1
|
Kusakabe Y, Matsumoto K, Tsuyuki T, Hayashi Y, Watanabe H. Baicalin target protein, Annexin A2, is a target of new antitumor drugs. Sci Rep 2024; 14:21814. [PMID: 39294172 PMCID: PMC11410801 DOI: 10.1038/s41598-024-68528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/24/2024] [Indexed: 09/20/2024] Open
Abstract
Baicalin is a flavonoid extracted from Scutellaria baicalensis Georgi. As it has significant antitumor and apoptosis-inducing effects, baicalin may be useful as a lead compound in new antitumor drug development. However, as the pharmacological actions of baicalin have yet to be elucidated, we isolated its target protein, which was successfully identified as Annexin A2. Annexin A2 forms a heterotetramer with S100A10 protein, which plays an important role in the plasminogen activator system. The heterotetramer bound to tissue plasminogen activator (tPA) activates the conversion of plasminogen to plasmin and promotes the expression of STAT-3 and NF-κB, which are target genes involved in the development of cancer. Moreover, NF-κB and STAT-3 induce the expression of cell inhibitors of apoptotic proteins and inhibit apoptosis. To examine whether these antitumor and apoptosis-inducing effects of baicalin are mediated by Annexin A2, we prepared Annexin A2 knockdown HepG2 cells. We compared mRNA expression by RT-qPCR and apoptosis by caspase-3 activity assays in Annexin A2 knockdown HepG2 cells. The results showed that the antitumor and apoptosis-inducing effects of baicalin are mediated by Annexin A2. The results of this study suggest that agents capable of inhibiting Annexin A2 may be useful candidates for the development of novel antitumor agents.
Collapse
Affiliation(s)
| | | | | | | | - Hideaki Watanabe
- Department of Dermatology, Showa University Northern Yokohama Hospital, Chigasakichuo, Tsuduki-ku, Yokohama City, Kanagawa, Japan.
| |
Collapse
|
2
|
Pei F, Tao Z, Lu Q, Fang T, Peng S. Octamer-binding transcription factor 4-positive circulating tumor cell predicts worse treatment response and survival in advanced cholangiocarcinoma patients who receive immune checkpoint inhibitors treatment. World J Surg Oncol 2024; 22:110. [PMID: 38664770 PMCID: PMC11044354 DOI: 10.1186/s12957-024-03369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/28/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Octamer-binding transcription factor 4-positive circulating tumor cell (OCT4+CTC) exhibits high stemness and invasive potential, which may influence the efficacy of immune checkpoint inhibitors (ICI). This study aimed to assess the prognostic role of OCT4+CTC in advanced cholangiocarcinoma (CCA) patients who received ICI treatment. METHODS In total, 40 advanced CCA patients who received ICI treatment were included, and CTC and OCT4 counts were detected via a Canpatrol system and an RNA in situ hybridization method before ICI treatment. Patients were subsequently divided into none CTC, OCT4-CTC, and OCT4+CTC groups. Patients were followed up for a median of 10.4 months. RESULTS The percentages of patients in none CTC, OCT4-CTC, and OCT4+CTC groups were 25.0%, 30.0%, and 45.0%, respectively. The proportion of patients with lymph node metastasis was highest in OCT4+CTC group, followed by none CTC group, and lowest in OCT4-CTC group (P = 0.025). The objective response rate (ORR) was lowest in OCT4+CTC group, moderate in OCT4-CTC group, and highest in none CTC group (P = 0.009), while disease control rate was not different among three groups (P = 0.293). In addition, progression-free survival (PFS) (P < 0.001) and overall survival (OS) (P = 0.001) were shorter in the OCT4+CTC group than in none CTC & OCT4-CTC group. Moreover, OCT4+CTC (versus none CTC) was independently linked with poorer PFS [hazard ratio (HR) = 6.752, P = 0.001] and OS (HR = 6.674, P = 0.003) in advanced CCA patients. CONCLUSION OCT4+CTC relates to lymph node metastasis and shows a good predictive value for poor treatment response and survival in advanced CCA patients who receive ICI treatment.
Collapse
Affiliation(s)
- Fei Pei
- Department of Hepatobiliary Pancreatic Surgery, Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No. 141 Tianjin Road, Huangshi, 435200, Hubei, China
| | - Zhen Tao
- Department of Hepatobiliary Pancreatic Surgery, Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No. 141 Tianjin Road, Huangshi, 435200, Hubei, China.
| | - Qi Lu
- Department of Hepatobiliary Pancreatic Surgery, Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No. 141 Tianjin Road, Huangshi, 435200, Hubei, China
| | - Tao Fang
- Department of Hepatobiliary Pancreatic Surgery, Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No. 141 Tianjin Road, Huangshi, 435200, Hubei, China
| | - Shasha Peng
- Department of Hepatobiliary Pancreatic Surgery, Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No. 141 Tianjin Road, Huangshi, 435200, Hubei, China
| |
Collapse
|
3
|
Gholamzad A, Khakpour N, Khosroshahi EM, Asadi S, Koohpar ZK, Matinahmadi A, Jebali A, Rashidi M, Hashemi M, Sadi FH, Gholamzad M. Cancer stem cells: The important role of CD markers, Signaling pathways, and MicroRNAs. Pathol Res Pract 2024; 256:155227. [PMID: 38490099 DOI: 10.1016/j.prp.2024.155227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/17/2024]
Abstract
For the first time, a subset of small cancer cells identified in acute myeloid leukemia has been termed Cancer Stem Cells (CSCs). These cells are notorious for their robust proliferation, self-renewal abilities, significant tumor-forming potential, spread, and resistance to treatments. CSCs are a global concern, as it found in numerous types of cancer, posing a real-world challenge today. Our review encompasses research on key CSC markers, signaling pathways, and MicroRNA in three types of cancer: breast, colon, and liver. These factors play a critical role in either promoting or inhibiting cancer cell growth. The reviewed studies have shown that as cells undergo malignant transformation, there can be an increase or decrease in the expression of different Cluster of Differentiation (CD) markers on their surface. Furthermore, alterations in essential signaling pathways, such as Wnt and Notch1, may impact CSC proliferation, survival, and movement, while also providing potential targets for cancer therapies. Additionally, some research has focused on MicroRNAs due to their dual role as potential therapeutic biomarkers and their ability to enhance CSCs' response to anti-cancer drugs. MicroRNAs also regulate a wide array of cellular processes, including the self-renewal and pluripotency of CSCs, and influence gene transcription. Thus, these studies indicate that MicroRNAs play a significant role in the malignancy of various tumors. Although the gathered information suggests that specific CSC markers, signaling pathways, and MicroRNAs are influential in determining the destiny of cancer cells and could be advantageous for therapeutic strategies, their precise roles and impacts remain incompletely defined, necessitating further investigation.
Collapse
Affiliation(s)
- Amir Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Niloofar Khakpour
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch,Islamic Azad University, Tonekabon, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus,Torun,Poland
| | - Ali Jebali
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Deprtment of Medical Nanotechnology,Faculty of Advanced Sciences and Technology,Tehran Medical Sciences,Islamic Azad University, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | | | - Mehrdad Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Quartieri M, Puspitasari A, Vitacchio T, Durante M, Tinganelli W. The role of hypoxia and radiation in developing a CTCs-like phenotype in murine osteosarcoma cells. Front Cell Dev Biol 2023; 11:1222809. [PMID: 38033871 PMCID: PMC10687637 DOI: 10.3389/fcell.2023.1222809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction: Cancer treatment has evolved significantly, yet concerns about tumor recurrence and metastasis persist. Within the dynamic tumor microenvironment, a subpopulation of mesenchymal tumor cells, known as Circulating Cancer Stem Cells (CCSCs), express markers like CD133, TrkB, and CD47, making them radioresistant and pivotal to metastasis. Hypoxia intensifies their stemness, complicating their identification in the bloodstream. This study investigates the interplay of acute and chronic hypoxia and radiation exposure in selecting and characterizing cells with a CCSC-like phenotype. Methods: LM8 murine osteosarcoma cells were cultured and subjected to normoxic (21% O2) and hypoxic (1% O2) conditions. We employed Sphere Formation and Migration Assays, Western Blot analysis, CD133 Cell Sorting, and CD133+ Fluorescent Activated Cell Sorting (FACS) analysis with a focus on TrkB antibody to assess the effects of acute and chronic hypoxia, along with radiation exposure. Results: Our findings demonstrate that the combination of radiation and acute hypoxia enhances stemness, while chronic hypoxia imparts a cancer stem-like phenotype in murine osteosarcoma cells, marked by increased migration and upregulation of CCSC markers, particularly TrkB and CD47. These insights offer a comprehensive understanding of the interactions between radiation, hypoxia, and cellular responses in the context of cancer treatment. Discussion: This study elucidates the complex interplay among radiation, hypoxia, and cellular responses, offering valuable insights into the intricacies and potential advancements in cancer treatment.
Collapse
Affiliation(s)
- Martina Quartieri
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Anggraeini Puspitasari
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Biology Division, Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Tamara Vitacchio
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Institut für Festkörperphysik, Technische Universität Darmstadt, Darmstadt, Germany
| | - Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| |
Collapse
|
5
|
Pu Y, Li L, Peng H, Liu L, Heymann D, Robert C, Vallette F, Shen S. Drug-tolerant persister cells in cancer: the cutting edges and future directions. Nat Rev Clin Oncol 2023; 20:799-813. [PMID: 37749382 DOI: 10.1038/s41571-023-00815-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
Drug-tolerant persister (DTP) cell populations were originally discovered in antibiotic-resistant bacterial biofilms. Similar populations with comparable features have since been identified among cancer cells and have been linked with treatment resistance that lacks an underlying genomic alteration. Research over the past decade has improved our understanding of the biological roles of DTP cells in cancer, although clinical knowledge of the role of these cells in treatment resistance remains limited. Nonetheless, targeting this population is anticipated to provide new treatment opportunities. In this Perspective, we aim to provide a clear definition of the DTP phenotype, discuss the underlying characteristics of these cells, their biomarkers and vulnerabilities, and encourage further research on DTP cells that might improve our understanding and enable the development of more effective anticancer therapies.
Collapse
Affiliation(s)
- Yi Pu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Lung Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Haoning Peng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - François Vallette
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France.
- Nantes Université, INSERM, U1307, CRCI2NA, Nantes, France.
| | - Shensi Shen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Einloth KR, Gayfield S, McMaster T, Didier A, Dworkin L, Creeden JF. The application, safety, and future of ex vivo immune cell therapies and prognosis in different malignancies. BIOIMPACTS : BI 2023; 13:439-455. [PMID: 38022382 PMCID: PMC10676524 DOI: 10.34172/bi.2023.27521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 12/01/2023]
Abstract
Introduction Immunotherapy has revolutionized how cancer is treated. Many of these immunotherapies rely on ex vivo expansion of immune cells, classically T cells. Still, several immunological obstacles remain, including tumor impermeability by immune cells and the immunosuppressive nature of the tumor microenvironment (TME). Logistically, high costs of treatment and variable clinical responses have also plagued traditional T cell-based immunotherapies. Methods To review the existing literature on cellular immunotherapy, the PubMed database was searched for publications using variations of the phrases "cancer immunotherapy", "ex vivo expansion", and "adoptive cell therapy". The Clinicaltrials.gov database was searched for clinical trials related to ex vivo cellular therapies using the same phrases. The National Comprehensive Cancer Network guidelines for cancer treatment were also referenced. Results To circumvent the challenges of traditional T cell-based immunotherapies, researchers have developed newer therapies including tumor infiltrating lymphocyte (TIL), chimeric antigen receptor (CAR), T cell receptor (TCR) modified T cell, and antibody-armed T cell therapies. Additionally, newer immunotherapeutic strategies have used other immune cells, including natural killer (NK) and dendritic cells (DC), to modulate the T cell immune response to cancers. From a prognostic perspective, circulating tumor cells (CTC) have been used to predict cancer morbidity and mortality. Conclusion This review highlights the mechanism and clinical utility of various types of ex vivo cellular therapies in the treatment of cancer. Comparing these therapies or using them in combination may lead to more individualized and less toxic chemotherapeutics.
Collapse
Affiliation(s)
- Katelyn R. Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Scott Gayfield
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Thomas McMaster
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Alexander Didier
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
7
|
Kwizera EA, Ou W, Lee S, Stewart S, Shamul JG, Xu J, Tait N, Tkaczuk KHR, He X. Greatly Enhanced CTC Culture Enabled by Capturing CTC Heterogeneity Using a PEGylated PDMS-Titanium-Gold Electromicrofluidic Device with Glutathione-Controlled Gentle Cell Release. ACS NANO 2022; 16:11374-11391. [PMID: 35797466 PMCID: PMC9649890 DOI: 10.1021/acsnano.2c05195] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The circulating tumor cells (CTCs, the root cause of cancer metastasis and poor cancer prognosis) are very difficult to culture for scale-up in vitro, which has hampered their use in cancer research/prognosis and patient-specific therapeutic development. Herein, we report a robust electromicrofluidic chip for not only efficient capture of heterogeneous (EpCAM+ and CD44+) CTCs with high purity but also glutathione-controlled gentle release of the CTCs with high efficiency and viability. This is enabled by coating the polydimethylsiloxane (PDMS) surface in the device with a 10 nm gold layer through a 4 nm titanium coupling layer, for convenient PEGylation and linkage of capture antibodies via the thiol-gold chemistry. Surprisingly, the percentage of EpCAM+ mammary CTCs can be as low as ∼35% (∼70% on average), showing that the commonly used approach of capturing CTCs with EpCAM alone may miss many EpCAM- CTCs. Furthermore, the CD44+ CTCs can be cultured to form 3D spheroids efficiently for scale-up. In contrast, the CTCs captured with EpCAM alone are poor in proliferation in vitro, consistent with the literature. By capture of the CTC heterogeneity, the percentage of stage IV patients whose CTCs can be successfully cultured/scaled up is improved from 12.5% to 68.8%. These findings demonstrate that the common practice of CTC capture with EpCAM alone misses the CTC heterogeneity including the critical CD44+ CTCs. This study may be valuable to the procurement and scale-up of heterogeneous CTCs, to facilitate the understanding of cancer metastasis and the development of cancer metastasis-targeted personalized cancer therapies conveniently via the minimally invasive liquid/blood biopsy.
Collapse
Affiliation(s)
- Elyahb A Kwizera
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Sojeong Lee
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Nancy Tait
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Katherine H R Tkaczuk
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
8
|
Khan SU, Fatima K, Malik F. Understanding the cell survival mechanism of anoikis-resistant cancer cells during different steps of metastasis. Clin Exp Metastasis 2022; 39:715-726. [PMID: 35829806 DOI: 10.1007/s10585-022-10172-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
Anchorage-independent survival of cancer cells is associated with metastasis as it enables cells to travel to secondary target sites. Tissue integrity is generally maintained by detachment-induced cell death called 'anoikis', but cancer cells undergoing the multistep metastatic process show resistance to anoikis. Anoikis resistance enables these cells to survive through the extracellular matrix (ECM) deprived phase, which starts when cancer cells detach and move into the circulation till cells reach to the secondary target site. Comprehensive analysis of the molecular and functional biology of anoikis resistance in cancer cells will provide crucial details about cancer metastasis, enabling us to identify novel therapeutic targets against cancer cell dissemination and ultimately secondary tumor formation. This review broadly summarizes recent advances in the understanding of cellular and molecular events leading to anoikis and anoikis resistance. It further elaborates more about the signaling cross-talk in anoikis resistance and its regulation during metastasis.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India
| | - Kaneez Fatima
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India
| | - Fayaz Malik
- Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, 190005, Srinagar, Jammu and Kashmir, India.
| |
Collapse
|
9
|
Zhou Y, Qiu S, Kim JT, Lee SB, Park HJ, Son MJ, Lee HJ, Chen J. Garcinone C Suppresses Tumorsphere Formation and Invasiveness by Hedgehog/Gli1 Signaling in Colorectal Cancer Stem-like Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7941-7952. [PMID: 35749593 DOI: 10.1021/acs.jafc.2c01891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hyperactivation of hedgehog signaling occurs in colorectal cancer stem-like cells (CSCs), a rare subpopulation, potentially involved in metastasis, chemotherapy resistance, and cancer relapse. Garcinone C, a xanthone isolated from mangosteen (Garcinia mangostana), suppresses colorectal cancer in vivo and in vitro by inhibiting Gli1-dependent noncanonical hedgehog signaling. Herein, we investigated the effect of garcinone C on cancer stemness and invasiveness in colorectal cancer; Gli1 was noted as pivotal in maintaining stemness and invasiveness in HCT116 and HT29 CSCs. Garcinone C inhibited the proliferation and self-renewal of HCT116 and HT29 CSCs. Colon cancer stemness markers such as CD44, CD133, ALDH1, and Nanog were significantly decreased by garcinone C. Computational studies showed that garcinone C showed a high affinity with the Gli1 protein ZF domain by forming hydrogen bonds with amino acid residues of ASP244, ARG223, and ASP216. Besides, MG132 blocked the effects of garcinone C on Gli1. Thus, garcinone C suppressed colorectal CSCs by binding to Gli1 and enhancing its degradation. MMP2 and MMP9 levels, invasive-related markers, were increased in HCT116 CSCs but decreased by garcinone C. E-cadherin level was reduced in HCT116 CSCs, while the presence of garcinone C was restored. Garcinone C inhibited the proliferation and invasiveness of colorectal CSCs by targeting Gli1-dependent Hh signaling. Garcinone C may be a potent natural agent against colorectal cancer relapse.
Collapse
Affiliation(s)
- Yimeng Zhou
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Shuai Qiu
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Seung Beom Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Ho Jin Park
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Moon Jeong Son
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou 510632, China
| |
Collapse
|
10
|
Wu M, Huang Y, Zhou Y, Zhao H, Lan Y, Yu Z, Jia C, Cong H, Zhao J. The Discovery of Novel Circulating Cancer-Related Cells in Circulation Poses New Challenges to Microfluidic Devices for Enrichment and Detection. SMALL METHODS 2022; 6:e2200226. [PMID: 35595707 DOI: 10.1002/smtd.202200226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/19/2022] [Indexed: 06/15/2023]
Abstract
Circulating tumor cells (CTCs) enumeration has been widely used as a surrogate predictive marker for early diagnoses, the evaluation of chemotherapy efficacy, and cancer prognosis. Microfluidic technologies for CTCs enrichment and detection have been developed and commercialized as automation platforms. Currently, in addition to CTCs, some new types of circulating cancer-related cells (e.g., CCSCs, CTECs, CAMLs, and heterotypic CTC clusters) in circulation are also reported to be correlated to cancer diagnosis, metastasis, or prognosis. And they widely differ from the conventional CTCs in positive markers, cellular morphology, or size, which presents a new technological challenge to microfluidic devices that use affinity-based capture methods or size-based filtration methods for CTCs detection. This review focuses on the biological and physical properties as well as clinical significance of the novel circulating cancer-related cells, and discusses the challenges of their discovery to microfluidic chip for enrichment. Finally, the current challenges of CTCs detection in clinical application and future opportunities are also discussed.
Collapse
Affiliation(s)
- Man Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuhang Huang
- Shanghai Normal University, Shanghai, 200030, China
| | - Yang Zhou
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuwei Lan
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhibin Yu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunping Jia
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
11
|
Pizon M, Schott D, Pachmann U, Schobert R, Pizon M, Wozniak M, Bobinski R, Pachmann K. Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (cCSCs) in Breast Cancer Patients. Cancers (Basel) 2022; 14:cancers14061476. [PMID: 35326627 PMCID: PMC8946779 DOI: 10.3390/cancers14061476] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Circulating cancer cells—and in particular their very rare subpopulation, circulating cancer stem cells (cCSCs)—are responsible for recurrence and metastasis. In this study, we present a novel process in which patient-derived xenograft (PDX) can be harvested on chorioallantoic membrane (CAM) from circulating cancer stem cells. In our opinion, the CAM-based PDX model using circulating cancer stem cells can provide a fast, low-cost, easy-to-use, and efficient preclinical platform for drug screening, therapy optimization, and biomarker discovery. Abstract Background: cCSCs are a small subset of circulating tumor cells with cancer stem cell features: resistance to cancer treatments and the capacity for generating metastases. PDX are an appreciated tool in oncology, providing biologically meaningful models of many cancer types, and potential platforms for the development of precision oncology approaches. Commonly, mouse models are used for the in vivo assessment of potential new therapeutic targets in cancers. However, animal models are costly and time consuming. An attractive alternative to such animal experiments is the chicken chorioallantoic membrane assay. Methods: In this study, primary cultures from cCSCs were established using the sphere-forming assay. Subsequently, tumorspheres were transplanted onto the CAM membrane of fertilized chicken eggs to form secondary microtumors. Results: We have developed an innovative in vitro platform for cultivation of cCSCs from peripheral blood of cancer patients. The number of tumorspheres increased significantly with tumor progression and aggressiveness of primary tumor. The number of tumorspheres was positively correlated with Ki-67, Her2 status, and grade score in primary breast tumors. The grafting of tumorspheres onto the CAM was successful and positively correlated with aggressiveness and proliferation capacity of the primary tumor. These tumors pathologically closely resembled the primary tumor. Conclusions: The number of tumorspheres cultured from peripheral blood and the success rate of establishing PDX directly reflect the aggressiveness and proliferation capacity of the primary tumor. A CAM-based PDX model using cCSC provides a fast, low-cost, easy to handle, and powerful preclinical platform for drug screening, therapy optimization, and biomarker discovery.
Collapse
Affiliation(s)
- Monika Pizon
- Department of Research and Development, Transfusion Center Bayreuth, 95448 Bayreuth, Germany; (D.S.); (U.P.); (K.P.)
- Correspondence:
| | - Dorothea Schott
- Department of Research and Development, Transfusion Center Bayreuth, 95448 Bayreuth, Germany; (D.S.); (U.P.); (K.P.)
| | - Ulrich Pachmann
- Department of Research and Development, Transfusion Center Bayreuth, 95448 Bayreuth, Germany; (D.S.); (U.P.); (K.P.)
| | - Rainer Schobert
- Department of Organic Chemistry, University of Bayreuth, 95440 Bayreuth, Germany;
| | - Marek Pizon
- Department of Cardiac Surgery, Clinic of Bayreuth, 95455 Bayreuth, Germany;
| | - Marta Wozniak
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Rafal Bobinski
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland;
| | - Katharina Pachmann
- Department of Research and Development, Transfusion Center Bayreuth, 95448 Bayreuth, Germany; (D.S.); (U.P.); (K.P.)
| |
Collapse
|
12
|
Thymoquinone Radiosensitizes Human Colorectal Cancer Cells in 2D and 3D Culture Models. Cancers (Basel) 2022; 14:cancers14061363. [PMID: 35326517 PMCID: PMC8945905 DOI: 10.3390/cancers14061363] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Resistance of cancer cells and normal tissue toxicity of ionizing radiation (IR) are known to limit the success of radiotherapy. There is growing interest in using IR with natural compounds to sensitize cancer cells and spare healthy tissues. Thymoquinone (TQ) was shown to radiosensitize several cancers, yet no studies have investigated its radiosensitizing effects on colorectal cancer (CRC). Here, we combined TQ with IR and determined its effects in two-dimensional (2D) and three-dimensional (3D) culture models derived from HCT116 and HT29 CRC cells, and in patient-derived organoids (PDOs). TQ sensitized CRC cells to IR and reduced cell viability and clonogenic survival and was non-toxic to non-tumorigenic intestinal cells. TQ sensitizing effects were associated with G2/M arrest and DNA damage as well as changes in key signaling molecules involved in this process. Combining a low dose of TQ (3 µM) with IR (2 Gy) inhibited sphere formation by 100% at generation 5 and this was associated with inhibition of stemness and DNA repair. These doses also led to ~1.4- to ~3.4-fold decrease in organoid forming ability of PDOs. Our findings show that combining TQ and IR could be a promising therapeutic strategy for eradicating CRC cells.
Collapse
|
13
|
Tieng FYF, Abu N, Nasir SN, Lee LH, Ab Mutalib NS. Liquid Biopsy-Based Colorectal Cancer Screening via Surface Markers of Circulating Tumor Cells. Diagnostics (Basel) 2021; 11:2136. [PMID: 34829483 PMCID: PMC8618170 DOI: 10.3390/diagnostics11112136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is ranked second for cancer-related deaths worldwide with approximately half of the patients being diagnosed at the late stages. The untimely detection of CRC results in advancement to the metastatic stage and nearly 90% of cancer-related deaths. The early detection of CRC is crucial to decrease its overall incidence and mortality rates. The recent introduction of circulating tumor cells (CTCs) has enabled a less invasive sampling method from liquid biopsies, besides revealing key information toward CRC metastasis. The current gold standard for CTC identification is the CellSearch® system (Veridex). This first-generation instrumentation relies on a single cell surface marker (CSM) to capture and count CTCs. Detection of CTCs allows the identification of patients at risk for metastasis, whereas CTC enumeration could improve risk assessment, monitoring of systemic therapy, and detection of therapy resistance in advanced metastatic CRC. In this review, we compared the pros and cons between single CSM-based CTC enrichment techniques and multi-marker-based systems. We also highlighted the challenges faced in the routine implementation of CSM-dependent CTC detection methods in CRC screening, prediction, prognosis, disease monitoring, and therapy selection toward precision medicine, as well as the dwelling on post-CTC analysis and characterization methods.
Collapse
Affiliation(s)
- Francis Yew Fu Tieng
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Siti Nurmi Nasir
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University of Malaysia, Subang Jaya 47500, Selangor, Malaysia
| | - Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University of Malaysia, Subang Jaya 47500, Selangor, Malaysia
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
14
|
Kalra RS, Soman GS, Parab PB, Mali AM, Varankar SS, Naik RR, Kamble SC, Dhanjal JK, Bapat SA. A monoclonal antibody against annexin A2 targets stem and progenitor cell fractions in tumors. Transl Oncol 2021; 15:101257. [PMID: 34715620 PMCID: PMC8564672 DOI: 10.1016/j.tranon.2021.101257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Development of a novel antibody (termed as mAb150) developed in our lab which targets annexin A2. Although there are earlier reports of another monoclonal antibody with the same target, the epitope recognized by mAb150 is novel. mAb150 is specifically recognized to target the achilles heel of cancer viz. cancer stem cells and progenitors that persist after treatments and potentially give rise to minimal residual disease.
The involvement of cancer stem cells (CSCs) in driving tumor dormancy and drug resistance is well established. Most therapeutic regimens however are ineffective in targeting these regenerative populations. We report the development and evaluation of a monoclonal antibody, mAb150, which targets the metastasis associated antigen, Annexin A2 (AnxA2) through recognition of a N-terminal epitope. Treatment with mAb150 potentiated re-entry of CSCs into the cell cycle that perturbed tumor dormancy and facilitated targeting of CSCs as was validated by in vitro and in vivo assays. Epigenetic potentiation further improved mAb150 efficacy in achieving total tumor regression by targeting regenerative populations to achieve tumor regression, specifically in high-grade serous ovarian adenocarcinoma.
Collapse
Affiliation(s)
- Rajkumar S Kalra
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - Gaurav S Soman
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Pradeep B Parab
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Avinash M Mali
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Sagar S Varankar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Wellcome-MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge, CB2 0AW
| | - Rutika R Naik
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India
| | - Swapnil C Kamble
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Department of Technology, Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India
| | - Jaspreet K Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India
| | - Sharmila A Bapat
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
15
|
Schuster E, Taftaf R, Reduzzi C, Albert MK, Romero-Calvo I, Liu H. Better together: circulating tumor cell clustering in metastatic cancer. Trends Cancer 2021; 7:1020-1032. [PMID: 34481763 PMCID: PMC8541931 DOI: 10.1016/j.trecan.2021.07.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/30/2023]
Abstract
Circulating tumor cells (CTCs) are vital components of liquid biopsies for diagnosis of residual cancer, monitoring of therapy response, and prognosis of recurrence. Scientific dogma focuses on metastasis mediated by single CTCs, but advancement of CTC detection technologies has elucidated multicellular CTC clusters, which are associated with unfavorable clinical outcomes and a 20- to 100-fold greater metastatic potential than single CTCs. While the mechanistic understanding of CTC cluster formation is still in its infancy, multiple cell adhesion molecules and tight junction proteins have been identified that underlie the outperforming attributes of homotypic and heterotypic CTC clusters, such as cell survival, cancer stemness, and immune evasion. Future directions include high-resolution characterization of CTCs at multiomic levels for diagnostic/prognostic evaluations and targeted therapies.
Collapse
Affiliation(s)
- Emma Schuster
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rokana Taftaf
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Carolina Reduzzi
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mary K Albert
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Isabel Romero-Calvo
- Biomedical Visualization Graduate Program, Department of Biomedical and Health Information Sciences, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Lurie Comprehensive Cancer Center and Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
16
|
Taftaf R, Liu X, Singh S, Jia Y, Dashzeveg NK, Hoffmann AD, El-Shennawy L, Ramos EK, Adorno-Cruz V, Schuster EJ, Scholten D, Patel D, Zhang Y, Davis AA, Reduzzi C, Cao Y, D'Amico P, Shen Y, Cristofanilli M, Muller WA, Varadan V, Liu H. ICAM1 initiates CTC cluster formation and trans-endothelial migration in lung metastasis of breast cancer. Nat Commun 2021; 12:4867. [PMID: 34381029 PMCID: PMC8358026 DOI: 10.1038/s41467-021-25189-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cell (CTC) clusters mediate metastasis at a higher efficiency and are associated with lower overall survival in breast cancer compared to single cells. Combining single-cell RNA sequencing and protein analyses, here we report the profiles of primary tumor cells and lung metastases of triple-negative breast cancer (TNBC). ICAM1 expression increases by 200-fold in the lung metastases of three TNBC patient-derived xenografts (PDXs). Depletion of ICAM1 abrogates lung colonization of TNBC cells by inhibiting homotypic tumor cell-tumor cell cluster formation. Machine learning-based algorithms and mutagenesis analyses identify ICAM1 regions responsible for homophilic ICAM1-ICAM1 interactions, thereby directing homotypic tumor cell clustering, as well as heterotypic tumor-endothelial adhesion for trans-endothelial migration. Moreover, ICAM1 promotes metastasis by activating cellular pathways related to cell cycle and stemness. Finally, blocking ICAM1 interactions significantly inhibits CTC cluster formation, tumor cell transendothelial migration, and lung metastasis. Therefore, ICAM1 can serve as a novel therapeutic target for metastasis initiation of TNBC. Circulating tumor cell (CTC) clusters are more efficient at mediating metastasis as compared to single cells and are associated with poor prognosis in breast cancer. Here, the authors show that ICAM1 is enriched in CTC clusters and its loss suppresses cell-cell interaction and CTC cluster formation, and propose ICAM1 as a therapeutic target for treating breast cancer metastasis.
Collapse
Affiliation(s)
- Rokana Taftaf
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xia Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Salendra Singh
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Yuzhi Jia
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nurmaa K Dashzeveg
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew D Hoffmann
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lamiaa El-Shennawy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Erika K Ramos
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Valery Adorno-Cruz
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma J Schuster
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David Scholten
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dhwani Patel
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Youbin Zhang
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew A Davis
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Carolina Reduzzi
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yue Cao
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX, USA
| | - Paolo D'Amico
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yang Shen
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX, USA
| | - Massimo Cristofanilli
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - William A Muller
- Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Vinay Varadan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
17
|
Cho HY, Choi JH, Lim J, Lee SN, Choi JW. Microfluidic Chip-Based Cancer Diagnosis and Prediction of Relapse by Detecting Circulating Tumor Cells and Circulating Cancer Stem Cells. Cancers (Basel) 2021; 13:1385. [PMID: 33803846 PMCID: PMC8003176 DOI: 10.3390/cancers13061385] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Detecting circulating tumor cells (CTCs) has been considered one of the best biomarkers in liquid biopsy for early diagnosis and prognosis monitoring in cancer. A major challenge of using CTCs is detecting extremely low-concentrated targets in the presence of high noise factors such as serum and hematopoietic cells. This review provides a selective overview of the recent progress in the design of microfluidic devices with optical sensing tools and their application in the detection and analysis of CTCs and their small malignant subset, circulating cancer stem cells (CCSCs). Moreover, discussion of novel strategies to analyze the differentiation of circulating cancer stem cells will contribute to an understanding of metastatic cancer, which can help clinicians to make a better assessment. We believe that the topic discussed in this review can provide brief guideline for the development of microfluidic-based optical biosensors in cancer prognosis monitoring and clinical applications.
Collapse
Affiliation(s)
- Hyeon-Yeol Cho
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea;
- Interdisciplinary Program for Bio-health Convergence, Kookmin University, Seoul 02707, Korea
| | - Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
- School of Chemical Engineering, Jeonbuk National University, Jeonju 54896, Korea
| | - Joungpyo Lim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| | - Sang-Nam Lee
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| |
Collapse
|
18
|
Du L, Cheng Q, Zheng H, Liu J, Liu L, Chen Q. Targeting stemness of cancer stem cells to fight colorectal cancers. Semin Cancer Biol 2021; 82:150-161. [PMID: 33631296 DOI: 10.1016/j.semcancer.2021.02.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/12/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Cancer initiating/ stem cells (CSCs) undergo self-renewal and differentiation that contributes to tumor initiation, recurrence and metastasis in colorectal cancer (CRC). Targeting of colorectal cancer stem cells (CCSCs) holds significant promise in eradicating cancer cells and ultimately curing patients with cancer. In this review, we will introduce the current progress of CCSC studies, including the specific surface markers of CCSCs, the intrinsic signaling pathways that regulate the stemness and differentiation characteristics of CCSCs, and the tumor organoid model for CCSC research. We will focus on how these studies will lead to the progress in targeting specific surface markers or signaling pathways on CCSCs by monoclonal antibodies, or by natural or synthetic compounds, or by immunotherapy. As CSCs are highly heterogeneous and plastic, we suggest that combinatory approaches that target the stemness network may represent an important strategy for eradicating cancers.
Collapse
Affiliation(s)
- Lei Du
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine. Beijing, 100101, China.
| | - Qi Cheng
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The Graduate University of Chinese Academy of Sciences. Beijing, 100049, China
| | - Hao Zheng
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jinming Liu
- The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lei Liu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine. Beijing, 100101, China
| | - Quan Chen
- The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
19
|
Fang G, Wu Y, Zhang X. Changes of Kinesin Family Member 18 Pathway in Epithelial Mesenchymal Transformation and Effect of Butyrate on Migration and Invasion of Colon Cancer Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Colon cancer is a common digestive system disease with an increasing incidence. Severe migration and invasion aggravates the deterioration of colon cancer patients. Previous studies have found that epithelial mesenchymal transition (EMT) is closely associated with early transference
of colon carcinoma and abnormal changes occur in KIF18 signaling pathway. Butyrate protects colonic mucosa with a considerable effect on the colon. This study predicts that butyrate may reverse EMT process of colon cancer cells through KIF18 signaling pathway, thereby inhibiting cell migration
and invasion. In this experiment, EMT model of colon cancer was used to investigate migration and invasion. Human colon cancer cell line SW1116 was cultured and assigned into control group (0 mmol/L butyrate), low concentration group (2 mmol/L), medium concentration group (4 mmol/L), and high
concentration group (10 mmol/L). After 72 hours, cell migration and invasion was analyzed by Transwell assays. E-cadherin, Vimentin, and KIF18 level was detected by Western blot and quantitative real-time PCR. After treatment, cell migration and invasion was significantly inhibited compared
to control dose-dependently. In addition, Vimentin and KIF18 mRNA level was significantly lower and E-cadherin mRNA was higher in treatment groups than control group in a dose-dependent manner (P < 0.05). Consistently, the profile of protein level of these molecules was similar to
mRNA expression profile. Electron microscope showed that after treatment with butyrate, the surface protuberances of colon cancer cells were abnormally increased, especially the vesicular protuberances, which were the microvilli of intestinal mucosal epithelium. In conclusion, KIF18 is crucial
in EMT of colon cancer cells. Butyrate may elevate E-cadherin and suppress Vimentin and KIF18 by activating KIF18 signaling, thus inhibiting invasion and migration.
Collapse
Affiliation(s)
- Guojiu Fang
- Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Yibin Wu
- Liver Surgery Department, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xueli Zhang
- Department of General Surgery, Shanghai Fengxian Central Hospital, Shanghai, 201400, China
| |
Collapse
|
20
|
Exploration of Circulating Tumour Cell (CTC) Biology: A Paradigm Shift in Liquid Biopsy. Indian J Clin Biochem 2020; 36:131-142. [PMID: 33867703 PMCID: PMC7994460 DOI: 10.1007/s12291-020-00923-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/01/2020] [Indexed: 01/07/2023]
Abstract
Circulating tumour cells (CTCs), are disseminated tumour cells found in the blood in solid tumour malignancies. Identification of CTCs act as emerging tools in the field of the Liquid Biopsy. Majority of the studies focused on detection and enumeration of CTCs due to technological challenges those results from the rarity of CTCs in the blood. Enumeration of CTCs has already proven their value as prognostic as well as predictive biomarkers for disease prognosis. However, recent advances in technology permitted to study the molecular and functional features of CTCs and these features have the potential to change the diagnostic, prognostic and predictive landscape in oncology. In this review, we summarize the paradigm shift in the field of liquid biopsy-based cancer diagnostics using CTC isolation and detection. We have discussed recent advances in the technologies for molecular characterization of CTCs which have aided a shift from CTC enumeration to an in-depth analysis of the CTC genome, transcriptomes, proteins, epigenomes along with various functional features. Finally, as a prognosticating strategy, the potentials of CTCs as a tool of liquid biopsy to predict micrometastasis, monitor prognosis and how to use them as an additional tool for cancer staging has been discussed.
Collapse
|
21
|
Papaccio F. Circulating cancer stem cells: an interesting niche to explore. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:253-258. [PMID: 36046778 PMCID: PMC9400729 DOI: 10.37349/etat.2020.00016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy
| |
Collapse
|
22
|
Xiao L, Jin H, Duan W, Hou Y. Roles of N-terminal Annexin A2 phosphorylation sites and miR-206 in colonic adenocarcinoma. Life Sci 2020; 253:117740. [PMID: 32376265 DOI: 10.1016/j.lfs.2020.117740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
AIMS Annexin A2 (ANXA2) is closely associated with tumor malignancy and its N-terminus includes a vital domain for its function. The aims are to explore the correlation between the sites (Tyr23, Ser1, Ser11 and Ser25) in the domain and its roles. MAIN METHODS We re-expressed ANXA2 with mutated sites in ANXA2-deleted human colonic adenocarcinoma cell line caco2 (ANXA2-/-caco2). A series of analyses were used to determine the correlation of each site with ANXA2 activation, cell malignancy enhancement and motility-associated microstructural development. Bioinformatics and luciferase reporter assays were employed to validate ANXA2-targeted miRNAs. KEY FINDINGS The in vitro results showed that all single and multiple mutations of the ANXA2 N-terminal sites inhibited ANXA2 phosphorylation at different levels and subsequently inhibited the proliferation, motility, and polymerization of F-actin and β-tubulin in caco2 cells. Motility-associated microstructures were significantly remodeled when these sites were mutated. The forced expression of miR-206 significantly suppressed the proliferation, motility and epithelial-mesenchymal transition (EMT) of caco2 cells. The in vivo results showed that all the ANXA2 N-terminal site mutations and forced expression of miR-206 significantly inhibited tumor growth. Overall, this study demonstrated that the sites of the ANXA2 N-terminus, especially Tyr23, play crucial roles in maintaining the high malignancy of colonic adenocarcinoma. Furthermore, miR-206 targets ANXA2 and plays a role as a cancer suppressor in colonic adenocarcinoma. SIGNIFICANCE Our study provided evidence that further elucidates the molecular mechanism of ANXA2 and its roles in colonic adenocarcinoma and suggested potential targets of ANXA2 for colonic adenocarcinoma therapy by using miR-206 as a novel strategy.
Collapse
Affiliation(s)
- Li Xiao
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China
| | - Huijuan Jin
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
23
|
Shifat-E-Rabbi M, Yin X, Fitzgerald CE, Rohde GK. Cell Image Classification: A Comparative Overview. Cytometry A 2020; 97:347-362. [PMID: 32040260 DOI: 10.1002/cyto.a.23984] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/18/2019] [Accepted: 01/18/2020] [Indexed: 12/13/2022]
Abstract
Cell image classification methods are currently being used in numerous applications in cell biology and medicine. Applications include understanding the effects of genes and drugs in screening experiments, understanding the role and subcellular localization of different proteins, as well as diagnosis and prognosis of cancer from images acquired using cytological and histological techniques. The article also reviews three main approaches for cell image classification most often used: numerical feature extraction, end-to-end classification with neural networks (NNs), and transport-based morphometry (TBM). In addition, we provide comparisons on four different cell imaging datasets to highlight the relative strength of each method. The results computed using four publicly available datasets show that numerical features tend to carry the best discriminative information for most of the classification tasks. Results also show that NN-based methods produce state-of-the-art results in the dataset that contains a relatively large number of training samples. Data augmentation or the choice of a more recently reported architecture does not necessarily improve the classification performance of NNs in the datasets with limited number of training samples. If understanding and visualization are desired aspects, TBM methods can offer the ability to invert classification functions, and thus can aid in the interpretation of results. These and other comparison outcomes are discussed with the aim of clarifying the advantages and disadvantages of each method. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Mohammad Shifat-E-Rabbi
- Imaging and Data Science Lab, Charlottesville, Virginia, 22903
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Xuwang Yin
- Imaging and Data Science Lab, Charlottesville, Virginia, 22903
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Cailey E Fitzgerald
- Imaging and Data Science Lab, Charlottesville, Virginia, 22903
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
| | - Gustavo K Rohde
- Imaging and Data Science Lab, Charlottesville, Virginia, 22903
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22903
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia, 22903
| |
Collapse
|
24
|
Li L, Mei H, Commey ANA. Application of RNA-sequencing to identify transcriptome modification by DCLK1 in colorectal cancer cells. Cancer Gene Ther 2019; 27:691-701. [PMID: 31636360 PMCID: PMC7170768 DOI: 10.1038/s41417-019-0144-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/02/2022]
Abstract
Doublecortin like kinase 1 (DCLK1) is a cancer stem cell marker for the colorectal cancer (CRC). It plays critical roles in the oncogenesis, progression and metastasis of CRC. DCLK1 can be an intriguing therapeutic target for CRC treatment. However, the molecular mechanism of how DCLK1 functions is unclear currently. In our research, we aim to apply RNA-Sequencing (RNA Seq) technology, a high throughput massively Next Generation Sequencing approach, to monitor transcriptome changes due to DCLK1 over-expression in the CRC cells. In order to achieve our goal, RNA from quadruplicate samples from two clones of isogenic DCLK1 stable over-expression cells and the parental wild type HCT116 cells was sent for RNA Seq on the Illumina NextSeq500 platform. Differentially expressed (DE) genes were evaluated by t-test (P <0.05 and fold-change ±1.5 or greater) using two methods: (1) FWER; and (2) Benjamani and Hochberg FDR (false discovery rate) which corrects for multiple comparisons. Gene networks and functional analysis were evaluated using Ingenuity Pathways Analysis (IPA). We identified 1463 DE genes common for both DCLK1 overexpression clone A and clone B cells. IPA results indicated that 72 canonical pathways were significantly modified by DCLK1 over-expression (P<0.05), among which 9 out of the top 10 pathways are involved in the cell cycle regulation, indicating that DCLK1 might play its tumorigenesis role via activation of pathways facilitating cell proliferation, repression of pathways inhibiting cells proliferation and function against pathways facilitating cell apoptosis. Cell cycle analysis results confirmed the IPA findings, which demonstrated that DCLK1 over-expression cells had much less G0/G1 cells but much more S and G2/M cells (P<0.05). In conclusion, DCLK1 over-expression significantly modified transcriptome profile of CRC cancer cells. Control of the cell cycle regulation might be one of the critical mechanism for DCLK1 function. Our findings provide more direct evidence for the development of DCLK1 as a therapeutic target for CRC treatment, and will be of great benefit for the discovery of novel therapeutic target within the DCLK1 molecular network for the treatment of colorectal cancer patients.
Collapse
Affiliation(s)
- Lianna Li
- Biology Department, Tougaloo College, 500 West County Line Road, Tougaloo, MS, 39174, USA.
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | | |
Collapse
|
25
|
He H, Xiao L, Cheng S, Yang Q, Li J, Hou Y, Song F, Su X, Jin H, Liu Z, Dong J, Zuo R, Song X, Wang Y, Zhang K, Duan W, Hou Y. Annexin A2 Enhances the Progression of Colorectal Cancer and Hepatocarcinoma via Cytoskeleton Structural Rearrangements. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2019; 25:950-960. [PMID: 31172894 DOI: 10.1017/s1431927619000679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Annexin A2 (ANXA2) is reported to be associated with cancer development. To investigate the roles ANXA2 plays during the development of cancer, the RNAi method was used to inhibit the ANXA2 expression in caco2 (human colorectal cancer cell line) and SMMC7721 (human hepatocarcinoma cell line) cells. The results showed that when the expression of ANXA2 was efficiently inhibited, the growth and motility of both cell lines were significantly decreased, and the development of the motility relevant microstructures, such as pseudopodia, filopodia, and the polymerization of microfilaments and microtubules were obviously inhibited. The cancer cell apoptosis was enhanced without obvious significance. The possible regulating pathway in the process was also predicted and discussed. Our results suggested that ANXA2 plays important roles in maintaining the malignancy of colorectal and hepatic cancer by enhancing the cell proliferation, motility, and development of the motility associated microstructures of cancer cells based on a possible complicated signal pathway.
Collapse
Affiliation(s)
- Huimin He
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Li Xiao
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Sinan Cheng
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Qian Yang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Jinmei Li
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Yifan Hou
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Fengying Song
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Xiaorong Su
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Huijuan Jin
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Zheng Liu
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Jing Dong
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Ruiye Zuo
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Xigui Song
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Yanyan Wang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Kun Zhang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Wei Duan
- School of Medicine, Deakin University,Waurn Ponds, VIC 3216,Australia
| | - Yingchun Hou
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| |
Collapse
|
26
|
Feng Y, Li Y, Li L, Wang X, Chen Z. Identification of specific modules and significant genes associated with colon cancer by weighted gene co‑expression network analysis. Mol Med Rep 2019; 20:693-700. [PMID: 31180534 DOI: 10.3892/mmr.2019.10295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/12/2019] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is one of the most commonly diagnosed malignancies and is a leading cause of cancer‑associated mortality. The aim of the present study was to investigate the molecular mechanisms underlying colon cancer and identify potentially significant genes associated with the disease using weighted gene co‑expression network analysis (WGCNA). The test datasets used were downloaded from The Cancer Genome Atlas (TCGA) database. WGCNA was applied to analyze microarray data obtained from colon adenocarcinoma samples to identify significant modules and highly associated genes. A gene co‑expression network was constructed and different gene modules were selected. Functional and pathway enrichment analyses were performed to investigate the molecular mechanisms of colon cancer. In addition, highly connected hub genes associated with the most significant module were selected for further analysis. Nine specific modules associated with colon cancer were identified, of which the turquoise module was observed to exhibit the greatest association with the disease. Pathway enrichment analysis of the turquoise module suggested that genes in the turquoise module were associated with 'RNA polymerase' and 'purine metabolism'. Furthermore, gene ontology enrichment analysis revealed the top 30 hub genes with a higher degree in the turquoise module, such as σ‑non‑opioid intracellular receptor 1, transmembrane protein 147 TMEM147) and carbamoyl‑phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase, were predominantly enriched in the biological processes 'translation' and 'gene expression'. Experimental verification demonstrated that the expression of TMEM147 in colon cancer was significantly increased compared with the control. Therefore, the results suggested that genes associated with RNA polymerase and the purine metabolic pathways may be substantially involved in the pathogenesis of colon cancer. Furthermore, TMEM147 may represent a biomarker for colon cancer.
Collapse
Affiliation(s)
- Ye Feng
- Department of Gastrointestinal Colorectal and Anal Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yanbo Li
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lin Li
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuefeng Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhi Chen
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
27
|
Li L, Jones K, Mei H. Doublecotin-Like Kinase 1 Increases Chemoresistance of Colorectal Cancer Cells through the Anti-Apoptosis Pathway. JOURNAL OF STEM CELL RESEARCH & THERAPY 2019; 9. [PMID: 31372308 DOI: 10.4172/2157-7633.1000447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Colorectal Cancer (CRC) is the third most common cancer diagnosed and the second leading cause of cancer-related deaths in the United States. Cancer Stem Cells (CSCs) are believed to be the primary reason for the recurrence of CRC. Specific stem cell marker, doublecortin-like kinase 1 (DCLK1) plays critical roles in the tumorigenesis and progression of CRC. Up-regulation of DCLK1 is correlated with poor prognosis. Whether DCLK1 is correlated with enhanced chemoresistance of CRC cells is unclear. We aim to reveal the association of DCLK1 with chemoresistance of CRC cells and the underlying molecular mechanisms. Methods Stable DCLK1 over-expression cells (DCLK1+) were established using the HCT116 cells (WT). DCLK1+ and WT cells were treated with 5-Fluorouracil (5-Fu) at different doses for 24 or 48 hours. MTT assay was used to evaluate cell viability and IC50 of 5-Fu was determined. Quantitative real-time PCR was applied to determine the gene expression of caspase-3 (casp-3), casp-4, and casp-10. Cleaved casp-3 expression was investigated using Western blot and immunofluorescence. Results Our results demonstrated that IC50 of 5-Fu for the DCLK1+ cells was significantly higher than that of the WT cells for both 24 and 48-hour treatment (p=0.002 and 0.048 respectively), indicating increased chemoresistance of the DCLK1+ cells. Gene expression of casp-3, casp-4, and casp-10 were significantly inhibited in the DCLK1+ cells after 5-Fu treatment compared to the WT cells (p=7.616e-08, 1.575e-05 and 5.307e-08, respectively). Cleaved casp-3 amount and casp-3 positive cells were significantly decreased in the DCLK1+ cells after 5-Fu treatment compared to the WT cells (p=0.015). Conclusions In conclusion, our results demonstrated that DCLK1 overexpression enhanced the chemoresistance of CRC cells to 5-Fu treatment by suppressing gene expression of key caspases in the apoptosis pathway and activation of the apoptosis pathway. DCLK1 can be an intriguing therapeutic target for the effective treatment of CRC patients.
Collapse
Affiliation(s)
- Lianna Li
- Biology Department, Tougaloo College, Tougaloo, USA
| | - Kierra Jones
- Biology Department, Tougaloo College, Tougaloo, USA
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, USA
| |
Collapse
|
28
|
Chang PH, Wu MH, Liu SY, Wang HM, Huang WK, Liao CT, Yen TC, Ng SH, Chen JS, Lin YC, Lin HC, Hsieh JCH. The Prognostic Roles of Pretreatment Circulating Tumor Cells, Circulating Cancer Stem-Like Cells, and Programmed Cell Death-1 Expression on Peripheral Lymphocytes in Patients with Initially Unresectable, Recurrent or Metastatic Head and Neck Cancer: An Exploratory Study of Three Biomarkers in One-time Blood Drawing. Cancers (Basel) 2019; 11:cancers11040540. [PMID: 30991692 PMCID: PMC6521270 DOI: 10.3390/cancers11040540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/21/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022] Open
Abstract
Circulating tumor cells (CTCs) and immune status are strongly related to cancer prognosis, although few studies have examined both factors. This prospective observational study (ClinicalTrials.gov: NCT02420600) evaluated whether CTCs, circulating cancer stem-like cells (cCSCs), and peripheral lymphocytes with/without Programmed cell death protein 1 (PD-1) expression were associated with prognosis among patients receiving palliative chemotherapy for initially unresectable, recurrent/metastatic head and neck squamous cell carcinoma (rmHNSCC). Thirty-four patients were enrolled between January 2015 and June 2016. Overall survival (OS) was associated with a higher CTC number (hazard ratio [HR]: 1.01, p = 0.0004) and cCSC ratio (HR: 29.903, p < 0.0001). Progression-free survival (PFS) was also associated with CTC number (HR: 1.013, p = 0.002) and cCSC ratio (HR: 10.92, p = 0.003). A CD8+ proportion of ≥ 17% was associated with improved OS (HR: 0.242, p = 0.004). A CD4: CD8 ratio of >1.2 was associated with poorer trend of PFS (HR: 2.12, p = 0.064). PD-1 expression was not associated with survival outcomes. Baseline CTCs, cCSC ratio, and CD8+ ratio may predict prognosis in rmHNSCC.
Collapse
Affiliation(s)
- Pei-Hung Chang
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
- Cancer Center, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Min-Hsien Wu
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
- Graduate Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan 333, Taiwan.
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| | - Sen-Yu Liu
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Hung-Ming Wang
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Wen-Kuan Huang
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Department of Oncology⁻Pathology, Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska, Karolinska University Hospital, SE-17176 Stockholm, Sweden.
| | - Chun-Ta Liao
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
| | - Tzu-Chen Yen
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Molecular Imaging Center, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
- Department of Nuclear Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
| | - Shu-Hang Ng
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Department of Diagnostic Radiology, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
- Department of Medical Imaging and Radiological Sciences, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
| | - Jen-Shi Chen
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Yung-Chang Lin
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Hung-Chih Lin
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| | - Jason Chia-Hsun Hsieh
- Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
- Circulating Tumor Cell Lab, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkuo 333, Taiwan.
| |
Collapse
|
29
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
30
|
Perumal V, Corica T, Dharmarajan AM, Sun Z, Dhaliwal SS, Dass CR, Dass J. Circulating Tumour Cells (CTC), Head and Neck Cancer and Radiotherapy; Future Perspectives. Cancers (Basel) 2019; 11:E367. [PMID: 30875950 PMCID: PMC6468366 DOI: 10.3390/cancers11030367] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023] Open
Abstract
Head and neck cancer is the seventh most common cancer in Australia and globally. Despite the current improved treatment modalities, there is still up to 50⁻60% local regional recurrence and or distant metastasis. High-resolution medical imaging technologies such as PET/CT and MRI do not currently detect the early spread of tumour cells, thus limiting the potential for effective minimal residual detection and early diagnosis. Circulating tumour cells (CTCs) are a rare subset of cells that escape from the primary tumour and enter into the bloodstream to form metastatic deposits or even re-establish themselves in the primary site of the cancer. These cells are more aggressive and accumulate gene alterations by somatic mutations that are the same or even greater than the primary tumour because of additional features acquired in the circulation. The potential application of CTC in clinical use is to acquire a liquid biopsy, by taking a reliable minimally invasive venous blood sample, for cell genotyping during radiotherapy treatment to monitor the decline in CTC detectability, and mutational changes in response to radiation resistance and radiation sensitivity. Currently, very little has been published on radiation therapy, CTC, and circulating cancer stem cells (CCSCs). The prognostic value of CTC in cancer management and personalised medicine for head and neck cancer radiotherapy patients requires a deeper understanding at the cellular level, along with other advanced technologies. With this goal, this review summarises the current research of head and neck cancer CTC, CCSC and the molecular targets for personalised radiotherapy response.
Collapse
Affiliation(s)
- Vanathi Perumal
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Tammy Corica
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
| | - Arun M Dharmarajan
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Zhonghua Sun
- Discipline of Medical Radiation Sciences, School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA 6102, Australia.
| | - Satvinder S Dhaliwal
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia.
| | - Crispin R Dass
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Joshua Dass
- Radiation Oncology, Sir Charles Gairdner Hospital, Cancer Centre, Nedlands, Perth, WA 6009, Australia.
| |
Collapse
|
31
|
van Schaijik B, Wickremesekera AC, Mantamadiotis T, Kaye AH, Tan ST, Stylli SS, Itinteang T. Circulating tumor stem cells and glioblastoma: A review. J Clin Neurosci 2019; 61:5-9. [PMID: 30622004 DOI: 10.1016/j.jocn.2018.12.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 12/22/2018] [Indexed: 12/26/2022]
Abstract
Glioblastoma (GB) is the most aggressive primary brain tumor in adults. The aggressive nature of GB has been attributed to the presence of cancer stem cells (CSCs) which drive tumorigenesis and are thought to be the root cause of the disease. Circulating tumor stem cells (CTSCs), which can be derived from CSCs, have been identified in numerous types of cancer including GB, have been proposed to contribute to local and distant recurrence. There are many technical difficulties in studying CTSCs, therefore there is a significant gap in the literature pertaining to how they arise and function, and how the understanding of the biology of CTSCs could elucidate the underlying cause of local recurrence and metastasis. An initial epithelial-to-mesenchymal transition (EMT) followed by mesenchymal-to-epithelial transition involving these primitive cells appear to be the critical processes underpinning metastasis. This review focuses on the association between CSCs undergoing EMT to become CTSCs, and how this could arise from the CSC subpopulation in GB, and contribute to the understanding of the pathogenesis and treatment.
Collapse
Affiliation(s)
| | - Agadha C Wickremesekera
- Gillies McIndoe Research Institute, Wellington, New Zealand; Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Theo Mantamadiotis
- Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Melbourne, Victoria 3000, Australia; Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand; Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Wellington, New Zealand.
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | | |
Collapse
|
32
|
Recent advances and perspectives on capture and concentration of label-free rare cells for biomedical science and engineering research. J Taiwan Inst Chem Eng 2018. [DOI: 10.1016/j.jtice.2018.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Cho HY, Hossain MK, Lee JH, Han J, Lee HJ, Kim KJ, Kim JH, Lee KB, Choi JW. Selective isolation and noninvasive analysis of circulating cancer stem cells through Raman imaging. Biosens Bioelectron 2018; 102:372-382. [DOI: 10.1016/j.bios.2017.11.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 01/06/2023]
|
34
|
Annexin A2-mediated cancer progression and therapeutic resistance in nasopharyngeal carcinoma. J Biomed Sci 2018; 25:30. [PMID: 29598816 PMCID: PMC5877395 DOI: 10.1186/s12929-018-0430-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/20/2018] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck cancer with poor clinical outcomes and insufficient treatments in Southeast Asian populations. Although concurrent chemoradiotherapy has improved recovery rates of patients, poor overall survival and low efficacy are still critical problems. To improve the therapeutic efficacy, we focused on a tumor-associated protein called Annexin A2 (ANXA2). This review summarizes the mechanisms by which ANXA2 promotes cancer progression (e.g., proliferation, migration, the epithelial-mesenchymal transition, invasion, and cancer stem cell formation) and therapeutic resistance (e.g., radiotherapy, chemotherapy, and immunotherapy). These mechanisms gave us a deeper understanding of the molecular aspects of cancer progression, and further provided us with a great opportunity to overcome therapeutic resistance of NPC and other cancers with high ANXA2 expression by developing this prospective ANXA2-targeted therapy.
Collapse
|
35
|
Correnti M, Raggi C. Stem-like plasticity and heterogeneity of circulating tumor cells: current status and prospect challenges in liver cancer. Oncotarget 2018; 8:7094-7115. [PMID: 27738343 PMCID: PMC5351693 DOI: 10.18632/oncotarget.12569] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/04/2016] [Indexed: 12/12/2022] Open
Abstract
Poor prognosis and high recurrence remain leading causes of primary liver cancerassociated mortality. The spread of circulating tumor cells (CTCs) in the blood plays a major role in the initiation of metastasis and tumor recurrence after surgery. Nevertheless, only a subset of CTCs can survive, migrate to distant sites and establish secondary tumors. Consistent with cancer stem cell (CSC) hypothesis, stem-like CTCs might represent a potential source for cancer relapse and distant metastasis. Thus, identification of stem-like metastasis-initiating CTC-subset may provide useful clinically prognostic information. This review will emphasize the most relevant findings of CTCs in the context of stem-like biology associated to liver carcinogenesis. In this view, the emerging field of stem-like CTCs may deliver substantial contribution in liver cancer field in order to move to personalized approaches for diagnosis, prognosis and therapy.
Collapse
Affiliation(s)
- Margherita Correnti
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Chiara Raggi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
36
|
Mason J, Blyth B, MacManus MP, Martin OA. Treatment for non-small-cell lung cancer and circulating tumor cells. Lung Cancer Manag 2017; 6:129-139. [PMID: 30643579 PMCID: PMC6310303 DOI: 10.2217/lmt-2017-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
Surgery is the main curative therapy for patients with localized non-small-cell lung cancer while radiotherapy (RT), alone or with concurrent platinum-based chemotherapy, remains the primary curative modality for locoregionally advanced non-small-cell lung cancer. The risk of distant metastasis is high after curative-intent treatment, largely attributable to the presence of undetected micrometastases, but which could also be related to treatment-related increases in circulating tumor cells (CTCs). CTC mobilization by RT or systemic therapies might either reflect efficient tumor destruction with improved prognosis, or might promote metastasis and thus represent a potential therapeutic target. RT may induce prometastatic biological alterations in CTC at the cellular level, which are detectable by 'liquid biopsies', though their rarity represents a major challenge. Improved methods of isolation and ex vivo propagation will be essential for the future of CTC research.
Collapse
Affiliation(s)
- Joel Mason
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - Benjamin Blyth
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Michael P MacManus
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Olga A Martin
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
37
|
Suppression of cancer stemness by upregulating Ligand-of-Numb protein X1 in colorectal carcinoma. PLoS One 2017; 12:e0188665. [PMID: 29190716 PMCID: PMC5708683 DOI: 10.1371/journal.pone.0188665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/10/2017] [Indexed: 01/27/2023] Open
Abstract
Cancer stem-like cells (CSCs) have been reported to play major roles in tumorigenesis, tumor relapse, and metastasis after therapy against colorectal carcinoma (CRC). Therefore, identification of colorectal CSC regulators could provide promising targets for CRC. Ligand-of-Numb protein X1 (LNX1) is one E3 ubiquitin ligase which mediates the ubiquitination and degradation of Numb. Although several studies indicate LNX1 could be a potential suppressor of cancer diseases, the functions of LNX1 in mediating cancer stemness remain poorly understood. In this study, LNX1 was identified as a negative regulator of cancer stemness in CRC, which was downregulated in colonospheres or side population (SP) cells. Furthermore, the coxsackievirus and adenovirus receptor (CXADR) was found to be one critical downstream mediator of cancer stemness regulated by LNX1. Interestingly, the anti-breast cancer drug tamoxifen was found to be an agonist of LNX1 and suppress cancer stemness in CRC. In sum, this study provided the evidences that LNX1 signaling plays important roles in regulating the stemness of colon cancer cells.
Collapse
|
38
|
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: 3.4] [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
|
39
|
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: 3.0] [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
|
40
|
Liu Y, Li H, Ban Z, Nai M, Yang L, Chen Y, Xu Y. Annexin A2 inhibition suppresses ovarian cancer progression via regulating β-catenin/EMT. Oncol Rep 2017; 37:3643-3650. [PMID: 28440436 DOI: 10.3892/or.2017.5578] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/16/2017] [Indexed: 01/23/2023] Open
Abstract
Annexin A2 is a member of the Annexin family that acts as a Ca2+-dependent phospholipid and membrane binding protein, which is associated with the survival and spread of multiple neoplasms. However, the function of Annexin A2 in ovarian cancer progression remains unclear. In this study, we aimed to investigate the role and underlying molecular mechanism of Annexin A2 in cell proliferation and invasion in ovarian cancer. We found that the mRNA expression of Annexin A2 was upregulated in ovarian cancer tissues and cell lines. In the loss-of-function of Annexin A2, β-catenin was indicated to be significantly suppressed and EMT constrained. Moreover, cell proliferation and invasion were both markedly inhibited by the downregulation of Annexin A2. Additionally, the overexpression of β-catenin obviously reversed the effect of Annexin A2 on EMT, and cell proliferation and invasion, indicating that Annexin A2 suppression regulated EMT through controlling β-catenin. Taken together, this study showed that Annexin A2 inhibition suppresses proliferation and invasion in ovarian cancer via β-catenin/EMT, proposing the potential role of Annexin A2 in the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Hongyu Li
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhenying Ban
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Manman Nai
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Li Yang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yannan Chen
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yiming Xu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
41
|
Electrical Impedance Spectroscopy for Detection of Cells in Suspensions Using Microfluidic Device with Integrated Microneedles. APPLIED SCIENCES-BASEL 2017. [DOI: 10.3390/app7020170] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Singh P, O'Connell M, Shubhashish S. Epigenetic regulation of human DCLK-1 gene during colon-carcinogenesis: clinical and mechanistic implications. Stem Cell Investig 2016; 3:51. [PMID: 27777940 DOI: 10.21037/sci.2016.09.07] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/08/2016] [Indexed: 12/26/2022]
Abstract
Colorectal carcinogenesis is a multi-step process. While ~25% of colorectal cancers (CRCs) arise in patients with a family history (genetic predisposition), ~75% of CRCs are due to age-associated accumulation of epigenetic alterations which can result in the suppression of key tumor suppressor genes leading to mutations and activation of oncogenic pathways. Sporadic colon-carcinogenesis is facilitated by many molecular pathways of genomic instability which include chromosomal instability (CIN), micro-satellite instability (MSI) and CpG island methylator phenotype (CIMP), leading towards loss of homeostasis and onset of neoplastic transformation. The unopposed activation of Wnt/β-catenin pathways, either due to loss of APC function or up-regulation of related stimulatory pathways, results in unopposed hyperproliferation of colonic crypts, considered the single most important risk factor for colon carcinogenesis. Hypermethylation of CpG islands within the promoters of specific genes can potentially inactivate DNA repair genes and/or critical tumor suppressor genes. Recently, CpG methylation of the 5' promoter of human (h) DCLK1 gene was reported in many human epithelial cancers, including colorectal cancers (CRCs), resulting in the loss of expression of the canonical long isoform of DCLK1 (DCLK1-L) in hCRCs. Instead, a shorter isoform of DCLK1 (DCLK1-S) was discovered to be expressed in hCRCs, from an alternate β promoter of DCLKL1-gene; the clinical and biological implications of these novel findings, in relation to recent publications is discussed.
Collapse
Affiliation(s)
- Pomila Singh
- Neuroscience and Cell Biology Department, University of Texas and Medical Branch, Galveston, TX, USA
| | - Malaney O'Connell
- Neuroscience and Cell Biology Department, University of Texas and Medical Branch, Galveston, TX, USA
| | - Sarkar Shubhashish
- Neuroscience and Cell Biology Department, University of Texas and Medical Branch, Galveston, TX, USA
| |
Collapse
|
43
|
Mirzaei A, Madjd Z, Kadijani AA, Tavakoli-Yaraki M, Modarresi MH, Verdi J, Akbari A, Tavoosidana G. Evaluation of circulating cellular DCLK1 protein, as the most promising colorectal cancer stem cell marker, using immunoassay based methods. Cancer Biomark 2016; 17:301-311. [DOI: 10.3233/cbm-160642] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Alireza Mirzaei
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azade Amini Kadijani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Tavoosidana
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Gao T, Wang M, Xu L, Wen T, Liu J, An G. DCLK1 is up-regulated and associated with metastasis and prognosis in colorectal cancer. J Cancer Res Clin Oncol 2016; 142:2131-40. [PMID: 27520310 DOI: 10.1007/s00432-016-2218-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE Metastasis is a primary cause of colorectal cancer (CRC)-related death, and cancer stem cells (CSCs) are thought to be majorly responsible for initiating metastatic behaviors. Doublecortin-like kinase 1 (DCLK1) was recently discovered to be a marker for gastrointestinal CSCs. Here, we aimed to explore whether DCLK1 is associated with CRC metastasis through clinical and in vitro investigations. METHODS The expression levels of DCLK1 mRNA and protein in human CRC tissues were analyzed through quantitative RT-PCR and immunohistochemistry staining, respectively. Human CRC cell line SW480 was selected to explore the effect of DCLK1 overexpression on cell migration and invasion. Besides, the associations between DCLK1 and epithelial-mesenchymal transition (EMT) were determined. RESULTS Compared to normal colorectal tissues, DCLK1 expression was significantly up-regulated in human CRC tissues and correlated well with high lymphatic metastasis and poor prognosis in patients. DCLK1 expression was inversely associated with overall survival in CRC patients. Overexpression of DCLK1 in SW480 cells markedly promoted cell migration and invasion. Furthermore, we validated that DCLK1 could facilitate EMT in cancer cells by up-regulation of the mesenchymal markers Vimentin and ZEB1 and down-regulation of the epithelial marker E-cadherin in SW480 cells. CONCLUSIONS DCLK1 up-regulation may play a contributory role in CRC metastasis and poor prognosis via activation of EMT. DCLK1 may serve as an independent predictor for CRC prognosis.
Collapse
Affiliation(s)
- Tianbo Gao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China
| | - Min Wang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China
| | - Lingling Xu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China
| | - Tao Wen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China
| | - Jian Liu
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China.
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gong Ti Nan Lu, Beijing, 100020, China.
| |
Collapse
|
45
|
Carnero A, Garcia-Mayea Y, Mir C, Lorente J, Rubio IT, LLeonart ME. The cancer stem-cell signaling network and resistance to therapy. Cancer Treat Rev 2016; 49:25-36. [PMID: 27434881 DOI: 10.1016/j.ctrv.2016.07.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 12/13/2022]
Abstract
The study of cancer stem cells (CSCs) has shown that tumors are driven by a subpopulation of self-renewing CSCs that retain the capacity to engender the various differentiated cell populations that form tumors. The characterization of CSCs has indicated that CSCs are remarkably resistant to conventional radio- and chemo-therapy. Clinically, the remaining populations of CSC are responsible for metastasis and recurrence in patients with cancer, which can lead to the disease becoming chronic and incurable. Therefore, the elimination of CSCs is an important goal of cancer treatments. Furthermore, CSCs are subject to strong regulation by the surrounding microenvironment, which also impacts tumor responses. In this review, we discuss the mechanisms by which pathways that are defective in CSCs influence ultimately therapeutic and clinical outcomes.
Collapse
Affiliation(s)
- A Carnero
- Instituto de Biomedicina de Sevilla (IBIS/HUVR/CSIC/Universidad de Sevilla), Molecular Biology of Cancer Group, Oncohematology and Genetic Department, Campus HUVR, Edificio IBIS, Avda. Manuel Siurot s/n. 41013, Sevilla, Spain
| | - Y Garcia-Mayea
- Vall d'Hebron Institut de Recerca (VHIR), Hospital Vall d'Hebron, Translational Research in Cancer Stem Cell Group, Pathology Department, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - C Mir
- Vall d'Hebron Institut de Recerca (VHIR), Hospital Vall d'Hebron, Translational Research in Cancer Stem Cell Group, Pathology Department, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - J Lorente
- Vall d'Hebron Institut de Recerca (VHIR), Hospital Vall d'Hebron, Translational Research in Cancer Stem Cell Group, Pathology Department, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - I T Rubio
- Vall d'Hebron Institut de Oncologia (VHIO), Hospital Vall d'Hebron, Breast Surgical Oncology Unit, Breast Cancer Center, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - M E LLeonart
- Vall d'Hebron Institut de Recerca (VHIR), Hospital Vall d'Hebron, Translational Research in Cancer Stem Cell Group, Pathology Department, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain.
| |
Collapse
|
46
|
Weygant N, Ge Y, Qu D, Kaddis JS, Berry WL, May R, Chandrakesan P, Bannerman-Menson E, Vega KJ, Tomasek JJ, Bronze MS, An G, Houchen CW. Survival of Patients with Gastrointestinal Cancers Can Be Predicted by a Surrogate microRNA Signature for Cancer Stem-like Cells Marked by DCLK1 Kinase. Cancer Res 2016; 76:4090-9. [PMID: 27287716 DOI: 10.1158/0008-5472.can-16-0029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/30/2016] [Indexed: 12/31/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a gastrointestinal (GI) tuft cell kinase that has been investigated as a biomarker of cancer stem-like cells in colon and pancreatic cancers. However, its utility as a biomarker may be limited in principle by signal instability and dilution in heterogeneous tumors, where the proliferation of diverse tumor cell lineages obscures the direct measurement of DCLK1 activity. To address this issue, we explored the definition of a miRNA signature as a surrogate biomarker for DCLK1 in cancer stem-like cells. Utilizing RNA/miRNA-sequencing datasets from the Cancer Genome Atlas, we identified a surrogate 15-miRNA expression signature for DCLK1 activity across several GI cancers, including colon, pancreatic, and stomach cancers. Notably, Cox regression and Kaplan-Meier analysis demonstrated that this signature could predict the survival of patients with these cancers. Moreover, we identified patient subgroups that predicted the clinical utility of this DCLK1 surrogate biomarker. Our findings greatly strengthen the clinical significance for DCLK1 expression across GI cancers. Further, they provide an initial guidepost toward the development of improved prognostic biomarkers or companion biomarkers for DCLK1-targeted therapies to eradicate cancer stem-like cells in these malignancies. Cancer Res; 76(14); 4090-9. ©2016 AACR.
Collapse
Affiliation(s)
- Nathaniel Weygant
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma
| | - Yang Ge
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, Beijing, China
| | - Dongfeng Qu
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma. United States Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma. The Peggy and Charles Stephenson Cancer Center, Oklahoma City, Oklahoma
| | - John S Kaddis
- City of Hope National Medical Center, Duarte, California
| | - William L Berry
- The University of Oklahoma Health Sciences Center Department of Cell Biology, Oklahoma City, Oklahoma
| | - Randal May
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma. United States Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Parthasarathy Chandrakesan
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma. United States Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma. The Peggy and Charles Stephenson Cancer Center, Oklahoma City, Oklahoma
| | | | - Kenneth J Vega
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma. United States Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - James J Tomasek
- The University of Oklahoma Health Sciences Center Department of Cell Biology, Oklahoma City, Oklahoma
| | - Michael S Bronze
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma
| | - Guangyu An
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, Beijing, China.
| | - Courtney W Houchen
- The University of Oklahoma Health Sciences Center Department of Medicine, Oklahoma City, Oklahoma. United States Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma. The Peggy and Charles Stephenson Cancer Center, Oklahoma City, Oklahoma. COARE Biotechnology Inc., Oklahoma City, Oklahoma.
| |
Collapse
|
47
|
Bamodu OA, Huang WC, Lee WH, Wu A, Wang LS, Hsiao M, Yeh CT, Chao TY. Aberrant KDM5B expression promotes aggressive breast cancer through MALAT1 overexpression and downregulation of hsa-miR-448. BMC Cancer 2016; 16:160. [PMID: 26917489 PMCID: PMC4768424 DOI: 10.1186/s12885-016-2108-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 02/01/2016] [Indexed: 12/17/2022] Open
Abstract
Background Triple negative breast cancers (TNBC) possess cell dedifferentiation characteristics, carry out activities connate to those of cancer stem cells (CSCs) and are associated with increased metastasis, as well as, poor clinical prognosis. The regulatory mechanism of this highly malignant phenotype is still poorly characterized. Accruing evidence support the role of non-coding RNAs (ncRNAs) as potent regulators of CSC and metastatic gene expression, with their dysregulation implicated in tumorigenesis and disease progression. Methods In this study, we investigated TNBC metastasis, metastasis-associated genes and potential inhibitory mechanisms using bioinformatics, tissue microarray analyses, immunoblotting, polymerase chain reaction, loss and gain of gene function assays and comparative analyses of data obtained. Results Compared with other breast cancer types, the highly metastatic MDA-MB-231 cells concurrently exhibited increased expression levels of Lysine-specific demethylase 5B protein (KDM5B) and long non-coding RNA (lncRNA), MALAT1, suggesting their functional association. KDM5B-silencing in the TNBC cells correlated with the upregulation of hsa-miR-448 and led to suppression of MALAT1 expression with decreased migration, invasion and clonogenic capacity in vitro, as well as, poor survival in vivo. This projects MALAT1 as a mediator of KDM5B oncogenic potential and highlights the critical role of this microRNA, lncRNA and histone demethylase in cancer cell motility and metastatic colonization. Increased expression of KDM5B correlating with disease progression and poor clinical outcome in breast cancer was reversed by hsa-miR-448. Conclusions Our findings demonstrate the critical role of KDM5B and its negative regulator hsa-miR-448 in TNBC metastasis and progression. Hsa-miR-448 disrupting KDM5B-MALAT1 signalling axis and associated activities in TNBC cells, projects it as a putative therapeutic factor for selective eradication of TNBC cells. KDM5B, MALAT1 and hsa-miR-448 are active looped components of the epigenetic poculo mortis in aggressive breast cancer. ![]()
Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2108-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan. .,Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Wen-Chien Huang
- Department of Thoracic Surgery, Mackay Memorial Hospital, Taipei, 10449, Taiwan.
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan.
| | - Alexander Wu
- Graduate Institute of Translational Medicine, Taipei Medical University, Taipei City, Taiwan. .,The PhD Program of Translational Medicine, Academia Sinica, Nankang, Taipei, Taiwan.
| | - Liang Shun Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan. .,Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| | - Chi-Tai Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan. .,Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Tsu-Yi Chao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan. .,Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan. .,Tri-Service General Hospital, Neihu District, Taipei City, Taiwan.
| |
Collapse
|
48
|
Lin S, Xu Y, Gan Z, Han K, Hu H, Yao Y, Huang M, Min D. Monitoring cancer stem cells: insights into clinical oncology. Onco Targets Ther 2016; 9:731-40. [PMID: 26929644 PMCID: PMC4755432 DOI: 10.2147/ott.s96645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) are a small, characteristically distinctive subset of tumor cells responsible for tumor initiation and progression. Several treatment modalities, such as surgery, glycolytic inhibition, driving CSC proliferation, immunotherapy, and hypofractionated radiotherapy, may have the potential to eradicate CSCs. We propose that monitoring CSCs is important in clinical oncology as CSC populations may reflect true treatment response and assist with managing treatment strategies, such as defining optimal chemotherapy cycles, permitting pretreatment cancer surveillance, conducting a comprehensive treatment plan, modifying radiation treatment, and deploying rechallenge chemotherapy. Then, we describe methods for monitoring CSCs.
Collapse
Affiliation(s)
- ShuChen Lin
- Department of Oncology, Shanghai Sixth People's Hospital East Campus, Shanghai Jiao Tong University, People's Republic of China
| | - YingChun Xu
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University, People's Republic of China
| | - ZhiHua Gan
- Department of Oncology, Shanghai Sixth People's Hospital East Campus, Shanghai Jiao Tong University, People's Republic of China
| | - Kun Han
- Department of Oncology, Shanghai Sixth People's Hospital East Campus, Shanghai Jiao Tong University, People's Republic of China
| | - HaiYan Hu
- Department of Oncology, The Sixth People's Hospital, Shanghai Jiao Tong University, People's Republic of China
| | - Yang Yao
- Department of Oncology, The Sixth People's Hospital, Shanghai Jiao Tong University, People's Republic of China
| | - MingZhu Huang
- Department of Medical Oncology, Cancer Hospital of Fudan University, Shanghai, People's Republic of China
| | - DaLiu Min
- Department of Oncology, Shanghai Sixth People's Hospital East Campus, Shanghai Jiao Tong University, People's Republic of China
| |
Collapse
|
49
|
Talukdar S, Emdad L, Das S, Sarkar D, Fisher P. Evolving Strategies for Therapeutically Targeting Cancer Stem Cells. Adv Cancer Res 2016; 131:159-91. [PMID: 27451127 DOI: 10.1016/bs.acr.2016.04.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer is a multifactor and multistep process that is affected intrinsically by the genetic and epigenetic makeup of tumor cells and extrinsically by the host microenvironment and immune system. A key component of cancer involves a unique subpopulation of highly malignant cancerous cells referred to as cancer stem cells (CSCs). CSCs are positioned at the apex of the tumor hierarchy with an ability to both self-renew and also generate non-CSC/differentiated progeny, which contribute to the majority of the tumor mass. CSCs undergo functional changes and show plasticity that is stimulated by specific microenvironmental cues and interactions in the tumor niche, which contribute to the complexity and heterogeneity of the CSC population. The prognostic value of CSCs in the clinic is evident since there are many examples in which CSCs serve as markers for poor patient prognosis. CSCs are innately resistant to many standard therapies and they display anoikis resistance, immune evasion, tumor dormancy, and field cancerization, which may result in metastasis and relapse. Many academic laboratories and biotechnology companies are currently focusing on strategies that target CSCs. Combination therapies, epigenetic modifiers, stemness inhibitors, CSC surface marker-based therapies, and immunotherapy-based CSC-targeting drugs are currently undergoing clinical trials. Potential new targets/strategies in CSC-targeted therapy include MDA-9/Syntenin (SDCBP), Patched (PTCH), epigenetic targets, noncoding RNAs, and differentiation induction. Defining ways of targeting and destroying CSCs holds potential to impact significantly on cancer therapy, including prevention of metastasis and cancer recurrence.
Collapse
|
50
|
Guo M, Dou J. Advances and perspectives of colorectal cancer stem cell vaccine. Biomed Pharmacother 2015; 76:107-20. [PMID: 26653557 DOI: 10.1016/j.biopha.2015.10.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is essentially an environmental and genetic disease featured by uncontrolled cell growth and the capability to invade other parts of the body by forming metastases, which inconvertibly cause great damage to tissues and organs. It has become one of the leading causes of cancer-related mortality in the developed countries such as United States, and approximately 1.2 million new cases are yearly diagnosed worldwide, with the death rate of more than 600,000 annually and incidence rates are increasing in most developing countries. Apart from the generally accepted theory that pathogenesis of colorectal cancer consists of genetic mutation of a certain target cell and diversifications in tumor microenvironment, the colorectal cancer stem cells (CCSCs) theory makes a different explanation, stating that among millions of colon cancer cells there is a specific and scanty cellular population which possess the capability of self-renewal, differentiation and strong oncogenicity, and is tightly responsible for drug resistance and tumor metastasis. Based on these characteristics, CCSCs are becoming a novel target cells both in the clinical and the basic studies, especially the study of CCSCs vaccines due to induced efficient immune response against CCSCs. This review provides an overview of CCSCs and preparation technics and targeting factors related to CCSCs vaccines in detail.
Collapse
Affiliation(s)
- Mei Guo
- Department of Pathogenic Biology and Immunology of Medical School, Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology of Medical School, Southeast University, Nanjing 210009, China.
| |
Collapse
|