1
|
Breast cancer tumor microenvironment affects Treg/IL-17-producing Treg/Th17 cell axis: Molecular and therapeutic perspectives. Mol Ther Oncolytics 2023; 28:132-157. [PMID: 36816749 PMCID: PMC9922830 DOI: 10.1016/j.omto.2023.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The tumor microenvironment (TME) comprises a variety of immune cells, among which T cells exert a prominent axial role in tumor development or anti-tumor responses in patients with breast cancer (BC). High or low levels of anti-inflammatory cytokines, such as transforming growth factor β, in the absence or presence of proinflammatory cytokines, such as interleukin-6 (IL-6), delineate the fate of T cells toward either regulatory T (Treg) or T helper 17 (Th17) cells, respectively. The transitional state of RORγt+Foxp3+ Treg (IL-17-producing Treg) resides in the middle of this reciprocal polarization, which is known as Treg/IL-17-producing Treg/Th17 cell axis. TME secretome, including microRNAs, cytokines, and extracellular vesicles, can significantly affect this axis. Furthermore, immune checkpoint inhibitors may be used to reconstruct immune cells; however, some of these novel therapies may favor tumor development. Therefore, understanding secretory and cell-associated factors involved in their differentiation or polarization and functions may be targeted for BC management. This review discusses microRNAs, cytokines, and extracellular vesicles (as secretome), as well as transcription factors and immune checkpoints (as cell-associated factors), which influence the Treg/IL-17-producing Treg/Th17 cell axis in BC. Furthermore, approved or ongoing clinical trials related to the modulation of this axis in the TME of BC are described to broaden new horizons of promising therapeutic approaches.
Collapse
|
2
|
Papavassiliou KA, Marinos G, Papavassiliou AG. Combining STAT3-Targeting Agents with Immune Checkpoint Inhibitors in NSCLC. Cancers (Basel) 2023; 15:cancers15020386. [PMID: 36672335 PMCID: PMC9857288 DOI: 10.3390/cancers15020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Despite recent therapeutic advances, non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related death. Signal transducer and activator of transcription 3 (STAT3) is a transcription factor (TF) with multiple tumor-promoting effects in NSCLC, including proliferation, anti-apoptosis, angiogenesis, invasion, metastasis, immunosuppression, and drug resistance. Recent studies suggest that STAT3 activation contributes to resistance to immune checkpoint inhibitors. Thus, STAT3 represents an attractive target whose pharmacological modulation in NSCLC may assist in enhancing the efficacy of or overcoming resistance to immune checkpoint inhibitors. In this review, we discuss the biological mechanisms through which STAT3 inhibition synergizes with or overcomes resistance to immune checkpoint inhibitors and highlight the therapeutic strategy of using drugs that target STAT3 as potential combination partners for immune checkpoint inhibitors in the management of NSCLC patients.
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, Medical School, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgios Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-210-746-2508
| |
Collapse
|
3
|
Designed miR-19a/b sponge induces apoptosis in lung cancer cells through the PI3K-PTEN-Akt pathway regulation. Mol Biol Rep 2022; 49:8485-8493. [PMID: 35767105 DOI: 10.1007/s11033-022-07670-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/01/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are one of the main factors in cancer development and can alter the activity of proto-oncogenic or tumor suppressor genes. The miR-17-92 cluster, which comprises miR-17, miR-18a, miR-19a/b, miR-20a, and miR-92a, has been identified as a biomarker in a variety of cancer types. Among them, miR-19a/b exerts an oncogenic effect by suppressing tumor suppressor genes, including PTEN and TP53INP1in numerous types of cancers, including NSCLC. An miRNA sponge is an mRNA with multiple repetitive sequences that prevents miRNAs from interacting with their targets, thereby inhibiting their action. METHODS AND RESULTS In this study, we designed an miR-19a/b sponge plasmid and transfected it into A549 lung cancer cell lines and analyzed its effects on PTEN and TP53INP1 gene expression as the main miR-19a/b target and apoptosis rate in these cell lines. CONCLUSIONS The findings revealed that miR-19a/b sponge significantly increased PTEN and TP53INP1 mRNA expression. The effect of the sponge on TP53INP1 was much greater than that on PTEN. This is because TP53INP1 is directly (sponge effect) and indirectly (AKT pathway is affected by the P53 gene) affected by this sponge. In addition, compared with the control group, the percentage of primary and secondary apoptosis increased significantly (P value < 0.0001).
Collapse
|
4
|
Parakh S, Ernst M, Poh AR. Multicellular Effects of STAT3 in Non-small Cell Lung Cancer: Mechanistic Insights and Therapeutic Opportunities. Cancers (Basel) 2021; 13:6228. [PMID: 34944848 PMCID: PMC8699548 DOI: 10.3390/cancers13246228] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and accounts for 85% of lung cancer cases. Aberrant activation of the Signal Transducer and Activator of Transcription 3 (STAT3) is frequently observed in NSCLC and is associated with a poor prognosis. Pre-clinical studies have revealed an unequivocal role for tumor cell-intrinsic and extrinsic STAT3 signaling in NSCLC by promoting angiogenesis, cell survival, cancer cell stemness, drug resistance, and evasion of anti-tumor immunity. Several STAT3-targeting strategies have also been investigated in pre-clinical models, and include preventing upstream receptor/ligand interactions, promoting the degradation of STAT3 mRNA, and interfering with STAT3 DNA binding. In this review, we discuss the molecular and immunological mechanisms by which persistent STAT3 activation promotes NSCLC development, and the utility of STAT3 as a prognostic and predictive biomarker in NSCLC. We also provide a comprehensive update of STAT3-targeting therapies that are currently undergoing clinical evaluation, and discuss the challenges associated with these treatment modalities in human patients.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, The Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, VIC 3084, Australia;
- Tumor Targeting Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
| | - Matthias Ernst
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
- Cancer and Inflammation Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| | - Ashleigh R. Poh
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
- Cancer and Inflammation Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| |
Collapse
|
5
|
Ye L, Morse LR, Falci SP, Olson JK, Shrivastava M, Nguyen N, Linnman C, Troy KL, Battaglino RA. hsa-MiR-19a-3p and hsa-MiR-19b-3p Are Associated with Spinal Cord Injury-Induced Neuropathic Pain: Findings from a Genome-Wide MicroRNA Expression Profiling Screen. Neurotrauma Rep 2021; 2:424-439. [PMID: 34755149 PMCID: PMC8570675 DOI: 10.1089/neur.2021.0011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuropathic pain in spinal cord injury (SCI) is associated with inflammation in both the peripheral and central nervous system (CNS), which may contribute to the initiation and maintenance of persistent pain. An understanding of factors contributing to neuroinflammation may lead to new therapeutic targets for neuropathic pain. Moreover, novel circulating biomarkers of neuropathic pain may facilitate earlier and more effective treatment. MicroRNAs (miRNAs) are short, non-coding single-stranded RNA that have emerged as important biomarkers and molecular mediators in physiological and pathological conditions. Using a genome-wide miRNA screening approach, we studied differential miRNA expression in plasma from 68 healthy, community-dwelling adults with and without SCI enrolled in ongoing clinical studies. We detected 2367 distinct miRNAs. Of these, 383 miRNAs were differentially expressed in acute SCI or chronic SCI versus no SCI and 71 were differentially expressed in chronic neuropathic pain versus no neuropathic pain. We selected homo sapiens (hsa)-miR-19a-3p and hsa-miR-19b-3p for additional analysis based on p-value, fold change, and their known role as regulators of neuropathic pain and neuroinflammation. Both hsa-miR-19a-3p and hsa-miR-19b-3p levels were significantly higher in those with chronic SCI and severe neuropathic pain versus those with chronic SCI and no neuropathic pain. In confirmatory studies, both hsa-miR-19a-3p and hsa-miR-19b-3p have moderate to strong discriminative ability to distinguish between those with and without pain. After adjusting for opioid use, hsa-miR-19b-3p levels were positively associated with pain interference with mood. Because hsa-miR-19 levels have been shown to change in response to exercise, folic acid, and resveratrol, these studies suggest that miRNAs are potential targets of therapeutic interventions.
Collapse
Affiliation(s)
- Liang Ye
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Leslie R Morse
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Scott P Falci
- Department of Neurological Surgery, Swedish Medical Center, Englewood, Colorado, USA
| | - Julie K Olson
- Department of Diagnostics and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Mayank Shrivastava
- Department of Diagnostics and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Nguyen Nguyen
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Clas Linnman
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Charlestown, Massachusetts, USA
| | - Karen L Troy
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Ricardo A Battaglino
- Department of Rehabilitation Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Keewan E, Matlawska-Wasowska K. The Emerging Role of Suppressors of Cytokine Signaling (SOCS) in the Development and Progression of Leukemia. Cancers (Basel) 2021; 13:4000. [PMID: 34439155 PMCID: PMC8393695 DOI: 10.3390/cancers13164000] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are pleiotropic signaling molecules that execute an essential role in cell-to-cell communication through binding to cell surface receptors. Receptor binding activates intracellular signaling cascades in the target cell that bring about a wide range of cellular responses, including induction of cell proliferation, migration, differentiation, and apoptosis. The Janus kinase and transducers and activators of transcription (JAK/STAT) signaling pathways are activated upon cytokines and growth factors binding with their corresponding receptors. The SOCS family of proteins has emerged as a key regulator of cytokine signaling, and SOCS insufficiency leads to constitutive activation of JAK/STAT signaling and oncogenic transformation. Dysregulation of SOCS expression is linked to various solid tumors with invasive properties. However, the roles of SOCS in hematological malignancies, such as leukemia, are less clear. In this review, we discuss the recent advances pertaining to SOCS dysregulation in leukemia development and progression. We also highlight the roles of specific SOCS in immune cells within the tumor microenvironment and their possible involvement in anti-tumor immunity. Finally, we discuss the epigenetic, genetic, and post-transcriptional modifications of SOCS genes during tumorigenesis, with an emphasis on leukemia.
Collapse
Affiliation(s)
- Esra’a Keewan
- Department of Pediatrics, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
7
|
El Founini Y, Chaoui I, Dehbi H, El Mzibri M, Abounader R, Guessous F. MicroRNAs: Key Regulators in Lung Cancer. Microrna 2021; 10:109-122. [PMID: 34047262 DOI: 10.2174/2211536610666210527102522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022]
Abstract
Noncoding RNAs have emerged as key regulators of the genome upon gene expression profiling and genome-wide sequencing. Among these noncoding RNAs, microRNAs are short noncoding RNAs that regulate a plethora of functions, biological processes and human diseases by targeting the messenger RNA stability through 3'UTR binding, leading to either mRNA cleavage or translation repression, depending on microRNA-mRNA complementarity degree. Additionally, strong evidence has suggested that dysregulation of miRNAs contribute to the etiology and progression of human cancers, such as lung cancer, the most common and deadliest cancer worldwide. Indeed, by acting as oncogenes or tumor suppressors, microRNAs control all aspects of lung cancer malignancy, including cell proliferation, survival, migration, invasion, angiogenesis, cancer stem cells, immune-surveillance escape, and therapy resistance; and their expressions are often associated with clinical parameters. Moreover, several deregulated microRNAs in lung cancer are carried by exosomes, microvesicles and secreted in body fluids, mainly the circulation where they conserve their stable forms. Subsequently, seminal efforts have been focused on extracellular microRNAs levels as noninvasive diagnostic and prognostic biomarkers in lung cancer. In this review, focusing on recent literature, we summarize the deregulation, mechanisms of action, functions and highlight clinical applications of miRNAs for better management and design of future lung cancer targeted therapies.
Collapse
Affiliation(s)
- Younes El Founini
- Unit of Biology and Medical Research, National Center of Energy, Sciences and Nuclear Techniques, Rabat, Morocco.,Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Morocco
| | - Imane Chaoui
- Unit of Biology and Medical Research, National Center of Energy, Sciences and Nuclear Techniques, Rabat, Morocco
| | - Hind Dehbi
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Morocco
| | - Mohammed El Mzibri
- Unit of Biology and Medical Research, National Center of Energy, Sciences and Nuclear Techniques, Rabat, Morocco
| | - Roger Abounader
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Fadila Guessous
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States.,Department of Biological Sciences, Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| |
Collapse
|
8
|
Peng T, Yang F, Sun Z, Yan J. miR-19a-3p Facilitates Lung Adenocarcinoma Cell Phenotypes by Inhibiting TEK. Cancer Biother Radiopharm 2021; 37:589-601. [PMID: 33493418 DOI: 10.1089/cbr.2020.4456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Both TEK and miR-19a-3p have been reported to regulate lung adenocarcinoma (LUAD) progression. However, the association between TEK and miR-19a-3p in LUAD remained unknown. This research aimed to investigate a novel miR-19a-3p/TEK interactome in LUAD cells. Methods: The mRNA expression and protein expression in the cell lines were determined using qPCR and Western blot assay, respectively. CCK-8 assay, EDU assay, flow cytometry cell apoptosis assay, scratch assay, and cell-to-extracellular matrix adhesion assay were performed to detect the proliferation, apoptosis, migration, and adhesion ability of A549 and H1975 cell lines. Results: Findings revealed that both mRNA and protein levels of TEK were downregulated in the LUAD tumor tissues and cell lines. It was also found that compared with the control group, the transfection of TEK overexpression plasmids into H1975 and A549 cell lines significantly inhibited cancerous phenotypes. However, experimental results indicated that by downregulating TEK, miR-19a-3p promoted LUAD cell phenotypes. Conclusion: This research demonstrated that an interactome existed between miR-19a-3p and TEK and that miR-19a-3p could suppress LUAD tumors by inhibiting TEK. This novel interactome could be used as a novel therapy target for LUAD.
Collapse
Affiliation(s)
- Tao Peng
- Department of Thoracic and Cardiovascular Surgery, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| | - Fan Yang
- Department of Thoracic and Cardiovascular Surgery, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| | - Zhanwen Sun
- Department of Thoracic and Cardiovascular Surgery, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| | - Jie Yan
- Department of Thoracic and Cardiovascular Surgery, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| |
Collapse
|
9
|
Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, Baligar P, Kumar AP, Goh BC, Wang L, Sethi G. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev 2020; 41:1291-1336. [PMID: 33289118 DOI: 10.1002/med.21761] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of the crucial transcription factors, responsible for regulating cellular proliferation, cellular differentiation, migration, programmed cell death, inflammatory response, angiogenesis, and immune activation. In this review, we have discussed the classical regulation of STAT3 via diverse growth factors, cytokines, G-protein-coupled receptors, as well as toll-like receptors. We have also highlighted the potential role of noncoding RNAs in regulating STAT3 signaling. However, the deregulation of STAT3 signaling has been found to be associated with the initiation and progression of both solid and hematological malignancies. Additionally, hyperactivation of STAT3 signaling can maintain the cancer stem cell phenotype by modulating the tumor microenvironment, cellular metabolism, and immune responses to favor drug resistance and metastasis. Finally, we have also discussed several plausible ways to target oncogenic STAT3 signaling using various small molecules derived from natural products.
Collapse
Affiliation(s)
- Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vipul Bhardwaj
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Akul Goel
- La Canada High School, La Canada Flintridge, California, USA
| | - Rajat Gupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Arundhiti Sharma
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Boon Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
- Department of Hematology-Oncology, National University Health System, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Kuang Z, Deng H, Xu L, Hu Q, Cai Y, Wang R, Luo S, Liu T, Hao W. A homogeneous immunoassay for detection of the interaction between two tumor biomarkers of IGF1R-β and SOCS1. Biotechnol Appl Biochem 2020; 68:769-775. [PMID: 32700452 DOI: 10.1002/bab.1989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022]
Abstract
The current protein interaction method is time consuming and cumbersome or the instrument is expensive. A new method that is convenient, fast, and high throughput needs to be studied urgently. The purpose of this study was to establish a homogeneous immunoassay to detect the interaction between insulin-like growth factor-1 receptor-β (IGF1R-β) and suppressor of cytokine signaling 1 (SOCS1). The recombinant vectors IGF1R-β/pENTER and SOCS1/pENTER were constructed and transfected into 293T cells. Based on homogeneous immunoassay technology, we established a suitable method. The signal intensity in the 293T lysate that overexpressed IGF1R-β and SOCS1, respectively, was compared with the signal intensity in the simultaneous expression of IGF1R-β and SOCS1. The interaction between IGF1R-β and SOCS1 was verified in vitro. The detection system for the interaction between IGF1R-β and SOCS1 was established. Compared with other methods, homogeneous immunoassay has the advantages of being rapid and sensitive, having higher sensitivity, and easy to operate. The interaction between IGF1R-β and SOCS1 was tested to verify the feasibility of this method and prove its practicability and sensitivity. This new method can be used as a high-throughput platform for protein-protein interaction, with the advantages of trace detection, short detective time, and high detective sensitivity.
Collapse
Affiliation(s)
- Zhenzhan Kuang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hao Deng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Lan Xu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Qianying Hu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yan Cai
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ruixue Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, People's Republic of China
| | - Shuhong Luo
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, People's Republic of China
| | - Tiancai Liu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
11
|
Zhang Y, Bewerunge-Hudler M, Schick M, Burwinkel B, Herpel E, Hoffmeister M, Brenner H. Blood-derived DNA methylation predictors of mortality discriminate tumor and healthy tissue in multiple organs. Mol Oncol 2020; 14:2111-2123. [PMID: 32506842 PMCID: PMC7463320 DOI: 10.1002/1878-0261.12738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/14/2020] [Accepted: 06/02/2020] [Indexed: 12/27/2022] Open
Abstract
Evidence has shown that certain methylation markers derived from blood can mirror corresponding methylation signatures in internal tissues. In the current study, we aimed to investigate two strong epigenetic predictors for life span, derived from blood DNA methylation data, in tissue samples of solid cancer patients. Using data from the Cancer Genome Atlas (TCGA) and the German DACHS study, we compared a mortality risk score (MRscore) and DNAmPhenoAge in paired tumor and adjacent normal tissue samples of patients with lung (N = 69), colorectal (n = 299), breast (n = 90), head/neck (n = 50), prostate (n = 50), and liver (n = 50) cancer. To explore the concordance across tissue and blood, we additionally assessed the two markers in blood samples of colorectal cancer (CRC) cases and matched controls (n = 93) in the DACHS+ study. The MRscore was significantly elevated in tumor tissues compared to normal tissues of all cancers except prostate cancer, for which an opposite pattern was observed. DNAmPhenoAge was consistently higher in all tumor tissues. The MRscore discriminated lung, colorectal, and prostate tumor tissues from normal tissues with very high accuracy [AUCs of 0.87, 0.99 (TCGA) /0.94 (DACHS), and 0.92, respectively]. DNAmPhenoAge accurately discriminated five types of tumor tissues from normal tissues (except prostate cancer), with AUCs of 0.82–0.93. The MRscore was also significantly higher in blood samples of CRC cases than in controls, with areas under the curve (AUC) of 0.74, whereas DNAmPhenoAge did not distinguish cases from controls, with AUC of 0.54. This study provides compelling evidence that blood‐derived DNAm markers could reflect methylation changes in less accessible tissues. Further research should explore the potential use of these findings for cancer diagnosis and early detection.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Bewerunge-Hudler
- Genomics and Proteomics Core Facilities, Microarray Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Schick
- Genomics and Proteomics Core Facilities, Microarray Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Burwinkel
- Molecular Epidemiology, German Cancer Research Center, Heidelberg, Germany.,Molecular Biology of Breast Cancer, Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Esther Herpel
- Department of General Pathology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,NCT Tissue Bank, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
12
|
Gutierrez E, Cahatol I, Bailey CAR, Lafargue A, Zhang N, Song Y, Tian H, Zhang Y, Chan R, Gu K, Zhang ACC, Tang J, Liu C, Connis N, Dennis P, Zhang C. Regulation of RhoB Gene Expression during Tumorigenesis and Aging Process and Its Potential Applications in These Processes. Cancers (Basel) 2019; 11:cancers11060818. [PMID: 31200451 PMCID: PMC6627600 DOI: 10.3390/cancers11060818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/01/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022] Open
Abstract
RhoB, a member of the Ras homolog gene family and GTPase, regulates intracellular signaling pathways by interfacing with epidermal growth factor receptor (EGFR), Ras, and phosphatidylinositol 3-kinase (PI3K)/Akt to modulate responses in cellular structure and function. Notably, the EGFR, Ras, and PI3K/Akt pathways can lead to downregulation of RhoB, while simultaneously being associated with an increased propensity for tumorigenesis. Functionally, RhoB, part of the Rho GTPase family, regulates intracellular signaling pathways by interfacing with EGFR, RAS, and PI3K/Akt/mammalian target of rapamycin (mTOR), and MYC pathways to modulate responses in cellular structure and function. Notably, the EGFR, Ras, and PI3K/Akt pathways can lead to downregulation of RhoB, while simultaneously being associated with an increased propensity for tumorigenesis. RHOB expression has a complex regulatory backdrop consisting of multiple histone deacetyltransferase (HDACs 1 and 6) and microRNA (miR-19a, -21, and -223)-mediated mechanisms of modifying expression. The interwoven nature of RhoB’s regulatory impact and cellular roles in regulating intracellular vesicle trafficking, cell motion, and the cell cycle lays the foundation for analyzing the link between loss of RhoB and tumorigenesis within the context of age-related decline in RhoB. RhoB appears to play a tissue-specific role in tumorigenesis, as such, uncovering and appreciating the potential for restoration of RHOB expression as a mechanism for cancer prevention or therapeutics serves as a practical application. An in-depth assessment of RhoB will serve as a springboard for investigating and characterizing this key component of numerous intracellular messaging and regulatory pathways that may hold the connection between aging and tumorigenesis.
Collapse
Affiliation(s)
- Eutiquio Gutierrez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA.
- Department of Internal Medicine, Harbor-UCLA Medical Center, 1000 W Carson Street, Torrance, CA 90509, USA.
| | - Ian Cahatol
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA
- Department of Graduate Medical Education, Community Memorial Health System, 147 N Brent Street, Ventura, CA 93003, USA
| | - Cedric A R Bailey
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E 2nd Street, Pomona, CA 91766, USA
- Department of Pathology and Immunology, Washington University School of Medicine, 509 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Audrey Lafargue
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Naming Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Ying Song
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Hongwei Tian
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Yizhi Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Ryan Chan
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Kevin Gu
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Angel C C Zhang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - James Tang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Chunshui Liu
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Nick Connis
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Phillip Dennis
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| | - Chunyu Zhang
- Department of Oncology, The Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, USA
| |
Collapse
|
13
|
Yeh SJ, Chang CA, Li CW, Wang LHC, Chen BS. Comparing progression molecular mechanisms between lung adenocarcinoma and lung squamous cell carcinoma based on genetic and epigenetic networks: big data mining and genome-wide systems identification. Oncotarget 2019; 10:3760-3806. [PMID: 31217907 PMCID: PMC6557199 DOI: 10.18632/oncotarget.26940] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the predominant type of lung cancer in the world. Lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC) are subtypes of NSCLC. We usually regard them as different disease due to their unique molecular characteristics, distinct cells of origin and dissimilar clinical response. However, the differences of genetic and epigenetic progression mechanism between LADC and LSCC are complicated to analyze. Therefore, we applied systems biology approaches and big databases mining to construct genetic and epigenetic networks (GENs) with next-generation sequencing data of LADC and LSCC. In order to obtain the real GENs, system identification and system order detection are conducted on gene regulatory networks (GRNs) and protein-protein interaction networks (PPINs) for each stage of LADC and LSCC. The core GENs were extracted via principal network projection (PNP). Based on the ranking of projection values, we got the core pathways in respect of KEGG pathway. Compared with the core pathways, we found significant differences between microenvironments, dysregulations of miRNAs, epigenetic modifications on certain signaling transduction proteins and target genes in each stage of LADC and LSCC. Finally, we proposed six genetic and epigenetic multiple-molecule drugs to target essential biomarkers in each progression stage of LADC and LSCC, respectively.
Collapse
Affiliation(s)
- Shan-Ju Yeh
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-An Chang
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Wei Li
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Lily Hui-Ching Wang
- Department of Medical Science, Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Bor-Sen Chen
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.,Department of Electrical Engineering, Yuan Ze University, Chungli 32003, Taiwan
| |
Collapse
|
14
|
Klimenko OV, Sidorov A. The full recovery of mice (Mus Musculus C57BL/6 strain) from virus-induced sarcoma after treatment with a complex of DDMC delivery system and sncRNAs. Noncoding RNA Res 2019; 4:69-78. [PMID: 31193489 PMCID: PMC6531865 DOI: 10.1016/j.ncrna.2019.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/22/2019] [Accepted: 03/22/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Virus-induced cellular genetic modifications result in the development of many human cancers. METHODS In our experiments, we used the RVP3 cell line, which produce primary mouse virus-induced sarcoma in 100% of cases. Inbreed 4-week-old female C57BL/6 mice were injected subcutaneously in the interscapular region with RVP3 cells. Three groups of mice were used. For treatment, one and/or two intravenous injections of a complex of small non-coding RNAs (sncRNAs) a-miR-155, piR-30074, and miR-125b with a 2-diethylaminoethyl-dextran methyl methacrylate copolymer (DDMC) delivery system were used. The first group consisted of untreated animals (control). The second group was treated with one injection of complex DDMC/sncRNAs (1st group). The third group was treated with two injections of complex DDMC/sncRNAs (2nd group). The tumors were removed aseptically, freed of necrotic material, and used with spleen and lungs for subsequent RT-PCR and immunofluorescence experiments, or stained with Leishman-Romanowski dye. RESULTS As a result, the mice fully recovered from virus-induced sarcoma after two treatments with a complex including the DDMC vector and a-miR-155, piR-30074, and miR-125b. In vitro studies showed genetic and morphological transformations of murine cancer cells after the injections. CONCLUSIONS Treatment of virus-induced sarcoma of mice with a-miR-155, piR-30074, and miR-125b as active component of anti-cancer complex and DDMC vector as delivery system due to epigenetic-regulated transformation of cancer cells into cells with non-cancerous physiology and morphology and full recovery of disease.
Collapse
Affiliation(s)
- Oxana V. Klimenko
- SID ALEX GROUP, Ltd., Kyselova 1185/2, Prague, 182 00, Czech Republic
| | | |
Collapse
|
15
|
Hu M, Jovanović B, Palić D. In silico prediction of MicroRNA role in regulation of Zebrafish (Danio rerio) responses to nanoparticle exposure. Toxicol In Vitro 2019; 60:187-202. [PMID: 31132477 DOI: 10.1016/j.tiv.2019.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/05/2019] [Accepted: 05/19/2019] [Indexed: 12/13/2022]
Abstract
The release of nanoparticles to the environment can affect health of the exposed organisms. MicroRNAs have been suggested as potential toxicology biomarkers, however the information about use of microRNA in aquatic organisms exposed to nanoparticles (NP) is limited. In silico analysis from publicly available gene expression data was performed. Data selection for the analysis was based on reported biological and pathological outcomes of NP induced toxicity in zebrafish. After identifying relevant genes, we constructed six miRNA-mRNA regulatory networks involved in nanoparticle induced toxicological responses in zebrafish. Based on our prediction and selection criteria we selected six miRNAs that overlapped in constructed networks with remarkable prediction score, and were validated by previous mammalian and zebrafish microRNA profiling studies: dre-miR-124, -144, -148, -155, -19a, -223. The results of this in silico analysis indicate that several highly conserved miRNAs likely have a regulatory role of organismal responses to nanoparticles, and can possibly be used as biomarkers of nanotoxicity in studies using zebrafish as model organism One health approaches.
Collapse
Affiliation(s)
- Moyan Hu
- Chair for Fish Diseases and Fisheries Biology, Faculty of Veterinary Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Boris Jovanović
- Department of Natural Resources Ecology and Management, Iowa State University, Ames, IA, USA
| | - Dušan Palić
- Chair for Fish Diseases and Fisheries Biology, Faculty of Veterinary Medicine, Ludwig Maximilian University of Munich, Munich, Germany.
| |
Collapse
|
16
|
MiR-19a as a prognostic indicator for cancer patients: a meta-analysis. Biosci Rep 2019; 39:BSR20182370. [PMID: 31015372 PMCID: PMC6522715 DOI: 10.1042/bsr20182370] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
MiR-19a was aberrantly expressed in various types of cancers and was observed to be potentially associated with the prognosis of cancer patients. The present analysis aims to elucidate its precise predictive value in various human malignancies. Online electronic searches of PubMed, Web of Science (WOS), Embase in English and VIP, Wanfang, SinoMed, and the China National Knowledge Infrastructure (CNKI) in Chinese up to September 8, 2018 were conducted. As a result, in overall analysis, a significant association was identified between miR-19a levels and OS (HRs = 2.31, CI: 1.11–4.83). The relation of miR-19a expression to OS was further recognized by fixed model within the studies of sample size less than 150 (HRs = 1.68, CI: 1.35–2.08), NOS scores greater than or equal to 8 (HRs = 1.53, CI: 1.13–2.06) or less than 8 (HRs = 1.89, CI: 1.58–2.27), specimen derived from tumor (HRs = 1.73, CI: 1.42–2.12) or blood (HRs = 1.87, CI: 1.46–2.40) and the patients of osteosarcoma (HRs = 7.17, CI: 5.04–10.21). Sensitivity analyses revealed no significant results. The association between miR-19a expression level and DFS was also found to be significant (HRs = 2.03, CI: 1.13–3.66). Correlations between miR-19a levels and clinicopathological features were examined and revealed that lymph node metastasis was significantly associated with miR-19a expression levels (OR = 0.565, CI: 0.346–0.921). Summarily, the over expression of miR-19a was an underlying risk of poor prognosis in many human malignancies, especially in osteosarcoma. Moreover, elevated miR-19a expression was linked to the potential of lymph node metastasis.
Collapse
|
17
|
Li X, Yan X, Wang F, Yang Q, Luo X, Kong J, Ju S. Down-regulated lncRNA SLC25A5-AS1 facilitates cell growth and inhibits apoptosis via miR-19a-3p/PTEN/PI3K/AKT signalling pathway in gastric cancer. J Cell Mol Med 2019; 23:2920-2932. [PMID: 30793479 PMCID: PMC6433659 DOI: 10.1111/jcmm.14200] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/17/2018] [Accepted: 01/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence has illustrated the vital roles of long non-coding RNAs (lncRNAs in gastric cancer (GC). Nevertheless, the majority of their roles and mechanisms in GC are still largely unknown. In this study, we investigate the roles of lncRNA SLC25A5-AS1 on tumourigenesis and explore its potential mechanisms in GC. The results showed that the expressions of SLC25A5-AS1 in GC were significantly lower than that of adjacent normal tissues, which were significantly associated with tumour size, TNM stage and lymph node metastasis. Moreover, SLC25A5-AS1 could inhibit GC cell proliferation, induce G1/G1 cell cycle arrest and cell apoptosis in vitro, as well as GC growth in vivo. Dual-luciferase reporter assay confirmed the direct interaction between SLC25A5-AS1 and miR-19a-3p, rescue experiment showed that co-transfection miR-19a-3p mimics and pcDNA-SLC25A5-AS1 could partially restore the ability of GC cell proliferation and the inhibition of cell apoptosis. The mechanism analyses further found that SLC25A5-AS1 might act as a competing endogenous RNAs (ceRNA), which was involved in the derepression of PTEN expression, a target gene of miR-19a-3p, and regulate malignant phenotype via PI3K/AKT signalling pathway in GC. Taken together, this study indicated that SLC25A5-AS1 was down-regulated in GC and functioned as a suppressor in the progression of GC. Moreover, it could act as a ceRNA to regulate cellular behaviours via miR-19a-3p/PTEN/PI3K/AKT signalling pathway. Thus, SLC25A5-AS1 might be served as a potential target for cancer therapeutics in GC.
Collapse
Affiliation(s)
- Xiwen Li
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
- Department of Clinical LaboratoryTraditional Chinese Medicine HospitalKunshanChina
| | - Xin Yan
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Feng Wang
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
| | - Qian Yang
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
| | - Xi Luo
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
| | - Jun Kong
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
| | - Shaoqing Ju
- Laboratory Medicine CenterAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
18
|
Luo H, Liang C. MicroRNA-148b inhibits proliferation and the epithelial-mesenchymal transition and increases radiosensitivity in non-small cell lung carcinomas by regulating ROCK1. Exp Ther Med 2018; 15:3609-3616. [PMID: 29545890 DOI: 10.3892/etm.2018.5845] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/07/2017] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for ~80% of all types of lung cancer, which has the highest morbidity and mortality of all types of cancer worldwide. It is important to identify novel biomarkers and the molecular mechanism of NSCLC to improve current treatments of NSCLC. The present study aimed to investigate the effect of miR-148b expression on the proliferation, epithelial-mesenchymal transition (EMT) and radiosensitivity of NSCLC cells. It was demonstrated that miR-148b expression was significantly decreased in NSCLC tissues and cell lines. A549 cells were then transfected with a miR-148b mimic and a miR-148b inhibitor. Transfection with the miR-148b mimic decreased proliferation whereas transfection with the miR-148b inhibitor increased the proliferation of A549 cells. Additionally, the miR-148b mimic increased E-cadherin expression and decreased N-cadherin and vimentin expression. By contrast, transfection with the miR-148b inhibitor decreased E-cadherin expression and increased N-cadherin and vimentin expression. Irradiation-induced cell death was significantly promoted by the miR-148b mimic but inhibited by the miR-148b inhibitor. The miR-148b mimic significantly decreased the expression of Rho-associated protein kinase 1 (ROCK1) and it was demonstrated that overexpression of ROCK1 significantly inhibited the effects of miR-148b on cell proliferation, the EMT and irradiation-induced cell death. Therefore, the current study revealed that miR-148b inhibited NSCLC cell proliferation and the EMT, and increased the radiosensitivity of NSCLC cells by inhibiting ROCK1 expression. Therefore, miR-148b/ROCK1 signaling may be a novel therapeutic target to inhibit the growth of NSCLC cells and enhance the effects of radiotherapy to treat patients with NSCLC.
Collapse
Affiliation(s)
- Haitao Luo
- Department of Oncology 2 Division, Foshan Nanhai District People's Hospital, Foshan, Guangdong 528200, P.R. China
| | - Caixia Liang
- Department of Respiratory Medicine, Foshan Nanhai District People's Hospital, Foshan, Guangdong 528200, P.R. China
| |
Collapse
|
19
|
Zhou Q, Huang SX, Zhang F, Li SJ, Liu C, Xi YY, Wang L, Wang X, He QQ, Sun CC, Li DJ. MicroRNAs: A novel potential biomarker for diagnosis and therapy in patients with non-small cell lung cancer. Cell Prolif 2017; 50. [PMID: 28990243 DOI: 10.1111/cpr.12394] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/09/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Lung cancer is still one of the most serious causes of cancer-related deaths all over the world. MicroRNAs (miRNAs) are defined as small non-coding RNAs which could play a pivotal role in post-transcriptional regulation of gene expression. Increasing evidence demonstrated dysregulation of miRNA expression associates with the development and progression of NSCLC. AIMS To emphasize a variety of tissue-specific miRNAs, circulating miRNAs and miRNA-derived exosomes could be used as potential diagnostic and therapeutic biomarkers in NSCLC patients. MATERIALS & METHODS In the current review, we paid attention to the significant discoveries of preclinical and clinical studies, which performed on tissue-specific miRNA, circulating miRNA and exosomal miRNA. The related studies were obtained through a systematic search of Pubmed, Web of Science, Embase. RESULTS A variety of tissue-specific miRNAs and circulating miRNAs with high sensitivity and specificity which could be used as potential diagnostic and therapeutic biomarkers in NSCLC patients. In addition, we emphasize that the miRNA-derived exosomes become novel diagnostic biomarkers potentially in these patients with NSCLC. CONCLUSION MiRNAs have emerged as non-coding RNAs, which have potential to be candidates for the diagnosis and therapy of NSCLC.
Collapse
Affiliation(s)
- Qun Zhou
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Shao-Xin Huang
- Department of Social Medicine and Public Health, School of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi, China
| | - Feng Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Shu-Jun Li
- Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, Wuhan, 430022, Hubei, China
| | - Cong Liu
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Yong-Yong Xi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Liang Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Xin Wang
- Department of Social Medicine and Public Health, School of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi, China
| | - Qi-Qiang He
- Department of School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| | - De-Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, China
| |
Collapse
|
20
|
Zhang L, Li J, Wang Q, Meng G, Lv X, Zhou H, Li W, Zhang J. The relationship between microRNAs and the STAT3-related signaling pathway in cancer. Tumour Biol 2017; 39:1010428317719869. [PMID: 28859543 DOI: 10.1177/1010428317719869] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs are non-coding RNAs that regulate gene expression by targeting messenger RNA molecules in 3' untranslated region. Mounting evidence indicates that microRNAs regulate several factors to influence various biological activities that are related to carcinogenesis, including signal transducer and activator of transcription 3, which is a transcription factor that also acts as an oncogene. MicroRNAs influence signal transducer and activator of transcription 3 either by directly targeting or via other pathway components upstream or downstream of signal transducer and activator of transcription 3 such as Janus kinases, members of the suppressor of cytokine signaling family, and other genes that regulate cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition. However, signal transducer and activator of transcription 3 activation changes the pattern of expression of microRNAs and mediates tumorigenesis. Moreover, the relationship between signal transducer and activator of transcription 3 and microRNAs varies among different kinds of cancers. A specific microRNA may act as an oncogene or tumor suppressor in different cancers, and microRNAs also directly or indirectly regulate signal transducer and activator of transcription 3 via pathways in the same cancers. In this review, we focus on the reciprocal regulation and roles of microRNAs and signal transducer and activator of transcription 3 in cancer, as well as describe current research progress on this relationship. A better understanding of this relationship may facilitate in the identification of targets for clinical therapeutics.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Junyao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Guangping Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Xuejiao Lv
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Hong Zhou
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Wei Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
21
|
miR-19a contributes to gefitinib resistance and epithelial mesenchymal transition in non-small cell lung cancer cells by targeting c-Met. Sci Rep 2017; 7:2939. [PMID: 28592790 PMCID: PMC5462753 DOI: 10.1038/s41598-017-01153-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 06/06/2016] [Indexed: 12/22/2022] Open
Abstract
Gefitinib, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, is used as a first-line treatment for advanced non-small cell lung cancer (NSCLC). However, most NSCLC patients inevitably develop gefitinib resistance, and the mechanisms underlying this resistance are not fully understood. In this study, we show that miR-19a is significantly down-regulated in gefitinib-resistant NSCLC cell lines compared with gefitinib-sensitive cell lines. In addition, the down-regulation of miR-19a suppressed the expression of epithelial markers but induced the expression levels of mesenchymal markers. A mechanistic analysis revealed that miR-19a regulated c-Met expression by directly targeting the c-Met 3′UTR. Overexpression of miR-19a decreased c-Met expression and re-sensitized gefitinib-resistant NSCLC cells in vitro and in vivo. Consistent with the in vitro findings, the miR-19a serum level was significantly decreased in NSCLC patients with acquired gefitinib resistance compared with the level observed prior to the acquisition of resistance in each patient, indicating that miR-19a expression may be a valuable biomarker for the prediction of acquired gefitinib resistance in a clinical setting. Our data demonstrate that the miR-19a/c-Met pathway plays a critical role in acquired resistance to gefitinib and that the manipulation of miR-19a might provide a therapeutic strategy for overcoming acquired gefitinib resistance.
Collapse
|
22
|
Chen CH, Li SH. A novel therapeutic approach for esophageal squamous cell carcinoma: suppressor of cytokine signaling-1 gene therapy. J Thorac Dis 2017; 9:1446-1449. [PMID: 28740654 PMCID: PMC5506160 DOI: 10.21037/jtd.2017.05.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Chang-Han Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Applied Chemistry, Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Taiwan.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
23
|
He M, Xue Y. MicroRNA-148a suppresses proliferation and invasion potential of non-small cell lung carcinomas via regulation of STAT3. Onco Targets Ther 2017; 10:1353-1361. [PMID: 28280370 PMCID: PMC5338933 DOI: 10.2147/ott.s123518] [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] [Indexed: 12/21/2022] Open
Abstract
Lung cancer has the highest morbidity and mortality in the world, and non-small cell lung carcinomas (NSCLC) account for 80% of cases of lung cancer. The mechanism of NSCLC is still largely unknown, and finding novel targets is of great importance for the treatment of NSCLC. The current study was designed to evaluate the role of miR-148a in NSCLC cell proliferation and invasion and to investigate the possible molecular mechanisms. We found that miR-148a expression was decreased in NSCLC tissues and cell lines. Upregulation of miR-148a significantly decreased A549 cell proliferation, and downregulation of miR-148a significantly increased A549 cell proliferation. Upregulation of miR-148a markedly increased apoptotic cell death and inhibited cell invasion potential. Upregulation of miR-148a significantly decreased signal transducer and activator of transcription 3 (STAT3) expression and 3′-untranslated region luciferase activity. Downregulation of miR-148a significantly increased STAT3 expression. Overexpression of STAT3 significantly inhibited the effect of miR-148a on cell viability and invasion potential. In conclusion, we found that miR-148a inhibited NSCLC cell proliferation and invasion potential through the inhibition of STAT3. Our findings highlight miR-148a/STAT3 axis as a novel therapeutic target for the inhibition of NSCLC growth.
Collapse
Affiliation(s)
- Mei He
- Department of Respiratory Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, People's Republic of China
| | - Yan Xue
- Department of Respiratory Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
24
|
Jiang M, Zhang WW, Liu P, Yu W, Liu T, Yu J. Dysregulation of SOCS-Mediated Negative Feedback of Cytokine Signaling in Carcinogenesis and Its Significance in Cancer Treatment. Front Immunol 2017; 8:70. [PMID: 28228755 PMCID: PMC5296614 DOI: 10.3389/fimmu.2017.00070] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/16/2017] [Indexed: 01/30/2023] Open
Abstract
Suppressor of cytokine signaling (SOCS) proteins are major negative feedback regulators of cytokine signaling mediated by the Janus kinase (JAK)-signal transducer and activator of transcription signaling pathway. In particular, SOCS1 and SOCS3 are strong inhibitors of JAKs and can play pivotal roles in the development and progression of cancers. The abnormal expression of SOCS1 and SOCS3 in cancer cells is associated with the dysregulation of cell growth, migration, and death induced by multiple cytokines and hormones in human carcinomas. In addition, the mechanisms involved in SOCS1- and SOCS3-regulated abnormal development and activation of immune cells in carcinogenesis, including T cells, macrophages, dendritic cells, and myeloid-derived suppressor cells, are still unclear. Therefore, this study aims to further discuss the molecules and signal pathways regulating the expression and function of SOCS1 and SOCS3 in various types of cancers and elucidate the feasibility and efficiency of SOCS-based target therapeutic strategy in anticancer treatment.
Collapse
Affiliation(s)
- Mengmeng Jiang
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wen-Wen Zhang
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Pengpeng Liu
- Cancer Molecular Diagnostic Center, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Wenwen Yu
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Liu
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jinpu Yu
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Cancer Molecular Diagnostic Center, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
25
|
Abba ML, Patil N, Leupold JH, Moniuszko M, Utikal J, Niklinski J, Allgayer H. MicroRNAs as novel targets and tools in cancer therapy. Cancer Lett 2017; 387:84-94. [DOI: 10.1016/j.canlet.2016.03.043] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/24/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
|
26
|
Elevated expression of microRNA-19a predicts a poor prognosis in patients with osteosarcoma. Pathol Res Pract 2016; 213:194-198. [PMID: 28214202 DOI: 10.1016/j.prp.2016.12.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/23/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022]
Abstract
MicroRNA (miR)-19a, a member of the miR-17-92 cluster, functions as an oncomiRNA in multiple kinds of cancers. However, its involvement in human osteosarcomas remains unclear. In this study, to analyze the expression pattern of miR-19a and to investigate its clinical implication in human osteosarcomas, quantitative reverse-transcription polymerase chain reaction was performed to detect expression levels of miR-19a in 166 self-pairs of osteosarcoma and noncancerous bone tissues. Associations between miR-19a expression and various clinicopathological parameters and patients' prognosis of osteosarcomas were further evaluated. As a results, miR-19a expression in osteosarcoma tissues was significantly higher than that in corresponding noncancerous bone tissues (P<0.001). Osteosarcoma patients with high miR-19a expression more frequently had large tumor size (P=0.03), advanced clinical stage (P=0.01), positive distant metastasis (P=0.008) and poor response to chemotherapy (P=0.01) than those with low miR-19a expression. Additionally, kaplan-Meier analysis showed that both overall and disease-free survivals of osteosarcoma patients with high miR-19a expression were shorter than those with low miR-19a expression (both P<0.001). Further multivariate analysis identified miR-19a expression as an independent prognostic factor for both overall (P=0.001) and disease-free (P=0.006) survivals. In conclusion, the aberrant expression of miR-19a may play a crucial role in development and progression of human osteosarcomas. MiR-19a may act as a novel prognostic marker for patients with this malignancy.
Collapse
|
27
|
Xue J, Yang J, Luo M, Cho WC, Liu X. MicroRNA-targeted therapeutics for lung cancer treatment. Expert Opin Drug Discov 2016; 12:141-157. [PMID: 27866431 DOI: 10.1080/17460441.2017.1263298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Lung cancer is one of the leading causes of cancer-related mortality worldwide. MicroRNAs (miRNAs) are endogenous non-coding small RNAs that repress the expression of a broad array of target genes. Many efforts have been made to therapeutically target miRNAs in cancer treatments using miRNA mimics and miRNA antagonists. Areas covered: This article summarizes the recent findings with the role of miRNAs in lung cancer, and discusses the potential and challenges of developing miRNA-targeted therapeutics in this dreadful disease. Expert opinion: The development of miRNA-targeted therapeutics has become an important anti-cancer strategy. Results from both preclinical and clinical trials of microRNA replacement therapy have shown some promise in cancer treatment. However, some obstacles, including drug delivery, specificity, off-target effect, toxicity mediation, immunological activation and dosage determination should be addressed. Several delivery strategies have been employed, including naked oligonucleotides, liposomes, aptamer-conjugates, nanoparticles and viral vectors. However, delivery remains a main challenge in miRNA-targeting therapeutics. Furthermore, immune-related serious adverse events are also a concern, which indicates the complexity of miRNA-based therapy in clinical settings.
Collapse
Affiliation(s)
- Jing Xue
- a Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , China.,b College of Life Science , Ningxia University , Yinchuan , China
| | - Jiali Yang
- a Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Meihui Luo
- a Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , China
| | - William C Cho
- c Department of Clinical Oncology , Queen Elizabeth Hospital , Kowloon , Hong Kong
| | - Xiaoming Liu
- a Center of Laboratory Medicine , General Hospital of Ningxia Medical University , Yinchuan , China.,b College of Life Science , Ningxia University , Yinchuan , China.,d Human Stem Cell Institute , General Hospital of Ningxia Medical University , Yinchuan , Ningxia , China
| |
Collapse
|
28
|
Vencken SF, Greene CM. Toll-Like Receptors in Cystic Fibrosis: Impact of Dysfunctional microRNA on Innate Immune Responses in the Cystic Fibrosis Lung. J Innate Immun 2016; 8:541-549. [PMID: 27043239 DOI: 10.1159/000444687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 02/15/2016] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that are particularly expressed in the sentinel and epithelial cells in the body, including the lung. They are central players in the innate immune system in response to microbial infection, and are the triggers of a complex pathway network that both promotes the inflammatory response and influences the adaptive immune response. These pathways are transiently and finely tuned by cellular factors, including a cell's microRNA response program. MicroRNAs are small, non-coding RNAs that specifically regulate gene expression. In this article, we review the disease-specific microRNA regulatory network of cystic fibrosis, a debilitating and ultimately fatal disease and, specifically, its effect on TLR signalling.
Collapse
Affiliation(s)
- Sebastian F Vencken
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | |
Collapse
|
29
|
Das SG, Romagnoli M, Mineva ND, Barillé-Nion S, Jézéquel P, Campone M, Sonenshein GE. miR-720 is a downstream target of an ADAM8-induced ERK signaling cascade that promotes the migratory and invasive phenotype of triple-negative breast cancer cells. Breast Cancer Res 2016; 18:40. [PMID: 27039296 PMCID: PMC4818899 DOI: 10.1186/s13058-016-0699-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/14/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ADAM8 (a disintegrin and metalloproteinase 8) protein promotes the invasive and metastatic phenotype of triple-negative breast cancer (TNBC) cells. High ADAM8 expression in breast cancer patients is an independent predictor of poor prognosis. Here, we investigated whether ADAM8 regulates specific miRNAs, their roles in aggressive phenotype, and potential use as biomarkers of disease. METHODS Microarray analysis was performed on RNA from MDA-MB-231 cells after transient ADAM8 knockdown using TaqMan miRNA cards. Changes in miRNA levels were confirmed using two ADAM8 siRNAs in TNBC cell lines. Kinase inhibitors, β1-integrin antagonist antibody, and different forms of ADAM8 were employed to elucidate the signaling pathway required for miR-720 expression. miR-720 levels were modulated using a specific antagomiR or a mimic, and effects on aggressive phenotype of TNBC cells were determined using Boyden chamber and 3D-Matrigel outgrowth assays. Plasma was isolated from mice before and after implantation of MDA-MB-231 cells and analyzed for miR-720 levels. Serum samples of TNBC patients were evaluated for their ADAM8 and miR-720 levels. RESULTS We identified 68 miRNAs differentially regulated upon ADAM8 knockdown, including decreased levels of secreted miR-720. Ectopic overexpression of wild-type ADAM8 or forms that lack metalloproteinase activity similarly induced miR-720 levels. The disintegrin and cysteine-rich domains of ADAM8 were shown to induce miR-720 via activation of a β1-integrin to ERK signaling cascade. Knockdown of miR-720 led to a significant decrease in migratory and invasive abilities of TNBC cells. Conversely, miR-720 overexpression rescued these properties. A profound increase in plasma levels of miR-720 was detected 7 days after TNBC cell inoculation into mouse mammary fat pads when tumors were barely palpable. Concordantly, miR-720 levels were found to be significantly higher in serum samples of TNBC patients with high ADAM8 expression. CONCLUSIONS We have shown for the first time that miR-720 is induced by ADAM8 signaling via ERK and plays an essential role in promoting the aggressive phenotype of TNBCs. miR-720 is elevated in serum of patients with ADAM8-high TNBC and, in a group with other miRNAs downstream of ADAM8, holds promise as a biomarker for early detection of or treatment response of ADAM8-positive TNBCs.
Collapse
Affiliation(s)
- Sonia G. Das
- />Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111 USA
| | - Mathilde Romagnoli
- />Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111 USA
- />Present address: Institut Curie, Centre de Recherche, UMR 144, 26 Rue d’Ulm, 75248 Paris, France
| | - Nora D. Mineva
- />Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111 USA
| | | | - Pascal Jézéquel
- />INSERM U892, IRT-UN, 8 quai Moncousu, 44007 Nantes Cedex, France
- />Institut de Cancérologie de Nantes, Centre de Lutte Contre le Cancer René Gauducheau, Boulevard Jacques Monod, 44 805 Saint-Herblain-Nantes Cedex, France
| | - Mario Campone
- />INSERM U892, IRT-UN, 8 quai Moncousu, 44007 Nantes Cedex, France
- />Institut de Cancérologie de Nantes, Centre de Lutte Contre le Cancer René Gauducheau, Boulevard Jacques Monod, 44 805 Saint-Herblain-Nantes Cedex, France
| | - Gail E. Sonenshein
- />Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111 USA
| |
Collapse
|
30
|
McCormick SM, Heller NM. Regulation of Macrophage, Dendritic Cell, and Microglial Phenotype and Function by the SOCS Proteins. Front Immunol 2015; 6:549. [PMID: 26579124 PMCID: PMC4621458 DOI: 10.3389/fimmu.2015.00549] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022] Open
Abstract
Macrophages are innate immune cells of dynamic phenotype that rapidly respond to external stimuli in the microenvironment by altering their phenotype to respond to and to direct the immune response. The ability to dynamically change phenotype must be carefully regulated to prevent uncontrolled inflammatory responses and subsequently to promote resolution of inflammation. The suppressor of cytokine signaling (SOCS) proteins play a key role in regulating macrophage phenotype. In this review, we summarize research to date from mouse and human studies on the role of the SOCS proteins in determining the phenotype and function of macrophages. We will also touch on the influence of the SOCS on dendritic cell (DC) and microglial phenotype and function. The molecular mechanisms of SOCS function in macrophages and DCs are discussed, along with how dysregulation of SOCS expression or function can lead to alterations in macrophage/DC/microglial phenotype and function and to disease. Regulation of SOCS expression by microRNA is discussed. Novel therapies and unanswered questions with regard to SOCS regulation of monocyte-macrophage phenotype and function are highlighted.
Collapse
Affiliation(s)
- Sarah M McCormick
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| | - Nicola M Heller
- Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA ; Anesthesiology and Critical Care Medicine, The Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|