1
|
Carbone L, Incognito GG, Incognito D, Nibid L, Caruso G, Berretta M, Taffon C, Palumbo M, Perrone G, Roviello F, Marrelli D. Clinical implications of epithelial-to-mesenchymal transition in cancers which potentially spread to peritoneum. Clin Transl Oncol 2025:10.1007/s12094-024-03837-2. [PMID: 39775727 DOI: 10.1007/s12094-024-03837-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a biological process by which epithelial cells increase their motility and acquire invasive capacity. It represents a crucial driver of cancer metastasis and peritoneal dissemination. EMT plasticity, with cells exhibiting hybrid epithelial/mesenchymal states, and its reverse process, mesenchymal-to-epithelial transition (MET), allows them to adapt to different microenvironments and evade therapeutic intervention. Resistance to conventional treatments, including chemotherapy, is a major problem. Therapies targeting EMT may inhibit tumour cell migration and invasion, while affecting normal cells and repair mechanisms, resulting in potential side effects. This paper addresses the question of the impact of EMT status on cancers with potential spread to the peritoneum, which has remained unclear in literature. Relevant studies were selected from 2000 to 2024. Three macrosections were analysed: (i) pathological characteristics, (ii) surgical implications and (iii) oncological therapies. The focus was on survival and peritoneal recurrence time in patients who underwent surgical treatment.
Collapse
Affiliation(s)
- Ludovico Carbone
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy.
| | - Giosuè Giordano Incognito
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Dalila Incognito
- Department of Human Pathology "G. Barresi", School of Specialization in Medical Oncology Unit, University of Messina, 98122, Messina, Italy
| | - Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Giuseppe Caruso
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Massimiliano Berretta
- Department of Human Pathology "G. Barresi", School of Specialization in Medical Oncology Unit, University of Messina, 98122, Messina, Italy
- Department of Clinical and Experimental Medicine, University of Messina, 98122, Messina, Italy
| | - Chiara Taffon
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Marco Palumbo
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Franco Roviello
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy
| | - Daniele Marrelli
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy
| |
Collapse
|
2
|
Malhotra K, Malik A, Almalki WH, Sahebkar A, Kesharwani P. Reactive Oxygen Species and its Manipulation Strategies in Cancer Treatment. Curr Med Chem 2025; 32:55-73. [PMID: 37303173 DOI: 10.2174/0929867330666230609110455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023]
Abstract
Cancer is one of the serious diseases of modern times, occurring in all parts of the world and shows a wide range of effects on the human body. Reactive Oxygen Species (ROS) such as oxide and superoxide ions have both advantages and disadvantages during the progression of cancer, dependent on their concentration. It is a necessary part of the normal cellular mechanisms. Changes in its normal level can cause oncogenesis and other relatable problems. Metastasis can also be controlled by ROS levels in the tumor cells, which can be prevented by the use of antioxidants. However, ROS is also used for the initiation of apoptosis in cells by different mediators. There exists a cycle between the production of oxygen reactive species, their effect on the genes, role of mitochondria and the progression of tumors. ROS levels cause DNA damage by the oxidation process, gene damage, altered expression of the genes and signalling mechanisms. They finally lead to mitochondrial disability and mutations, resulting in cancer. This review summarizes the important role and activity of ROS in developing different types of cancers like cervical, gastric, bladder, liver, colorectal and ovarian cancers.
Collapse
Affiliation(s)
- Kabil Malhotra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arzoo Malik
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
3
|
Fujimoto H, Yoshihara M, Rodgers R, Iyoshi S, Mogi K, Miyamoto E, Hayakawa S, Hayashi M, Nomura S, Kitami K, Uno K, Sugiyama M, Koya Y, Yamakita Y, Nawa A, Enomoto A, Ricciardelli C, Kajiyama H. Tumor-associated fibrosis: a unique mechanism promoting ovarian cancer metastasis and peritoneal dissemination. Cancer Metastasis Rev 2024; 43:1037-1053. [PMID: 38546906 PMCID: PMC11300578 DOI: 10.1007/s10555-024-10169-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 08/06/2024]
Abstract
Epithelial ovarian cancer (EOC) is often diagnosed in advanced stage with peritoneal dissemination. Recent studies indicate that aberrant accumulation of collagen fibers in tumor stroma has a variety of effects on tumor progression. We refer to remodeled fibrous stroma with altered expression of collagen molecules, increased stiffness, and highly oriented collagen fibers as tumor-associated fibrosis (TAF). TAF contributes to EOC cell invasion and metastasis in the intraperitoneal cavity. However, an understanding of molecular events involved is only just beginning to emerge. Further development in this field will lead to new strategies to treat EOC. In this review, we focus on the recent findings on how the TAF contributes to EOC malignancy. Furthermore, we will review the recent initiatives and future therapeutic strategies for targeting TAF in EOC.
Collapse
Affiliation(s)
- Hiroki Fujimoto
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Masato Yoshihara
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Raymond Rodgers
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - Shohei Iyoshi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Kazumasa Mogi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Emiri Miyamoto
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sae Hayakawa
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Maia Hayashi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynaecology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Mai Sugiyama
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Koya
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiko Yamakita
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Nawa
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia.
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
4
|
Wu T, Du X, Liu HH, Liu LY, Yang YK, Wang SJ, Duan CL. Bioactive solanidane steroidal alkaloids from Solanum lyratum. Fitoterapia 2024; 175:105916. [PMID: 38527590 DOI: 10.1016/j.fitote.2024.105916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/07/2024] [Accepted: 03/17/2024] [Indexed: 03/27/2024]
Abstract
Six previously unreported solanidane steroidal alkaloids, namely lyrasolanosides A-F, were isolated from Solanum lyratum. In addition, five known steroidal alkaloids were also identified. The structures of these compounds were determined through the use of NMR, HRESIMS,UV, IR and ECD analysis. To assess their bioactivities, the cytotoxic effects of the six previously unreported compounds were evaluated on A549 cells. The results revealed that lyrasolanoside B (2) exhibited the highest potency among them. Lyrasolanoside B (2) exhibited significant inhibition of cell migration, invasion, and adhesion dramatically. Mechanistically, it was found to suppress the activity of JAK2/STAT3 signaling pathway by downregulating the expression of phosphorylated JAK2/STAT3 in an exosome-dependent manner. In addition, lyrasolanoside B (2) was found to significantly upregulate the expression of E-cadherin and downregulate the expression of N-cadherin and vimentin. These findings indicate that lyrasolanoside B (2) inhibits the metastasis of A549 cells by suppressing exosome-mediated EMT. These findings suggest that lyrasolanoside B (2) may inhibit the metastasis of lung cancer by regulating A549-derived exosomes.
Collapse
Affiliation(s)
- Tong Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiao Du
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hai-Hui Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liang-Yu Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu-Ke Yang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Su-Juan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Chang-Ling Duan
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
5
|
Liu G, Li B, Qin S, Nice EC, Yang J, Yang L, Huang C. Redox signaling-mediated tumor extracellular matrix remodeling: pleiotropic regulatory mechanisms. Cell Oncol (Dordr) 2024; 47:429-445. [PMID: 37792154 DOI: 10.1007/s13402-023-00884-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The extracellular matrix (ECM), a fundamental constituent of all tissues and organs, is crucial for shaping the tumor microenvironment. Dysregulation of ECM remodeling has been closely linked to tumor initiation and progression, where specific signaling pathways, including redox signaling, play essential roles. Reactive oxygen species (ROS) are risk factors for carcinogenesis whose excess can facilitate the oxidative damage of biomacromolecules, such as DNA and proteins. Emerging evidence suggests that redox effects can aid the modification, stimulation, and degradation of ECM, thus affecting ECM remodeling. These alterations in both the density and components of the ECM subsequently act as critical drivers for tumorigenesis. In this review, we provide an overview of the functions and primary traits of the ECM, and it delves into our current understanding of how redox reactions participate in ECM remodeling during cancer progression. We also discuss the opportunities and challenges presented by clinical strategies targeting redox-controlled ECM remodeling to overcome cancer. CONCLUSIONS The redox-mediated ECM remodeling contributes importantly to tumor survival, progression, metastasis, and poor prognosis. A comprehensive investigation of the concrete mechanism of redox-mediated tumor ECM remodeling and the combination usage of redox-targeted drugs with existing treatment means may reveal new therapeutic strategy for future antitumor therapies.
Collapse
Affiliation(s)
- Guowen Liu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Jinlin Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Li Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China.
| |
Collapse
|
6
|
Jahani S, Zare N, Mirzaei Y, Arefnezhad R, Zarei H, Goleij P, Bagheri N. Mesenchymal stem cells and ovarian cancer: Is there promising news? J Cell Biochem 2023; 124:1437-1448. [PMID: 37682985 DOI: 10.1002/jcb.30471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023]
Abstract
Ovarian cancer (OC) is described as a heterogeneous complex condition with high mortality, weak prognosis, and late-stage presentation. OC has several subgroups based on different indices, like the origin and histopathology. The current treatments against OC include surgery followed by chemotherapy and radiotherapy; however, these methods have represented diverse side effects without enough effectiveness on OC. Recently, mesenchymal stem cell (MSC)-based therapy has acquired particular attention for treating diverse problems, such as cancer. These multipotent stem cells can be obtained from different sources, such as the umbilical cord, adipose tissues, bone marrow, and placenta, and their efficacy has been investigated against OC. Hence, in this narrative review, we aimed to review and discuss the present studies about the effects of various sources of MSCs on OC with a special focus on involved mechanisms.
Collapse
Affiliation(s)
| | - Nabi Zare
- Coenzyme R Research Institute, Tehran, Iran
| | - Yousef Mirzaei
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | | | - Hooman Zarei
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Sana Institute of Higher Education, Sari, Iran
- International Network of Stem Cell (INSC), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
7
|
Świerczewska M, Sterzyńska K, Ruciński M, Andrzejewska M, Nowicki M, Januchowski R. The response and resistance to drugs in ovarian cancer cell lines in 2D monolayers and 3D spheroids. Biomed Pharmacother 2023; 165:115152. [PMID: 37442067 DOI: 10.1016/j.biopha.2023.115152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer is the most common type of gynecologic cancer. One of the leading causes of high mortality is chemoresistance, developed primarily or during treatment. Different mechanisms of drug resistance appear at the cellular and cancer tissue organization levels. We examined the differences in response to the cytotoxic drugs CIS, MTX, DOX, VIN, PAC, and TOP using 2D (two-dimensional) and 3D (three-dimensional) culture methods. We tested the drug-sensitive ovarian cancer cell line W1 and established resistant cell lines to appropriate cytotoxic drugs. The following qualitative and quantitative methods were used to assess: 1) morphology - inverted microscope and hematoxylin & eosin staining; 2) viability - MTT assay; 3) gene expression - a quantitative polymerase chain reaction; 4) identification of proteins - immunohistochemistry, and immunofluorescence. Our results indicate that the drug-sensitive and drug-resistant cells cultured in 3D conditions exhibit stronger resistance than the cells cultured in 2D conditions. A traditional 2D model shows that drug resistance of cancer cells is caused mainly by changes in the expression of genes encoding ATP-binding cassette transporter proteins, components of the extracellular matrix, "new" established genes related to drug resistance in ovarian cancer cell lines, and universal marker of cancer stem cells. Whereas in a 3D model, the drug resistance in spheroids can be related to other mechanisms such as the structure of the spheroid (dense or loose), the cell type (necrotic, quiescent, proliferating cells), drug concentrations or drug diffusion into the dense cellular/ECM structure.
Collapse
Affiliation(s)
- Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Marcin Ruciński
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Małgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Radosław Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland.
| |
Collapse
|
8
|
Pharmacological targeting of the mitochondrial calcium-dependent potassium channel KCa3.1 triggers cell death and reduces tumor growth and metastasis in vivo. Cell Death Dis 2022; 13:1055. [PMID: 36539400 PMCID: PMC9768205 DOI: 10.1038/s41419-022-05463-8] [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: 07/27/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Ion channels are non-conventional, druggable oncological targets. The intermediate-conductance calcium-dependent potassium channel (KCa3.1) is highly expressed in the plasma membrane and in the inner mitochondrial membrane (mitoKCa3.1) of various cancer cell lines. The role mitoKCa3.1 plays in cancer cells is still undefined. Here we report the synthesis and characterization of two mitochondria-targeted novel derivatives of a high-affinity KCa3.1 antagonist, TRAM-34, which retain the ability to block channel activity. The effects of these drugs were tested in melanoma, pancreatic ductal adenocarcinoma and breast cancer lines, as well as in vivo in two orthotopic models. We show that the mitochondria-targeted TRAM-34 derivatives induce release of mitochondrial reactive oxygen species, rapid depolarization of the mitochondrial membrane, fragmentation of the mitochondrial network. They trigger cancer cell death with an EC50 in the µM range, depending on channel expression. In contrast, inhibition of the plasma membrane KCa3.1 by membrane-impermeant Maurotoxin is without effect, indicating a specific role of mitoKCa3.1 in determining cell fate. At sub-lethal concentrations, pharmacological targeting of mitoKCa3.1 significantly reduced cancer cell migration by enhancing production of mitochondrial reactive oxygen species and nuclear factor-κB (NF-κB) activation, and by downregulating expression of Bcl-2 Nineteen kD-Interacting Protein (BNIP-3) and of Rho GTPase CDC-42. This signaling cascade finally leads to cytoskeletal reorganization and impaired migration. Overexpression of BNIP-3 or pharmacological modulation of NF-κB and CDC-42 prevented the migration-reducing effect of mitoTRAM-34. In orthotopic models of melanoma and pancreatic ductal adenocarcinoma, the tumors at sacrifice were 60% smaller in treated versus untreated animals. Metastasis of melanoma cells to lymph nodes was also drastically reduced. No signs of toxicity were observed. In summary, our results identify mitochondrial KCa3.1 as an unexpected player in cancer cell migration and show that its pharmacological targeting is efficient against both tumor growth and metastatic spread in vivo.
Collapse
|
9
|
Clemente-González C, Carnero A. Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation. Cancers (Basel) 2022; 14:5930. [PMID: 36497411 PMCID: PMC9738438 DOI: 10.3390/cancers14235930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/18/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
During tumor growth, the delivery of oxygen to cells is impaired due to aberrant or absent vasculature. This causes an adaptative response that activates the expression of genes that control several essential processes, such as glycolysis, neovascularization, immune suppression, and the cancer stemness phenotype, leading to increased metastasis and resistance to therapy. Hypoxic tumor cells also respond to an altered hypoxic microenvironment by secreting vesicles, factors, cytokines and nucleic acids that modify not only the immediate microenvironment but also organs at distant sites, allowing or facilitating the attachment and growth of tumor cells and contributing to metastasis. Hypoxia induces the release of molecules of different biochemical natures, either secreted or inside extracellular vesicles, and both tumor cells and stromal cells are involved in this process. The mechanisms by which these signals that can modify the premetastatic niche are sent from the primary tumor site include changes in the extracellular matrix, recruitment and activation of different stromal cells and immune or nonimmune cells, metabolic reprogramming, and molecular signaling network rewiring. In this review, we will discuss how hypoxia might alter the premetastatic niche through different signaling molecules.
Collapse
Affiliation(s)
- Cynthia Clemente-González
- Instituto de Biomedicina de Sevilla (IBIS), Consejo Superior de Investigaciones Científicas, Hospital Universitario Virgen del Rocío (HUVR), Universidad de Sevilla, 41013 Seville, Spain
- CIBERONC (Centro de Investigación Biomédica en Red Cáncer), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Consejo Superior de Investigaciones Científicas, Hospital Universitario Virgen del Rocío (HUVR), Universidad de Sevilla, 41013 Seville, Spain
- CIBERONC (Centro de Investigación Biomédica en Red Cáncer), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
10
|
Ashraf R, Kumar S. Mfn2-mediated mitochondrial fusion promotes autophagy and suppresses ovarian cancer progression by reducing ROS through AMPK/mTOR/ERK signaling. Cell Mol Life Sci 2022; 79:573. [PMID: 36308626 PMCID: PMC11803038 DOI: 10.1007/s00018-022-04595-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/24/2022]
Abstract
Mitochondrial dynamics are balanced fission and fusion events that regulate mitochondrial morphology, and alteration in these events results in mitochondrial dysfunction and contributes to many diseases, including tumorigenesis. Ovarian cancer (OC) cells exhibit fragmented mitochondria, but the mechanism by which mitochondrial dynamics regulators contribute to OC is considerably less clear. Here, we elucidated the potential role of Mfn2-mediated mitochondrial fusion in OC and present evidence that genetic or pharmacological activation of Mfn2 leads to mitochondrial fusion and reduces ROS generation, which correlates with reduced cell proliferation, invasion, migration, and EMT in OC cells. Also, increased mitochondrial fusion promotes the F-actin remodeling, reduces lamellipodia formation, and thus reduces EMT. Increased expression of Mfn2 triggers AMPK, promotes autophagy, reduces ROS, and suppresses OC progression by downregulating the p-mTOR (2481 and 2448) and p-ERK axis. OC patients with higher Mfn2 expression have better survival than those with lower Mfn2 levels. Our findings demonstrate that restoration of Mfn2-mediated mitochondrial fusion suppressed OC progression and suggest that this process could be a potential strategy in OC treatment.
Collapse
Affiliation(s)
- Rahail Ashraf
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karkambadi Road, Rami Reddy Nagar, Mangalam, Tirupati, Andhra Pradesh, 517507, India
| | - Sanjay Kumar
- Division of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karkambadi Road, Rami Reddy Nagar, Mangalam, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
11
|
Ghoneum A, Almousa S, Warren B, Abdulfattah AY, Shu J, Abouelfadl H, Gonzalez D, Livingston C, Said N. Exploring the clinical value of tumor microenvironment in platinum-resistant ovarian cancer. Semin Cancer Biol 2021; 77:83-98. [PMID: 33476723 PMCID: PMC8286277 DOI: 10.1016/j.semcancer.2020.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Platinum resistance in epithelial ovarian cancer (OvCa) is rising at an alarming rate, with recurrence of chemo-resistant high grade serous OvCa (HGSC) in roughly 75 % of all patients. Additionally, HGSC has an abysmal five-year survival rate, standing at 39 % and 17 % for FIGO stages III and IV, respectively. Herein we review the crucial cellular interactions between HGSC cells and the cellular and non-cellular components of the unique peritoneal tumor microenvironment (TME). We highlight the role of the extracellular matrix (ECM), ascitic fluid as well as the mesothelial cells, tumor associated macrophages, neutrophils, adipocytes and fibroblasts in platinum-resistance. Moreover, we underscore the importance of other immune-cell players in conferring resistance, including natural killer cells, myeloid-derived suppressive cells (MDSCs) and T-regulatory cells. We show the clinical relevance of the key platinum-resistant markers and their correlation with the major pathways perturbed in OvCa. In parallel, we discuss the effect of immunotherapies in re-sensitizing platinum-resistant patients to platinum-based drugs. Through detailed analysis of platinum-resistance in HGSC, we hope to advance the development of more effective therapy options for this aggressive disease.
Collapse
Affiliation(s)
- Alia Ghoneum
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Sameh Almousa
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Bailey Warren
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Ammar Yasser Abdulfattah
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Alexandria University School of Medicine, Alexandria, Egypt
| | - Junjun Shu
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; The Third Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hebatullah Abouelfadl
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Department of Genetics, Animal Health Research Institute, Dokki, Egypt
| | - Daniela Gonzalez
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Christopher Livingston
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Neveen Said
- Departments of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Departments of Urology, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA; Comprehensive Cancer Center, Winston Salem, NC, 27157, USA.
| |
Collapse
|
12
|
Sarmiento-Salinas FL, Perez-Gonzalez A, Acosta-Casique A, Ix-Ballote A, Diaz A, Treviño S, Rosas-Murrieta NH, Millán-Perez-Peña L, Maycotte P. Reactive oxygen species: Role in carcinogenesis, cancer cell signaling and tumor progression. Life Sci 2021; 284:119942. [PMID: 34506835 DOI: 10.1016/j.lfs.2021.119942] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major causes of death in the world and its global burden is expected to continue increasing. In several types of cancers, reactive oxygen species (ROS) have been extensively linked to carcinogenesis and cancer progression. However, studies have reported conflicting evidence regarding the role of ROS in cancer, mostly dependent on the cancer type or the step of the tumorigenic process. We review recent studies describing diverse aspects of the interplay of ROS with cancer in the different stages of cancer progression, with a special focus on their role in carcinogenesis, their importance for cancer cell signaling and their relationship to the most prevalent cancer risk factors.
Collapse
Affiliation(s)
- Fabiola Lilí Sarmiento-Salinas
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Andrea Perez-Gonzalez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Adilene Acosta-Casique
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Adrián Ix-Ballote
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico; Posgrado en Ciencias y Tecnologías Biomédicas, Instituto Nacional de Astrofísica, Óptica y Electrónica, Puebla, Mexico
| | - Alfonso Diaz
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Samuel Treviño
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | | | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco, Puebla, Mexico.
| |
Collapse
|
13
|
Insights into the Role of Oxidative Stress in Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8388258. [PMID: 34659640 PMCID: PMC8516553 DOI: 10.1155/2021/8388258] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022]
Abstract
Oxidative stress (OS) arises when the body is subjected to harmful endogenous or exogenous factors that overwhelm the antioxidant system. There is increasing evidence that OS is involved in a number of diseases, including ovarian cancer (OC). OC is the most lethal gynecological malignancy, and risk factors include genetic factors, age, infertility, nulliparity, microbial infections, obesity, smoking, etc. OS can promote the proliferation, metastasis, and therapy resistance of OC, while high levels of OS have cytotoxic effects and induce apoptosis in OC cells. This review focuses on the relationship between OS and the development of OC from four aspects: genetic alterations, signaling pathways, transcription factors, and the tumor microenvironment. Furthermore, strategies to target aberrant OS in OC are summarized and discussed, with a view to providing new ideas for clinical treatment.
Collapse
|
14
|
Cai J, Cui Y, Yang J, Wang S. Epithelial-mesenchymal transition: When tumor cells meet myeloid-derived suppressor cells. Biochim Biophys Acta Rev Cancer 2021; 1876:188564. [PMID: 33974950 DOI: 10.1016/j.bbcan.2021.188564] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous myeloid cell population characterized by protumoral functions in the tumor immune network. An increasing number of studies have focused on the biological functions of MDSCs in tumor immunity. Epithelial-mesenchymal transition (EMT) is a cellular plasticity process accompanied by a loss of epithelial phenotypes and an acquisition of mesenchymal phenotypes. In general, tumor cells that undergo EMT are more likely to invade and metastasize. Recently, extensive evidence suggests that EMT is closely related to a highly immunosuppressive environment. This review will summarize the immunosuppressive capacities of MDSC subsets and their distinct role in tumor EMT and further discuss immunotherapy for tumor EMT by targeting MDSCs.
Collapse
Affiliation(s)
- Jingshan Cai
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yudan Cui
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
15
|
Non-Coding RNAs as Biomarkers of Tumor Progression and Metastatic Spread in Epithelial Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13081839. [PMID: 33921525 PMCID: PMC8069230 DOI: 10.3390/cancers13081839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Despite advances in cancer research in recent years, efficient predictive biomarkers of tumor progression and metastatic spread for ovarian cancer are still missing. Therefore, we critically address recent findings in the field of non-coding RNAs (microRNAs and long non-coding RNAs) and DNA methylation in ovarian cancer patients as promising novel biomarkers of ovarian cancer progression. Abstract Ovarian cancer is one of the most common causes of death among gynecological malignancies. Molecular changes occurring in the primary tumor lead to metastatic spread into the peritoneum and the formation of distant metastases. Identification of these changes helps to reveal the nature of metastases development and decipher early biomarkers of prognosis and disease progression. Comparing differences in gene expression profiles between primary tumors and metastases, together with disclosing their epigenetic regulation, provides interesting associations with progression and metastasizing. Regulatory elements from the non-coding RNA families such as microRNAs and long non-coding RNAs seem to participate in these processes and represent potential molecular biomarkers of patient prognosis. Progress in therapy individualization and its proper targeting also rely upon a better understanding of interactions among the above-listed factors. This review aims to summarize currently available findings of microRNAs and long non-coding RNAs linked with tumor progression and metastatic process in ovarian cancer. These biomolecules provide promising tools for monitoring the patient’s response to treatment, and further they serve as potential therapeutic targets of this deadly disease.
Collapse
|
16
|
Zhang X, Liu X, Cui W, Zhang R, Liu Y, Li Y, Hao J. Sohlh2 alleviates malignancy of EOC cells under hypoxia via inhibiting the HIF1α/CA9 signaling pathway. Biol Chem 2021; 401:263-271. [PMID: 31318683 DOI: 10.1515/hsz-2019-0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/08/2019] [Indexed: 01/26/2023]
Abstract
Epithelial ovarian cancer (EOC) is the most common and deadly ovarian cancer. Most of the patients have abdominal/pelvic invasion and metastasis at the time of diagnosis, but the underlying mechanism remains unclear. Insufficiency of blood perfusion and diffusion within most solid tumors can lead to a hypoxic tumor microenvironment and promotes tumor malignancy. In the present study, we detected the role of the spermatogenesis- and oogenesis-specific basic helix-loop-helix (bHLH) transcription factor 2 (sohlh2) on migration, invasion and epithelial-mesenchymal transition (EMT) of EOC cell lines under hypoxia in vitro. We also investigated the possible mechanism underlying it. The results showed that sohlh2 inhibited the migration, invasion and EMT of EOC cells and might function through suppression of the hypoxia-inducible factor 1α (HIF1α)/carbonic anhydrase 9 (CA9) signaling pathway. Our results may open a new avenue for the further development of diagnostic tools and novel therapeutics that will benefit EOC patients.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Xinyu Liu
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Weiwei Cui
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Ruihong Zhang
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Yang Liu
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Yongkun Li
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| | - Jing Hao
- Key Laboratory of The Ministry of Education for Experimental Teratology, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
17
|
Nowacka M, Sterzynska K, Andrzejewska M, Nowicki M, Januchowski R. Drug resistance evaluation in novel 3D in vitro model. Biomed Pharmacother 2021; 138:111536. [PMID: 34311534 DOI: 10.1016/j.biopha.2021.111536] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 01/09/2023] Open
Abstract
Ovarian cancer rates the highest mortality among all gynecological malignancies. The main reason for high mortality is the development of drug resistance. It can be related to changes in the expression of many drug resistance genes as well as expression of extracellular matrix proteins and cell density in the tumor. We developed a simple two-dimensional and three-dimensional model of drug sensitive A2780 and resistant to cisplatin and paclitaxel variants of ovarian cancer cell line. Using MTT assay, we compared drug resistance in two-dimensional and three-dimensional cell culture conditions. Real-time polymerase chain reaction analysis was used to compare the expression of drug resistance genes. The expression of proteins in spheroids was determined by immunohistochemistry. We observed a moderate increase in cisplatin resistance and a significant increase in paclitaxel resistance between two-dimensional and three-dimensional cell culture conditions. Our findings show that changes in the expression of drug resistance genes may play a crucial role in the drug resistance of cancer cells in traditional cell culture. On the other hand, the drug resistance in spheroids may result from different mechanisms such as cell density in the spheroid, extracellular matrix proteins expression and drug capacity to diffuse into the spheroid.
Collapse
Affiliation(s)
- Marta Nowacka
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Karolina Sterzynska
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Malgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, PL-61-781 Poznan, Poland.
| | - Radoslaw Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Gora, Zyty 28 St, 65-046 Zielona Gora, Poland.
| |
Collapse
|
18
|
Sharma A, Kaur H, De R, Srinivasan R, Pal A, Bhattacharyya S. Knockdown of E-cadherin induces cancer stem-cell-like phenotype and drug resistance in cervical cancer cells. Biochem Cell Biol 2021; 99:587-595. [PMID: 33677985 DOI: 10.1139/bcb-2020-0592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cervical cancer is one of the leading causes of mortality amongst women in developing countries, and resistance to therapy is the main reason for treatment failure. Recent advances suggest that cancer stem cells (CSCs) are critically involved in regulating the chemo-resistant behavior of cervical cancer cells. In our study, cells with the CSC phenotype were isolated, and we examined the expression levels of stem cell markers and genes associated with epithelial-mesenchymal transition (EMT) using different assays. However, the cells with the CSC phenotype could not be cultured for further cytotoxicity studies, so we established a model of CSC in cervical cancer cells. We performed siRNA-mediated knockdown of E-cadherin in these cells, and studied them for EMT-associated stem-cell-like properties. We also performed dose-dependent cell viability assays using clinically relevant drugs such as cisplatin, cyclopamine, and GANT58 to analyze the drug resistant behavior of these cancer cells. We found that knockdown of E-cadherin induces EMT in cervical cancer cells, imparting stem-cell like characteristics along with enhanced tumorsphere formation, cell migration, invasiveness, and drug resistance. This is the first study to establish a CSC model in cervical cancer cells by knockdown of E-cadherin, which can be used to develop anti-cancer therapies.
Collapse
Affiliation(s)
- Anuka Sharma
- Department of Biophysics, PGIMER, Chandigarh, India
| | | | - Renaissa De
- Department of Biophysics, PGIMER, Chandigarh, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecologic Pathology, PGIMER, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, PGIMER, Chandigarh, India
| | | |
Collapse
|
19
|
Wei L, He Y, Bi S, Li X, Zhang J, Zhang S. miRNA‑199b‑3p suppresses growth and progression of ovarian cancer via the CHK1/E‑cadherin/EMT signaling pathway by targeting ZEB1. Oncol Rep 2021; 45:569-581. [PMID: 33416170 PMCID: PMC7757082 DOI: 10.3892/or.2020.7895] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/01/2020] [Indexed: 11/05/2022] Open
Abstract
Ovarian cancer is one of the most common gynecological malignancies and its pathogenesis and progression are regulated by multiple genes. MicroRNAs (miRNAs) are endogenous non‑coding RNAs that regulate body function by altering post‑transcriptional gene expression. Previous studies have suggested that miRNAs are closely associated with the pathogenesis and progression of several malignancies, including breast cancer, hepatocellular carcinoma and glioma, among others. Therefore, miRNAs are promising novel targets for the diagnosis, treatment and determination of prognostic factors in patients with ovarian cancer. In the present study, the role of miRNA‑133b‑3p in ovarian cancer progression and its possible mechanism of action were investigated. The results demonstrated that the expression of miRNA‑199b‑3p and zinc finger E‑box binding homeobox (ZEB)1 were increased in patients with ovarian cancer. The overall survival (OS) and disease‑free survival (DFS) of patients with ovarian cancer and high miRNA‑199b‑3p expression were prolonged compared with those of patients with low miRNA‑199b‑3p expression. Additionally, the OS and DFS of patients with ovarian cancer and low ZEB1 expression were longer compared with those of patients with high ZEB1 expression. Furthermore, miRNA‑199b‑3p overexpression reduced cell proliferation and promoted apoptosis in an in vitro model of ovarian cancer. miRNA‑199b‑3p overexpression also suppressed ZEB1 and checkpoint kinase 1 expression and induced E‑cadherin expression and epithelial‑to‑mesenchymal transition in this model. Furthermore, the effects of miRNA‑199b‑3p‑mediated apoptosis and migration were attenuated by ZEB1 and E‑cadherin, respectively. The results of the present study indicated that miRNA‑199b‑3p suppressed ovarian cancer progression by targeting ZEB1, which may represent a promising therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Liqun Wei
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Yuanqi He
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Shuhong Bi
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Xiaoxiao Li
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Jianzhong Zhang
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Shihong Zhang
- Department of Gynecology and Obstetrics, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200, P.R. China
- Department of Gynecology and Obstetrics, Affiliated Hospital of Beihua University, Jilin, Jilin 132001, P.R. China
| |
Collapse
|
20
|
Perrone MG, Luisi O, De Grassi A, Ferorelli S, Cormio G, Scilimati A. Translational Theragnosis of Ovarian Cancer: where do we stand? Curr Med Chem 2020; 27:5675-5715. [PMID: 31419925 DOI: 10.2174/0929867326666190816232330] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/13/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ovarian cancer is the second most common gynecologic malignancy, accounting for approximately 220,000 deaths annually worldwide. Despite radical surgery and initial high response rates to platinum- and taxane-based chemotherapy, most patients experience a relapse, with a median progression-free survival of only 18 months. Overall survival is approximately 30% at 5 years from the diagnosis. In comparison, patients out from breast cancer are more than 80 % after ten years from the disease discovery. In spite of a large number of published fundamental and applied research, and clinical trials, novel therapies are urgently needed to improve outcomes of the ovarian cancer. The success of new drugs development in ovarian cancer will strongly depend on both fully genomic disease characterization and, then, availability of biomarkers able to identify women likely to benefit from a given new therapy. METHODS In this review, the focus is given to describe how complex is the diseases under the simple name of ovarian cancer, in terms of cell tumor types, histotypes, subtypes, and specific gene mutation or differently expressed in the tumor with respect the healthy ovary. The first- and second-line pharmacological treatment clinically used over the last fifty years are also described. Noteworthy achievements in vitro and in vivo tested new drugs are also summarized. Recent literature related to up to date ovarian cancer knowledge, its detection by biomarkers and chemotherapy was searched from several articles on Pubmed, Google Scholar, MEDLINE and various Governmental Agencies till April 2019. RESULTS The papers referenced by this review allow a deep analysis of status of the art in the classification of the several types of ovarian cancer, the present knowledge of diagnosis based on biomarkers and imaging techniques, and the therapies developed over the past five decades. CONCLUSION This review aims at stimulating more multi-disciplinary efforts to identify a panel of novel and more specific biomarkers to be used to screen patients for a very early diagnosis, to have prognosis and therapy efficacy indications. The desired final goal would be to have available tools allowing to reduce the recurrence rate, increase both the disease progression free interval and of course the overall survival at five years from the diagnosis that today is still very low.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Oreste Luisi
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Oncologico "Giovanni Paolo II" Bari, Italy
| | - Antonio Scilimati
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
21
|
Ye M, Zhou J, Gao Y, Pan S, Zhu X. The prognostic value of the lysyl oxidase family in ovarian cancer. J Clin Lab Anal 2020; 34:e23538. [PMID: 33058284 PMCID: PMC7755792 DOI: 10.1002/jcla.23538] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Our study intended to evaluate the prognostic value of lysyl oxidase (LOX) and its four relevant members, the lysyl oxidase-like genes (LOXL1-4), in ovarian cancer (OC) patients. MATERIAL AND METHODS The Kaplan-Meier plotter (KM plotter) database was used to investigate the prognostic power of the LOX family for OC patients. Overall survival (OS) and progression-free survival (PFS) were the clinical endpoints. The prognostic roles of the LOX family in OC patients were also analyzed according to various clinicopathological characteristics, including histological subtypes, clinical stages, pathological grades, and chemotherapeutic treatments. RESULTS Overexpression of LOX, LOXL1, LOXL2, and LOXL3 mRNA indicated poor OS and PFS in OC patients, particularly in serous and grade II + III OC patients. Overexpression of LOXL4 mRNA resulted in worse PFS in OC patients. Overexpression of LOX and LOXL1 mRNA showed worse OS and PFS in stage III + IV OC patients, and overexpression of LOXL3 mRNA indicated worse OS and PFS in stage I + II OC patients. Overexpression of LOX, LOXL3, and LOXL4 mRNA indicated worse OS and PFS among OC patients who received platinum, taxol, and taxol + platinum chemotherapy. Overexpression of LOXL1 and LOXL2 mRNA was related to lower OS and PFS in OC patients who received platinum chemotherapy. CONCLUSION LOX, LOXL1, LOXL2, and LOXL3 may become potential predictive markers for negative outcomes in OC patients. Moreover, the LOX family can serve as new molecular predictors for the efficiency of platinum-based chemotherapy in OC patients.
Collapse
Affiliation(s)
- Miaomiao Ye
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Junhan Zhou
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ying Gao
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuya Pan
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xueqiong Zhu
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
22
|
Zhuo S, Chen Z, Yang Y, Zhang J, Tang J, Yang K. Clinical and Biological Significances of a Ferroptosis-Related Gene Signature in Glioma. Front Oncol 2020; 10:590861. [PMID: 33330074 PMCID: PMC7718027 DOI: 10.3389/fonc.2020.590861] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/23/2020] [Indexed: 01/10/2023] Open
Abstract
Ferroptosis is a form of cell death characterized by non-apoptosis induced by small molecules in tumors. Studies have demonstrated that ferroptosis regulates the biological behaviors of tumors. Therefore, genes that control ferroptosis can be a promising candidate bioindicator in tumor therapy. Herein, functions of ferroptosis-related genes in glioma were investigated. We systematically assessed the relationship between ferroptosis-related genes expression profiles and prognosis in glioma patients based on The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) RNA sequencing datasets. Using the non-negative matrix factorization (NMF) clustering method, 84 ferroptosis-related genes in the RNA sequencing data were distinctly classified into two subgroups (named cluster 1 and cluster 2) in glioma. The least absolute shrinkage and selection operator (LASSO) was used to develop a 25 gene risk signature. The relationship between the gene risk signature and clinical features in glioma was characterized. Results show that the gene risk signature associated with clinical features can be as an independent prognostic indicator in glioma patients. Collectively, the ferroptosis-related risk signature presented in this study can potentially predict the outcome of glioma patients.
Collapse
Affiliation(s)
- Shenghua Zhuo
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Zhimin Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yibei Yang
- Department of Physical Education, Hainan Normal University, Haikou, China
| | - Jinben Zhang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Jianming Tang
- Department of Radiation Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Kun Yang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical College, Haikou, China
| |
Collapse
|
23
|
Liu Y, Xiang D, Zhang H, Yao H, Wang Y. Hypoxia-Inducible Factor-1: A Potential Target to Treat Acute Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8871476. [PMID: 33282113 PMCID: PMC7685819 DOI: 10.1155/2020/8871476] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extrapulmonary injury factors. Presently, excessive inflammation in the lung and the apoptosis of alveolar epithelial cells are considered to be the key factors in the pathogenesis of ALI. Hypoxia-inducible factor-1 (HIF-1) is an oxygen-dependent conversion activator that is closely related to the activity of reactive oxygen species (ROS). HIF-1 has been shown to play an important role in ALI and can be used as a potential therapeutic target for ALI. This manuscript will introduce the progress of HIF-1 in ALI and explore the feasibility of applying inhibitors of HIF-1 to ALI, which brings hope for the treatment of ALI.
Collapse
Affiliation(s)
- Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115 MA, USA
| | - Hanlin Yao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| |
Collapse
|
24
|
Ye M, Song Y, Pan S, Chu M, Wang ZW, Zhu X. Evolving roles of lysyl oxidase family in tumorigenesis and cancer therapy. Pharmacol Ther 2020; 215:107633. [PMID: 32693113 DOI: 10.1016/j.pharmthera.2020.107633] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
The lysyl oxidase (LOX) family is comprised of LOX and four LOX-like proteins (LOXL1, LOXL2, LOXL3, and LOXL4), and mainly functions in the remodeling of extracellular matrix (ECM) and the cross-linking of collagen and elastic fibers. Recently, a growing body of research has demonstrated that LOX family is critically involved in the regulation of cancer cell proliferation, migration, invasion and metastasis. In this review, we discuss the roles of LOX family members in the development and progression of different types of human cancers. Furthermore, we also describe the potential inhibitors of LOX family proteins and highlight that LOX family might be an important therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shuya Pan
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Man Chu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China..
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
25
|
Yan W, Xie M, Li R, Hu H, Tang B, Shen J. Identification and Validation of Reference Genes Selection in Ovarian Cancer Exposed to Hypoxia. Onco Targets Ther 2020; 13:7423-7431. [PMID: 32801765 PMCID: PMC7395691 DOI: 10.2147/ott.s249733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Hypoxia-mediated tumor metastasis, progression and drug resistance are major clinical challenges in ovarian cancer. Meanwhile, the genetic basis of these traits is still not clear. RT-qPCR, as an efficient and sensitive gene expression technique, has been widely used for gene analyses, providing a basis for in-depth understanding of molecular changes in different microenvironments. However, there is currently a lack of suitable reference genes to normalize the data associated with hypoxia in ovarian cancer cells. METHODS A systematic method is needed to select the most suitable reference gene. Here, eight candidate reference genes (GAPDH, β-actin, 18S RNA, TUBB, PPIA, TBP, RPL13A and SDHA) from humans were selected to assess their expression levels in SKOV3 cells under hypoxia. The geNorm and NormFinder programs were utilized to evaluate the expression stabilities of these selected candidate reference genes. RESULTS Interestingly, 18S RNA was considered to be an ideal reference gene for the normalization of target gene expression under hypoxic conditions. Furthermore, this result was confirmed in another two ovarian cancer cell line, CAOV3 and OVCAR3 cell line. Finally, these results suggest that appropriate reference genes should be selected before performing gene expression analysis during hypoxic environmental exposure. CONCLUSION 18S RNA can be used as an appropriate reference gene for the study of gene expression in ovarian cancer samples under hypoxia by RT-qPCR.
Collapse
Affiliation(s)
- Wenying Yan
- Department of Gynecology, Wangjiang Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Mei Xie
- Department of Respiratory and Critical Care Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Rong Li
- Department of Gynecology, Sichuan Maternal and Child Health Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Hongmei Hu
- Department of Gynecology, Sichuan Maternal and Child Health Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Biao Tang
- Department of Gynecology, Sichuan Maternal and Child Health Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Jie Shen
- Department of Orthopaedics, The First Hospital Affiliated to AMU (Southwest Hospital), Chongqing, People’s Republic of China
| |
Collapse
|
26
|
Roque R, Costa Sousa F, Figueiredo-Dias M. Epithelial-mesenchymal interconversions in ovarian cancer: The levels and functions of E-cadherin in intraabdominal dissemination. Oncol Rev 2020; 14:475. [PMID: 32676171 PMCID: PMC7358986 DOI: 10.4081/oncol.2020.475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
The metastatic process of ovarian cancer (OC) is almost exclusively defined by direct shedding of tumor cells into the abdominal cavity, followed by clustering into multicellular aggregates and posterior peritoneal anchorage. This process relies on dynamic intercellular interactions which are modified by epithelial- mesenchymal interconversions and, therefore, E-cadherin expression variability. Although widely accepted as a tumor suppressor in many types of cancer, E-cadherin is currently known to have a dynamic expression and a much more complex role in OC. First, high E-cadherin expression is considered a sign of metaplasia in the normal ovarian epithelium, due to its association with epithelial growth factor receptor (EGFR) mediated cell proliferation. Subsequently, it is the decreased expression of E-cadherin that allows the acquisition of a more invasive phenotype, leading to the spread of primary tumor cells into the peritoneal fluid. This downregulation seems to depend on complex regulatory mechanisms, from molecular proteolysis to microenvironment interference and epigenetic regulation. E-cadherin cleavage and its resulting fragments appear to be essential to the process of dissemination and even to the formation of multicellular aggregates. Paradoxically, the maintenance of some E-cadherin expression seems to promote intercellular adhesion, resistance, and survival while decreasing cancer response to chemotherapy. Multiple studies have shown that reversing epithelial-mesenchymal transaction (EMT) and increasing E-cadherin expression prevents OC intraperitoneal dissemination, but findings that simultaneously correlate E-cadherin downregulation to higher chemotherapy sensitivity should not be ignored. Nevertheless, EMT and E-cadherin seem to have a potential interest as therapeutic targets in novel approaches to OC treatment.
Collapse
Affiliation(s)
| | - Filipa Costa Sousa
- Universitary Clinic of Gynecology, Faculty of Medicine, University of Coimbra
- Gynecology Department, Universitary Hospital Center of Coimbra, Portugal
| | - Margarida Figueiredo-Dias
- Universitary Clinic of Gynecology, Faculty of Medicine, University of Coimbra
- Gynecology Department, Universitary Hospital Center of Coimbra, Portugal
| |
Collapse
|
27
|
Inhibition of miR-214-3p Aids in Preventing Epithelial Ovarian Cancer Malignancy by Increasing the Expression of LHX6. Cancers (Basel) 2019; 11:cancers11121917. [PMID: 31810245 PMCID: PMC6966693 DOI: 10.3390/cancers11121917] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
In human epithelial ovarian cancer (EOC), various miRNAs can function as either oncogenes or tumor suppressor genes. We investigated miRNAs known to be involved in EOC progression and analyzed their expression in tissues and serum-derived exosomes from benign serous cystadenoma, borderline serous tumor, low-grade serous ovarian cancer, and high-grade serous ovarian cancer patients (HGSO). The HGSO group was divided based on the platinum-free interval, which is defined as the duration from the completion of platinum-based chemotherapy to recurrence. We also analyzed the mRNA levels of target genes that candidate miRNAs might regulate in patient tissues. miR-214-3p was highly expressed in tissues and exosomes derived from EOC with high malignancy and also found to regulate the expression of LIM homeobox domain 6 (LHX6) mRNA. Serum exosomal levels of miR-214-3p were significantly increased in platinum-resistant HGSO (25.2-fold, p < 0.001) compared to the exosomal expression of benign tumor patients. On transfection of miR-214-3p inhibitor in EOC cells, cell proliferation was inhibited while apoptotic cell death was increased. Collectively, we suggest that miR-214-3p in serum exosomes can be a potential biomarker for the diagnosis and prognosis of ovarian tumor, and its inhibition can be a supportive treatment for EOC.
Collapse
|
28
|
Michalcova K, Roychoudhury S, Halenar M, Tvrda E, Kovacikova E, Vasicek J, Chrenek P, Baldovska S, Sanislo L, Kren V, Kolesarova A. In vitro response of human ovarian cancer cells to dietary bioflavonoid isoquercitrin. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2019; 54:752-757. [PMID: 31271108 DOI: 10.1080/03601234.2019.1633214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Isoquercitrin is a dietary bioflavonoid used as a food supplement. We studied the mechanism underlying its effect in human ovarian cancer cells using OVCAR-3 cell line. Viability, survival, apoptosis, release of human transforming growth factor-β1 (TGF-β1) and TGF-β1 receptor, and intracellular reactive oxygen species (ROS) generation by OVCAR-3 cells were examined after isoquercitrin treatment at concentrations 5, 10, 25, 50, and 100 μg mL-1. AlamarBlue assay revealed that isoquercitrin did not cause any significant change (P > 0.05) in cell viability as compared to control. Apoptotic assay using flow cytometry did not find any significant change (P > 0.05) in the proportion of live, dead and apoptotic cells as compared to control. ELISA also showed that the release of human TGF-β1 and TGF-β1 receptor were not significantly (P > 0.05) affected by isoquercitrin as compared to control. Chemiluminescence assay demonstrated that lower concentrations (5, 10, and 25 μg mL-1) were able to exhibit beneficial effects by inhibiting the generation of intracellular ROS. In contrast, elevated concentrations of 50 and 100 μg mL-1 led to oxidative stress (P < 0.05). We concluded that the beneficial effect of isoquercitrin on ovarian cancer cells may be mediated by an antioxidative pathway that involves inhibition of intracellular ROS generation, thereby limiting oxidative stress.
Collapse
Affiliation(s)
- Katarina Michalcova
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| | - Shubhadeep Roychoudhury
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
- Department of Life Science and Bioinformatics , Assam University , Silchar , India
| | - Marek Halenar
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| | - Eva Tvrda
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| | - Eva Kovacikova
- Research Centre AgroBioTech , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| | - Jaromir Vasicek
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
- Research Institute for Animal Production Nitra , National Agricultural and Food Centre , Lužianky , Slovak Republic
| | - Peter Chrenek
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
- Research Institute for Animal Production Nitra , National Agricultural and Food Centre , Lužianky , Slovak Republic
| | - Simona Baldovska
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| | - Luboslav Sanislo
- St. Elizabeth Cancer Institute Hospital , Bratislava , Slovak Republic
| | - Vladimir Kren
- Institute of Microbiology , Czech Academy of Sciences , Prague 4 , Czech Republic
| | - Adriana Kolesarova
- Faculty of Biotechnology and Food Sciences , Slovak University of Agriculture in Nitra , Nitra , Slovak Republic
| |
Collapse
|
29
|
Ma W, Li T, Wu S, Li J, Wang X, Li H. LOX and ACSL5 as potential relapse markers for pancreatic cancer patients. Cancer Biol Ther 2019; 20:787-798. [PMID: 30712446 DOI: 10.1080/15384047.2018.1564565] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is one of the most malignant diseases and has a poor prognosis. The screening and validation of biomarkers with predictive value for prognosis and treatment efficacy are important. To identify potential prognostic markers of pancreatic cancer patients, we conducted a study that included 99 pancreatic cancer patients. Three patients with PFS>18 months were enrolled in the treat group, and three patients with PFS<12 months were enrolled in the control group. Differentially expressed genes (DEGs) between these two groups were analyzed by whole-genome expression microarray. A total of 178 DEGs were identified, including 110 up-regulated and 68 down-regulated genes. Next, 24 candidate genes were selected for validation by qPCR based on fold change and previous studies. The results showed that the mRNA levels of four candidate genes, including ACSL5, SLC44A4, LOX, and TOX3, were correlated with PFS. Immunohistochemical staining was performed to validate the protein expression levels of these four markers. The results showed that patients with LOX high, ACSL5 low and TOX3 low expression had a significantly shorter PFS than those with LOX low, ACSL5 high and TOX3 high expression. Multivariable analysis revealed differentiation, tumor stage, LOX expression, and ACSL5 expression were independent prognostic factors for PFS. Then, we use the TCGA database to explore the underlying mechanism of LOX influence pancreatic cancer progression. Protein-protein interaction network of ACSL5 was established by STRING to uncover the potential regulation mechanism. Our findings reveal that LOX and ACSL5 are potential prognostic markers for the prognosis of pancreatic cancer patients.
Collapse
Affiliation(s)
- Weidong Ma
- a Department of Pancreatic Cancer , National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Ting Li
- b Department of Gastrointestinal Cancer Biology , Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Si Wu
- b Department of Gastrointestinal Cancer Biology , Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Jian Li
- a Department of Pancreatic Cancer , National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Xiuchao Wang
- a Department of Pancreatic Cancer , National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Hui Li
- b Department of Gastrointestinal Cancer Biology , Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| |
Collapse
|
30
|
Mutual Expression of ALDH1A1, LOX, and Collagens in Ovarian Cancer Cell Lines as Combined CSCs- and ECM-Related Models of Drug Resistance Development. Int J Mol Sci 2018; 20:ijms20010054. [PMID: 30583585 PMCID: PMC6337354 DOI: 10.3390/ijms20010054] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022] Open
Abstract
A major contributor leading to treatment failure of ovarian cancer patients is the drug resistance of cancer cell. CSCs- (cancer stem cells) and ECM (extracellular matrix)-related models of drug resistance are described as independently occurring in cancer cells. Lysyl oxidase (LOX) is another extracellular protein involved in collagen cross-linking and remodeling of extracellular matrix and has been correlated with tumor progression. The expression of LOX, COL1A2, COL3A1, and ALDH1A1 was performed in sensitive (A2780, W1) and resistant to paclitaxel (PAC) (A2780PR1 and W1PR2) and topotecan (TOP) (W1TR) cell lines at the mRNA (real-time PCR analysis) and protein level (Western blot and immunofluorescence analysis). The ALDH1A1 activity was measured with the ALDEFLUOR test and flow cytometry analysis. The protein expression in ovarian cancer tissues was determined by immunohistochemistry. We observed an increased expression of LOX and collagens in PAC and TOP resistant cell lines. Subpopulations of ALDH1A1 positive and negative cells were also noted for examined cell lines. Additionally, the coexpression of LOX with ALDH1A1 and COL1A2 with ALDH1A1 was observed. The expression of LOX, collagens, and ALDH1A1 was also detected in ovarian cancer lesions. In our study LOX, ALDH1A1 and collagens were found to be coordinately expressed by cells resistant to PAC (LOX, ALDH1A1, and COL1A2) or to TOP (LOX and ALDH1A1). This represents the study where molecules related with CSCs (ALDH1A1) and ECM (LOX, collagens) models of drug resistance are described as occurring simultaneously in ovarian cancer cells treated with PAC and TOP.
Collapse
|
31
|
Xu Y, Wang X, Huang Y, Ma Y, Jin X, Wang H, Wang J. Inhibition of lysyl oxidase expression by dextran sulfate affects invasion and migration of gastric cancer cells. Int J Mol Med 2018; 42:2737-2749. [PMID: 30226558 PMCID: PMC6192768 DOI: 10.3892/ijmm.2018.3855] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 08/23/2018] [Indexed: 12/15/2022] Open
Abstract
In the present study, the effect of dextran sulfate (DS) on the metastasis and invasion of human gastric cancer cells and its key underlying mechanism were investigated. The levels of hypoxia‑inducible factor 1α (HIF‑1α), transforming growth factor β (TGF‑β) and lysyl oxidase (LOX) expression were evaluated in human gastric cancer and peritumoral tissues by immunohistochemical analysis. Cell proliferation and apoptosis were also examined using the Cell Counting Kit‑8 assay and flow cytometry. The effect of DS on the invasion and migration of BGC‑823 cells was assessed using a Transwell assay. BGC‑823 cells were divided into the control (phosphate‑buffered saline‑treated) and experimental (DS‑treated) groups, and cultured for different times under hypoxic conditions. Subsequently, LOX and TGF‑β expression levels in the cells were measured by immunocytochemistry, immunofluorescence, reverse transcription‑quantitative polymerase chain reaction and western blot analysis. HIF‑1α, TGF‑β and LOX expression levels were significantly higher in human gastric cancer tissues as compared with that in adjacent tissues. DS influenced cell proliferation and apoptosis in a dose‑dependent manner. Furthermore, DS reduced the number of invaded and migrated cells. Under hypoxic conditions, DS reduced HIF‑1α, TGF‑β and LOX expression levels in BGC‑823 cells. After 12 h, the effect of combination of DS and β‑aminopropionitrile (BAPN) on LOX and TGF‑β protein levels was more significant compared with that of DS or BAPN alone. Therefore, DS may inhibit the invasion and migration of human gastric cancer cells under hypoxic conditions by influencing LOX.
Collapse
Affiliation(s)
- Yuanyi Xu
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750001, P.R. China
| | - Xiaofei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yunning Huang
- Department of Gastrointestinal Surgery, Ningxia People's Hospital, Yinchuan, Ningxia 750021, P.R. China
| | - Yanmei Ma
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750001, P.R. China
| | - Xiu Jin
- Department of Pathology, Affiliated Hospital of Jining Medical College, Jining, Shandong 272029, P.R. China
| | - Honghong Wang
- Department of Pathology, Ningxia People's Hospital, Yinchuan, Ningxia 750021, P.R. China
| | - Juan Wang
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750001, P.R. China
| |
Collapse
|
32
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018; 10:E302. [PMID: 30200478 PMCID: PMC6162424 DOI: 10.3390/cancers10090302] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
33
|
Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
|
34
|
Jin J, Cao L, Wang L, Wang Z, Ding W, Mao L, Suo S, Zhuang Z, Tong H. Expression of hypoxia-inducible factor-1a predicts benefit from rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in diffuse large B-cell lymphoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4472-4482. [PMID: 31949844 PMCID: PMC6962976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/21/2018] [Indexed: 06/10/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) has been identified as an unfavorable prognostic factor in most solid tumors. However, HIF-1α was suggested to predict improved survival in Western patients with diffuse large B-cell lymphoma (DLBCL) under rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) treatment. We studied HIF-1α protein expression by immunohistochemical staining of 155 paraffin-embedded specimens from Chinese patients with DLBCL treated with R-CHOP or CHOP. Results were correlated with patient outcome. HIF-1α expression had no impact on survival for the patients treated with CHOP. In the R-CHOP-treated group, however, HIF-1α expression was significantly correlated with superior OS and EFS (P = 0.048 and 0.040, respectively). Moreover, HIF-1α expression maintained independent prognostic value for OS (RR, 0.41; 95% CI, 0.19-0.92; P = 0.030) and EFS (RR, 0.53; 95% CI, 0.31-0.90; P = 0.020) when it was adjusted by IPI stratification. Therefore, HIF-1α expression benefits from R-CHOP in DLBCL.
Collapse
Affiliation(s)
- Jie Jin
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, People’s Republic of China
- Institute of Hematology, Zhejiang University School of MedicineHangzhou, People’s Republic of China
| | - Lihong Cao
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, People’s Republic of China
- Institute of Hematology, Zhejiang University School of MedicineHangzhou, People’s Republic of China
- Department of Hematology, Shulan (Hangzhou) HospitalHangzhou, People’s Republic of China
| | - Lijun Wang
- Department of Pathology, The Fourth Affiliated Hospital of Zhejiang UniversityYiwu 322000, People’s Republic of China
| | - Zhaoming Wang
- Department of Pathology, The First Affiliated Hospital of Zhejiang UniversityHangzhou, People’s Republic of China
| | - Wei Ding
- Department of Pathology, The First Affiliated Hospital of Zhejiang UniversityHangzhou, People’s Republic of China
| | - Liping Mao
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, People’s Republic of China
- Institute of Hematology, Zhejiang University School of MedicineHangzhou, People’s Republic of China
| | - Shanshan Suo
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, People’s Republic of China
- Institute of Hematology, Zhejiang University School of MedicineHangzhou, People’s Republic of China
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesda, MD, USA
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, People’s Republic of China
- Institute of Hematology, Zhejiang University School of MedicineHangzhou, People’s Republic of China
| |
Collapse
|
35
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
36
|
Ghoneum A, Afify H, Salih Z, Kelly M, Said N. Role of tumor microenvironment in the pathobiology of ovarian cancer: Insights and therapeutic opportunities. Cancer Med 2018; 7:5047-5056. [PMID: 30133163 PMCID: PMC6198242 DOI: 10.1002/cam4.1741] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/15/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is the fifth most common cancer affecting women and at present, stands as the most lethal gynecologic malignancy. The poor disease outcome is due to the nonspecific symptoms and the lack of effective treatment at advanced stages. Thus, it is of utmost importance to understand ovarian carcinoma through several lenses and to dissect the role that the unique peritoneal tumor microenvironment plays in ovarian cancer progression and metastasis. This review seeks to highlight several determinants of this unique tumor microenvironment, their influence on disease outcome and ongoing clinical trials targeting these determinants.
Collapse
Affiliation(s)
- Alia Ghoneum
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Hesham Afify
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Ziyan Salih
- Department of Pathology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Michael Kelly
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Neveen Said
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina.,Department of Pathology, Wake Forest University School of Medicine, Winston Salem, North Carolina.,Department of Urology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
37
|
The Role of Inflammation and Inflammatory Mediators in the Development, Progression, Metastasis, and Chemoresistance of Epithelial Ovarian Cancer. Cancers (Basel) 2018; 10:cancers10080251. [PMID: 30061485 PMCID: PMC6116184 DOI: 10.3390/cancers10080251] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Inflammation plays a role in the initiation and development of many types of cancers, including epithelial ovarian cancer (EOC) and high grade serous ovarian cancer (HGSC), a type of EOC. There are connections between EOC and both peritoneal and ovulation-induced inflammation. Additionally, EOCs have an inflammatory component that contributes to their progression. At sites of inflammation, epithelial cells are exposed to increased levels of inflammatory mediators such as reactive oxygen species, cytokines, prostaglandins, and growth factors that contribute to increased cell division, and genetic and epigenetic changes. These exposure-induced changes promote excessive cell proliferation, increased survival, malignant transformation, and cancer development. Furthermore, the pro-inflammatory tumor microenvironment environment (TME) contributes to EOC metastasis and chemoresistance. In this review we will discuss the roles inflammation and inflammatory mediators play in the development, progression, metastasis, and chemoresistance of EOC.
Collapse
|
38
|
Ghoneum A, Afify H, Salih Z, Kelly M, Said N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget 2018; 9:22832-22849. [PMID: 29854318 PMCID: PMC5978268 DOI: 10.18632/oncotarget.25126] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/21/2018] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is the fifth most common cancer affecting the female population and at present, stands as the most lethal gynecologic malignancy. Poor prognosis and low five-year survival rate are attributed to nonspecific symptoms and below par diagnostic criteria at early phases along with a lack of effective treatment at advanced stages. It is thus of utmost importance to understand ovarian carcinoma through several lenses including its molecular pathogenesis, epidemiology, histological subtypes, hereditary factors, diagnostic approaches and methods of treatment. Above all, it is crucial to dissect the role that the unique peritoneal tumor microenvironment plays in ovarian cancer progression and metastasis. This review seeks to highlight several important aspects of ovarian cancer pathobiology as a means to provide the necessary background to approach ovarian malignancies in the future.
Collapse
Affiliation(s)
- Alia Ghoneum
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Hesham Afify
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Ziyan Salih
- Department of Cancer Pathology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Michael Kelly
- Department of Cancer Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Neveen Said
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Department of Cancer Pathology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Department of Cancer Urology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
39
|
Li Y, Zhang Y, Liu X, Wang M, Wang P, Yang J, Zhang S. Lutein inhibits proliferation, invasion and migration of hypoxic breast cancer cells via downregulation of HES1. Int J Oncol 2018; 52:2119-2129. [PMID: 29620169 DOI: 10.3892/ijo.2018.4332] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/14/2018] [Indexed: 11/06/2022] Open
Abstract
An intratumoral hypoxic microenvironment is frequently observed in solid tumors, including breast cancer. Lutein, a plant-derived compound and non-vitamin A carotenoid, has been demonstrated to possess multiple protective properties including anti-inflammation, anti-oxidative stress and antitumor effects. The main objective of the present research was to elucidate the involvement of lutein in the production of reactive oxygen species (ROS) under hypoxia, the activation of hairy and enhancer of split 1 (HES1), and the proliferation, invasion and migration of breast cancer cells. The human breast cancer cell lines MDA‑MB‑157 and MCF‑7 were exposed to hypoxic conditions and various concentrations of lutein. An MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay was performed to examine cell proliferation, and Annexin V-fluorescein isothiocyanate/propidium iodide staining was performed to analyze the apoptosis ratio. The levels of hypoxia inducible factor-1α (HIF‑1α), NOTCH signaling molecules, HES1 and epithelial-mesenchymal transition (EMT)-associated factors were examined by reverse transcription-quantitative polymerase chain reaction and western blot analysis. Wound healing and Transwell invasion assays were used to detect the invasion and migration of breast cancer cells. Intracellular ROS levels were examined using 2,7-dichlorodihydrofluorescein-diacetate and flow cytometry. The results revealed that cell proliferation was inhibited by lutein in a dose-dependent manner, and the apoptosis ratio gradually increased with lutein treatment under hypoxia as evident from flow cytometry-based analysis. Exposure to lutein inhibited hypoxia-mediated activation of HIF‑1α, NOTCH signaling and HES1 expression, and suppressed the hypoxia-induced expression of EMT-associated factors. Lutein markedly inhibited the invasion and migration of breast cancer cells under hypoxia. Hypoxia-induced production of ROS was also decreased by lutein. Furthermore, the ROS scavenger N‑acetylcysteine also suppressed hypoxia inducible factor 1α and HES1 expression in breast cancer cells during hypoxia, but hydrogen peroxide (H2O2) levels were increased. Taken together, the results of the present study suggested that lutein may be a novel candidate for the chemoprevention of breast cancer. Furthermore, HES1 may be crucial in mediating the involvement of lutein in the suppression of hypoxia-driven ROS-induced breast cancer progression.
Collapse
Affiliation(s)
- Yuan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuekun Zhang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaosa Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Mingchen Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Peng Wang
- Department of Basic Medicine, Nursing College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jianbo Yang
- Medical Laboratories, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Shanfeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
40
|
Zhang D, Kaushiva A, Xi Y, Wang T, Li N. Non-herbal tea consumption and ovarian cancer risk: a systematic review and meta-analysis of observational epidemiologic studies with indirect comparison and dose–response analysis. Carcinogenesis 2018; 39:808-818. [DOI: 10.1093/carcin/bgy048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/17/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Dongyu Zhang
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, CB, Chapel Hill, NC, USA
| | - Alpana Kaushiva
- Department of Epidemiology, University of Illinois at Chicago School of Public Health, Chicago, IL, USA
| | - Yuzhi Xi
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, CB, Chapel Hill, NC, USA
| | - Tengteng Wang
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, CB, Chapel Hill, NC, USA
| | - Nan Li
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
41
|
Lu J, Qian Y, Jin W, Tian R, Zhu Y, Wang J, Meng X, Wang R. Hypoxia-inducible factor-1α regulates epithelial-to-mesenchymal transition in paraquat-induced pulmonary fibrosis by activating lysyl oxidase. Exp Ther Med 2017; 15:2287-2294. [PMID: 29467842 PMCID: PMC5792814 DOI: 10.3892/etm.2017.5677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 10/25/2017] [Indexed: 12/20/2022] Open
Abstract
Pulmonary fibrosis (PF) is one of the most prevalent causes of death following paraquat (PQ) poisoning. As demonstrated in previous studies by the present authors, epithelial-to-mesenchymal transition (EMT) is associated with PQ-induced PF. In addition, hypoxia-inducible factor-1α (HIF-1α) and lysyl oxidase (LOX) promote EMT following PQ poisoning. However, the association between HIF-1α- and LOX-mediated regulation of EMT remains unclear. The present study investigated the association between HIF-1α and LOX with regard to PQ-induced EMT. A549 and RLE-6TN cells were treated with PQ, and HIF-1α and LOX expression was silenced with short interfering RNAs. Changes in the expression of HIF-1α, LOX, β-catenin and EMT-related makers were detected using real-time quantitative polymerase chain reaction, immunofluorescence, and western blotting. HIF-1α and LOX were associated with PQ-induced EMT, and their expression levels were significantly increased (P<0.05). LOX expression was significantly decreased following PQ poisoning when HIF-1α expression was inhibited (P<0.05). However, the level of HIF-1α did not change significantly when LOX was silenced. The expression level of β-catenin and the degree of EMT were significantly decreased following HIF-1α and LOX silencing in both cell lines (P<0.05). The association between HIF-1α and LOX in regulating EMT during PQ-induced PF may be unidirectional. HIF-1α may regulate PQ-induced EMT through the LOX/β-catenin pathway.
Collapse
Affiliation(s)
- Jian Lu
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yongbing Qian
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Wei Jin
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Rui Tian
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| | - Yong Zhu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, P.R. China
| | - Jinfeng Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, P.R. China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, P.R. China
| | - Ruilan Wang
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, P.R. China
| |
Collapse
|
42
|
Redox regulation in tumor cell epithelial-mesenchymal transition: molecular basis and therapeutic strategy. Signal Transduct Target Ther 2017; 2:17036. [PMID: 29263924 PMCID: PMC5661624 DOI: 10.1038/sigtrans.2017.36] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is recognized as a driving force of cancer cell metastasis and drug resistance, two leading causes of cancer recurrence and cancer-related death. It is, therefore, logical in cancer therapy to target the EMT switch to prevent such cancer metastasis and recurrence. Previous reports have indicated that growth factors (such as epidermal growth factor and fibroblast growth factor) and cytokines (such as the transforming growth factor beta (TGF-β) family) are major stimulators of EMT. However, the mechanisms underlying EMT initiation and progression remain unclear. Recently, emerging evidence has suggested that reactive oxygen species (ROS), important cellular secondary messengers involved in diverse biological events in cancer cells, play essential roles in the EMT process in cancer cells by regulating extracellular matrix (ECM) remodeling, cytoskeleton remodeling, cell–cell junctions, and cell mobility. Thus, targeting EMT by manipulating the intracellular redox status may hold promise for cancer therapy. Herein, we will address recent advances in redox biology involved in the EMT process in cancer cells, which will contribute to the development of novel therapeutic strategies by targeting redox-regulated EMT for cancer treatment.
Collapse
|
43
|
Bilyk O, Coatham M, Jewer M, Postovit LM. Epithelial-to-Mesenchymal Transition in the Female Reproductive Tract: From Normal Functioning to Disease Pathology. Front Oncol 2017; 7:145. [PMID: 28725636 PMCID: PMC5497565 DOI: 10.3389/fonc.2017.00145] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/21/2017] [Indexed: 12/15/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a physiological process that is vital throughout the human lifespan. In addition to contributing to the development of various tissues within the growing embryo, EMT is also responsible for wound healing and tissue regeneration later in adulthood. In this review, we highlight the importance of EMT in the development and normal functioning of the female reproductive organs (the ovaries and the uterus) and describe how dysregulation of EMT can lead to pathological conditions, such as endometriosis, adenomyosis, and carcinogenesis. We also summarize the current literature relating to EMT in the context of ovarian and endometrial carcinomas, with a particular focus on how molecular mechanisms and the tumor microenvironment can govern cancer cell plasticity, therapy resistance, and metastasis.
Collapse
Affiliation(s)
- Olena Bilyk
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Mackenzie Coatham
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Michael Jewer
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | | |
Collapse
|
44
|
E-cadherin expression as a prognostic factor in patients with ovarian cancer: a meta-analysis. Oncotarget 2017; 8:81052-81061. [PMID: 29113366 PMCID: PMC5655261 DOI: 10.18632/oncotarget.18898] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/12/2017] [Indexed: 01/11/2023] Open
Abstract
The prognostic role of epithelial cadherin (E-cadherin) downregulation in ovarian cancer has been assessed for years while the results remain inconclusive. The aim of our study was to assess this issue. Eligible studies were identified through searches of PubMed, EMBASE and Cochrane Database. In total, 1562 patients from 17 studies were included to assess the association between E-cadherin expression and overall survival/progression-free survival and clinicopathological characteristics of ovarian cancer patients. Hazard ratios (HRs) or odds ratios (ORs) with 95% confidence interval (95% CI) were calculated to estimate the effect. The quality of 17 studies was evaluated using the Newcastle Ottawa Quality Assessment Scale. We also performed subgroup analysis, publication bias and sensitivity analysis in this meta-analysis. The results showed that negative E-cadherin expression significantly predicted poor overall survival of ovarian cancer patients (HR = 1.90, 95% CI = 1.50–2.40). However, negative E-cadherin was not associated with poor progression-free survival (HR = 1.19, 95% CI = 0.86–1.64). Moreover, Negative E-cadherin expression was distinctly associated with FIGO stage (OR = 0.42, 95% CI = 0.31–0.57), tumor grade (OR = 0.48, 95% CI = 0.34–0.67), metastasis (OR = 0.13, 95% CI = 0.07–0.26) and recurrence (OR = 0.48, 95% CI = 0.29–0.79). This meta-analysis revealed that negative E-cadherin expression might be a predicative factor of poor prognosis in ovarian cancer patients.
Collapse
|
45
|
De Donato M, Petrillo M, Martinelli E, Filippetti F, Zannoni GF, Scambia G, Gallo D. Uncovering the role of nuclear Lysyl oxidase (LOX) in advanced high grade serous ovarian cancer. Gynecol Oncol 2017; 146:170-178. [PMID: 28495238 DOI: 10.1016/j.ygyno.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/28/2017] [Accepted: 05/01/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Lysyl oxidase (LOX) is an enzyme that catalyzes the cross-linking of collagen and elastin in the extracellular matrix, thus controlling the tensile strength of tissues. Along with this primary function, there are evidences supporting a role for LOX in many critical biological functions, including gene expression regulation, cell growth, adhesion and migration. Accordingly, recent studies have supported a pivotal role for LOX in cancer progression and metastasis. The current study aimed at investigating the prognostic significance and the functional role of intracellular LOX in ovarian cancer. METHODS To this end, we analyzed LOX expression by immunohistochemistry in archived tumor material from advanced high grade serous ovarian cancer (HGSOC) patients (n=70) and correlated data with clinicopathological parameters and with response to chemotherapy. In vitro experiments were also used to investigate the functional consequences of LOX expression on behavioral aspects of HGSOC cells. RESULTS Our results showed that nuclear LOX expression is associated with unfavorable outcome in advanced HGSOC, being an independent prognostic factor for disease recurrence. Besides, high nuclear levels were seen to be associated with resistance to first-line chemotherapy. Through gene expression modulation experiments in HGSOC cell lines, we demonstrate that LOX positively regulates cell proliferation, migration and anchorage-independent growth. CONCLUSIONS Collectively, our data suggest that LOX functions as a tumor promoter in HGSOC and positively regulates several aspects of the metastatic cascade.
Collapse
Affiliation(s)
- Marta De Donato
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Marco Petrillo
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Enrica Martinelli
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Flavia Filippetti
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Gian Franco Zannoni
- Department of Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Scambia
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Daniela Gallo
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
46
|
Liu D, Zhang R, Wu J, Pu Y, Yin X, Cheng Y, Wu J, Feng C, Luo Y, Zhang J. Interleukin-17A promotes esophageal adenocarcinoma cell invasiveness through ROS-dependent, NF-κB-mediated MMP-2/9 activation. Oncol Rep 2017; 37:1779-1785. [PMID: 28184939 DOI: 10.3892/or.2017.5426] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/28/2016] [Indexed: 11/06/2022] Open
Abstract
Interleukin-17A (IL-17A), a pro-inflammatory cytokine secreted primarily by Th17 cells, has been proved to be involved in the microenvironment of certain inflammation-related tumors. However, the role of IL-17A in cancer development has always been controversial. In this study, we investigated the effect of IL-17A on the regulation of esophageal adenocarcinoma (EAC) cell invasiveness and related molecular mechanism. Surface IL-17 receptor (IL-17R) expression on human EAC cell line OE19 was examined using flow cytometry. The effect of IL-17A on cell proliferation was measured by MTT assay. Cell migration and invasive ability in vitro were assessed by wound-healing and Matrigel-coated Transwell invasion assay. Intracellular reactive oxygen species (ROS) levels were determined by flow cytometry and fluorescence microscope. The protein expression levels of MMP-2, MMP-9, NF-κB and p-IκB-α were detected by western blotting. Our results showed that IL-17A promoted migration and invasion of OE19 cells in a dose-dependent manner, however it had less effect on OE19 cell proliferation. Furthermore, IL-17A treatment significantly upregulated the expression of MMP-2 and MMP-9, stimulated intracellular ROS production, increased IκB-α phosphorylation and NF-κB nuclear translocation. Nevertheless, IL-17A-induced expression of MMP-2/9 and OE19 cell invasiveness were both inhibited by pretreatment with N-acetyl-L-cysteine (NAC, a ROS scavenger) or pyrrolidine dithiocarbamate (PDTC, a NF-κB inhibitor). In conclusion, these findings demonstrate that IL-17A can promote the migration and invasiveness of EAC cells through ROS/NF-κB/MMP-2/9 signaling pathway activation, indicating that IL-17A may be a potential therapeutic target for EAC.
Collapse
Affiliation(s)
- Dong Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Rong Zhang
- Department of Gastroenterology, Shaanxi Provincal People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jie Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yansong Pu
- Department of General Surgery, Shaanxi Provincal People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiaoran Yin
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yan Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jing Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Cheng Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yumei Luo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jun Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
47
|
Broughton LJ, Giuntini F, Savoie H, Bryden F, Boyle RW, Maraveyas A, Madden LA. Duramycin-porphyrin conjugates for targeting of tumour cells using photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 163:374-84. [DOI: 10.1016/j.jphotobiol.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/27/2022]
|
48
|
Nuclear expression of lysyl oxidase enzyme is an independent prognostic factor in rectal cancer patients. Oncotarget 2016; 8:60015-60024. [PMID: 28947950 PMCID: PMC5601118 DOI: 10.18632/oncotarget.9623] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
Emerging evidence has implicated a pivotal role for lysyl oxidase (LOX) in cancer progression and metastasis. Whilst the majority of work has focused on the extracellular matrix cross-linking role of LOX, the exact function of intracellular LOX localisation remains unclear. In this study, we analysed the LOX expression patterns in the nuclei of rectal cancer patient samples and determined the clinical significance of this expression. Nuclear LOX expression was significantly increased in patient lymph node metastases compared to their primary tumours. High nuclear LOX expression in tumours was correlated with a high rate of distant metastasis and increased recurrence. Multivariable analysis showed that high nuclear LOX expression was also correlated with poor overall survival and disease free survival. Furthermore, we are the first to identify LOX enzyme isoforms (50 kDa and 32 kDa) within the nucleus of colon cancer cell lines by confocal microscopy and Western blot. Our results show a powerful link between nuclear LOX expression in tumours and patient survival, and offer a promising prognostic biomarker for rectal cancer patients.
Collapse
|
49
|
Wang L, Li H, Li M, Wang S, Jiang X, Li Y, Ping G, Cao Q, Liu X, Fang W, Chen G, Yang J, Wu C. SL4, a chalcone-based compound, induces apoptosis in human cancer cells by activation of the ROS/MAPK signalling pathway. Cell Prolif 2015; 48:718-28. [PMID: 26500049 PMCID: PMC6496741 DOI: 10.1111/cpr.12226] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/08/2015] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES SL4, a chalcone-based compound, exhibits clearly inhibitory effects on HIF-1 and has been shown to effectively suppress tumour invasion and angiogenesis in vitro and in vivo. Here, studies were conducted to determine SL4's anti-apoptotic effects and its underlying mechanisms, in human cancer cells. MATERIALS AND METHODS Cytotoxicity, apoptotic induction and its involved mechanisms of SL4 were investigated using normal cells, cancer cells and mouse xenograft models. The role of reactive oxygen species (ROS) and mitogen-activated protein kinase (MAPK) signalling in SL4-induced apoptosis was explored by manipulating specific scavenger or signalling inhibitors, in cultured cells. RESULTS SL4 significantly inhibited cell population growth of human cancer cell lines but exhibited lower cytotoxicity against normal cells. In addition, SL4 effectively induced apoptosis of Hep3B and MDA-MB-435 cells by activating procaspase-8, -9 and -3, and down-regulating expression levels of XIAP, but did not affect HIF-1 apoptosis-related targets, Survivin and Bcl-XL. Further study showed that SL4 also reduced mitochondrial membrane potential and promoted generation of ROS. ROS generation and apoptotic induction by SL4 were blocked by NAC, a scavenger of ROS, suggesting SL4-induced apoptosis via ROS accumulation. We also found that MAPKs, JNK and p38, but not ERK1/2, to be critical mediators in SL4-induced apoptosis. SP600125 and SB203580, specific inhibitors of JNK kinase and p38 kinase, significantly retarded apoptosis induced by SL4. Moreover, anti-oxidant NAC blocked activation of JNK and p38 induced by SL4, indicating that ROS may act as upstream signalling of JNK and p38 activation. It is noteworthy that animal studies revealed dramatic reduction (49%) in tumour volume after 11 days SL4 treatment. CONCLUSIONS These data demonstrate that SL4 induced apoptosis in human cancer cells through activation of the ROS/MAPK signalling pathway, suggesting that it may be a novel lead compound, as a cancer drug candidate, with polypharmacological characteristics.
Collapse
Affiliation(s)
- L.‐H. Wang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - H.‐H. Li
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - M. Li
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - S. Wang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - X.‐R. Jiang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - Y. Li
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - G.‐F. Ping
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - Q. Cao
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - X. Liu
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - W.‐H. Fang
- Key Laboratory of Structure‐Based Drugs Design & Discovery of Ministry of EducationShenyang Pharmaceutical UniversityShenyang110016China
| | - G.‐L. Chen
- Key Laboratory of Structure‐Based Drugs Design & Discovery of Ministry of EducationShenyang Pharmaceutical UniversityShenyang110016China
| | - J.‐Y. Yang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - C.‐F. Wu
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| |
Collapse
|
50
|
Cho A, Howell VM, Colvin EK. The Extracellular Matrix in Epithelial Ovarian Cancer - A Piece of a Puzzle. Front Oncol 2015; 5:245. [PMID: 26579497 PMCID: PMC4629462 DOI: 10.3389/fonc.2015.00245] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/15/2015] [Indexed: 02/04/2023] Open
Abstract
Epithelial ovarian cancer is the fifth leading cause of cancer-related deaths in women and the most lethal gynecological malignancy. Extracellular matrix (ECM) is an integral component of both the normal and tumor microenvironment. ECM composition varies between tissues and is crucial for maintaining normal function and homeostasis. Dysregulation and aberrant deposition or loss of ECM components is implicated in ovarian cancer progression. The mechanisms by which tumor cells induce ECM remodeling to promote a malignant phenotype are yet to be elucidated. A thorough understanding of the role of the ECM in ovarian cancer is needed for the development of effective biomarkers and new therapies.
Collapse
Affiliation(s)
- Angela Cho
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, NSW, Australia
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
| | - Viive M. Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| | - Emily K. Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|