1
|
Charbit H, Lavon I. Investigating Expression Dynamics of miR-21 and miR-10b in Glioblastoma Cells In Vitro: Insights into Responses to Hypoxia and Secretion Mechanisms. Int J Mol Sci 2024; 25:7984. [PMID: 39063226 PMCID: PMC11277016 DOI: 10.3390/ijms25147984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma poses significant challenges in oncology, with bevacizumab showing promise as an antiangiogenic treatment but with limited efficacy. microRNAs (miRNAs) 10b and 21 have emerged as potential biomarkers for bevacizumab response in glioblastoma patients. This study delves into the expression dynamics of miR-21 and miR-10b in response to hypoxia and explores their circulation mechanisms. In vitro experiments exposed glioma cells (A172, U87MG, U251) and human umbilical vein endothelial cells (HUVEC) to hypoxic conditions (1% oxygen) for 24 h, revealing heightened levels of miR-10b and miR-21 in glioblastoma cells. Manipulating miR-10b expression in U87MG, demonstrating a significant decrease in VEGF alpha (VEGFA) following miR-10b overexpression under hypoxic conditions. Size exclusion chromatography illustrated a notable shift towards miR-21 and miR-10b exosomal packaging during hypoxia. A proposed model suggests that effective bevacizumab treatment reduces VEGFA levels, heightening hypoxia and subsequently upregulating miR-21 and miR-10b expression. These miRNAs, released via exosomes, might impact various cellular processes, with miR-10b notably contributing to VEGFA level reduction. However, post-treatment increases in miR-10b and miR-21 could potentially restore cells to normoxic conditions through the downregulation of VEGF. This study highlights the intricate feedback loop involving miR-10b, miR-21, and VEGFA in glioblastoma treatment, underscoring the necessity for personalized therapeutic strategies. Further research should explore clinical implications for personalized glioma treatments.
Collapse
Affiliation(s)
| | - Iris Lavon
- Leslie and Michael Gaffin Center for Neuro-Oncology, Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
2
|
Abdallah M, Aziz IH, Alsammarraie AZ. Assessment of miRNA-10b Expression Levels as a Potential Precursor to Metastasis in Localized and Locally Advanced/Metastatic Breast Cancer among Iraqi Patients. Int J Breast Cancer 2024; 2024:2408355. [PMID: 38450330 PMCID: PMC10917482 DOI: 10.1155/2024/2408355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 03/08/2024] Open
Abstract
Breast cancer (BC) stands as the most prevalent form of carcinoma among women, ranking as the second leading cause of cancer-related mortality in the female population. The objective of this study is to assess the expression of miR-10b and determine its diagnostic and prognostic significance in breast cancer patients across various disease stages. The investigation was carried out in Baghdad at the Oncology Teaching Hospital within Baghdad Medical City and the Oncology Unit at Al-Yarmouk Teaching Hospital. A total of 150 samples were included and divided into two groups: the blood group consisting of 90 samples (including control subjects, localized BC patients, and those with metastatic and locally advanced BC) and the tissue group comprising 60 samples (representing both benign and malignant BC cases). The study spanned from March 2022 to January 2023, with patients' ages ranging from 24 to 75 years. The primary focus of this investigation was to identify the gene expression of miRNA-10b in all sample types. This was achieved by measuring gene expression levels and normalizing them to the level of a housekeeping gene (U6), and quantification was carried out considering the ΔCt value and the fold change method (2-ΔΔCt). The results revealed an upregulated fold expression of miRNA-10b, particularly in locally advanced and metastatic BC, where the expression was significantly higher compared to the other groups, with a fold expression of 1.770 ± 0.1070. In localized breast cancer, the fold expression was 1.624 ± 0.064, and in malignant tissue, it measured 1.546 ± 0.06754, all relative to apparently healthy control subjects. In summary, our research provides compelling evidence supporting the classification of miRNA-10b as an oncogenic factor in BC. The central involvement of miRNA-10b in the tumorigenic processes of BC highlights its reference for developing novel targeted therapeutic interventions and detection biomarkers for BC treatment. Notably, elevated expression of miRNA-10b was observed in BC tissues, correlating with an unfavorable distant metastasis-free survival outcome.
Collapse
Affiliation(s)
- Mays Abdallah
- Institute of Genetic Engineering and Biotechnology, University of Baghdad, Baghdad, Iraq
| | - Ismail H. Aziz
- Institute of Genetic Engineering and Biotechnology, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
3
|
Abdullaev B, Rasyid SA, Ali E, Al-Dhalimy AMB, Mustafa YF, Fenjan MN, Misra N, Al-Musawi SG, Alawadi A, Alsalamy A. Effective exosomes in breast cancer: focusing on diagnosis and treatment of cancer progression. Pathol Res Pract 2024; 253:154995. [PMID: 38113765 DOI: 10.1016/j.prp.2023.154995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Breast cancer (BC) is the most prevalent aggressive malignant tumor in women worldwide and develops from breast tissue. Although cutting-edge treatment methods have been used and current mortality rates have decreased, BC control is still not satisfactory. Clarifying the underlying molecular mechanisms will help clinical options. Extracellular vesicles known as exosomes mediate cellular communication by delivering a variety of biomolecules, including proteins, oncogenes, oncomiRs, and even pharmacological substances. These transferable bioactive molecules can alter the transcriptome of target cells and affect signaling pathways that are related to tumors. Numerous studies have linked exosomes to BC biology, including therapeutic resistance and the local microenvironment. Exosomes' roles in tumor treatment resistance, invasion, and BC metastasis are the main topics of discussion in this review.
Collapse
Affiliation(s)
- Bekhzod Abdullaev
- Research Department of Biotechnology, New Uzbekistan University, Tashkent, Uzbekistan; Department of Oncology, School of Medicine, Central Asian University, Tashkent, Uzbekistan.
| | - Sri Anggarini Rasyid
- Faculty of Science and Technology, Mandala Waluya University, Kendari, South East Sulawesi, Indonesia.
| | - Eyhab Ali
- college of chemistry, Al-Zahraa University for Women, Karbala, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Iraq
| | - Mohammed N Fenjan
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Neeti Misra
- Department of Management, Uttaranchal Institute of Management, Uttaranchal University, India
| | | | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Iraq
| |
Collapse
|
4
|
H. Al-Zuaini H, Rafiq Zahid K, Xiao X, Raza U, Huang Q, Zeng T. Hypoxia-driven ncRNAs in breast cancer. Front Oncol 2023; 13:1207253. [PMID: 37583933 PMCID: PMC10424730 DOI: 10.3389/fonc.2023.1207253] [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: 04/17/2023] [Accepted: 07/06/2023] [Indexed: 08/17/2023] Open
Abstract
Low oxygen tension, or hypoxia is the driving force behind tumor aggressiveness, leading to therapy resistance, metastasis, and stemness in solid cancers including breast cancer, which now stands as the leading cause of cancer-related mortality in women. With the great advancements in exploring the regulatory roles of the non-coding genome in recent years, the wide spectrum of hypoxia-responsive genome is not limited to just protein-coding genes but also includes multiple types of non-coding RNAs, such as micro RNAs, long non-coding RNAs, and circular RNAs. Over the years, these hypoxia-responsive non-coding molecules have been greatly implicated in breast cancer. Hypoxia drives the expression of these non-coding RNAs as upstream modulators and downstream effectors of hypoxia inducible factor signaling in the favor of breast cancer through a myriad of molecular mechanisms. These non-coding RNAs then contribute in orchestrating aggressive hypoxic tumor environment and regulate cancer associated cellular processes such as proliferation, evasion of apoptotic death, extracellular matrix remodeling, angiogenesis, migration, invasion, epithelial-to-mesenchymal transition, metastasis, therapy resistance, stemness, and evasion of the immune system in breast cancer. In addition, the interplay between hypoxia-driven non-coding RNAs as well as feedback and feedforward loops between these ncRNAs and HIFs further contribute to breast cancer progression. Although the current clinical implications of hypoxia-driven non-coding RNAs are limited to prognostics and diagnostics in breast cancer, extensive explorations have established some of these hypoxia-driven non-coding RNAs as promising targets to treat aggressive breast cancers, and future scientific endeavors hold great promise in targeting hypoxia-driven ncRNAs at clinics to treat breast cancer and limit global cancer burden.
Collapse
Affiliation(s)
| | - Kashif Rafiq Zahid
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiangyan Xiao
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Qiyuan Huang
- Department of Clinical Biobank Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
5
|
Wang L, Qiao C, Cao L, Cai S, Ma X, Song X, Jiang Q, Huang C, Wang J. Significance of HOXD transcription factors family in progression, migration and angiogenesis of cancer. Crit Rev Oncol Hematol 2022; 179:103809. [PMID: 36108961 DOI: 10.1016/j.critrevonc.2022.103809] [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: 03/04/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 10/31/2022] Open
Abstract
The transcription factors (TFs) of the HOX family play significant roles during early embryonic development and cellular processes. They also play a key role in tumorigenesis as tumor oncogenes or suppressors. Furthermore, TFs of the HOXD geFIne cluster affect proliferation, migration, and invasion of tumors. Consequently, dysregulated activity of HOXD TFs has been linked to clinicopathological characteristics of cancer. HOXD TFs are regulated by non-coding RNAs and methylation of DNA on promoter and enhancer regions. In addition, HOXD genes modulate the biological function of cancer cells via the MEK and AKT signaling pathways, thus, making HOXD TFs, a suitable molecular marker for cancer prognosis and therapy. In this review, we summarized the roles of HOXD TFs in different cancers and highlighted its potential as a diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Lumin Wang
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Institute of precision medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Chenyang Qiao
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Li Cao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Shuang Cai
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Xiaoping Ma
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Xinqiu Song
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an, Shaanxi, PR China
| | - Qiuyu Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China.
| | - Jinhai Wang
- Gastroenterology department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Institute of precision medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
6
|
Sacco A, Martelli F, Pal A, Saraceno C, Benussi L, Ghidoni R, Rongioletti M, Squitti R. Regulatory miRNAs in Cardiovascular and Alzheimer's Disease: A Focus on Copper. Int J Mol Sci 2022; 23:3327. [PMID: 35328747 PMCID: PMC8948703 DOI: 10.3390/ijms23063327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), are key regulators of differentiation and development. In the cell, transcription factors regulate the production of miRNA in response to different external stimuli. Copper (Cu) is a heavy metal and an essential micronutrient with widespread industrial applications. It is involved in a number of vital biological processes encompassing respiration, blood cell line maturation, and immune responses. In recent years, the link between deregulation of miRNAs' functionality and the development of various pathologies as well as cardiovascular diseases (CVDs) has been extensively studied. Alzheimer's disease (AD) is the most common cause of dementia in the elderly with a complex disease etiology, and its link with Cu abnormalities is being increasingly studied. A direct interaction between COMMD1, a regulator of the Cu pathway, and hypoxia-inducible factor (HIF) HIF-1a does exist in ischemic injury, but little information has been collected on the role of Cu in hypoxia associated with AD thus far. The current review deals with this matter in an attempt to structurally discuss the link between miRNA expression and Cu dysregulation in AD and CVDs.
Collapse
Affiliation(s)
- Anna Sacco
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy;
| | - Amit Pal
- Department of Biochemistry, AIIMS, Kalyani 741245, India;
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (C.S.); (L.B.); (R.G.)
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (C.S.); (L.B.); (R.G.)
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (C.S.); (L.B.); (R.G.)
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita Fatebenefratelli Hospital, Isola Tiberina, 00186 Rome, Italy;
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (C.S.); (L.B.); (R.G.)
| |
Collapse
|
7
|
He G, Peng X, Wei S, Yang S, Li X, Huang M, Tang S, Jin H, Liu J, Zhang S, Zheng H, Fan Q, Liu J, Yang L, Li H. Exosomes in the hypoxic TME: from release, uptake and biofunctions to clinical applications. Mol Cancer 2022; 21:19. [PMID: 35039054 PMCID: PMC8762953 DOI: 10.1186/s12943-021-01440-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a remarkable trait of the tumor microenvironment (TME). When facing selective pressure, tumor cells show various adaptive characteristics, such as changes in the expression of cancer hallmarks (increased proliferation, suppressed apoptosis, immune evasion, and so on) and more frequent cell communication. Because of the adaptation of cancer cells to hypoxia, exploring the association between cell communication mediators and hypoxia has become increasingly important. Exosomes are important information carriers in cell-to-cell communication. Abundant evidence has proven that hypoxia effects in the TME are mediated by exosomes, with the occasional formation of feedback loops. In this review, we equally focus on the biogenesis and heterogeneity of cancer-derived exosomes and their functions under hypoxia and describe the known and potential mechanism ascribed to exosomes and hypoxia. Notably, we call attention to the size change of hypoxic cancer cell-derived exosomes, a characteristic long neglected, and propose some possible effects of this size change. Finally, jointly considering recent developments in the understanding of exosomes and tumors, we describe noteworthy problems in this field that urgently need to be solved for better research and clinical application.
Collapse
Affiliation(s)
- Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shilei Tang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Sheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyu Zheng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
8
|
Abstract
Hypoxia is defined as a cellular stress condition caused by a decrease in oxygen below physiologically normal levels. Cells in the core of a rapidly growing solid tumor are faced with the challenge of inadequate supply of oxygen through the blood, owing to improper vasculature inside the tumor. This hypoxic microenvironment inside the tumor initiates a gene expression program that alters numerous signaling pathways, allowing the cancer cell to eventually evade adverse conditions and attain a more aggressive phenotype. A multitude of studies covering diverse aspects of gene regulation has tried to uncover the mechanisms involved in hypoxia-induced tumorigenesis. The role of epigenetics in executing widespread and dynamic changes in gene expression under hypoxia has been gaining an increasing amount of support in recent years. This chapter discusses, in detail, various epigenetic mechanisms driving the cellular response to hypoxia in cancer.
Collapse
Affiliation(s)
- Deepak Pant
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Srinivas Abhishek Mutnuru
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India
| | - Sanjeev Shukla
- Epigenetics and RNA Processing Lab (ERPL), Indian Institute of Science Education and Research Bhopal, Bhopal, India.
| |
Collapse
|
9
|
Ahmed KA, Hasib TA, Paul SK, Saddam M, Mimi A, Saikat ASM, Faruque HA, Rahman MA, Uddin MJ, Kim B. Potential Role of CCN Proteins in Breast Cancer: Therapeutic Advances and Perspectives. Curr Oncol 2021; 28:4972-4985. [PMID: 34940056 PMCID: PMC8700172 DOI: 10.3390/curroncol28060417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
CCNs are a specific type of matricellular protein, which are essential signaling molecules, and play multiple roles in multicellular eukaryotes. This family of proteins consists of six separate members, which exist only in vertebrates. The architecture of CCN proteins is multi-modular comprising four distinct modules. CCN Proteins achieve their primary functional activities by binding with several integrin7 receptors. The CCN family has been linked to cell adhesion, chemotaxis and migration, mitogenesis, cell survival, angiogenesis, differentiation, tumorigenesis, chondrogenesis, and wound healing, among other biological interactions. Breast cancer is the most commonly diagnosed cancer worldwide and CCN regulated breast cancer stands at the top. A favorable or unfavorable association between various CCNs has been reported in patients with breast carcinomas. The pro-tumorigenic CCN1, CCN2, CCN3, and CCN4 may lead to human breast cancer, although the anti-tumorigenic actions of CCN5 and CCN6 are also present. Several studies have been conducted on CCN proteins and cancer in recent years. CCN1 and CCN3 have been shown to exhibit a dual nature of tumor inhibition and tumor suppression to some extent in quiet recent time. Pharmacological advances in treating breast cancer by targeting CCN proteins are also reported. In our study, we intend to provide an overview of these research works while keeping breast cancer in focus. This information may facilitate early diagnosis, early prognosis and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Kazi Ahsan Ahmed
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
- Bio-Science Research Initiative, Gopalganj 8100, Bangladesh
| | - Tasnin Al Hasib
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
- Bio-Science Research Initiative, Gopalganj 8100, Bangladesh
| | - Shamrat Kumar Paul
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
- Bio-Science Research Initiative, Gopalganj 8100, Bangladesh
| | - Md. Saddam
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
- Bio-Science Research Initiative, Gopalganj 8100, Bangladesh
| | - Afsana Mimi
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
| | - Abu Saim Mohammad Saikat
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.); (A.M.); (A.S.M.S.)
| | - Hasan Al Faruque
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Md. Ataur Rahman
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (M.A.R.); (M.J.U.); (B.K.)
| | - Md. Jamal Uddin
- ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh; (K.A.A.); (T.A.H.); (S.K.P.); (H.A.F.)
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Women’s University, Seoul 03760, Korea
- Correspondence: (M.A.R.); (M.J.U.); (B.K.)
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (M.A.R.); (M.J.U.); (B.K.)
| |
Collapse
|
10
|
Qian C, Liu Q. FOXO3a inhibits nephroblastoma cell proliferation, migration and invasion, and induces apoptosis through downregulating the Wnt/β‑catenin signaling pathway. Mol Med Rep 2021; 24:796. [PMID: 34515328 PMCID: PMC8446726 DOI: 10.3892/mmr.2021.12436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Forkhead transcription factor O subfamily 3A (FOXO3a) is an important tumor suppressor gene that is expressed in renal tissue and has been reported to be downregulated in clear cell renal cell carcinoma (CCRCC). Notably, the overexpression of FOXO3a was previously discovered to inhibit the progression of CCRCC. However, the expression levels of FOXO3a in nephroblastoma cell lines remain unknown. The present study aimed to investigate the expression levels of FOXO3a in nephroblastoma cell lines and to determine the mechanism of action of the biological functions of FOXO3a. Western blotting and reverse transcription‑quantitative PCR were used to analyze the expression levels of FOXO3a in nephroblastoma cell lines. Subsequently, the effects of the overexpression of FOXO3a and the genetic knockdown of the Wnt/β‑catenin signaling protein Axin‑2 on the biological functions were determined through Cell Counting Kit‑8, cell colony formation assays, scratch and Transwell assay and flow cytometric analysis experiments. The expression levels of FOXO3a were discovered to be downregulated in nephroblastoma cell lines. The overexpression of FOXO3a inhibited the proliferation, invasion and migration of nephroblastoma cells, while inducing apoptosis. Furthermore, the overexpression of FOXO3a downregulated the expression levels of β‑catenin and Cyclin‑D1 proteins involved in the Wnt/β‑catenin signaling pathway. Cell proliferation and the migration and invasion ability of 17‑94 cells in shRNA‑Axin2‑2 group were promoted. Cell apoptosis was predominantly increased by overexpressed FOXO3a, which was reversed by shRNA‑Axin2‑1. The biological effects of overexpressing FOXO3a on nephroblastoma were reversed after activation of Wnt/β‑catenin. In conclusion, the findings of the present study suggested that FOXO3a may inhibit nephroblastoma cell proliferation, migration and invasion, while inducing apoptosis, by downregulating the Wnt/β‑catenin signaling pathway. These results may provide a novel method for the early diagnosis and precise treatment of nephroblastoma.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Pediatric Surgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277102, P.R. China
| | - Qiang Liu
- Department of Urinary Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| |
Collapse
|
11
|
Chao CT, Yeh HY, Tsai YT, Chiang CK, Chen HW. A combined microRNA and target protein-based panel for predicting the probability and severity of uraemic vascular calcification: a translational study. Cardiovasc Res 2021; 117:1958-1973. [PMID: 32866261 DOI: 10.1093/cvr/cvaa255] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/24/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Vascular calcification (VC) increases the future risk of cardiovascular events in uraemic patients, but effective therapies are still unavailable. Accurate identification of those at risk of developing VC using pathogenesis-based biomarkers is of particular interest and may facilitate individualized risk stratification. We aimed to uncover microRNA (miRNA)-target protein-based biomarker panels for evaluating uraemic VC probability and severity. METHODS AND RESULTS We created a three-tiered in vitro VC model and an in vivo uraemic rat model receiving high phosphate diet to mimic uraemic VC. RNAs from the three-tiered in vitro and in vivo uraemic VC models underwent miRNA and mRNA microarray, with results screened for differentially expressed miRNAs and their target genes as biomarkers. Findings were validated in original models and additionally in an ex vivo VC model and human cells, followed by functional assays of identified miRNAs and target proteins, and tests of sera from end-stage renal disease (ESRD) and non-dialysis-dependent chronic kidney disease (CKD) patients without and with VC. Totally 122 down-regulated and 119 up-regulated miRNAs during calcification progression were identified initially; further list narrowing based on miRNA-mRNA pairing, anti-correlation, and functional enrichment left 16 and 14 differentially expressed miRNAs and mRNAs. Levels of four miRNAs (miR-10b-5p, miR-195, miR-125b-2-3p, and miR-378a-3p) were shown to decrease throughout all models tested, while one mRNA (SULF1, a potential target of miR-378a-3p) exhibited the opposite trend concurrently. Among 96 ESRD (70.8% with VC) and 59 CKD patients (61% with VC), serum miR-125b2-3p and miR-378a-3p decreased with greater VC severity, while serum SULF1 levels increased. Adding serum miR-125b-2-3p, miR-378a-3p, and SULF1 into regression models for VC substantially improved performance compared to using clinical variables alone. CONCLUSION Using a translational approach, we discovered a novel panel of biomarkers for gauging the probability/severity of uraemic VC based on miRNAs/target proteins, which improved the diagnostic accuracy.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Biomarkers/blood
- Cells, Cultured
- Disease Models, Animal
- Female
- Gene Expression Profiling
- Gene Regulatory Networks
- Humans
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Organ Culture Techniques
- Predictive Value of Tests
- Protein Interaction Maps
- Proteome
- Proteomics
- Rats, Sprague-Dawley
- Risk Assessment
- Risk Factors
- Severity of Illness Index
- Signal Transduction
- Sulfotransferases/blood
- Transcriptome
- Translational Research, Biomedical
- Uremia/complications
- Uremia/genetics
- Uremia/metabolism
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Rats
Collapse
Affiliation(s)
- Chia-Ter Chao
- Division of Nephrology, Department of Medicine, National Taiwan University Hospital Bei-Hu Branch, No. 87, Neijiang Street, Wanhua District, Taipei 10845, Taiwan
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
| | - Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, No.70, Linxi Road, Shilin District, Taipei 11102, Taiwan
| | - You-Tien Tsai
- Division of Nephrology, Department of Medicine, National Taiwan University Hospital Bei-Hu Branch, No. 87, Neijiang Street, Wanhua District, Taipei 10845, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
- Department of Integrative Diagnostics and Therapeutics, National Taiwan University Hospital, No. 7, Zhongshan South Road, Zhongzheng District, Taipei 10002, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
| |
Collapse
|
12
|
Das A, Haque I, Ray P, Ghosh A, Dutta D, Quadir M, De A, Gunewardena S, Chatterjee I, Banerjee S, Weir S, Banerjee SK. CCN5 activation by free or encapsulated EGCG is required to render triple-negative breast cancer cell viability and tumor progression. Pharmacol Res Perspect 2021; 9:e00753. [PMID: 33745223 PMCID: PMC7981588 DOI: 10.1002/prp2.753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG) has been considered an anticancer agent despite conflicting and discrepant bioavailability views. EGCG impairs the viability and self-renewal capacity of triple-negative breast cancer (TNBC) cells and makes them sensitive to estrogen via activating ER-α. Surprisingly, the mechanism of EGCG's action on TNBC cells remains unclear. CCN5/WISP-2 is a gatekeeper gene that regulates viability, ER-α, and stemness in TNBC and other types of cancers. This study aimed to investigate whether EGCG (free or encapsulated in nanoparticles) interacts with the CCN5 protein by emphasizing its bioavailability and enhancing its anticancer effect. We demonstrate that EGCG activates CCN5 to inhibit in vitro cell viability through apoptosis, the sphere-forming ability via reversing TNBC cells' stemness, and suppressing tumor growth in vivo. Moreover, we found EGCG-loaded nanoparticles to be functionally more active and superior in their tumor-suppressing ability than free-EGCG. Together, these studies identify EGCG (free or encapsulated) as a novel activator of CCN5 in TNBC cells and hold promise as a future therapeutic option for TNBC with upregulated CCN5 expression.
Collapse
Affiliation(s)
- Amlan Das
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Present address:
National Institute of Biomedical GenomicsKalyaniWest BengalIndia
| | - Inamul Haque
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
| | - Priyanka Ray
- Department of Chemical Biochemical Environmental Engineering (CBEEUniversity of MarylandBaltimoreMDUSA
| | - Arnab Ghosh
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Debasmita Dutta
- Department of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoNDUSA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoNDUSA
| | - Archana De
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Indranil Chatterjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Present address:
Department of Life SciencesCentral University of Tamil NaduThiruvarurIndia
| | - Snigdha Banerjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Scott Weir
- Department of PharmacologyToxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Sushanta K. Banerjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
- Lead contact, SKB, Cancer Research UnitKansas CityMOUSA
| |
Collapse
|
13
|
Haque I, Kawsar HI, Motes H, Sharma M, Banerjee S, Banerjee SK, Godwin AK, Huang CH. Downregulation of miR-506-3p Facilitates EGFR-TKI Resistance through Induction of Sonic Hedgehog Signaling in Non-Small-Cell Lung Cancer Cell Lines. Int J Mol Sci 2020; 21:E9307. [PMID: 33291316 PMCID: PMC7729622 DOI: 10.3390/ijms21239307] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutation eventually develop resistance to EGFR-targeted tyrosine kinase inhibitors (TKIs). Treatment resistance remains the primary obstacle to the successful treatment of NSCLC. Although drug resistance mechanisms have been studied extensively in NSCLC, the regulation of these mechanisms has not been completely understood. Recently, increasing numbers of microRNAs (miRNAs) are implicated in EGFR-TKI resistance, indicating that miRNAs may serve as novel targets and may hold promise as predictive biomarkers for anti-EGFR therapy. MicroRNA-506 (miR-506) has been identified as a tumor suppressor in many cancers, including lung cancer; however, the role of miR-506 in lung cancer chemoresistance has not yet been addressed. Here we report that miR-506-3p expression was markedly reduced in erlotinib-resistant (ER) cells. We identified Sonic Hedgehog (SHH) as a novel target of miR-506-3p, aberrantly activated in ER cells. The ectopic overexpression of miR-506-3p in ER cells downregulates SHH signaling, increases E-cadherin expression, and inhibits the expression of vimentin, thus counteracting the epithelial-mesenchymal transition (EMT)-mediated chemoresistance. Our results advanced our understanding of the molecular mechanisms underlying EGFR-TKI resistance and indicated that the miR-506/SHH axis might represent a novel therapeutic target for future EGFR mutated lung cancer treatment.
Collapse
Affiliation(s)
- Inamul Haque
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hameem I Kawsar
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hannah Motes
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Kirksville College of Osteopathic Medicine, Andrew Taylor Still University, Jefferson St, Kirksville, MO 63501, USA
| | - Mukut Sharma
- Research Service, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
| | - Snigdha Banerjee
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sushanta K Banerjee
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Chao H Huang
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Shi L, Tao C, Tang Y, Xia Y, Li X, Wang X. Hypoxia-induced hsa_circ_0000826 is linked to liver metastasis of colorectal cancer. J Clin Lab Anal 2020; 34:e23405. [PMID: 32633429 PMCID: PMC7521269 DOI: 10.1002/jcla.23405] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/20/2020] [Accepted: 05/08/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND hsa_circ_0000826 has been previously linked to CRC through the competing endogenous RNA network; however, the upstream driver of hsa_circ_0000826 elevation remains unknown. In this study, we aim to elucidate the effect of hypoxia-induced hsa_circ_0000826 on CRC tumorigenesis and metastasis. METHODS RNA scope assay was used to evaluate the expression of hsa_circ_0000826 in CRC cells under hypoxia condition. The effects of hsa_circ_0000826 on phenotypes of CRC cells were evaluated through cell migration and invasion assay. The nude, AOM-DSS model mice and APCMin /+ mice were used to investigate the relationship between circ_0000826, hypoxia, and CRC in mice. A total of 100 CRC tissue samples, as well as the paired adjacent tissues, were collected, and qRT-PCR assay was used to detect the expression of hsa_circ_0000826 in these samples. RESULTS Hypoxia-induced hsa_circ_0000826 overexpression can increase the malignant phenotypes, tumor formation, and metastasis capability of CRC cells in vitro. mmu_circ_0000826 levels were significantly increased in the CRC tissues from AOM-DSS and APC mice model under hypoxia conditions. Further, the hypoxia-induced upregulation of mmu_circ_0000826 can also promote CRC tumorigenesis and liver metastasis in vivo. The expression of hsa_circ_0000826 in serum was significantly increased in CRC tissues in 100-pair of CRC and according to the adjacent normal tissues by qRT-PCR assays. Moreover, the expression levels of hsa_circ_0000826 in serum of patient with liver metastasis were significantly increased than those without metastasis. CONCLUSION Our results suggested that hsa_circ_0000826 was induced by the hypoxia in CRC, which can be a potential biomarker of CRC liver metastasis.
Collapse
Affiliation(s)
- Li Shi
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Key Laboratory of Liver TransplantationChinese Academy of Medical SciencesNanjingChina
- NHC Key Laboratory of Living Donor Liver TransplantationNanjing Medical UniversityNanjingChina
| | - Chengzhe Tao
- School of Public HealthNanjing Medical UniversityNanjingChina
| | - Yining Tang
- School of PharmacyNanjing Medical UniversityNanjingChina
| | - Yongxiang Xia
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Key Laboratory of Liver TransplantationChinese Academy of Medical SciencesNanjingChina
- NHC Key Laboratory of Living Donor Liver TransplantationNanjing Medical UniversityNanjingChina
| | - Xiangcheng Li
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Key Laboratory of Liver TransplantationChinese Academy of Medical SciencesNanjingChina
- NHC Key Laboratory of Living Donor Liver TransplantationNanjing Medical UniversityNanjingChina
| | - Xuehao Wang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Key Laboratory of Liver TransplantationChinese Academy of Medical SciencesNanjingChina
- NHC Key Laboratory of Living Donor Liver TransplantationNanjing Medical UniversityNanjingChina
| |
Collapse
|
15
|
Song ZB, Yang HP, Xu AQ, Zhan ZM, Song Y, Li ZY. Connective tissue growth factor as an unfavorable prognostic marker promotes the proliferation, migration, and invasion of gliomas. Chin Med J (Engl) 2020; 133:670-678. [PMID: 32197031 PMCID: PMC7190229 DOI: 10.1097/cm9.0000000000000683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In consideration of the difficulty in diagnosing high heterogeneous glioma, valuable prognostic markers are urgent to be investigated. This study aimed to verify that connective tissue growth factor (CTGF) is associated with the clinical prognosis of glioma, also to analyze the effect of CTGF on the biological function. METHODS In this study, glioma and non-tumor tissue samples were obtained in 2012 to 2014 from the Department of Neurosurgery of Nanfang Hospital of Southern Medical University, Guangzhou, China. Based on messenger RNA (mRNA) data from the Cancer Genome Atlas (TCGA) and CCGA dataset, combined with related clinical information, we detected the expression of CTGF mRNA in glioma and assessed its effect on the prognosis of glioma patients. High expression of CTGF mRNA and protein in glioma were verified by reverse transcription-polymerase chain reaction, immunohistochemistry, and Western blotting. The role of CTGF in the proliferation, migration, and invasion of gliomas were respectively identified by methylthiazoletetrazolium assay, Transwell and Boyden assay in vitro. The effect on glioma cell circle was assessed by flow cytometry. For higher expression of CTGF in glioblastoma (GBM), the biological function of CTGF in GBM was investigated by gene ontology (GO) analysis. RESULTS In depth analysis of TCGA data revealed that CTGF mRNA was highly expressed in glioma (GBM, n = 163; lowly proliferative glioma [LGG], n = 518; non-tumor brain tissue, n = 207; LGG, t = 2.410, GBM, t = 2.364, P < 0.05). CTGF mRNA and protein expression in glioma (86%) was significantly higher than that in non-tumor tissues (18%) verified by collected samples. Glioma patients with higher expression of CTGF showed an obviously poorer overall survival (35.4 and 27.0 months compared to 63.3 and 55.1 months in TCGA and Chinese Glioma Genome Atlas (CGGA) databases separately, CGGA: χ = 7.596, P = 0.0059; TCGA: χ = 10.46, P = 0.0012). Inhibiting CTGF expression could significantly suppress the proliferation, migration, and invasion of gliomas. CTGF higher expression had been observed in GBM, and GO analysis demonstrated that the function of CTGF in GBM was mainly associated with metabolism and energy pathways (P < 0.001). CONCLUSIONS CTGF is highly expressed in glioma, especially GBM, as an unfavorable and independent prognostic marker for glioma patients and facilitates the progress of glioma.
Collapse
Affiliation(s)
- Zi-Bin Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hui-Ping Yang
- The First Clinical Medical Institute of Southern Medical University, Guangzhou, Guangdong 510515, China
| | - An-Qi Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zheng-Ming Zhan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhi-Yong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
16
|
Pedroza-Torres A, Romero-Córdoba SL, Justo-Garrido M, Salido-Guadarrama I, Rodríguez-Bautista R, Montaño S, Muñiz-Mendoza R, Arriaga-Canon C, Fragoso-Ontiveros V, Álvarez-Gómez RM, Hernández G, Herrera LA. MicroRNAs in Tumor Cell Metabolism: Roles and Therapeutic Opportunities. Front Oncol 2019; 9:1404. [PMID: 31921661 PMCID: PMC6917641 DOI: 10.3389/fonc.2019.01404] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Dysregulated metabolism is a common feature of cancer cells and is considered a hallmark of cancer. Altered tumor-metabolism confers an adaptive advantage to cancer cells to fulfill the high energetic requirements for the maintenance of high proliferation rates, similarly, reprogramming metabolism confers the ability to grow at low oxygen concentrations and to use alternative carbon sources. These phenomena result from the dysregulated expression of diverse genes, including those encoding microRNAs (miRNAs) which are involved in several metabolic and tumorigenic pathways through its post-transcriptional-regulatory activity. Further, the identification of key actionable altered miRNA has allowed to propose novel targeted therapies to modulated tumor-metabolism. In this review, we discussed the different roles of miRNAs in cancer cell metabolism and novel miRNA-based strategies designed to target the metabolic machinery in human cancer.
Collapse
Affiliation(s)
- Abraham Pedroza-Torres
- Cátedra CONACyT-Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Sandra L Romero-Córdoba
- Departamento de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico
| | - Montserrat Justo-Garrido
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Iván Salido-Guadarrama
- Biología Computacional, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Rubén Rodríguez-Bautista
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerologia, Mexico City, Mexico
| | - Sarita Montaño
- Laboratorio de Bioinformática, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa (FCQB-UAS), Culiacán, Mexico
| | - Rodolfo Muñiz-Mendoza
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | | | - Greco Hernández
- Laboratorio de Traducción y Cáncer, Unidad de Investigaciones Biomedicas en Cáncer, Instituto Nacional de Cancerolgía, Mexico City, Mexico
| | - Luis A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
17
|
Gupta V, Bhavanasi S, Quadir M, Singh K, Ghosh G, Vasamreddy K, Ghosh A, Siahaan TJ, Banerjee S, Banerjee SK. Protein PEGylation for cancer therapy: bench to bedside. J Cell Commun Signal 2019; 13:319-330. [PMID: 30499020 PMCID: PMC6732144 DOI: 10.1007/s12079-018-0492-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022] Open
Abstract
PEGylation is a biochemical modification process of bioactive molecules with polyethylene glycol (PEG), which lends several desirable properties to proteins/peptides, antibodies, and vesicles considered to be used for therapy or genetic modification of cells. However, PEGylation of proteins is a complex process and can be carried out using more than one strategy that depends on the nature of the protein and the desired application. Proteins of interest are covalently conjugated or non-covalently complexed with inert PEG strings. Purification of PEGylated protein is another critical step, which is mainly carried out based on electrostatic interactions or molecular sizes using chromatography. Several PEGylated drugs are being used for diseases like anemia, kidney disease, multiple sclerosis, hemophilia and cancers. With the advancement and increased specificity of the PEGylation process, the world of drug therapy, and specifically cancer therapy could benefit by utilizing this technique to create more stable and non-immunogenic therapies. In this article we describe the structure and functions of PEGylation and how this chemistry helps in drug discovery. Moreover, special emphasis has been given to CCN-family proteins that can be targeted or used as therapy to prevent or block cancer progression through PEGylation technology.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sneha Bhavanasi
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, 58108, USA.
| | - Kevin Singh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
| | - Gaurav Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
| | - Kritin Vasamreddy
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Teruna J Siahaan
- School of Pharmacy-Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, 66047, USA
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, 64128, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
18
|
Tian XP, Wang CY, Jin XH, Li M, Wang FW, Huang WJ, Yun JP, Xu RH, Cai QQ, Xie D. Acidic Microenvironment Up-Regulates Exosomal miR-21 and miR-10b in Early-Stage Hepatocellular Carcinoma to Promote Cancer Cell Proliferation and Metastasis. Am J Cancer Res 2019; 9:1965-1979. [PMID: 31037150 PMCID: PMC6485281 DOI: 10.7150/thno.30958] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/27/2019] [Indexed: 12/18/2022] Open
Abstract
Rationale: The incidence of hepatocellular carcinoma is rising worldwide. It is predicted that nearly half of the early-stage hepatocellular carcinoma (E-HCC) patients will develop recurrence. Dysregulated pH, a hallmark of E-HCC, is correlated with poor prognosis. The acidic microenvironment has been shown to promote the release of exosomes, the membrane vesicles recognized as intercellular communicators associated with tumor progression, recurrence, and metastasis. We, therefore, aimed to identify exosomes induced by acidic microenvironment that may regulate E-HCC progression and to explore their mechanisms and clinical significance in E-HCCs. Methods: miRNA microarray analysis and LASSO logistic statistic model were used to identify the main functional exosomal miRNAs. Invasion and scratch assays were performed to examine the migration and invasion of HCC cells. Immunoblotting and immunofluorescence were employed to detect the epithelial-to-mesenchymal transition (EMT) in HCC cells. Chromatin immunoprecipitation (ChIP) was used to analyze the binding of HIF-1α and HIF-2α to promoter regions of miR-21 and miR-10b. Results: The acidic microenvironment in HCC was correlated with poor prognosis of patients. Exosomes from HCC cells cultured in the acidic medium could promote cell proliferation, migration, and invasion of recipient HCC cells. We identified miR-21 and miR-10b as the most important functional miRNAs in acidic HCC-derived exosomes. Also, the acidic microenvironment triggered the activation of HIF-1α and HIF-2α and stimulated exosomal miR-21 and miR-10b expression substantially promoting HCC cell proliferation, migration, and invasion both in vivo and in vitro. In E-HCC patients, serum exosomal miR-21 and miR-10b levels were associated with advanced tumor stage and HIF-1α and HIF-2α expression and were independent prognostic factors for disease-free survival of E-HCC patients. Most importantly, we developed a nano-drug to target exosomal miR-21 and/or miR-10b and examined its therapeutic effects against HCC in vivo. Conclusion: Our findings suggested that the exosomal miR-21 and miR-10b induced by acidic microenvironment in HCC promote cancer cell proliferation and metastasis and may serve as prognostic molecular markers and therapeutic targets for HCC.
Collapse
|
19
|
Yang W, Ma J, Zhou W, Cao B, Zhou X, Zhang H, Zhao Q, Hong L, Fan D. Reciprocal regulations between miRNAs and HIF-1α in human cancers. Cell Mol Life Sci 2019; 76:453-471. [PMID: 30317527 PMCID: PMC11105242 DOI: 10.1007/s00018-018-2941-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023]
Abstract
Hypoxia inducible factor-1α (HIF-1α) is a central molecule involved in mediating cellular processes. Alterations of HIF-1α and hypoxically regulated microRNAs (miRNAs) are correlated with patients' outcome in various cancers, indicating their crucial roles on cancer development. Recently, an increasing number of studies have revealed the intricate regulations between miRNAs and HIF-1α in modulating a wide variety of processes, including proliferation, metastasis, apoptosis, and drug resistance, etc. miRNAs are a class of small noncoding RNAs which function as negative regulators by directly targeting mRNAs. Evidence shows that miRNAs can be regulated by HIF-1α at transcriptional level. In turn, HIF-1α itself can be modulated by many miRNAs whose alterations have been implicated in tumorigenesis, thus forming a reciprocal regulation network. These findings add a new layer of complexity to our understanding of HIF-1α regulatory networks. Here, we will provide a comprehensive overview of the current advances about the bidirectional interactions between HIF-1α and miRNAs in human cancers. Besides, the review will summarize the roles of miRNAs/HIF-1α crosstalk according to various cellular processes. Finally, the potential values of miRNAs/HIF-1α loops in clinical applications are discussed.
Collapse
Affiliation(s)
- Wanli Yang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Jiaojiao Ma
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Bo Cao
- Air Force Military Medical University, Xi'an, China
| | - Xin Zhou
- Air Force Military Medical University, Xi'an, China
| | - Hongwei Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Qingchuan Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Liu Hong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
20
|
Samadi P, Saki S, Dermani FK, Pourjafar M, Saidijam M. Emerging ways to treat breast cancer: will promises be met? Cell Oncol (Dordr) 2018; 41:605-621. [PMID: 30259416 DOI: 10.1007/s13402-018-0409-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer among women and it is responsible for more than 40,000 deaths in the United States and more than 500,000 deaths worldwide each year. In previous decades, the development of improved screening, diagnosis and treatment methods has led to decreases in BC mortality rates. More recently, novel targeted therapeutic options, such as the use of monoclonal antibodies and small molecule inhibitors that target specific cancer cell-related components, have been developed. These components include ErbB family members (HER1, HER2, HER3 and HER4), Ras/MAPK pathway components (Ras, Raf, MEK and ERK), VEGF family members (VEGFA, VEGFB, VEGFC, VEGF and PGF), apoptosis and cell cycle regulators (BAK, BAX, BCL-2, BCL-X, MCL-1 and BCL-W, p53 and PI3K/Akt/mTOR pathway components) and DNA repair pathway components such as BRCA1. In addition, long noncoding RNA inhibitor-, microRNA inhibitor/mimic- and immunotherapy-based approaches are being developed for the treatment of BC. Finally, a novel powerful technique called CRISPR-Cas9-based gene editing is emerging as a precise tool for the targeted treatment of cancer, including BC. CONCLUSIONS Potential new strategies that are designed to specifically target BC are presented. Several clinical trials using these strategies are already in progress and have shown promising results, but inherent limitations such as off-target effects and low delivery efficiencies still have to be resolved. By improving the clinical efficacy of current therapies and exploring new ones, it is anticipated that novel ways to overcome BC may become attainable.
Collapse
Affiliation(s)
- Pouria Samadi
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sahar Saki
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Karimi Dermani
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Pourjafar
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
21
|
Yao L, Liu Y, Cao Z, Li J, Huang Y, Hu X, Shao Z. MicroRNA-493 is a prognostic factor in triple-negative breast cancer. Cancer Sci 2018; 109:2294-2301. [PMID: 29777630 PMCID: PMC6029816 DOI: 10.1111/cas.13644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/22/2023] Open
Abstract
Breast cancer is one of the most common malignant diseases in women. Triple‐negative breast cancer (TNBC) shows higher aggressiveness and recurrence rates than other subtypes, and there are no effective targets or tailored treatments for TNBC patients. Thus, finding effective prognostic markers for TNBC could help clinicians in their ability to care for their patients. We used tissue microarrays (TMAs) to detect microRNA‐493 (miR‐493) expression in breast cancer samples. A miRCURY LNA detection probe specific for miR‐493 was used in in situ hybridization assays. Staining results were reviewed by two independent pathologists and classified as high or low expression of miR‐493. Kaplan–Meier survival plots and multivariate Cox analysis were carried out to clarify the relationship between miR‐493 and survival. In the Kaplan–Meier analysis, patients with high miR‐493 expression had better disease‐free survival than patients with low miR‐493 expression. After adjusting for common clinicopathological factors in breast cancer, the expression level of miR‐493 was still a significant prognostic factor in breast cancer. Further subtype analysis revealed that miR‐493 expression levels were only significantly prognostic in TNBC patients. These results were validated in the Molecular Taxonomy of Breast Cancer International Consortium database for overall survival. We proved the prognostic role of miR‐493 in TNBC by using one of the largest breast cancer TMAs available and validated it in a large public RNA sequencing database.
Collapse
Affiliation(s)
- Ling Yao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yirong Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhigang Cao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Junjing Li
- Department of Breast Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yanni Huang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhiming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
22
|
|
23
|
Tantawy M, Elzayat MG, Yehia D, Taha H. Identification of microRNA signature in different pediatric brain tumors. Genet Mol Biol 2018; 41:27-34. [PMID: 29658967 PMCID: PMC5901491 DOI: 10.1590/1678-4685-gmb-2016-0334] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 08/21/2017] [Indexed: 12/02/2022] Open
Abstract
Understanding pediatric brain tumor biology is essential to help on disease
stratification, and to find novel markers for early diagnosis. MicroRNA (miRNA)
expression has been linked to clinical outcomes and tumor biology. Here, we
aimed to detect the expression of different miRNAs in different pediatric brain
tumor subtypes to discover biomarkers for early detection and develop novel
therapies. Expression of 82 miRNAs was detected in 120 pediatric brain tumors
from fixed-formalin paraffin-embedded tissues, low-grade glioma, high-grade
glioma, ependymoma, and medulloblastoma, using quantitative real-time PCR.
Low-expression of miR-221, miR-9, and miR-181c/d and over-expression of miR-101,
miR-222, miR-139, miR-1827, and miR-34c was found in medulloblastoma; low
expression of miR-10a and over-expression of miR-10b and miR-29a in ependymoma;
low expression of miR-26a and overexpression of miR-19a/b, miR-24, miR-27a, miR-
584, and miR-527 in low-grade glioma. Cox regression showed differential miRNA
expression between responders and non-responders. The most specific were miR-10a
and miR-29a low expression in LGG non-responders, miR-135a and miR-146b
over-expression in ependymoma non-responders, and miR-135b overexpression in
medulloblastoma non-responders. MicroRNAs are differentially expressed in
subtypes of brain tumors suggesting that they may help diagnosis. A greater
understanding of aberrant miRNA in pediatric brain tumors may support
development of novel therapies.
Collapse
Affiliation(s)
- Marwa Tantawy
- Research Department, Children's Cancer Hospital Egypt, Cairo, Egypt
| | - Mariam G Elzayat
- Research Department, Children's Cancer Hospital Egypt, Cairo, Egypt
| | - Dina Yehia
- Research Department, Children's Cancer Hospital Egypt, Cairo, Egypt
| | - Hala Taha
- Pathology Department, Children's Cancer Hospital Egypt, Cairo, Egypt.,Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
24
|
Maity G, Haque I, Ghosh A, Dhar G, Gupta V, Sarkar S, Azeem I, McGregor D, Choudhary A, Campbell DR, Kambhampati S, Banerjee SK, Banerjee S. The MAZ transcription factor is a downstream target of the oncoprotein Cyr61/CCN1 and promotes pancreatic cancer cell invasion via CRAF-ERK signaling. J Biol Chem 2018; 293:4334-4349. [PMID: 29414775 PMCID: PMC5868262 DOI: 10.1074/jbc.ra117.000333] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/01/2018] [Indexed: 01/18/2023] Open
Abstract
Myc-associated zinc-finger protein (MAZ) is a transcription factor with dual roles in transcription initiation and termination. Deregulation of MAZ expression is associated with the progression of pancreatic ductal adenocarcinoma (PDAC). However, the mechanism of action of MAZ in PDAC progression is largely unknown. Here, we present evidence that MAZ mRNA expression and protein levels are increased in human PDAC cell lines, tissue samples, a subcutaneous tumor xenograft in a nude mouse model, and spontaneous cancer in the genetically engineered PDAC mouse model. We also found that MAZ is predominantly expressed in pancreatic cancer stem cells. Functional analysis indicated that MAZ depletion in PDAC cells inhibits invasive phenotypes such as the epithelial-to-mesenchymal transition, migration, invasion, and the sphere-forming ability of PDAC cells. Mechanistically, we detected no direct effects of MAZ on the expression of K-Ras mutants, but MAZ increased the activity of CRAF-ERK signaling, a downstream signaling target of K-Ras. The MAZ-induced activation of CRAF-ERK signaling was mediated via p21-activated protein kinase (PAK) and protein kinase B (AKT/PKB) signaling cascades and promoted PDAC cell invasiveness. Moreover, we found that the matricellular oncoprotein cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) regulates MAZ expression via Notch-1-sonic hedgehog signaling in PDAC cells. We propose that Cyr61/CCN1-induced expression of MAZ promotes invasive phenotypes of PDAC cells not through direct K-Ras activation but instead through the activation of CRAF-ERK signaling. Collectively, these results highlight key molecular players in PDAC invasiveness and may help inform therapeutic strategies to improve clinical management and outcomes of PDAC.
Collapse
Affiliation(s)
- Gargi Maity
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
| | - Inamul Haque
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
| | - Arnab Ghosh
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Gopal Dhar
- From the Cancer Research Unit, Veterans Affairs Medical Center
| | | | - Sandipto Sarkar
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Imaan Azeem
- From the Cancer Research Unit, Veterans Affairs Medical Center
| | - Douglas McGregor
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
- the Pathology Department, Veterans Affairs Medical Center, Kansas City, Missouri 64128
| | - Abhishek Choudhary
- the Gastroenterology Department, Veterans Affairs Medical Center, Kansas City, Missouri 64128
| | - Donald R Campbell
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the University of Missouri Kansas City and Saint Luke's Hospital of Kansas City, Kansas City, Missouri, and
| | - Suman Kambhampati
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Sarah Cannon Cancer Center at HCA Midwest Health, Kansas City, Missouri 64131
| | - Sushanta K Banerjee
- From the Cancer Research Unit, Veterans Affairs Medical Center,
- the Department of Pathology and Laboratory Medicine, and
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Snigdha Banerjee
- From the Cancer Research Unit, Veterans Affairs Medical Center,
- the Department of Pathology and Laboratory Medicine, and
| |
Collapse
|
25
|
Ullmann P, Qureshi-Baig K, Rodriguez F, Ginolhac A, Nonnenmacher Y, Ternes D, Weiler J, Gäbler K, Bahlawane C, Hiller K, Haan S, Letellier E. Hypoxia-responsive miR-210 promotes self-renewal capacity of colon tumor-initiating cells by repressing ISCU and by inducing lactate production. Oncotarget 2018; 7:65454-65470. [PMID: 27589845 PMCID: PMC5323168 DOI: 10.18632/oncotarget.11772] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
Low oxygen concentrations (hypoxia) are known to affect the cellular metabolism and have been suggested to regulate a subpopulation of cancer cells with tumorigenic properties, the so-called tumor-initiating cells (TICs). To better understand the mechanism of hypoxia-induced TIC activation, we set out to study the role of hypoxia-responsive miRNAs in recently established colon cancer patient-derived TICs. We were able to show that low oxygen concentrations consistently lead to the upregulation of miR-210 in different primary TIC-enriched cultures. Both stable overexpression of miR-210 and knockdown of its target gene ISCU resulted in enhanced TIC self-renewal. We could validate the tumorigenic properties of miR- 210 in in vivo experiments by showing that ectopic expression of miR-210 results in increased tumor incidence. Furthermore, enhanced miR-210 expression correlated with reduced TCA cycle activity and increased lactate levels. Importantly, by blocking lactate production via inhibition of LDHA, we could reverse the promoting effect of miR-210 on self-renewal capacity, thereby emphasizing the regulatory impact of the glycolytic phenotype on colon TIC properties. Finally, by assessing expression levels in patient tissue, we could demonstrate the clinical relevance of the miR-210/ISCU signaling axis for colorectal carcinoma. Taken together, our study highlights the importance of hypoxia-induced miR-210 in the regulation of colon cancer initiation.
Collapse
Affiliation(s)
- Pit Ullmann
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Komal Qureshi-Baig
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Fabien Rodriguez
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | | | - Dominik Ternes
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jil Weiler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karoline Gäbler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Christelle Bahlawane
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karsten Hiller
- Luxembourg Centre for Systems Biomedicine, L-4367 Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
26
|
Choudhry H, Harris AL. Advances in Hypoxia-Inducible Factor Biology. Cell Metab 2018; 27:281-298. [PMID: 29129785 DOI: 10.1016/j.cmet.2017.10.005] [Citation(s) in RCA: 587] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/03/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022]
Abstract
Hypoxia-inducible factor (HIF), a central regulator for detecting and adapting to cellular oxygen levels, transcriptionally activates genes modulating oxygen homeostasis and metabolic activation. Beyond this, HIF influences many other processes. Hypoxia, in part through HIF-dependent mechanisms, influences epigenetic factors, including DNA methylation and histone acetylation, which modulate hypoxia-responsive gene expression in cells. Hypoxia profoundly affects expression of many noncoding RNAs classes that have clinicopathological implications in cancer. HIF can regulate noncoding RNAs production, while, conversely, noncoding RNAs can modulate HIF expression. There is recent evidence for crosstalk between circadian rhythms and hypoxia-induced signaling, suggesting involvement of molecular clocks in adaptation to fluxes in nutrient and oxygen sensing. HIF induces increased production of cellular vesicles facilitating intercellular communication at a distance-for example, promoting angiogenesis in hypoxic tumors. Understanding the complex networks underlying cellular and genomic regulation in response to hypoxia via HIF may identify novel and specific therapeutic targets.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Cancer Metabolism and Epigenetic Unit, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Oncology, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK.
| |
Collapse
|
27
|
Haque I, Ghosh A, Acup S, Banerjee S, Dhar K, Ray A, Sarkar S, Kambhampati S, Banerjee SK. Leptin-induced ER-α-positive breast cancer cell viability and migration is mediated by suppressing CCN5-signaling via activating JAK/AKT/STAT-pathway. BMC Cancer 2018; 18:99. [PMID: 29370782 PMCID: PMC5785848 DOI: 10.1186/s12885-018-3993-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
Background In menopausal women, one of the critical risk factors for breast cancer is obesity/adiposity. It is evident from various studies that leptin, a 16 kDa protein hormone overproduced in obese people, plays the critical role in neovascularization and tumorigenesis in breast and other organs. However, the mechanisms by which obesity influences the breast carcinogenesis remained unclear. In this study, by analyzing different estrogen receptor-α (ER-α)-positive and ER-α-negative BC cell lines, we defined the role of CCN5 in the leptin-mediated regulation of growth and invasive capacity. Methods We analyzed the effect of leptin on cell viability of ER-α-positive MCF-7 and ZR-75-1 cell lines and ER-α-negative MDA-MB-231 cell line. Additionally, we also determined the effect of leptin on the epithelial-mesenchymal transition (EMT) bio-markers, in vitro invasion and sphere-formation of MCF-7 and ZR-75-1 cell lines. To understand the mechanism, we determined the impact of leptin on CCN5 expression and the functional role of CCN5 in these cells by the treatment of human recombinant CCN5 protein(hrCCN5). Moreover, we also determined the role of JAK-STAT and AKT in the regulation of leptin-induced suppression of CCN5 in BC cells. Results Present studies demonstrate that leptin can induce cell viability, EMT, sphere-forming ability and migration of MCF-7 and ZR-75-1 cell lines. Furthermore, these studies found that leptin suppresses the expression of CCN5 at the transcriptional level. Although the CCN5 suppression has no impact on the constitutive proliferation of MCF-7 and ZR-75-1 cells, it is critical for leptin-induced viability and necessary for EMT, induction of in vitro migration and sphere formation, as the hrCCN5 treatment significantly inhibits the leptin-induced viability, EMT, migration and sphere-forming ability of these cells. Mechanistically, CCN5-suppression by leptin is mediated via activating JAK/AKT/STAT-signaling pathways. Conclusions These studies suggest that CCN5 serves as a gatekeeper for leptin-dependent growth and progression of luminal-type (ER-positive) BC cells. Leptin may thus need to destroy the CCN5-barrier to promote BC growth and progression via activating JAK/AKT/STAT signaling. Therefore, these observations suggest a therapeutic potency of CCN5 by restoration or treatment in obese-related luminal-type BC growth and progression.
Collapse
Affiliation(s)
- Inamul Haque
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Seth Acup
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA. .,Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA. .,Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA.
| | - Kakali Dhar
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Present Address: Syngene International Ltd, Clinical Development, Tower 1, Semicon Park, Phase II, Electronics City, Hosur Road, Bangalore, Karnataka, 560100, India.,Present Address: Saint James School of Medicine, Anguilla, British West Indies, USA
| | - Amitabha Ray
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Present Address: Syngene International Ltd, Clinical Development, Tower 1, Semicon Park, Phase II, Electronics City, Hosur Road, Bangalore, Karnataka, 560100, India.,Present Address: Saint James School of Medicine, Anguilla, British West Indies, USA
| | - Sandipto Sarkar
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA. .,Department of Medicine, University of Kansas Medical Center, Kansas City, KS, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA. .,Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA. .,Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA.
| |
Collapse
|
28
|
Ghosh P, Banerjee S, Maity G, De A, Banerjee SK. Detection of CCN1 and CCN5 mRNA in Human Cancer Samples Using a Modified In Situ Hybridization Technique. Methods Mol Biol 2018; 1489:495-504. [PMID: 27734400 DOI: 10.1007/978-1-4939-6430-7_41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
In situ hybridization is an ideal tool for the detection and localization of mRNA expression of specific gene(s) in tissue sections and cell lines for prognosis, predictive markers, and highlighted potential therapeutic targets. Given the importance of CCN1 and CCN5 in breast and pancreatic cancer progression, these two secretory proteins could be novel therapeutic targets. Thus, evaluating the distribution of mRNA of these targets using in situ hybridization could be important preclinical tools. This chapter describes a detailed in situ hybridization technique for the detection of CCN1 and CCN5 in formalin-fixed, paraffin-embedded patient samples of breast and pancreatic cancers.
Collapse
Affiliation(s)
- Priyanka Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Division of Hematology and Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Division of Hematology and Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gargi Maity
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA.,Division of Hematology and Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Archana De
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, USA. .,Division of Hematology and Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA. .,Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
29
|
Hypoxia-induced microRNA-590-5p promotes colorectal cancer progression by modulating matrix metalloproteinase activity. Cancer Lett 2017; 416:31-41. [PMID: 29247825 DOI: 10.1016/j.canlet.2017.12.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/04/2017] [Accepted: 12/10/2017] [Indexed: 12/20/2022]
Abstract
Hypoxia leads to cancer progression and promotes the metastatic potential of cancer cells. MicroRNAs (miRNAs) are small non-coding RNA that have emerged as key players involved in cancer development and progression. Hypoxia alters a set of hypoxia-mediated miRNAs expression during tumor development and it may function as oncogenes or tumor-suppressors. However, the roles and molecular mechanisms of hypoxia-regulatory miRNAs in colorectal cancer (CRC) progression remain poorly understood. Here we firstly identified miR-590-5p as hypoxia-sensitive miRNAs which was upregulated in colon cancer cells under hypoxia. Hypoxia-induced miR-590-5p suppressed the expression of RECK, in turn, promoting cell invasiveness and migratory abilities via activation of matrix metalloproteinases (MMPs) and filopodia protrusion in vitro. Inhibition of miR-590-5p suppressed tumor growth and metastasis in mouse xenograft and CRC liver metastasis models via inhibition of MMPs activity. Clinical analysis revealed higher miR-590-5p expression in CRC, compared to normal specimens. Furthermore, miR-590-5p expression was significantly increased in liver metastasis as compared to their matched primary CRC. Taken together, our findings provide the first evidence that miR-590-5p may have potential as a therapeutic target for CRC patients with metastasis.
Collapse
|
30
|
Gupta V, Haque I, Chakraborty J, Graff S, Banerjee S, Banerjee SK. Racial disparity in breast cancer: can it be mattered for prognosis and therapy. J Cell Commun Signal 2017; 12:119-132. [PMID: 29188479 DOI: 10.1007/s12079-017-0416-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/10/2017] [Indexed: 01/01/2023] Open
Abstract
Breast cancer (BC) has emerged as a deadly disease that affects the lives of millions of women worldwide. It is the second leading cause of cancer-related deaths in the United States. Advancements in BC screening, preventive measures and treatment have resulted in significant decline in BC related deaths. However, unacceptable levels of racial disparity have been consistently reported, especially in African-American (AA) women compared to European American (EA). AA women go through worse prognosis, shorter survival time and higher mortality rates, despite higher cancer incidence reported in EA. These disparities are independent of socioeconomic status, access to healthcare or age, or even the stage of BC. Recent race-specific genetic and epigenetic studies have reported biological causes, which form the crux of this review. However, the developments are just the tip of the iceberg. Prioritizing primary research towards studying race-specific tumor microenvironment and biological composition of the host system in delineating the cause of these disparities is utmost necessary to ameliorate the disparity and design appropriate diagnosis/treatment regimen for AA women suffering from BC. In this review article, we discuss emerging trends and exciting discoveries that reveal how genetic/epigenetic circuitry contributed to racial disparity and discussed the strategies that may help in future therapeutic development.
Collapse
Affiliation(s)
- Vijayalaxmi Gupta
- Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA.,Department of Pathology and Integrative Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Inamul Haque
- Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA.,Department of Pathology and Integrative Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jinia Chakraborty
- Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA.,Blue Valley West High School, Overland Park, KS, USA
| | - Stephanie Graff
- Sarah Cannon Cancer Center at HCA Midwest Health, Kansas City, MO, USA
| | - Snigdha Banerjee
- Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA. .,Department of Pathology and Integrative Science, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Sushanta K Banerjee
- Cancer Research Unit, Research Division 151, VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA. .,Department of Pathology and Integrative Science, University of Kansas Medical Center, Kansas City, KS, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
31
|
Identification of differentially expressed genes regulated by molecular signature in breast cancer-associated fibroblasts by bioinformatics analysis. Arch Gynecol Obstet 2017; 297:161-183. [PMID: 29063236 DOI: 10.1007/s00404-017-4562-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Breast cancer is a severe risk to public health and has adequately convoluted pathogenesis. Therefore, the description of key molecular markers and pathways is of much importance for clarifying the molecular mechanism of breast cancer-associated fibroblasts initiation and progression. Breast cancer-associated fibroblasts gene expression dataset was downloaded from Gene Expression Omnibus database. METHODS A total of nine samples, including three normal fibroblasts, three granulin-stimulated fibroblasts and three cancer-associated fibroblasts samples, were used to identify differentially expressed genes (DEGs) between normal fibroblasts, granulin-stimulated fibroblasts and cancer-associated fibroblasts samples. The gene ontology (GO) and pathway enrichment analysis was performed, and protein-protein interaction (PPI) network of the DEGs was constructed by NetworkAnalyst software. RESULTS Totally, 190 DEGs were identified, including 66 up-regulated and 124 down-regulated genes. GO analysis results showed that up-regulated DEGs were significantly enriched in biological processes (BP), including cell-cell signalling and negative regulation of cell proliferation; molecular function (MF), including insulin-like growth factor II binding and insulin-like growth factor I binding; cellular component (CC), including insulin-like growth factor binding protein complex and integral component of plasma membrane; the down-regulated DEGs were significantly enriched in BP, including cell adhesion and extracellular matrix organization; MF, including N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase activity and calcium ion binding; CC, including extracellular space and extracellular matrix. WIKIPATHWAYS analysis showed the up-regulated DEGs were enriched in myometrial relaxation and contraction pathways. WIKIPATHWAYS, REACTOME, PID_NCI and KEGG pathway analysis showed the down-regulated DEGs were enriched endochondral ossification, TGF beta signalling pathway, integrin cell surface interactions, beta1 integrin cell surface interactions, malaria and glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulphate. The top 5 up-regulated hub genes, CDKN2A, MME, PBX1, IGFBP3, and TFAP2C and top 5 down-regulated hub genes VCAM1, KRT18, TGM2, ACTA2, and STAMBP were identified from the PPI network, and subnetworks revealed these genes were involved in significant pathways, including myometrial relaxation and contraction pathways, integrin cell surface interactions, beta1 integrin cell surface interaction. Besides, the target hsa-mirs for DEGs were identified. hsa-mir-759, hsa-mir-4446-5p, hsa-mir-219a-1-3p and hsa-mir-26a-5p were important miRNAs in this study. CONCLUSIONS We pinpoint important key genes and pathways closely related with breast cancer-associated fibroblasts initiation and progression by a series of bioinformatics analysis on DEGs. These screened genes and pathways provided for a more detailed molecular mechanism underlying breast cancer-associated fibroblasts occurrence and progression, holding promise for acting as molecular markers and probable therapeutic targets.
Collapse
|
32
|
Deficiency of CCN5/WISP-2-Driven Program in breast cancer Promotes Cancer Epithelial cells to mesenchymal stem cells and Breast Cancer growth. Sci Rep 2017; 7:1220. [PMID: 28450698 PMCID: PMC5430628 DOI: 10.1038/s41598-017-00916-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer progression and relapse is conceivably due to tumor initiating cells (TICs)/cancer stem cells. EMT (epithelial-mesenchymal-transition)-signaling regulates TICs’ turnover. However, the mechanisms associated with this episode are unclear. We show that, in triple-negative-breast cancer (TNBC) cells enriched with TICs, CCN5 significantly blocks cellular growth via apoptosis, reversing EMT-signaling and impairing mammosphere formation, thereby blocking the tumor-forming ability and invasive capacity of these cells. To corroborate these findings, we isolated tumor-initiating side populations (SP) and non-side population (NSP or main population) from MCF-7 cell line, and evaluated the impact of CCN5 on these subpopulations. CCN5 was overexpressed in the NSP but downregulated in the SP. Characteristically, NSP cells are ER-α positive and epithelial type with little tumorigenic potency, while SP cells are very similar to triple-negative ones that do not express ER-α- and Her-2 and are highly tumorigenic in xenograft models. The overexpression of CCN5 in SP results in EMT reversion, ER-α upregulation and delays in tumor growth in xenograft models. We reasoned that CCN5 distinguishes SP and NSP and could reprogram SP to NSP transition, thereby delaying tumor growth in the xenograft model. Collectively, we reveal how CCN5-signaling underlies the driving force to prevent TNBC growth and progression.
Collapse
|
33
|
Expression of the circulating and the tissue microRNAs after surgery, chemotherapy, and radiotherapy in mice mammary tumor. Tumour Biol 2016; 37:14225-14234. [DOI: 10.1007/s13277-016-5292-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/18/2016] [Indexed: 12/18/2022] Open
|
34
|
Wang G, Gu J, Gao Y. MicroRNA target for MACC1 and CYR61 to inhibit tumor growth in mice with colorectal cancer. Tumour Biol 2016; 37:13983-13993. [PMID: 27492459 DOI: 10.1007/s13277-016-5252-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/15/2016] [Indexed: 12/30/2022] Open
Abstract
Cysteine-rich protein 61 (CYR61) and metastasis associated in colon cancer (MACC1) protein promoted human colorectal cancer (CRC) cell metastasis and closely related to the patient's prognosis in colorectal cancer. The purpose of this article is to investigate whether CYR61 and MACC1 can serve as dual potential targets for gene therapy of human CRC. In this study, microRNA (miRNA) targeting for both CYR61 and MACC1 was used to investigate the mechanism and therapeutic effects for CRC cells and mice with CRC. We observed that silencing miRNA for CYR61 and MACC1 inhibited the epithelial-mesenchymal transition (EMT) process, and co-treatment strengthened this effect. MTT assay showed that the growth of colorectal tumor cells was decreased due to miRNA treatment. Apoptosis assay revealed that miRNA for CYR61 and MACC1 promoted CRC cells apoptotic. The animals' study results showed that the expression levels of CYR61 and MACC1 were significantly decreased after miRNA-100 and miRNA-143 treatment, respectively. The expression levels of apoptosis-promoting protein were increased significantly after treatment with miRNA-100 and miRNA-143, which suggested that both miRNA-100 and miRNA-143 may induce apoptosis by mitochondria-dependent pathway. In addition, metastasis and invasion assays showed that miRNA-100 and miRNA-143 treatment inhibited obviously migratory and invasive abilities of CRC cells. Furthermore, our data also showed that the tumor growth was significantly inhibited and survival rate of tumor-bearing mice was greatly improved by common treatments of miRNA-100 and miRNA-143. In conclusion, the abilities of apoptosis, metastasis, and invasion in CRC tumor cells were significantly suppressed by miRNA-100 and miRNA-143 targeting CYR61 and MACC1, respectively. As a result, CYR61 and MACC1 may serve as potential targets for gene therapy in human CRC treatments.
Collapse
Affiliation(s)
- Guiqi Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Hebei Medical University, No. 89, Donggang Road, Shijiazhuang, 050031, China
| | - Jingfeng Gu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Hebei Medical University, No. 89, Donggang Road, Shijiazhuang, 050031, China.
| | - Yingchao Gao
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Hebei Medical University, No. 89, Donggang Road, Shijiazhuang, 050031, China
| |
Collapse
|
35
|
Mammary Development and Breast Cancer: A Wnt Perspective. Cancers (Basel) 2016; 8:cancers8070065. [PMID: 27420097 PMCID: PMC4963807 DOI: 10.3390/cancers8070065] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
The Wnt pathway has emerged as a key signaling cascade participating in mammary organogenesis and breast oncogenesis. In this review, we will summarize the current knowledge of how the pathway regulates stem cells and normal development of the mammary gland, and discuss how its various components contribute to breast carcinoma pathology.
Collapse
|
36
|
Hawa Z, Haque I, Ghosh A, Banerjee S, Harris L, Banerjee SK. The miRacle in Pancreatic Cancer by miRNAs: Tiny Angels or Devils in Disease Progression. Int J Mol Sci 2016; 17:E809. [PMID: 27240340 PMCID: PMC4926343 DOI: 10.3390/ijms17060809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/04/2016] [Accepted: 05/19/2016] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with increasing incidence and high mortality. Surgical resection is the only potentially curative treatment of patients with PDAC. Because of the late presentation of the disease, about 20 percent of patients are candidates for this treatment. The average survival of resected patients is between 12 and 20 months, with a high probability of relapse. Standard chemo and radiation therapies do not offer significant improvement of the survival of these patients. Furthermore, novel treatment options aimed at targeting oncogenes or growth factors in pancreatic cancer have proved unsuccessful. Thereby, identifying new biomarkers that can detect early stages of this disease is of critical importance. Among these biomarkers, microRNAs (miRNAs) have supplied a profitable recourse and become an attractive focus of research in PDAC. MiRNAs regulate many genes involved in the development of PDAC through mRNA degradation or translation inhibition. The possibility of intervention in the molecular mechanisms of miRNAs regulation could begin a new generation of PDAC therapies. This review summarizes the reports describing miRNAs involvement in cellular processes involving pancreatic carcinogenesis and their utility in diagnosis, survival and therapeutic potential in pancreatic cancer.
Collapse
Affiliation(s)
- Zuhair Hawa
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
| | - Inamul Haque
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - LaCoiya Harris
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66205, USA.
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| |
Collapse
|
37
|
Wang Y, Li Z, Zhao X, Zuo X, Peng Z. miR-10b promotes invasion by targeting HOXD10 in colorectal cancer. Oncol Lett 2016; 12:488-494. [PMID: 27347170 PMCID: PMC4907168 DOI: 10.3892/ol.2016.4628] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/08/2016] [Indexed: 12/22/2022] Open
Abstract
Studies have shown that homeobox D10 (HOXD10) is the target gene of microRNA-10b (miR-10b) and is closely associated with the inhibition of cell migration and invasion. Ras homolog family member C (RhoC) has been reported to promote tumor metastasis in various types of cancer. The effect of miR-10b on colorectal cancer (CRC) metastasis and the associated molecular mechanisms remain elusive. The present study aimed to investigate whether miR-10b could promote invasion by targeting HOXD10 in CRC by exploring the association between miR-10b and HOXD10 expression in CRC patients. The findings revealed that miR-10b levels were elevated in the CRC specimens and significantly correlated with advanced clinical stage and lymph node metastasis. In addition, HOXD10 was a direct target of miR-10b, and the increased expression of RhoC and downregulation of HOXD10 correlated with the increased expression level of miR-10b. HOXD10 protein level was also markedly attenuated in lymph node metastasis-positive tumor tissues compared with lymph node metastasis-free tumor tissues. These findings suggest that miR-10b may stimulate the upregulation of RhoC through targeting HOXD10, thus promoting the invasion and migration in CRC tumor.
Collapse
Affiliation(s)
- Yunfeng Wang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Zhen Li
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Xuhong Zhao
- Department of Central Experiment Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Xiaoming Zuo
- Department of Pathology, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Zhihai Peng
- Department of General Surgery, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
38
|
Zhang X, Cheng J, Fu L, Li Q. Overexpression of tissue microRNA10b may help predict glioma prognosis. J Clin Neurosci 2016; 29:59-63. [PMID: 26988656 DOI: 10.1016/j.jocn.2015.10.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/14/2015] [Accepted: 10/25/2015] [Indexed: 12/19/2022]
Abstract
We investigated the relationship between microRNA-10b (miR-10b) expression and prognosis in human glioma patients. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis was used to characterize the expression patterns of miR-10b in 128 glioma and 20 normal brain tissues. Clinical information - age, sex, Karnofsky Performance Status (KPS) and World Health Organization (WHO) grade - were also collected. The associations between miR-10b expression and the clinicopathological factors and outcome of glioma patients were statistically analyzed. Expression levels of miR-10b in glioma tissue were significantly higher than in normal brain tissue (P<0.001). High-grade glioma (WHO grade III and IV) had much higher miR-10b expression levels than low-grade tumors (WHO grade I and II). Additionally, the increased miR-10b expression in the glioma tissues was significantly associated with a low KPS (P=0.03). Kaplan-Meier survival curves and Cox regression analyses showed that overexpression of miR-10b (P=0.01) and high grade (P=0.02) were independent factors predicting poor outcome for glioma patients. Furthermore, subgroup analyses showed that the miR-10b expression level was significantly associated with poor overall survival in glioma patients with high grades (P<0.001). Up-regulation of miR-10b may have value in predicting clinical outcome in glioma patients, particularly for those with high pathological grades.
Collapse
Affiliation(s)
- Xinxin Zhang
- Shandong Cancer Hospital and Institute, 440 Jiyan, Jinan 250117, Shandong Province, China
| | - Jian Cheng
- The Second Hospital of Shandong University, Shandong Province, China
| | - Ling Fu
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Qingshui Li
- Shandong Cancer Hospital and Institute, 440 Jiyan, Jinan 250117, Shandong Province, China.
| |
Collapse
|
39
|
The interplay between microRNAs and Twist1 transcription factor: a systematic review. Tumour Biol 2016; 37:7007-19. [PMID: 26880587 DOI: 10.1007/s13277-016-4960-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/02/2016] [Indexed: 12/11/2022] Open
Abstract
Twist1 (also known as Twist) is a transcription factor that belongs to the family of basic helix-loop-helix (bHLH) proteins. It functions as a negative regulator of epithelial gene expression and a positive regulator of mesenchymal gene expression, thereby leading to induction of the epithelial mesenchymal transition (EMT), a process in which epithelial cells acquire the motile and migratory characteristics of mesenchymal cells. In addition to regulating the expression of protein-coding genes, Twist1 regulates the expression of microRNAs (miRNAs), adding a regulatory layer to EMT induction. Interestingly, the mRNA of Twist1 represents a downstream target of miRNAs, indicating an intricate network between miRNAs and Twist1. This network was shown to play multiple roles in cancer cell migration, invasion, and metastasis. The network can induce angiogenesis, protect cells from oncogene-induced apoptosis and senescence, enhance cancer cell resistance to conventional therapies, and increase cancer stem cell (CSC) populations. Recently, miRNAs have attracted considerable attention as potential promising tools in cancer therapies. Thus, this systematic review was conducted to clarify the reciprocal link between Twist1 and miRNAs in order to provide potential candidate miRNAs for diagnostic and therapeutic approaches in cancer treatment.
Collapse
|
40
|
MicroRNA-101 targets von Hippel-Lindau tumor suppressor (VHL) to induce HIF1α mediated apoptosis and cell cycle arrest in normoxia condition. Sci Rep 2016; 6:20489. [PMID: 26841847 PMCID: PMC4740907 DOI: 10.1038/srep20489] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/05/2016] [Indexed: 01/04/2023] Open
Abstract
The activation/inactivation of HIF1α is precisely regulated in an oxygen-dependent manner. HIF1α is essential for hypoxia induced apoptosis and cell cycle arrest. Several recent studies indicated that the expression of miRNAs can be modulated by hypoxia. However, the involvement of miRNAs in the regulation of HIF1α induction remains elusive. In present study, we demonstrated that miR-101 was rapidly and transiently induced after hypoxia in breast cancer cells. Over-expression of miR-101 significantly inhibited cell proliferation in breast cancer cells through increased apoptosis and cell cycle arrest in normoxia condition. This inhibitory phenomenon seems due to miR-101-mediated induction of HIF1α, because we identified that VHL, a negative regulator of HIF1α, is a novel target of miR-101 and over-expression of miR-101 decreased VHL levels and subsequently stabilized HIF1α and induced its downstream target VEGFA. Furthermore, we demonstrated that siRNA-mediated knockdown of VHL or HIF1α overexpression could also induce apoptosis and cell cycle arrest whereas enforced expression of VHL, administration of anti-miR-101 oligos or treatment of 2-MeOE2, an inhibitor of HIF1α, could rescue cells from such inhibition. These results reveal a novel regulatory mechanism of HIF1α induction in normoxia and suggest that miR-101 mediated proliferation inhibition may through HIF1α mediated apoptosis and cell cycle arrest.
Collapse
|
41
|
Zhang P, Hong H, Sun X, Jiang H, Ma S, Zhao S, Zhang M, Wang Z, Jiang C, Liu H. MicroRNA-10b regulates epithelial-mesenchymal transition by modulating KLF4/Notch1/E-cadherin in cisplatin-resistant nasopharyngeal carcinoma cells. Am J Cancer Res 2016; 6:141-156. [PMID: 27186392 PMCID: PMC4859649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/21/2015] [Indexed: 06/05/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an initiating event in tumor cell invasion and metastasis that contributes to therapeutic resistance to compounds including cisplatin. MicroRNAs (miRNAs) have been associated with EMT as well as resistance to standard therapies. However, the underlying mechanisms by which miRNAs control the development of resistance to cisplatin (DDP), and the accompanying EMT-like properties are required to elucidate. Here we show that microRNA-10b (miR-10b) is up-regulated in HNE1/DDP cells, and inhibition of miR-10b expression reversed the EMT phenotype. However, over-expression of miR-10b was able to promote the acquisition of an EMT phenotype in HNE1 cells. Additionally, we identified that miR-10b expression inversely correlates with KLF4, which then controls expression of Notch1. Knock-down of Notch1 inhibited cell migration, invasion, and reversed EMT in HNE1/DDP cells, which was dependent on miR-10b. In summary, our results reveal that miR-10b regulates EMT by modulating KLF4/Notch1/E-cadherin expression, which promotes invasion and migration of nasal pharyngeal carcinoma cells.
Collapse
Affiliation(s)
- Pei Zhang
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
| | - Haiyu Hong
- Department of Otolaryngology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, China
| | - Xiaojin Sun
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
| | - Hao Jiang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical CollegeNo 287, Chang-Huai Road, Bengbu 233000, Anhui, P. R. China
| | - Shiyin Ma
- Department of ENT, The First Affiliated Hospital of Bengbu Medical CollegeNo 287, Chang-Huai Road, Bengbu 233000, Anhui, P. R. China
| | - Surong Zhao
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
| | - Mengxiao Zhang
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
| | - Zhiwei Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA 02215, USA
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow UniversitySuzhou, China
| | - Chenchen Jiang
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
- Priority Research Center for Cancer Research, University of NewcastleNewcastle, NSW 2308, Australia
| | - Hao Liu
- Faculty of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical PharmaceuticalsBengbu 233030, Anhui, P. R. China
| |
Collapse
|
42
|
Abba ML, Patil N, Leupold JH, Allgayer H. MicroRNA Regulation of Epithelial to Mesenchymal Transition. J Clin Med 2016; 5:jcm5010008. [PMID: 26784241 PMCID: PMC4730133 DOI: 10.3390/jcm5010008] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a central regulatory program that is similar in many aspects to several steps of embryonic morphogenesis. In addition to its physiological role in tissue repair and wound healing, EMT contributes to chemo resistance, metastatic dissemination and fibrosis, amongst others. Classically, the morphological change from epithelial to mesenchymal phenotype is characterized by the appearance or loss of a group of proteins which have come to be recognized as markers of the EMT process. As with all proteins, these molecules are controlled at the transcriptional and translational level by transcription factors and microRNAs, respectively. A group of developmental transcription factors form the backbone of the EMT cascade and a large body of evidence shows that microRNAs are heavily involved in the successful coordination of mesenchymal transformation and vice versa, either by suppressing the expression of different groups of transcription factors, or otherwise acting as their functional mediators in orchestrating EMT. This article dissects the contribution of microRNAs to EMT and analyzes the molecular basis for their roles in this cellular process. Here, we emphasize their interaction with core transcription factors like the zinc finger enhancer (E)-box binding homeobox (ZEB), Snail and Twist families as well as some pluripotency transcription factors.
Collapse
Affiliation(s)
- Mohammed L Abba
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Nitin Patil
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Jörg Hendrik Leupold
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Heike Allgayer
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| |
Collapse
|
43
|
Liu ZJ, Semenza GL, Zhang HF. Hypoxia-inducible factor 1 and breast cancer metastasis. J Zhejiang Univ Sci B 2015; 16:32-43. [PMID: 25559953 DOI: 10.1631/jzus.b1400221] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulating evidence has shown that the hypoxic microenvironment, which is critical during cancer development, plays a key role in regulating breast cancer progression and metastasis. The effects of hypoxia-inducible factor 1 (HIF-1), a master regulator of the hypoxic response, have been extensively studied during these processes. In this review, we focus on the roles of HIF-1 in regulating breast cancer cell metastasis, specifically its effects on multiple key steps of metastasis, such as epithelial-mesenchymal transition (EMT), invasion, extravasation, and metastatic niche formation. We also discuss the roles of HIF-1-regulated non-coding RNAs in breast cancer metastasis, and therapeutic opportunities for breast cancer through targeting the HIF-1 pathway.
Collapse
Affiliation(s)
- Zhao-Ji Liu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
44
|
Wang W, Luo YP. MicroRNAs in breast cancer: oncogene and tumor suppressors with clinical potential. J Zhejiang Univ Sci B 2015; 16:18-31. [PMID: 25559952 DOI: 10.1631/jzus.b1400184] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRs) are small single-stranded RNA molecules, which function as key negative regulators of post-transcriptional modulation in almost all biological processes. Abnormal expression of microRNAs has been observed in various types of cancer including breast cancer. Great efforts have been made to identify an association between microRNA expression profiles and breast cancer, and to understand the functional role and molecular mechanism of aberrant-expressed microRNAs. As research progressed, 'oncogenic microRNAs' and 'tumor suppressive microRNAs' became a focus of interest. The potential of candidate microRNAs from both intercellular (tissue) and extracellular (serum) sources for clinical diagnosis and prognosis was revealed, and treatments involving microRNA achieved some amazing curative effects in cancer disease models. In this review, advances from the most recent studies of microRNAs in one of the most common cancers, breast cancer, are highlighted, especially the functions of specifically selected microRNAs. We also assess the potential value of these microRNAs as diagnostic and prognostic markers, and discuss the possible development of microRNA-based therapies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | | |
Collapse
|
45
|
Aspirin blocks growth of breast tumor cells and tumor-initiating cells and induces reprogramming factors of mesenchymal to epithelial transition. J Transl Med 2015; 95:702-17. [PMID: 25867761 DOI: 10.1038/labinvest.2015.49] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 02/10/2015] [Accepted: 02/16/2015] [Indexed: 12/19/2022] Open
Abstract
Acetylsalicylic acid (ASA), also known as aspirin, a classic, nonsteroidal, anti-inflammatory drug (NSAID), is widely used to relieve minor aches and pains and to reduce fever. Epidemiological studies and other experimental studies suggest that ASA use reduces the risk of different cancers including breast cancer (BC) and may be used as a chemopreventive agent against BC and other cancers. These studies have raised the tempting possibility that ASA could serve as a preventive medicine for BC. However, lack of in-depth knowledge of the mechanism of action of ASA reshapes the debate of risk and benefit of using ASA in prevention of BC. Our studies, using in vitro and in vivo tumor xenograft models, show a strong beneficial effect of ASA in the prevention of breast carcinogenesis. We find that ASA not only prevents breast tumor cell growth in vitro and tumor growth in nude mice xenograft model through the induction of apoptosis, but also significantly reduces the self-renewal capacity and growth of breast tumor-initiating cells (BTICs)/breast cancer stem cells (BCSCs) and delays the formation of a palpable tumor. Moreover, ASA regulates other pathophysiological events in breast carcinogenesis, such as reprogramming the mesenchymal to epithelial transition (MET) and delaying in vitro migration in BC cells. The tumor growth-inhibitory and reprogramming roles of ASA could be mediated through inhibition of TGF-β/SMAD4 signaling pathway that is associated with growth, motility, invasion, and metastasis in advanced BCs. Collectively, ASA has a therapeutic or preventive potential by attacking possible target such as TGF-β in breast carcinogenesis.
Collapse
|
46
|
Nagpal N, Ahmad HM, Chameettachal S, Sundar D, Ghosh S, Kulshreshtha R. HIF-inducible miR-191 promotes migration in breast cancer through complex regulation of TGFβ-signaling in hypoxic microenvironment. Sci Rep 2015; 5:9650. [PMID: 25867965 PMCID: PMC4394754 DOI: 10.1038/srep09650] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 03/10/2015] [Indexed: 12/13/2022] Open
Abstract
The molecular mechanisms of hypoxia induced breast cell migration remain incompletely understood. Our results show that hypoxia through hypoxia-inducible factor (HIF) brings about a time-dependent increase in the level of an oncogenic microRNA, miR-191 in various breast cancer cell lines. miR-191 enhances breast cancer aggressiveness by promoting cell proliferation, migration and survival under hypoxia. We further established that miR-191 is a critical regulator of transforming growth factor beta (TGFβ)-signaling and promotes cell migration by inducing TGFβ2 expression under hypoxia through direct binding and indirectly by regulating levels of a RNA binding protein, human antigen R (HuR). The levels of several TGFβ pathway genes (like VEGFA, SMAD3, CTGF and BMP4) were found to be higher in miR-191 overexpressing cells. Lastly, anti-miR-191 treatment given to breast tumor spheroids led to drastic reduction in spheroid tumor volume. This stands as a first report of identification of a microRNA mediator that links hypoxia and the TGFβ signaling pathways, both of which are involved in regulation of breast cancer metastasis. Together, our results show a critical role of miR-191 in hypoxia-induced cancer progression and suggest that miR-191 inhibition may offer a novel therapy for hypoxic breast tumors.
Collapse
Affiliation(s)
- Neha Nagpal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, India-110016
| | - Hafiz M. Ahmad
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India- 110067
| | - Shibu Chameettachal
- Department of Textile Technology, Indian Institute of Technology Delhi, India-110016
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, India-110016
| | - Sourabh Ghosh
- Department of Textile Technology, Indian Institute of Technology Delhi, India-110016
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, India-110016
- ;
| |
Collapse
|
47
|
MicroRNA-10b and minichromosome maintenance complex component 5 gene as prognostic biomarkers in breast cancer. Tumour Biol 2015; 36:4487-94. [PMID: 25596707 DOI: 10.1007/s13277-015-3090-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/08/2015] [Indexed: 01/20/2023] Open
Abstract
The aim of this study is to identify micro-ribonucleic acid (microRNA) and its target, in addition to their relationship to the outcome in breast cancer (BC). To achieve this aim, we investigated microRNA-10b (miR-10b) and minichromosome maintenance complex component 5 (MCM5 mRNA) expression in 230 breast tissue samples by real-time PCR and semiquantitative conventional RT-PCR, respectively. Relapse-free survival (RFS) associated with miRNA-10b and MCM5 mRNA were tested by Kaplan-Meier survival analysis. The impact of miRNA-10b andMCM5 mRNA expression on the survival was evaluated by Cox proportional hazard regression model. The expression of miRNA-10b and MCM5 mRNA was positive in 86.4 and 79.7 % breast cancer patients, respectively. The overall concordance rate between miRNA-10b and MCM5 RNA was 90.4 %. The median follow-up period was 50 months. The survival analysis showed that high levels of both miR-10b and MCM5 were associated with short relapse free survival of BC. We identified MCM5 mRNA expression changes consistent with the miRNA-10b target regulation. Thus, we could consider miRNA-10b and MCM5 mRNA as prognostic markers and potential therapeutic targets in breast cancer to be applied to other patient data sets.
Collapse
|
48
|
Xiao G, Tang Z, Yuan X, Yuan J, Zhao J, Zhang Z, He Z, Liu J. The expression of Wnt-1 inducible signaling pathway protein-2 in astrocytoma: Correlation between pathological grade and clinical outcome. Oncol Lett 2014; 9:235-240. [PMID: 25435966 PMCID: PMC4246620 DOI: 10.3892/ol.2014.2663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 10/15/2014] [Indexed: 01/16/2023] Open
Abstract
Wnt-1 inducible signaling pathway protein-2 (WISP-2) is a member of the CCN family, which is critical for the control of cell morphology, motion, adhesion and other processes involved in tumorigenesis. The expression pattern and clinical significance of WISP-2 in astrocytomas remains unclear. In this study, reverse transcription-polymerase chain reaction was performed to systematically investigate the expression of WISP-2 in 47 astrocytoma tissues of different pathological grades and 10 normal brain tissues. The mRNA expression levels of WISP-2 in the astrocytoma tissues were observed to be significantly higher than those in the normal brain tissues. Furthermore, the upregulation of WISP-2 was found to be associated with astrocytomas of higher pathological grades. Subsequently, 154 astrocytoma and 15 normal brain tissues were analyzed using immunohistochemistry and similar results were obtained. Univariate and multivariate survival analyses were used to determine the correlations between WISP-2 expression and overall survival (OS) and progression-free survival (PFS). The results indicated that the expression of WISP-2 was found to negatively correlate with patient PFS and OS. These results demonstrated that the WISP-2 protein is involved in the pathogenesis and progression of human astrocytomas and may serve as a malignant biomarker of this disease.
Collapse
Affiliation(s)
- Gelei Xiao
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xianrui Yuan
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| | - Jian Yuan
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| | - Jie Zhao
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| | - Zhiping Zhang
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| | - Zhengwen He
- Department of Neurosurgery, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jingping Liu
- The Institute of Skull Base Surgery and Neurooncology at Hunan, Xiangya Hospital, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
49
|
Haque I, Banerjee S, De A, Maity G, Sarkar S, Majumdar M, Jha SS, McGragor D, Banerjee SK. CCN5/WISP-2 promotes growth arrest of triple-negative breast cancer cells through accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation. Oncogene 2014; 34:3152-63. [PMID: 25132260 DOI: 10.1038/onc.2014.250] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 04/18/2014] [Accepted: 07/03/2014] [Indexed: 02/07/2023]
Abstract
The matricellular protein CCN5/WISP-2 represents a promising target in triple-negative breast cancer (TNBC) because treatment or induced activation of CCN5 in TNBC cells promotes cell growth arrest at the G0/G1 phase, reduces cell proliferation and delays tumor growth in the xenograft model. Our studies found that the p27(Kip1) tumor suppressor protein is upregulated and relocalized to the nucleus from cytoplasm by CCN5 in these cells and that these two events (upregulation and relocalization of p27(Kip1)) are critical for CCN5-induced growth inhibition of TNBC cells. In the absence of CCN5, p27(Kip1) resides mostly in the cytoplasm, which is associated with the aggressive nature of cancer cells. Mechanistically, CCN5 inhibits Skp2 expression, which seems to stabilize the p27(Kip1) protein in these cells. On the other hand, CCN5 also recruits FOXO3a to mediate the transcriptional regulation of p27(Kip1). The recruitment of FOXO3a is achieved by the induction of its expression and activity through shifting from cytoplasm to the nucleus. Our data indicate that CCN5 blocks PI3K/AKT signaling to dephosphorylate at S318, S253 and Thr32 in FOXO3a for nuclear relocalization and activation of FOXO3a. Moreover, inhibition of α6β1 receptors diminishes CCN5 action on p27(Kip1) in TNBC cells. Collectively, these data suggest that CCN5 effectively inhibits TNBC growth through the accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation, and thus, activation of CCN5 may have the therapeutic potential to kill TNBC.
Collapse
Affiliation(s)
- I Haque
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - A De
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - G Maity
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Sarkar
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| | - M Majumdar
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S S Jha
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - D McGragor
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S K Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA [3] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
50
|
Gits CMM, van Kuijk PF, de Rijck JCWM, Muskens N, Jonkers MBE, van IJcken WF, Mathijssen RHJ, Verweij J, Sleijfer S, Wiemer EAC. MicroRNA response to hypoxic stress in soft tissue sarcoma cells: microRNA mediated regulation of HIF3α. BMC Cancer 2014; 14:429. [PMID: 24927770 PMCID: PMC4065608 DOI: 10.1186/1471-2407-14-429] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 05/29/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hypoxia is often encountered in solid tumors and known to contribute to aggressive tumor behavior, radiation- and chemotherapy resistance resulting in a poor prognosis for the cancer patient. MicroRNAs (miRNAs) play a role in the regulation of the tumor cell response to hypoxia, however, not much is known about the involvement of miRNAs in hypoxic signalling pathways in soft tissue sarcomas (STS). METHOD A panel of twelve STS cell lines was exposed to atmospheric oxygen concentrations (normoxia) or 1% oxygen (hypoxia) for up to 48 h. Hypoxic conditions were verified and miRNA expression profiles were assessed by LNA™ oligonucleotide microarrays and RT-PCR after 24 h. The expression of target genes regulated by hypoxia responsive miRNAs is examined by end-point PCR and validated by luciferase reporter constructs. RESULTS Exposure of STS cell lines to hypoxic conditions gave rise to upregulation of Hypoxia Inducible Factor (HIF) 1α protein levels and increased mRNA expression of HIF1 target genes CA9 and VEGFA. Deregulation of miRNA expression after 24 h of hypoxia was observed. The most differentially expressed miRNAs (p<0.001) in response to hypoxia were miR-185-3p, miR-485-5p, miR-216a-5p (upregulated) and miR-625-5p (downregulated). The well-known hypoxia responsive miR-210-3p could not be reliably detected by the microarray platform most likely for technical reasons, however, its upregulation upon hypoxic stress was apparent by qPCR. Target prediction algorithms identified 11 potential binding sites for miR-485-5p and a single putative miR-210-3p binding site in the 3'UTR of HIF3α, the least studied member of the HIF family. We showed that HIF3α transcripts, expressing a 3'UTR containing the miR-485-5p and miR-210-3p target sites, are expressed in all sarcoma cell lines and upregulated upon hypoxia. Additionally, luciferase reporter constructs containing the 3'UTR of HIF3α were used to demonstrate regulation of HIF3α by miR-210-3p and miR-485-5p. CONCLUSION Here we provide evidence for the miRNA mediated regulation of HIF3α by hypoxia responsive miRNAs in STS, which may help to tightly regulate and fine-tune the hypoxic response. This provides a better insight into the mechanisms underlying the hypoxic response in STS and may ultimately yield information on novel prognostic and predictive markers or targets for treatment.
Collapse
Affiliation(s)
- Caroline MM Gits
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Patricia F van Kuijk
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jonneke CWM de Rijck
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nikky Muskens
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Moniek BE Jonkers
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Wilfred F van IJcken
- Center for Biomics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron HJ Mathijssen
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jaap Verweij
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Erik AC Wiemer
- Department of Medical Oncology, Erasmus University Medical Center – Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rm Be422, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|