18081604] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 12/12/2022] Open
Abstract
The importance of Wnt/β-catenin signaling in cancer stem cells (CSCs) has been acknowledged; however, the mechanism through which it regulates the biological function of CSCs and promotes cancer progression remains elusive. Hence, to understand the intricate mechanism by which Wnt controls stemness, the specific downstream target genes of Wnt were established by analyzing the genetic signatures of multiple types of metastatic cancers based on gene set enrichment. By focusing on the molecular function of Wnt target genes, the biological roles of Wnt were interpreted in terms of CSC dynamics from initiation to metastasis. Wnt signaling participates in cancer initiation by generating CSCs from normal stem cells or non-CSCs and augmenting persistent growth at the primary region, which is resistant to anti-cancer therapy. Moreover, it assists CSCs in invading nearby tissues and in entering the blood stream, during which the negative feedback of the Wnt signaling pathway maintains CSCs in a dormant state that is suitable for survival. When CSCs arrive at distant organs, another burst of Wnt signaling induces CSCs to succeed in re-initiation and colonization. This comprehensive understanding of Wnt target genes provides a plausible explanation for how Wnt allows CSCs variation during cancer progression.
Collapse
|
47
|
Zhang S, Zhang G, Guo H. DCAMKL1 is associated with the malignant status and poor outcome in bladder cancer. Tumour Biol 2017. [PMID: 28621231 DOI: 10.1177/1010428317703822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DCAMKL1 (doublecortin and CaM kinase-like 1) has been found to be overexpressed and function as an oncogene in several types of cancer, but there are limited reports on the role of DCAMKL1 in bladder cancer. The messenger RNA and protein expression of DCAMKL1 in bladder cancer tissues and cell lines was measured by quantitative reverse transcription polymerase chain reaction, western blotting, or immunohistochemistry. The correlation between DCAMKL1 protein expression and clinicopathological characteristics was analyzed. Univariate and multivariate Cox regression models were adopted to evaluate prognostic significance of DCAMKL1 in bladder cancer patients. In our results, DCAMKL1 messenger RNA and protein were overexpressed in bladder cancer tissues compared with adjacent normal tissues. DCAMKL1 protein overexpression was positively associated with clinical stage, muscularis invasion, lymph node metastasis, and distant metastasis. The univariate and multivariate analyses suggested DCAMKL1 protein overexpression was an unfavorable prognostic factor in bladder cancer patients. In conclusion, DCAMKL1 is an independent poor prognostic factor for bladder cancer patients.
Collapse
Affiliation(s)
- Shiqing Zhang
- 1 Department of Urology, Jining No. 1 People's Hospital, Jining, China
| | - Guoqing Zhang
- 2 Department of Planning Immunization, Center for Disease Control and Prevention, Jining, China
| | - Hongbo Guo
- 1 Department of Urology, Jining No. 1 People's Hospital, Jining, China
| |
Collapse
|
48
|
Ikezono Y, Koga H, Akiba J, Abe M, Yoshida T, Wada F, Nakamura T, Iwamoto H, Masuda A, Sakaue T, Yano H, Tsuruta O, Torimura T. Pancreatic Neuroendocrine Tumors and EMT Behavior Are Driven by the CSC Marker DCLK1. Mol Cancer Res 2017; 15:744-752. [PMID: 28179411 DOI: 10.1158/1541-7786.mcr-16-0285] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/22/2016] [Accepted: 01/08/2017] [Indexed: 11/16/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1), a marker for intestinal and pancreatic cancer stem cells, is highly expressed in neuroblastomas. This study was conducted to assess DCLK1 expression levels in pancreatic neuroendocrine tumor (PNET) tissues and to explore the roles of this molecule in clinical tissue from multiple PNET patients, cells (BON1, QGP1, and CM) and tumor xenografts. Immunohistochemically, all PNET tissues highly and diffusely expressed DCLK1 as a full-length isoform, identical to that detected in primary liver NETs. A DCLK1-overexpressing PNET cell line (QGP1-DCLK1) exhibited epithelial-mesenchymal transition (EMT)-related gene signatures, and robust upregulation of Slug (SNAI2), N-Cadherin (CDH2), and Vimentin (VIM) was validated by real-time PCR and immunoblotting. QGP1-DCLK1 cells had increased cell migration in a wound-healing assay and formed significantly larger xenograft tumors in nude mice. The factors involved in the formation of the fast-growing tumors included p-FAK (on Tyr925), p-ERK1/2, p-AKT, Paxillin, and Cyclin D1, which upon knockdown or pharmacologic inhibition of DCLK1 abolished the expression of these molecules. In conclusion, robust and ubiquitous expression of DCLK1 was first demonstrated here in human PNET tissue specimens and cells. DCLK1 characterized the PNET cell behavior, inducing p-FAK/SLUG-mediated EMT. These findings suggest the possibility of developing novel therapeutic strategies against PNETs by targeting DCLK1.Implications: Evidence here reveals that human PNETs diffusely and robustly express the cancer stem cell marker DCLK1, which drives SLUG-mediated EMT, and suggests that NETs share biological features for druggable targets with other tumors, including neuroblastoma that also highly expresses DCLK1. Mol Cancer Res; 15(6); 744-52. ©2017 AACR.
Collapse
Affiliation(s)
- Yu Ikezono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan. .,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Mitsuhiko Abe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takafumi Yoshida
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Fumitaka Wada
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Osamu Tsuruta
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| |
Collapse
|
49
|
Babaian A, Mager DL. Endogenous retroviral promoter exaptation in human cancer. Mob DNA 2016; 7:24. [PMID: 27980689 PMCID: PMC5134097 DOI: 10.1186/s13100-016-0080-x] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022] Open
Abstract
Cancer arises from a series of genetic and epigenetic changes, which result in abnormal expression or mutational activation of oncogenes, as well as suppression/inactivation of tumor suppressor genes. Aberrant expression of coding genes or long non-coding RNAs (lncRNAs) with oncogenic properties can be caused by translocations, gene amplifications, point mutations or other less characterized mechanisms. One such mechanism is the inappropriate usage of normally dormant, tissue-restricted or cryptic enhancers or promoters that serve to drive oncogenic gene expression. Dispersed across the human genome, endogenous retroviruses (ERVs) provide an enormous reservoir of autonomous gene regulatory modules, some of which have been co-opted by the host during evolution to play important roles in normal regulation of genes and gene networks. This review focuses on the “dark side” of such ERV regulatory capacity. Specifically, we discuss a growing number of examples of normally dormant or epigenetically repressed ERVs that have been harnessed to drive oncogenes in human cancer, a process we term onco-exaptation, and we propose potential mechanisms that may underlie this phenomenon.
Collapse
Affiliation(s)
- Artem Babaian
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z1L3 Canada ; Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| | - Dixie L Mager
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z1L3 Canada ; Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
50
|
Peng S, Hu GQ. Colorectal cancer stem cells. Shijie Huaren Xiaohua Zazhi 2016; 24:3953-3962. [DOI: 10.11569/wcjd.v24.i28.3953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive system, and its initiation, promotion and prognosis are closely related to cancer stem cells (CSCs). CSCs are defined as a minority population of cancer cells with self-renewal ability, multi-lineage differentiation potential and highly aggressive behaviors, which have been identified in many types of cancers including CRC as one of the key mediators driving cancer metastasis and progression. The presence of these CSCs can be attributed to the failure of cancer treatments as these cells are believed to exhibit therapy resistance. Here, we review the current understanding of colorectal CSCs, with an emphasis on candidate markers, biological properties, related signaling pathways, and clinical applications.
Collapse
|