1
|
Wang X, Li Z, Shen J, Liu L. Targeting protein tyrosine phosphatase non-receptor type 6 (PTPN6) as a therapeutic strategy in acute myeloid leukemia. Cell Biol Toxicol 2024; 41:11. [PMID: 39707066 PMCID: PMC11662038 DOI: 10.1007/s10565-024-09965-3] [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: 07/06/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by the clonal expansion of myeloid progenitor cells. Despite advancements in treatment, the prognosis for AML patients remains poor, highlighting the need for novel therapeutic targets. Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6), also known as SHP-1, is a critical regulator of hematopoietic cell signaling and has been implicated in various leukemias. This study investigates the therapeutic potential of targeting PTPN6 in AML. We employed both in vitro and in vivo models to evaluate the effects of PTPN6 inhibition on AML cell proliferation, apoptosis, and differentiation. Our results demonstrate that PTPN6 inhibition leads to a significant reduction in AML cell viability, induces apoptosis, and promotes differentiation of leukemic cells into mature myeloid cells. Mechanistic studies revealed that PTPN6 inhibition disrupts key signaling pathways involved in AML pathogenesis, including the JAK/STAT and PI3K/AKT pathways. Furthermore, the combination of PTPN6 inhibitors with standard chemotherapeutic agents exhibited a synergistic effect, enhancing the overall therapeutic efficacy. These findings suggest that PTPN6 is a promising therapeutic target in AML and warrants further investigation into the development of PTPN6 inhibitors for clinical application in AML treatment.
Collapse
Affiliation(s)
- Xiaoou Wang
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China
| | - Zhenggang Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Jing Shen
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, 110001, People's Republic of China.
| | - Lin Liu
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China.
| |
Collapse
|
2
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
3
|
Lv Y, Guo S, Jin L, Wang K, Li Y, Li H, Lu Y, Liu H. MiR-5195-3p predicts clinical prognosis and represses colorectal cancer progression by targeting TLR4/MyD88 signaling. Cell Div 2024; 19:29. [PMID: 39390599 PMCID: PMC11468180 DOI: 10.1186/s13008-024-00133-x] [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: 05/28/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Recent studies have highlighted the role of miR-5195-3p in suppressing cell growth in various cancers. However, the specific functional impact of miR-5195-3p in colorectal cancer (CRC) remain to be fully clarified. The importance of miR-5195-3p in CRC was evaluated, aiming to uncover its underlying molecular mechanism and identify it as a potential therapeutic target for CRC. RESULTS Our research has shown that miR-5195-3p is markedly under-expressed in CRC tissues and cell cultures, with its reduced presence associated with a higher TNM stage, lymphatic invasion, and unfavorable survival outcome. Ectopic miR-5195-3p expression curtailed proliferation, migration, and invasion of SW1116 and HT29 cells. Additionally, we discovered that miR-5195-3p directly targets and negatively influences Toll-like receptor 4 (TLR4) in CRC cells. Moreover, an inverse relationship was noted between miR-5195-3p and TLR4 expression in CRC tissue samples. Notably, restoring TLR4 expression counteracted miR-5195-3p's suppressive impact on cell growth, motility, invasiveness, epithelial-mesenchymal transition (EMT), and the TLR4/MyD88 signaling pathway in SW1116 and HT29 cells. CONCLUSIONS MiR-5195-3p suppresses the CRC cellular functions through the downregulation of TLR4/MyD88 signaling, indicating that targeting miR-5195-3p might offer a viable therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Yandong Lv
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Shuwei Guo
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Lingtong Jin
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Kai Wang
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Yongsheng Li
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Haonan Li
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Yikang Lu
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China
| | - Hongzhou Liu
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, No.110 Yanan South Road, Luzhou District, 046000, Changzhi City, Shanxi Province, P. R. China.
| |
Collapse
|
4
|
Guo Y, Bao J, Lin D, Hong K, Cen K, Sun J, Wang Z, Wu Z. Novel immune checkpoint-related gene model to predict prognosis and treatment responsiveness in low-grade gliomas. Heliyon 2023; 9:e20178. [PMID: 37809899 PMCID: PMC10559968 DOI: 10.1016/j.heliyon.2023.e20178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Recently, studies have shown that immune checkpoint-related genes (ICGs) are instrumental in maintaining immune homeostasis and can be regarded as potential therapeutic targets. However, the prognostic applications of ICGs require further elucidation in low-grade glioma (LGG) cases. In the present study, a unique prognostic gene signature in LGG has been identified and validated as well based on ICGs as a means of facilitating clinical decision-making. The RNA-seq data as well as corresponding clinical data of LGG samples have been retrieved utilizing the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. ICG-defined non-negative matrix factorization (NMF) clustering was performed to categorize patients with LGG into two molecular subtypes with different prognoses, clinical traits, and immune microenvironments. In the TCGA database, a signature integrating 8 genes has been developed utilizing the LASSO Cox method and validated in the GEO database. The signature developed is superior to other well-recognized signatures in terms of predicting the survival probability of patients with LGG. This 8-gene signature was then subsequently applied to categorize patients into high- and low-risk groups, and differences between them in terms of gene alteration frequency were observed. There were remarkable variations in IDH1 (91% and 64%) across low-as well as high-risk groups. Additionally, various analyses like function enrichment, tumor immune microenvironment, and chemotherapy drug sensitivity revealed significant variations across high- and low-risk populations. Overall, this 8-gene signature may function as a useful tool for prognosis and immunotherapy outcome predictions among LGG patients.
Collapse
Affiliation(s)
- Yangyang Guo
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Jingxia Bao
- Department of Breast Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Danfeng Lin
- Department of Breast Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Kai Hong
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Kenan Cen
- The Affiliated Hospital of Medical School of Ningbo University, Jiangbei District, Ningbo, 315020, Zhejiang, People's Republic of China
| | - Jie Sun
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Zhepei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Zhixuan Wu
- Department of Breast Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| |
Collapse
|
5
|
Lučić I, Kurtović M, Mlinarić M, Piteša N, Čipak Gašparović A, Sabol M, Milković L. Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches. Int J Mol Sci 2023; 24:10683. [PMID: 37445860 DOI: 10.3390/ijms241310683] [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: 05/06/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer (BC) and ovarian cancer (OC) are among the most common and deadly cancers affecting women worldwide. Both are complex diseases with marked heterogeneity. Despite the induction of screening programs that increase the frequency of earlier diagnosis of BC, at a stage when the cancer is more likely to respond to therapy, which does not exist for OC, more than 50% of both cancers are diagnosed at an advanced stage. Initial therapy can put the cancer into remission. However, recurrences occur frequently in both BC and OC, which are highly cancer-subtype dependent. Therapy resistance is mainly attributed to a rare subpopulation of cells, named cancer stem cells (CSC) or tumor-initiating cells, as they are capable of self-renewal, tumor initiation, and regrowth of tumor bulk. In this review, we will discuss the distinctive markers and signaling pathways that characterize CSC, their interactions with the tumor microenvironment, and the strategies they employ to evade immune surveillance. Our focus will be on identifying the common features of breast cancer stem cells (BCSC) and ovarian cancer stem cells (OCSC) and suggesting potential therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Lučić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Matea Kurtović
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Monika Mlinarić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Nikolina Piteša
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ana Čipak Gašparović
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Lidija Milković
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
6
|
Jin J, Yang Z, Liu H, Guo M, Chen B, Zhu H, Wang Y, Lin J, Wang S, Chen S. Effects of acupuncture on the miR-146a-mediated IRAK1/TRAF6/NF-κB signaling pathway in rats with sarcopenia induced by D-galactose. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:47. [PMID: 36819511 PMCID: PMC9929824 DOI: 10.21037/atm-22-6082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Background Sarcopenia during aging is closely linked to sterile, low-grade, chronic inflammation. However, considering the increasingly aging global population, the effectiveness of existing treatments for sarcopenia is not exact, and acupuncture, as an effective anti-inflammatory therapy, has the potential to treat it. Methods Fifty Sprague-Dawley rats were randomly allocated into five groups, including Control group, D-galactose (D-gal) group, D-gal + acupuncture (DA) group, D-gal + non-acupoint (DN) group and D-gal amino acid mixture (DAA) group. An aging rat was model constructed using D-gal for 12 weeks. Rats in the control group received 0.9% physiological saline daily. Treatment groups were acupunctured or given amino acid mixture interventions daily, and lasted for last 4 consecutive weeks. The effects of acupuncture were evaluated by the hematoxylin and eosin staining (H&E), transmission electron microscopic (TEM) examination and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. The anti-inflammatory mechanism of acupuncture was studied by using the expressions of microRNA-146a (miR-146a) mediated nuclear factor-kappa B (NF-κB) signaling pathway-related proteins were detected by immunofluorescence, western blotting, quantitative real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). Results Rats injected by D-galactose (D-gal) revealed apparent skeletal muscle atrophy with significantly reduced cross-sectional area and fiber diameter. In contrast, acupuncture treatment alleviated these hallmarks of skeletal muscle atrophy and mitigated the mitochondrial aberrations and skeletal muscle apoptosis in D-gal rats. In addition, acupuncture also downgraded the overexpression of inflammatory factors in skeletal muscle, influenced miR-146a and the target genes level, and inhibited NF-κB nuclear translation in D-gal rats. Conclusions Acupuncture may ameliorate skeletal muscle atrophy, and its effects may be associated with the control of mitochondrial function regulation and the suppression of inflammation.
Collapse
Affiliation(s)
- Jing Jin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhengyu Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haichao Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingling Guo
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Borui Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haoming Zhu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianping Lin
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shizhong Wang
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shaoqing Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
7
|
TLR4 Agonist and Hypoxia Synergistically Promote the Formation of TLR4/NF-κB/HIF-1α Loop in Human Epithelial Ovarian Cancer. Anal Cell Pathol (Amst) 2022; 2022:4201262. [PMID: 35464826 PMCID: PMC9023210 DOI: 10.1155/2022/4201262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 12/04/2022] Open
Abstract
Inflammation and hypoxia are involved in numerous cancer progressions. Reportedly, the toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) pathway and hypoxia-inducible factor-1α (HIF-1α) are activated and closely related to the chemoresistance and poor prognosis of epithelial ovarian cancer (EOC). However, the potential correlation between TLR4/NF-κB and HIF-1α remains largely unknown in EOC. In our study, the possible positive correlation among TLR4, NF-κB, and HIF-1α proteins was investigated in the EOC tissues. Our in vitro results demonstrated that LPS can induce and activate HIF-1α through the TLR4/NF-κB signaling in A2780 and SKOV3 cells. Moreover, hypoxia-induced TLR4 expression and the downstream transcriptional activity of NF-κB were HIF-1α-dependent. The cross talk between the TLR4/NF-κB signaling pathway and HIF-1α was also confirmed in the nude mice xenograft model. Therefore, we first proposed the formation of a TLR4/NF-κB/HIF-1α loop in EOC. The positive feedback loop enhanced the susceptibility and responsiveness to inflammation and hypoxia, which synergistically promote the initiation and progression of EOC. The novel mechanism may act as a future therapeutic candidate for the treatment of EOC.
Collapse
|
8
|
Wilczyński JR, Wilczyński M, Paradowska E. Cancer Stem Cells in Ovarian Cancer-A Source of Tumor Success and a Challenging Target for Novel Therapies. Int J Mol Sci 2022; 23:ijms23052496. [PMID: 35269636 PMCID: PMC8910575 DOI: 10.3390/ijms23052496] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells. The aim of this review is to show the most contemporary knowledge concerning the biology of ovarian cancer stem cells (OCSCs) and their impact on chemo-resistance and prognosis in ovarian cancer patients, as well as to present the treatment options targeted exclusively on the OCSCs. The review presents data concerning the role of cancer stem cells in general and then concentrates on OCSCs. The surface and intracellular OCSCs markers and their meaning both for cancer biology and clinical prognosis, signaling pathways specifically activated in OCSCs, the genetic and epigenetic regulation of OCSCs function including the recent studies on the non-coding RNA regulation, cooperation between OCSCs and the tumor microenvironment (ovarian cancer niche) including very specific environment such as ascites fluid, the role of shear stress, autophagy and metabolic changes for the function of OCSCs, and finally mechanisms of OCSCs escape from immune surveillance, are described and discussed extensively. The possibilities of anti-OCSCs therapy both in experimental settings and in clinical trials are presented, including the recent II phase clinical trials and immunotherapy. OCSCs are a unique population of cancer cells showing a great plasticity, self-renewal potential and resistance against anti-cancer treatment. They are responsible for the progression and recurrence of the tumor. Several completed and ongoing clinical trials have tested different anti-OCSCs drugs which, however, have shown unsatisfactory efficacy in most cases. We propose a novel approach to ovarian cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
- Correspondence:
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| |
Collapse
|
9
|
Zhang Z, Huang Q, Yu L, Zhu D, Li Y, Xue Z, Hua Z, Luo X, Song Z, Lu C, Zhao T, Liu Y. The Role of miRNA in Tumor Immune Escape and miRNA-Based Therapeutic Strategies. Front Immunol 2022; 12:807895. [PMID: 35116035 PMCID: PMC8803638 DOI: 10.3389/fimmu.2021.807895] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
Tumor immune escape is a critical step in the malignant progression of tumors and one of the major barriers to immunotherapy, making immunotherapy the most promising therapeutic approach against tumors today. Tumor cells evade immune surveillance by altering the structure of their own, or by causing abnormal gene and protein expression, allowing for unrestricted development and invasion. These genetic or epigenetic changes have been linked to microRNAs (miRNAs), which are important determinants of post-transcriptional regulation. Tumor cells perform tumor immune escape by abnormally expressing related miRNAs, which reduce the killing effect of immune cells, disrupt the immune response, and disrupt apoptotic pathways. Consequently, there is a strong trend toward thoroughly investigating the role of miRNAs in tumor immune escape and utilizing them in tumor treatment. However, because of the properties of miRNAs, there is an urgent need for a safe, targeted and easily crossed biofilm vehicle to protect and deliver them in vivo, and exosomes, with their excellent biological properties, have successfully beaten traditional vehicles to provide strong support for miRNA therapy. This review summarizes the multiple roles of miRNAs in tumor immune escape and discusses their potential applications as an anti-tumor therapy. Also, this work proposes exosomes as a new opportunity for miRNA therapy, to provide novel ideas for the development of more effective tumor-fighting therapeutic approaches based on miRNAs.
Collapse
Affiliation(s)
- Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qingcai Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiqian Song
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yuanyan Liu, ; Cheng Lu, ; Ting Zhao,
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yuanyan Liu, ; Cheng Lu, ; Ting Zhao,
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yuanyan Liu, ; Cheng Lu, ; Ting Zhao,
| |
Collapse
|
10
|
Capilliposide from Lysimachia capillipes promotes terminal differentiations and reverses paclitaxel resistance in A2780T cells of human ovarian cancer by regulating Fos/Jun pathway. CHINESE HERBAL MEDICINES 2021; 14:111-116. [PMID: 36120123 PMCID: PMC9476669 DOI: 10.1016/j.chmed.2021.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/01/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022] Open
Abstract
Objective To investigate the potential effect of Lysimachia capillipes capilliposide (LCC) on the chemo sensitivity and the stemness of human ovarian cancer cells. Methods Cell Counting Kit-8 (CCK8) was used to measure the IC50 values. The apoptosis of cells was measured through flow cytometry. Evaluation of the stemness and differentiation markers was performed by the immunoblotting and the immunostaining assays. RNA-seq was performed through the Illumina HiSeq PE150 platform and differentially expressed genes (DEGs) were screened out through the bioinformation analysis. Overexpression or knockdown of Fos gene was achieved by shRNA transfection. Results Pre-exposure of A2780T cells with 10 μg/mL LCC sensitized them to paclitaxel, of which the IC50 value reduced from 8.644 μmol/L (95%CI: 7.315–10.082 μmol/L) to 2.5 μmol/L (95%CI: 2.233–2.7882 μmol/L). Exposure with LCC enhanced the paclitaxel-induced apoptosis and inhibited the colony formation of A2780T cells. LCC exposure reduced the expression of cancer stemness markers, ALDH1, Myd88 and CD44, while promoting that of terminal differentiation markers, NFATc1, Cathepsin K and MMP9. RNA-seq analysis revealed that the expressions of FOS and JUN were upregulated in LCC-treated A2780T cells. A2780T cells overexpressing Fos gene displayed increased paclitaxel-sensitivity and reduced cell stemness, and shared common phenotypes with LCC-treated A2780T cells. Conclusion These findings suggested that LCC promoted terminal differentiations of ovarian cancer cells and sensitized them to paclitaxel through activating the Fos/Jun pathway. LCC might become a novel therapy that targets at cancer stem cells and enhances the chemotherapeutic effect of ovarian cancer treatments.
Collapse
|
11
|
Bates M, Spillane CD, Gallagher MF, McCann A, Martin C, Blackshields G, Keegan H, Gubbins L, Brooks R, Brooks D, Selemidis S, O’Toole S, O’Leary JJ. The role of the MAD2-TLR4-MyD88 axis in paclitaxel resistance in ovarian cancer. PLoS One 2020; 15:e0243715. [PMID: 33370338 PMCID: PMC7769460 DOI: 10.1371/journal.pone.0243715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/25/2020] [Indexed: 01/29/2023] Open
Abstract
Despite the use of front-line anticancer drugs such as paclitaxel for ovarian cancer treatment, mortality rates have remained almost unchanged for the past three decades and the majority of patients will develop recurrent chemoresistant disease which remains largely untreatable. Overcoming chemoresistance or preventing its onset in the first instance remains one of the major challenges for ovarian cancer research. In this study, we demonstrate a key link between senescence and inflammation and how this complex network involving the biomarkers MAD2, TLR4 and MyD88 drives paclitaxel resistance in ovarian cancer. This was investigated using siRNA knockdown of MAD2, TLR4 and MyD88 in two ovarian cancer cell lines, A2780 and SKOV-3 cells and overexpression of MyD88 in A2780 cells. Interestingly, siRNA knockdown of MAD2 led to a significant increase in TLR4 gene expression, this was coupled with the development of a highly paclitaxel-resistant cell phenotype. Additionally, siRNA knockdown of MAD2 or TLR4 in the serous ovarian cell model OVCAR-3 resulted in a significant increase in TLR4 or MAD2 expression respectively. Microarray analysis of SKOV-3 cells following knockdown of TLR4 or MAD2 highlighted a number of significantly altered biological processes including EMT, complement, coagulation, proliferation and survival, ECM remodelling, olfactory receptor signalling, ErbB signalling, DNA packaging, Insulin-like growth factor signalling, ion transport and alteration of components of the cytoskeleton. Cross comparison of the microarray data sets identified 7 overlapping genes including MMP13, ACTBL2, AMTN, PLXDC2, LYZL1, CCBE1 and CKS2. These results demonstrate an important link between these biomarkers, which to our knowledge has never before been shown in ovarian cancer. In the future, we hope that triaging patients into alterative treatment groups based on the expression of these three biomarkers or therapeutic targeting of the mechanisms they are involved in will lead to improvements in patient outcome and prevent the development of chemoresistance.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| | - Cathy D. Spillane
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
| | - Michael F. Gallagher
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
| | - Amanda McCann
- College of Health Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| | - Gordon Blackshields
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| | - Helen Keegan
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| | - Luke Gubbins
- College of Health Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Robert Brooks
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Doug Brooks
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology, Bundoora, Australia
| | - Sharon O’Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J. O’Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland
- Trinity St James’s Cancer Institute, Dublin, Ireland
- Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Yuan J, Li T, Yi K, Hou M. The suppressive role of miR-362-3p in epithelial ovarian cancer. Heliyon 2020; 6:e04258. [PMID: 32671239 PMCID: PMC7347651 DOI: 10.1016/j.heliyon.2020.e04258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/01/2020] [Accepted: 06/16/2020] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer is a common cancer worldwide. Epithelial ovarian cancer (EOC) is the most common subtype of ovarian cancer. This study was designed to explore the function of miR-362-3p in EOC. QRT-PCR analysis was used to test miR-362-3p levels in EOC tissues and cell lines. Cell viability was tested via MTT assay. Transwell systems were applied to assay cell migration. The target gene of miR-362-3p was evaluated using dual luciferase reporter assays. The MyD88 protein in EOC cells was tested via western blot. Our data showed that miR-362-3p was expressed at low levels in EOC tissues and cells. miR-362-3p inhibited cell proliferation and migration, bound the 3′-untranslated region (UTR) of MyD88, and inhibited MyD88 expression. MyD88 was inversely correlated with miR-362-3p in EOC, and MyD88 overexpression partly reduced the anti-proliferative effect of miR-362-3p in EOC cells. In conclusion, our data showed that miR-362-3p has an anti-proliferative effect on EOC.
Collapse
Affiliation(s)
- Jialing Yuan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| | - Tao Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| | - Ke Yi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| | - Minmin Hou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| |
Collapse
|
13
|
Syed SN, Brüne B. MicroRNAs as Emerging Regulators of Signaling in the Tumor Microenvironment. Cancers (Basel) 2020; 12:E911. [PMID: 32276464 PMCID: PMC7225969 DOI: 10.3390/cancers12040911] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
A myriad of signaling molecules in a heuristic network of the tumor microenvironment (TME) pose a challenge and an opportunity for novel therapeutic target identification in human cancers. MicroRNAs (miRs), due to their ability to affect signaling pathways at various levels, take a prominent space in the quest of novel cancer therapeutics. The role of miRs in cancer initiation, progression, as well as in chemoresistance, is being increasingly investigated. The canonical function of miRs is to target mRNAs for post-transcriptional gene silencing, which has a great implication in first-order regulation of signaling pathways. However, several reports suggest that miRs also perform non-canonical functions, partly due to their characteristic non-coding small RNA nature. Examples emerge when they act as ligands for toll-like receptors or perform second-order functions, e.g., to regulate protein translation and interactions. This review is a compendium of recent advancements in understanding the role of miRs in cancer signaling and focuses on the role of miRs as novel regulators of the signaling pathway in the TME.
Collapse
Affiliation(s)
- Shahzad Nawaz Syed
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany
| |
Collapse
|
14
|
Lupi LA, Delella FK, Cucielo MS, Romagnoli GG, Kaneno R, Nunes IDS, Domeniconi RF, Martinez M, Martinez FE, Fávaro WJ, Chuffa LGDA. P-MAPA and Interleukin-12 Reduce Cell Migration/Invasion and Attenuate the Toll-Like Receptor-Mediated Inflammatory Response in Ovarian Cancer SKOV-3 Cells: A Preliminary Study. Molecules 2019; 25:E5. [PMID: 31861351 PMCID: PMC6982916 DOI: 10.3390/molecules25010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/28/2019] [Accepted: 12/09/2019] [Indexed: 12/29/2022] Open
Abstract
Immunotherapies have emerged as promising complementary treatments for ovarian cancer (OC), but its effective and direct role on OC cells is unclear. This study examined the combinatory effects of the protein aggregate magnesium-ammonium phospholinoleate-palmitoleate anhydride, known as P-MAPA, and the human recombinant interleukin-12 (hrIL-12) on cell migration/invasion, apoptosis, toll-like receptor (TLR)-mediated inflammation, and cytokine/chemokine profile in human OC cell line SKOV-3. P-MAPA and IL-12 showed cancer cell toxicity under low doses after 48 h. Although apoptosis/necrosis and the cell cycle were unchanged by the treatments, P-MAPA enhanced the sensitivity to paclitaxel (PTX) and P-MAPA associated with IL-12 significantly reduced the migratory potential and invasion capacity of SKOV-3 cells. P-MAPA therapy reduced TLR2 immunostaining and the myeloid differentiation factor 88 (MyD88), but not the TLR4 levels. Moreover, the combination of P-MAPA with IL-12 attenuated the levels of MyD88, interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-kB p65). The IL-12 levels were increased and P-MAPA stimulated the secretion of cytokines IL-3, IL-9, IL-10, and chemokines MDC/CCL22 and, regulated on activation, normal T cells expressed and secreted (RANTES)/CCL5. Conversely, combination therapy reduced the levels of IL-3, IL-9, IL-10, MDC/CCL22, and RANTES/CCL5. Collectively, P-MAPA and IL-12 reduce cell dynamics and effectively target the TLR-related downstream molecules, eliciting a protective effect against chemoresistance. P-MAPA also stimulates the secretion of anti-inflammatory molecules, possibly having an immune response in the OC microenvironment.
Collapse
Affiliation(s)
- Luiz Antonio Lupi
- Department of Anatomy, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (L.A.L.); (M.S.C.); (R.F.D.); (F.E.M.)
| | - Flávia Karina Delella
- Department of Morphology, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil;
| | - Maira Smaniotto Cucielo
- Department of Anatomy, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (L.A.L.); (M.S.C.); (R.F.D.); (F.E.M.)
| | - Graziela Gorete Romagnoli
- Department of Microbiology and Immunology, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (G.G.R.); (R.K.)
| | - Ramon Kaneno
- Department of Microbiology and Immunology, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (G.G.R.); (R.K.)
| | | | - Raquel Fantin Domeniconi
- Department of Anatomy, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (L.A.L.); (M.S.C.); (R.F.D.); (F.E.M.)
| | - Marcelo Martinez
- Department of Morphology and Pathology, Federal University of São Carlos, 13565-905 São Paulo, Brazil;
| | - Francisco Eduardo Martinez
- Department of Anatomy, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (L.A.L.); (M.S.C.); (R.F.D.); (F.E.M.)
| | - Wagner José Fávaro
- Department of Structural and Functional Biology, UNICAMP-University of Campinas, Campinas, 13083-970 São Paulo, Brazil;
| | - Luiz Gustavo de Almeida Chuffa
- Department of Anatomy, UNESP-São Paulo State University, Institute of Biosciences, Botucatu, 18618-689 São Paulo, Brazil; (L.A.L.); (M.S.C.); (R.F.D.); (F.E.M.)
| |
Collapse
|
15
|
Nuclear Factor κB Signaling and Its Related Non-coding RNAs in Cancer Therapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:208-217. [PMID: 31841993 PMCID: PMC6920321 DOI: 10.1016/j.omtn.2019.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022]
Abstract
Nuclear factor κB (NF-κB) acts as a nuclear factor that is composed of five main subunits. It is a pluripotent and crucial dimer transcription factor that has a close relationship with many serious illnesses, especially its influences on cell proliferation, inflammation, and cancer initiation and progression. NF-κB acts as part of the signaling pathway and determines its effect on the expression of several other genes, such as epidermal growth factor receptor (EGFR), p53, signal transducer and activator of transcription 3 (STAT3), and non-coding RNA (ncRNA). Continuous activation of the NF-κB signaling pathway has been seen in many cancer types. While the NF-κB signaling pathway is tightly regulated in physiological settings, quite frequently it is constitutively activated in cancer, and the molecular biology mechanism underlying the deregulated activation of NF-κB signaling remains unclear. In this review, we discuss the regulatory role and possible clinical significance of ncRNA (microRNA [miRNA] and long non-coding RNA [lncRNA]) in NF-κB signaling in cancer, including in the conversion of inflammation to carcinogenesis. Non-coding RNA plays an essential and complex role in the NF-κB signaling pathway. NF-κB activation can also induce the ncRNA status. Targeting NF-κB signaling by ncRNA is becoming a promising strategy of drug development and cancer treatment.
Collapse
|
16
|
Zhu G, Cheng Z, Huang Y, Zheng W, Yang S, Lin C, Ye J. MyD88 mediates colorectal cancer cell proliferation, migration and invasion via NF‑κB/AP‑1 signaling pathway. Int J Mol Med 2019; 45:131-140. [PMID: 31746347 PMCID: PMC6889924 DOI: 10.3892/ijmm.2019.4390] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
The role of myeloid differentiation factor 88 (MyD88) in malignant tumors is largely unknown. Therefore, in this study, we aimed to examine the function and underlying mechanism of MyD88 in colorectal carcinoma in vitro using SW480 and HCT116 cell lines and in vivo using a nude mouse model. SW480 and HCT116 cells were infected with a lentiviral-based effective MyD88 siRNA virus. CCK-8 and colony formation assay were used to assess cell proliferation. Transwell and scratch assays were used to test the migration of colorectal cancer cells, and the Transwell assay was further used to analyze the invasiveness of colorectal cancer cells. Western blotting was performed to analyze the underlying mechanism of MyD88 regulation. In vitro experiments demonstrated that silencing MyD88 in SW480 and HCT116 cells markedly suppressed growth and invasion. Furthermore, MyD88 knockdown affected the MyD88-NF-κB/AP-1 signaling pathways in SW480 and HCT116 cells. In vivo, MyD88 knockdown inhibited tumor growth in a HCT116 cell subcutaneous nude model. We found that knockdown of the MyD88 gene can affect proliferation, invasion, and migration of colorectal cancer cells. We further verified that MyD88 knockdown can reduce the activity of NF-κB and AP-1 pathways. These results show that MyD88 gene plays an important role in promoting colorectal cancer, and thus can be exploited as a potential diagnostic and prognostic biomarker for colorectal cancer.
Collapse
Affiliation(s)
- Guangwei Zhu
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zhibin Cheng
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yongjian Huang
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Wei Zheng
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Shugang Yang
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Chunlin Lin
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Jianxin Ye
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
17
|
Ignacio RMC, Lee ES, Son DS. Potential Roles of Innate Immune Chemokine and Cytokine Network on Lipopolysaccharide-Based Therapeutic Approach in Ovarian Cancer. Immune Netw 2019; 19:e22. [PMID: 31281719 PMCID: PMC6597445 DOI: 10.4110/in.2019.19.e22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer (OC), the deadliest gynecological cancer, results in poor overall survival, urgently requiring a novel therapeutic approach. As cumulative exposures to endotoxins decreased OC risk epidemiologically, we evaluated if LPS, a Toll-like receptor 4 agonist known as active component of endotoxins, could increase survival in the murine peritoneal dissemination model of SKOV-3 OC cells. LPS significantly increased the mean survival time of more than 116 days compared with 63 days in the control. Furthermore, no tumor burden was present in three mice among eight LPS-treated mice. SKOV-3 cells were not responsive to LPS and showed unaltered chemokine signature. Rather than direct effects to OC cells, LPS was found to increase proinflammatory chemokines and cytokines, such as CXCL1, CXCL8, TNF, and IL-1B, in innate immune system. Taken together, LPS is likely to potentiate the cytotoxic-related innate immunogenicity via proinflammatory chemokines and cytokines, which attenuates the peritoneal dissemination of OC.
Collapse
Affiliation(s)
- Rosa Mistica C Ignacio
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
18
|
Zhu Y, Zhang H, Zhang G, Shi Y, Huang J. Co-expression of CD44/MyD88 is a poor prognostic factor in advanced epithelial ovarian cancer. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:91. [PMID: 31019941 DOI: 10.21037/atm.2019.01.28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Cluster of differentiation 44 (CD44)/myeloid differentiation factor 88 (MyD88) is the molecular characterization of EOC stem cells. An important characteristic of CD44+/MyD88+ epithelial ovarian cancer (EOC) cells, which differentiate them from the CD44-/MyD88- EOC cells, is the presence of a functional TLR4-MyD88-NFkB pathway. The aim of our study is to investigate the clinical significance of CD44/MyD88 co-expression in EOC. Methods A total of 138 specimens of ovarian tissues was detected CD44 and MyD88 expression by immunocytochemistry, including EOC (N=108), borderline tumors (N=10), benign cysts (N=10) and normal ovarian tissue (N=10). The association of CD44/MyD88 co-expression with clinicopathological factors and outcomes was analyzed. Results The expression of CD44 was showed distinct difference in EOC (53 of 108, 49.1%), in borderline tumors (3 of 10, 30.0%), in benign cysts (2 of 10, 20.0%) and normal ovarian (2 of 10, 20.0%). A total of 41 (38.0%) cancers showed a combined expression of CD44/MyD88. The expression of CD44 and MyD88 had definitely correlativity (r=0.21, P=0.026). CD44/MyD88 co-expression was associated with tumor progression, metastasis, and recurrence in advanced EOC, and an independent prognostic factor for disease-free survival and overall survival. Conclusions CD44/MyD88 co-expression has been shown to contribute to EOC progression and outcome directly and has a promising as a therapeutic target in EOC.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Gynaecologic Oncology, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.,Department of Ultrasound, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Hongtao Zhang
- Department of Obstetrics and Gynecology, Sichuan Jinxin Women and Children's Hospital, Chengdu 610000, China
| | - Guonan Zhang
- Department of Gynaecologic Oncology, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Yu Shi
- Department of Gynaecologic Oncology, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jianming Huang
- Department of Gynaecologic Oncology, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.,Department of Biochemistry & Molecular Biology, Sichuan Cancer Hospital & Institute, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| |
Collapse
|
19
|
McGrath SE, Annels N, Madhuri TK, Tailor A, Butler-Manuel SA, Morgan R, Pandha H, Michael A. Engrailed-2 (EN2) - a novel biomarker in epithelial ovarian cancer. BMC Cancer 2018; 18:943. [PMID: 30285763 PMCID: PMC6171236 DOI: 10.1186/s12885-018-4816-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/13/2018] [Indexed: 11/11/2022] Open
Abstract
Background Epithelial ovarian cancer is a common malignancy, with no clinically approved diagnostic biomarker. Engrailed-2 (EN2) is a homeodomain-containing transcription factor, essential during embryological neural development, which is dysregulated in several cancer types. We evaluated the expression of EN2 in Epithelial ovarian cancer, and reviewed its role as a biomarker. Methods We evaluated 8 Epithelial ovarian cancer cell lines, along with > 100 surgical specimens from the Royal Surrey County Hospital (2009–2014). In total, 108 tumours and 5 normal tissue specimens were collected. En2 mRNA was evaluated by semi-quantitative RT-PCR. Histological sub-type, and platinum-sensitive/−resistant status were compared. Protein expression was assessed in cell lines (immunofluorescence), and in > 150 tumours (immunohistochemistry). Results En2 mRNA expression was elevated in serous ovarian tumours compared with normal ovary (p < 0.001), particularly in high-grade serous ovarian cancer (p < 0.0001) and in platinum-resistant tumours (p = 0.0232). Median Overall Survival and Progression-free Survival were reduced with high En2 expression (OS = 28 vs 42 months, p = 0.0329; PFS = 8 vs 27 months; p = 0.0004). Positive cytoplasmic EN2 staining was demonstrated in 78% of Epithelial ovarian cancers, with absence in normal ovary. EN2 positive high-grade serous ovarian cancer patients had a shorter PFS (10 vs 17.5 months; p = 0.0103). Conclusion The EN2 transcription factor is a novel ovarian cancer biomarker. It demonstrates prognostic value, correlating with worse Overall Survival and Progression-free Survival. It is hoped that further work will validate its use as a biomarker, and provide insight into the role of EN2 in the development, progression and spread of ovarian cancer.
Collapse
Affiliation(s)
- Sophie Elena McGrath
- Oncology, School of Biosciences and Medicine, FHMS, University of Surrey, Leggett Building, Daphne Jackson Road, Guildford, GU2 7WG, UK.,Present Address: The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SM2 5PT, UK
| | - Nicola Annels
- Oncology, School of Biosciences and Medicine, FHMS, University of Surrey, Leggett Building, Daphne Jackson Road, Guildford, GU2 7WG, UK
| | | | - Anil Tailor
- Royal Surrey County Hospital, Egerton Road, Guildford, GU2 7XX, UK
| | | | - Richard Morgan
- Oncology, School of Biosciences and Medicine, FHMS, University of Surrey, Leggett Building, Daphne Jackson Road, Guildford, GU2 7WG, UK.,Present Address: The Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire, BD7 1DP, UK
| | - Hardev Pandha
- Oncology, School of Biosciences and Medicine, FHMS, University of Surrey, Leggett Building, Daphne Jackson Road, Guildford, GU2 7WG, UK
| | - Agnieszka Michael
- Oncology, School of Biosciences and Medicine, FHMS, University of Surrey, Leggett Building, Daphne Jackson Road, Guildford, GU2 7WG, UK.
| |
Collapse
|
20
|
McCloskey CW, Rodriguez GM, Galpin KJC, Vanderhyden BC. Ovarian Cancer Immunotherapy: Preclinical Models and Emerging Therapeutics. Cancers (Basel) 2018; 10:cancers10080244. [PMID: 30049987 PMCID: PMC6115831 DOI: 10.3390/cancers10080244] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has emerged as one of the most promising approaches for ovarian cancer treatment. The tumor microenvironment (TME) is a key factor to consider when stimulating antitumoral responses as it consists largely of tumor promoting immunosuppressive cell types that attenuate antitumor immunity. As our understanding of the determinants of the TME composition grows, we have begun to appreciate the need to address both inter- and intra-tumor heterogeneity, mutation/neoantigen burden, immune landscape, and stromal cell contributions. The majority of immunotherapy studies in ovarian cancer have been performed using the well-characterized murine ID8 ovarian carcinoma model. Numerous other animal models of ovarian cancer exist, but have been underutilized because of their narrow initial characterizations in this context. Here, we describe animal models that may be untapped resources for the immunotherapy field because of their shared genomic alterations and histopathology with human ovarian cancer. We also shed light on the strengths and limitations of these models, and the knowledge gaps that need to be addressed to enhance the utility of preclinical models for testing novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Curtis W McCloskey
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Galaxia M Rodriguez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Kristianne J C Galpin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
21
|
Wu K, Zhang H, Fu Y, Zhu Y, Kong L, Chen L, Zhao F, Yu L, Chen X. TLR4/MyD88 signaling determines the metastatic potential of breast cancer cells. Mol Med Rep 2018; 18:3411-3420. [PMID: 30066873 PMCID: PMC6102647 DOI: 10.3892/mmr.2018.9326] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The influence of Toll-like receptor (TLR)4/myeloid differentiation factor (MyD)88 signaling on the invasion and metastasis of cancer cells has been previously reported. The purpose of the present study was to determine the role of TLR4/MyD88 in breast cancer cell migration and invasion, and to discover novel therapeutic targets for breast cancer treatment. TLR4, MyD88 and high mobility group box 1 (HMGB1) mRNA expression levels were assessed in highly invasive human MDA-MB-231 breast cancer cells, breast cancer cells with a low rate of invasion (MCF-7) and normal human MDA-Kb2 mammary gland cells by reverse transcription-quantitative polymerase chain reaction. The protein expression levels of these markers were detected by western blotting and immunofluorescence. Randomly selected breast cancer and paracarcinoma tissues were used to measure TLR4 and MyD88 protein expression levels by immunohistochemistry. The mRNA and protein expression levels of TLR4 and MyD88 were significantly higher in MDA-MB-231 cells compared with either MCF-7 cells or MDA-Kb2 cells. The mRNA and protein expression levels of HMGB1 were comparable in the two breast cancer cell lines, with no statistical difference (P>0.05). TLR4 and MyD88 protein expression levels were also significantly higher in breast cancer tissues compared with paracarcinoma tissues (P<0.05). TLR4 and MyD88 protein expression levels were positively correlated with axillary lymph node metastasis and histological grade (P<0.05). TLR4/MyD88 expression levels were positively correlated with the metastasis of breast cancer cells. TLR4/MyD88 may be useful as a novel biomarker to evaluate the prognosis and treatment of patients with breast cancer.
Collapse
Affiliation(s)
- Kunlin Wu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Huihao Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yajuan Fu
- Southern Biomedical Research Center, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Lingjun Kong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ling Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Feng Zhao
- First Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Liangfei Yu
- First Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
22
|
Wu J, Ding J, Yang J, Guo X, Zheng Y. MicroRNA Roles in the Nuclear Factor Kappa B Signaling Pathway in Cancer. Front Immunol 2018; 9:546. [PMID: 29616037 PMCID: PMC5868594 DOI: 10.3389/fimmu.2018.00546] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/02/2018] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor kappa B (NF-κB) is a pluripotent and crucial dimer transcription factor that orchestrates various physiological and pathological processes, especially cell proliferation, inflammation, and cancer development and progression. NF-κB expression is transient and tightly regulated in normal cells, but it is activated in cancer cells. Recently, numerous studies have demonstrated microRNAs (miRNAs) play a vital role in the NF-κB signaling pathway and NF-κB-associated immune responses, radioresistance and drug resistance of cancer, some acting as inhibitors and the others as activators. Although it is still in infancy, targeting NF-κB or the NF-κB signaling pathway by miRNAs is becoming a promising strategy of cancer treatment.
Collapse
Affiliation(s)
- Jin’en Wu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute (CAAS), Lanzhou, China
| | - Juntao Ding
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jing Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute (CAAS), Lanzhou, China
| | - Xiaola Guo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute (CAAS), Lanzhou, China
| | - Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute (CAAS), Lanzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
Block MS, Vierkant RA, Rambau PF, Winham SJ, Wagner P, Traficante N, Tołoczko A, Tiezzi DG, Taran FA, Sinn P, Sieh W, Sharma R, Rothstein JH, Ramón Y Cajal T, Paz-Ares L, Oszurek O, Orsulic S, Ness RB, Nelson G, Modugno F, Menkiszak J, McGuire V, McCauley BM, Mack M, Lubiński J, Longacre TA, Li Z, Lester J, Kennedy CJ, Kalli KR, Jung AY, Johnatty SE, Jimenez-Linan M, Jensen A, Intermaggio MP, Hung J, Herpel E, Hernandez BY, Hartkopf AD, Harnett PR, Ghatage P, García-Bueno JM, Gao B, Fereday S, Eilber U, Edwards RP, de Sousa CB, de Andrade JM, Chudecka-Głaz A, Chenevix-Trench G, Cazorla A, Brucker SY, Alsop J, Whittemore AS, Steed H, Staebler A, Moysich KB, Menon U, Koziak JM, Kommoss S, Kjaer SK, Kelemen LE, Karlan BY, Huntsman DG, Høgdall E, Gronwald J, Goodman MT, Gilks B, García MJ, Fasching PA, de Fazio A, Deen S, Chang-Claude J, Candido Dos Reis FJ, Campbell IG, Brenton JD, Bowtell DD, Benítez J, Pharoah PDP, Köbel M, Ramus SJ, Goode EL. MyD88 and TLR4 Expression in Epithelial Ovarian Cancer. Mayo Clin Proc 2018; 93:307-320. [PMID: 29502561 PMCID: PMC5870793 DOI: 10.1016/j.mayocp.2017.10.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/14/2017] [Accepted: 10/16/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To evaluate myeloid differentiation primary response gene 88 (MyD88) and Toll-like receptor 4 (TLR4) expression in relation to clinical features of epithelial ovarian cancer, histologic subtypes, and overall survival. PATIENTS AND METHODS We conducted centralized immunohistochemical staining, semi-quantitative scoring, and survival analysis in 5263 patients participating in the Ovarian Tumor Tissue Analysis consortium. Patients were diagnosed between January 1, 1978, and December 31, 2014, including 2865 high-grade serous ovarian carcinomas (HGSOCs), with more than 12,000 person-years of follow-up time. Tissue microarrays were stained for MyD88 and TLR4, and staining intensity was classified using a 2-tiered system for each marker (weak vs strong). RESULTS Expression of MyD88 and TLR4 was similar in all histotypes except clear cell ovarian cancer, which showed reduced expression compared with other histotypes (P<.001 for both). In HGSOC, strong MyD88 expression was modestly associated with shortened overall survival (hazard ratio [HR], 1.13; 95% CI, 1.01-1.26; P=.04) but was also associated with advanced stage (P<.001). The expression of TLR4 was not associated with survival. In low-grade serous ovarian cancer (LGSOC), strong expression of both MyD88 and TLR4 was associated with favorable survival (HR [95% CI], 0.49 [0.29-0.84] and 0.44 [0.21-0.89], respectively; P=.009 and P=.02, respectively). CONCLUSION Results are consistent with an association between strong MyD88 staining and advanced stage and poorer survival in HGSOC and demonstrate correlation between strong MyD88 and TLR4 staining and improved survival in LGSOC, highlighting the biological differences between the 2 serous histotypes.
Collapse
Affiliation(s)
| | | | - Peter F Rambau
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada; Pathology Department, Catholic University of Health and Allied Sciences-Bugando, Mwanza, Tanzania
| | - Stacey J Winham
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Philipp Wagner
- Tübingen University Hospital, Department of Women's Health, Tübingen, Germany
| | - Nadia Traficante
- Sir Peter MacCallum Department of Oncology, the University of Melbourne, Parkville, Victoria, Australia
| | - Aleksandra Tołoczko
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Daniel G Tiezzi
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Florin Andrei Taran
- Tübingen University Hospital, Department of Women's Health, Tübingen, Germany
| | - Peter Sinn
- Department of Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Weiva Sieh
- Department of Population Health Science and Policy, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Raghwa Sharma
- Pathology West ICPMR Westmead, Westmead Hospital, the University of Sydney, Sydney, Australia; University of Western Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Joseph H Rothstein
- Department of Population Health Science and Policy, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Luis Paz-Ares
- H12O-CNIO Lung Cancer Clinical Research Unit, Madrid, Spain; Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Oleg Oszurek
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Sandra Orsulic
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Gregg Nelson
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA; Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA; Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Janusz Menkiszak
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| | - Valerie McGuire
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA
| | - Bryan M McCauley
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Marie Mack
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Jan Lubiński
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | | | - Zheng Li
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN; Department of Gynecologic Oncology, the Third Affiliated Hospital of Kunming Medical University (Yunnan Tumor Hospital), Kunming, China
| | - Jenny Lester
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Catherine J Kennedy
- Centre for Cancer Research, the Westmead Institute for Medical Research, the University of Sydney, Sydney, New South Wales, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
| | | | - Audrey Y Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sharon E Johnatty
- Department of Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Allan Jensen
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria P Intermaggio
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | - Jillian Hung
- Centre for Cancer Research, the Westmead Institute for Medical Research, the University of Sydney, Sydney, New South Wales, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Esther Herpel
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany; Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | | | - Andreas D Hartkopf
- Tübingen University Hospital, Department of Women's Health, Tübingen, Germany
| | - Paul R Harnett
- Centre for Cancer Research, the Westmead Institute for Medical Research, the University of Sydney, Sydney, New South Wales, Australia; Crown Princess Mary Cancer Centre, Westmead Hospital, the University of Sydney, Sydney, Australia
| | - Prafull Ghatage
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Bo Gao
- Centre for Cancer Research, the Westmead Institute for Medical Research, the University of Sydney, Sydney, New South Wales, Australia; Crown Princess Mary Cancer Centre, Westmead Hospital, the University of Sydney, Sydney, Australia
| | - Sian Fereday
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Ursula Eilber
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert P Edwards
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Christiani B de Sousa
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jurandyr M de Andrade
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Anita Chudecka-Głaz
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| | | | - Alicia Cazorla
- Pathology Department, Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Y Brucker
- Tübingen University Hospital, Department of Women's Health, Tübingen, Germany
| | - Jennifer Alsop
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Alice S Whittemore
- Department of Health Research and Policy and Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA
| | - Helen Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, Edmonton, Alberta, Canada
| | - Annette Staebler
- Tübingen University Hospital, Institute of Pathology, Tübingen, Germany
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY
| | - Usha Menon
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | | | - Stefan Kommoss
- Tübingen University Hospital, Department of Women's Health, Tübingen, Germany
| | - Susanne K Kjaer
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Linda E Kelemen
- Department of Public Health Sciences, Medical University of South Carolina and Hollings Cancer Center, Charleston, SC
| | - Beth Y Karlan
- Women's Cancer Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Translational and Applied Genomics, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Estrid Høgdall
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark; Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Blake Gilks
- Genetic Pathology Evaluation Centre, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - María José García
- Human Genetics Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Peter A Fasching
- David Geffen School of Medicine, Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles; University Breast Center Franconia, Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Anna de Fazio
- Centre for Cancer Research, the Westmead Institute for Medical Research, the University of Sydney, Sydney, New South Wales, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Suha Deen
- Department of Histopathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francisco J Candido Dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ian G Campbell
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - James D Brenton
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK; Cambridge Experimental Cancer Medicine Centre, Cambridge, UK
| | - David D Bowtell
- Sir Peter MacCallum Department of Oncology, the University of Melbourne, Parkville, Victoria, Australia; Cancer Genomics Program, Research Department, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Garvan Institute, Sydney, New South Wales, Australia
| | - Javier Benítez
- Human Genetics Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul D P Pharoah
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK; Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Susan J Ramus
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia; Garvan Institute, Sydney, New South Wales, Australia
| | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN.
| |
Collapse
|
24
|
Hao B, Chen Z, Bi B, Yu M, Yao S, Feng Y, Yu Y, Pan L, Di D, Luo G, Zhang X. Role of TLR4 as a prognostic factor for survival in various cancers: a meta-analysis. Oncotarget 2018; 9:13088-13099. [PMID: 29560134 PMCID: PMC5849198 DOI: 10.18632/oncotarget.24178] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/04/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Accumulating evidence showed that high expression of toll like receptor 4 (TLR4) was significantly associated with the outcome of patients with solid cancers. However, other studies failed to draw a similar conclusion. Thus, a systematic meta-analysis was performed to assess the prognostic value of TLR4 in solid tumors. RESULTS Data from 15 studies and 1294 patients were enrolled. Among the 15 studies, 14 studies demonstrated the association between overall survival(OS) and TLR4 expression, and 7 studies described the relationship between disease-free survival(DFS) and TLR4 expression. High expression of TLR4 was significantly associated with poor OS (pooled hazard ratio (HR) = 2.05; 95% confidence interval (CI) (1.49, 2,49), P < 0.001). The results of meta regression analysis indicated that the subgroups of ethnic (PD = 0.924), tumor type (PD = 0.669), HR obtained method (PD = 0.945), analysis type (PD = 0.898), and cut-off value(PD = 0.835) were not the resource of heterogeneity. Moreover, patients with elevated TLR4 had a significantly worse DFS (pooled HR = 1.79; 95% CI (1.11, 2.88), P < 0.05). MATERIALS AND METHODS We searched PubMed, Embase and the Cochrane Library (last update by April 18, 2017) to identify literatures evaluating the value of TLR4 in cancer patients. Combined hazard ratios (HRs) for OS and DFS were assessed using fixed-effects models and random effects models respectively. CONCLUSIONS The meta-analysis suggests that elevated expression of TLR4 is associated with poor OS and shorter DFS of patients with solid tumors. The results indicate that TLR4, as a novel prognostic biomarker in solid tumors, could potentially help to improve treatment decision-making of solid tumors in clinical.
Collapse
Affiliation(s)
- Bo Hao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Zhen Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Baochen Bi
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Miaomei Yu
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Shuang Yao
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Yuehua Feng
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Yang Yu
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Lili Pan
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Dongmei Di
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Guanghua Luo
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Xiaoying Zhang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| |
Collapse
|
25
|
MyD88 is an essential component of retinoic acid-induced differentiation in human pluripotent embryonal carcinoma cells. Cell Death Differ 2017; 24:1975-1986. [PMID: 28885616 DOI: 10.1038/cdd.2017.124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/02/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
We have previously reported that myeloid differentiation primary response gene 88 (MyD88) is downregulated during all-trans retinoic acid (RA)-induced differentiation of pluripotent NTera2 human embryonal carcinoma cells (hECCs), whereas its maintained expression is associated with RA differentiation resistance in nullipotent 2102Ep hECCs. MyD88 is the main adapter for toll-like receptor (TLR) signalling, where it determines the secretion of chemokines and cytokines in response to pathogens. In this study, we report that loss of MyD88 is essential for RA-facilitated differentiation of hECCs. Functional analysis using a specific MyD88 peptide inhibitor (PepInh) demonstrated that high MyD88 expression in the self-renewal state inhibits the expression of a specific set of HOX genes. In NTera2 cells, MyD88 is downregulated during RA-induced differentiation, a mechanism that could be broadly replicated by MyD88 PepInh treatment of 2102Ep cells. Notably, MyD88 inhibition transitioned 2102Ep cells into a stable, self-renewing state that appears to be primed for differentiation upon addition of RA. At a molecular level, MyD88 inhibition combined with RA treatment upregulated HOX, RA signalling and TLR signalling genes. These events permit differentiation through a standard downregulation of Oct4-Sox2-Nanog mechanism. In line with its role in regulating secretion of specific proteins, conditioned media experiments demonstrated that differentiated (MyD88 low) NTera2 cell media was sufficient to differentiate NTera2 cells. Protein array analysis indicated that this was owing to secretion of factors known to regulate angiogenesis, neurogenesis and all three branches of TGF-β Superfamily signalling. Collectively, these data offer new insights into RA controlled differentiation of pluripotent cells, with notable parallels to the ground state model of embryonic stem cell self-renewal. These data may provide insights to facilitate improved differentiation protocols for regenerative medicine and differentiation-therapies in cancer treatment.
Collapse
|
26
|
Sun NK, Huang SL, Chang TC, Chao CCK. TLR4 and NFκB signaling is critical for taxol resistance in ovarian carcinoma cells. J Cell Physiol 2017; 233:2489-2501. [PMID: 28771725 DOI: 10.1002/jcp.26125] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/01/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Nian-Kang Sun
- Department of Biochemistry and Molecular Biology, College of Medicine; Chang Gung University; Taoyuan, Taiwan Republic of China
- Division of Biomedical Sciences; Chang Gung University of Science and Technology; Taoyuan, Taiwan Republic of China
- Department of Obstetrics and Gynaecology; Chang Gung Memorial Hospital Linkou Medical Centre; Taoyuan, Taiwan Republic of China
| | - Shang-Lang Huang
- Department of Biochemistry and Molecular Biology, College of Medicine; Chang Gung University; Taoyuan, Taiwan Republic of China
| | - Ting-Chang Chang
- Department of Obstetrics and Gynaecology; Chang Gung Memorial Hospital Linkou Medical Centre; Taoyuan, Taiwan Republic of China
| | - Chuck C.-K. Chao
- Department of Biochemistry and Molecular Biology, College of Medicine; Chang Gung University; Taoyuan, Taiwan Republic of China
- Department of Obstetrics and Gynaecology; Chang Gung Memorial Hospital Linkou Medical Centre; Taoyuan, Taiwan Republic of China
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan, Taiwan Republic of China
- Liver Research Center; Chang Gung Memorial Hospital at Linkou; Taoyuan, Taiwan Republic of China
| |
Collapse
|
27
|
Wang C, Armasu SM, Kalli KR, Maurer MJ, Heinzen EP, Keeney GL, Cliby WA, Oberg AL, Kaufmann SH, Goode EL. Pooled Clustering of High-Grade Serous Ovarian Cancer Gene Expression Leads to Novel Consensus Subtypes Associated with Survival and Surgical Outcomes. Clin Cancer Res 2017. [PMID: 28280090 DOI: 10.1158/1078-0432.ccr-17-0246] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Here we assess whether molecular subtyping identifies biological features of tumors that correlate with survival and surgical outcomes of high-grade serous ovarian cancer (HGSOC).Experimental Design: Consensus clustering of pooled mRNA expression data from over 2,000 HGSOC cases was used to define molecular subtypes of HGSOCs. This de novo classification scheme was then applied to 381 Mayo Clinic HGSOC patients with detailed survival and surgical outcome information.Results: Five molecular subtypes of HGSOC were identified. In the pooled dataset, three subtypes were largely concordant with prior studies describing proliferative, mesenchymal, and immunoreactive tumors (concordance > 70%), and the group of tumors previously described as differentiated type was segregated into two new types, one of which (anti-mesenchymal) had downregulation of genes that were typically upregulated in the mesenchymal subtype. Molecular subtypes were significantly associated with overall survival (P < 0.001) and with rate of optimal surgical debulking (≤1 cm, P = 1.9E-4) in the pooled dataset. Among stage III-C or IV Mayo Clinic patients, molecular subtypes were also significantly associated with overall survival (P = 0.001), as well as rate of complete surgical debulking (no residual disease; 16% in mesenchymal tumors compared with >28% in other subtypes; P = 0.02).Conclusions: HGSOC tumors may be categorized into five molecular subtypes that associate with overall survival and the extent of residual disease following debulking surgery. Because mesenchymal tumors may have features that were associated with less favorable surgical outcome, molecular subtyping may have future utility in guiding neoadjuvant treatment decisions for women with HGSOC. Clin Cancer Res; 23(15); 4077-85. ©2017 AACR.
Collapse
Affiliation(s)
- Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Sebastian M Armasu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Ethan P Heinzen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Gary L Keeney
- Department of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - William A Cliby
- Department of Gynecologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
28
|
Wang C, Armasu SM, Kalli KR, Maurer MJ, Heinzen EP, Keeney GL, Cliby WA, Oberg AL, Kaufmann SH, Goode EL. Pooled Clustering of High-Grade Serous Ovarian Cancer Gene Expression Leads to Novel Consensus Subtypes Associated with Survival and Surgical Outcomes. Clin Cancer Res 2017; 23:4077-4085. [PMID: 28280090 DOI: 10.1158/1078-0432.ccr-17-0246] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/13/2017] [Accepted: 03/06/2017] [Indexed: 01/27/2023]
Abstract
Purpose: Here we assess whether molecular subtyping identifies biological features of tumors that correlate with survival and surgical outcomes of high-grade serous ovarian cancer (HGSOC).Experimental Design: Consensus clustering of pooled mRNA expression data from over 2,000 HGSOC cases was used to define molecular subtypes of HGSOCs. This de novo classification scheme was then applied to 381 Mayo Clinic HGSOC patients with detailed survival and surgical outcome information.Results: Five molecular subtypes of HGSOC were identified. In the pooled dataset, three subtypes were largely concordant with prior studies describing proliferative, mesenchymal, and immunoreactive tumors (concordance > 70%), and the group of tumors previously described as differentiated type was segregated into two new types, one of which (anti-mesenchymal) had downregulation of genes that were typically upregulated in the mesenchymal subtype. Molecular subtypes were significantly associated with overall survival (P < 0.001) and with rate of optimal surgical debulking (≤1 cm, P = 1.9E-4) in the pooled dataset. Among stage III-C or IV Mayo Clinic patients, molecular subtypes were also significantly associated with overall survival (P = 0.001), as well as rate of complete surgical debulking (no residual disease; 16% in mesenchymal tumors compared with >28% in other subtypes; P = 0.02).Conclusions: HGSOC tumors may be categorized into five molecular subtypes that associate with overall survival and the extent of residual disease following debulking surgery. Because mesenchymal tumors may have features that were associated with less favorable surgical outcome, molecular subtyping may have future utility in guiding neoadjuvant treatment decisions for women with HGSOC. Clin Cancer Res; 23(15); 4077-85. ©2017 AACR.
Collapse
Affiliation(s)
- Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Sebastian M Armasu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Ethan P Heinzen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Gary L Keeney
- Department of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - William A Cliby
- Department of Gynecologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
29
|
Wei L, Yin F, Zhang W, Li L. STROBE-compliant integrin through focal adhesion involve in cancer stem cell and multidrug resistance of ovarian cancer. Medicine (Baltimore) 2017; 96:e6345. [PMID: 28328815 PMCID: PMC5371452 DOI: 10.1097/md.0000000000006345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be the root of carcinoma relapse and drug resistance in ovarian cancer. Hunting for the potential CSC genes and explain their functions would be a feasible strategy to meet the challenge of the drug resistance in ovarian cancer. In this study, we performed bioinformatic approaches such as biochip data extraction and pathway enrichment analyses to elucidate the mechanism of the CSC genes in regulation of drug resistance. Potential key genes, integrins, were identified to be related to CSC in addition to their associations with drug resistance and prognosis in ovarian cancer. A total of 36 ovarian CSC genes involved in regulation of drug resistance were summarized, and potential drug resistance-related CSC genes were identified based on 3 independent microarrays retrieved from the Gene Expression Omnibus (GEO) Profiles. Pathway enrichment of CSC genes associated with drug resistance in ovarian cancer indicated that focal adhesion signaling might play important roles in CSC genes-mediated drug resistance. Integrins are members of the adhesion molecules family, and integrin subunit alpha 1, integrin subunit alpha 5, and integrin subunit alpha 6 (ITGA6) were identified as central CSC genes and their expression in side population cells, cisplatin-resistant SKOV3 (SKOV3/DDP2) cells, and cisplatin-resistant A2780 (A2780/DDP) cells were dysregulated as measured by real-time quantitative polymerase chain reaction. The high expression of ITGA6 in 287 ovarian cancer patients of TCGA cohort was significantly associated with poorer progression-free survival. This study provide the basis for further understanding of CSC genes in regulation of drug resistance in ovarian cancer, and integrins could be a potential biomarker for prognosis of ovarian cancer.
Collapse
Affiliation(s)
- Luwei Wei
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University
| | - Fuqiang Yin
- Life Sciences Institute, Guangxi Medical University
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, PR China
| | - Wei Zhang
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University
| | - Li Li
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, PR China
| |
Collapse
|
30
|
The inflammatory microenvironment in epithelial ovarian cancer: a role for TLR4 and MyD88 and related proteins. Tumour Biol 2016; 37:13279-13286. [PMID: 27460076 DOI: 10.1007/s13277-016-5163-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/12/2016] [Indexed: 12/20/2022] Open
Abstract
The tumor-associated inflammatory microenvironment may play a pivotal role in epithelial ovarian cancer (EOC) carcinogenesis and outcomes, but a detailed profile in patient-derived tumors is needed. Here, we investigated the expression of TLR4- and MyD88-associated markers in tumors from over 500 EOC patients using immunohistochemical staining. We demonstrate that high expression of TLR4 and MyD88 predicts poorer overall survival in patients with EOC; most likely, this is due to their association with serous histology and features of high tumor burden and aggressiveness, including stage, grade, and ascites at surgery. Combined TLR4 and MyD88 expression appears to serve as an independent risk factor for shortened survival time, even after covariate adjustment (both moderate HR 1.1 [95 % CI 0.7-1.8], both strong HR 2.1 [95 % CI 1.1-3.8], both weak as referent; p = 0.027). We reveal that in EOC tissues with elevated expression of both TLR4 and MyD88 and activated NF-κB signaling pathway, expression of hsp60, hsp70, beta 2 defensin, and HMGB1 are also enriched. In total, these results suggest that activation of TLR4/MyD88/NF-κB signaling by endogenous ligands may contribute to an inflammatory microenvironment that drives a more aggressive phenotype with poorer clinical outcome in EOC patients.
Collapse
|
31
|
English DP, Menderes G, Black J, Schwab CL, Santin AD. Molecular diagnosis and molecular profiling to detect treatment-resistant ovarian cancer. Expert Rev Mol Diagn 2016; 16:769-82. [PMID: 27169329 DOI: 10.1080/14737159.2016.1188692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Epithelial ovarian cancer remains the gynecologic tumor with the highest rate of recurrence after initial optimal cytoreductive surgery followed by adjuvant chemotherapy. Unfortunately, with the development of recurrent ovarian cancer often comes the discovery of chemo-resistant disease. The absence of improvement in long term survival, notwithstanding the use of newer agents as is seen in other cancers, emphasizes the need for improved understanding of the processes that lead to chemo-resistant disease. AREAS COVERED This review will cover the following topics: 1. Molecular and cellular mechanisms in platinum and paclitaxel resistance 2. Other molecular mediators of chemo-resistance 3. Expression of stem cell markers in ovarian cancer and relationship to chemo-resistance 4. MicroRNA and long non-coding RNA expression in chemo-resistant ovarian cancer 5. Determination of chromosomal aberrations as markers of chemo-resistance 6. Molecular profiling in chemo-resistant disease. A standard MEDLINE search was performed using the key words; ovarian cancer, chemo-resistant disease, molecular profiling, cancer stem cells and chemotherapy. Expert Commentary: Over the next few years the challenge remains to precisely determine the mechanisms responsible for the onset and maintenance of chemo-resistance and to effectively target these mechanisms.
Collapse
Affiliation(s)
- Diana P English
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , Stanford University , Stanford , CA , USA
| | - Gulden Menderes
- b Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology , Yale University School of Medicine , New Haven , CT , USA
| | - Jonathan Black
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , Stanford University , Stanford , CA , USA
| | - Carlton L Schwab
- b Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology , Yale University School of Medicine , New Haven , CT , USA
| | - Alessandro D Santin
- b Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
32
|
Liu H, Zhang G, Huang J, Ma S, Mi K, Cheng J, Zhu Y, Zha X, Huang W. Atractylenolide I modulates ovarian cancer cell-mediated immunosuppression by blocking MD-2/TLR4 complex-mediated MyD88/NF-κB signaling in vitro. J Transl Med 2016; 14:104. [PMID: 27118139 PMCID: PMC4847224 DOI: 10.1186/s12967-016-0845-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/30/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND TLR4/MD-2 complex-mediated MyD88-dependent activation of NF-κB and Akt promotes tumor-associated immunosuppression in epithelial ovarian cancer (EOC) via induction of immunesuppressive cytokines and indoleamine 2,3-dioxygenase (IDO). Atractylenolide I (AO-1) is a naturally occurring sesquiterpene lactone known to change the conformational ensemble of human MD-2 on EOC cells. This study examined the modulation by AO-1 of TLR4/MD-2 complex-mediated MyD88/NF-κB signaling. METHODS The expression and activation of NF-κB, Akt and IDO1 by MyD88(+) EOC SKOV3 cells was determined using western blot; the TLR4/MD-2 complex on SKOV3 cells and the phenotype of T lymphocytes were determined using flow cytometry; IDO activity was evaluated by measuring L-kynurenine; Immunesuppressive cytokines were detected using ELISA; T-cell proliferation to mitogen stimulation was assessed by MTT assay; the cytotoxicity of lymphocytes and NK cells was measured using LDH-cytotoxicity assay. RESULTS AO-1 could down-regulate expression of TLR4/MD-2 complex, resulting in downregulation of MyD88/NF-κB signaling and activation of NF-κB, Akt and IDO1 and secretion of IL-6, TGF-β1, VEGF and IL-17A by EOC SKOV3 cells, and further reduce increased levels of regulatory T cells (Treg cells) and improve decreased proliferative response and antitumor cytotoxicity of T lymphocytes exposed to EOC SKOV3 cell supernatant. CONCLUSION AO-1 may reverse EOC cell-mediated immunosuppression through blocking TLR4/MD-2 complex-mediated MyD88/NF-κB signaling.
Collapse
Affiliation(s)
- Hong Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3 of South People's Road, Chengdu, 610041, China.,Department of Gynecologic Oncology, Sichuan Cancer Hospital, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Guonan Zhang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Jianming Huang
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Shiqi Ma
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Kun Mi
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Jia Cheng
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Yi Zhu
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, No.55, Section 4 of South People's Road, Chengdu, 610041, China.,Department of Ultrasound, Sichuan Cancer Hospital, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Xiao Zha
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, No.55, Section 4 of South People's Road, Chengdu, 610041, China
| | - Wei Huang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3 of South People's Road, Chengdu, 610041, China.
| |
Collapse
|
33
|
miR-146a Attenuates Inflammatory Pathways Mediated by TLR4/NF-κB and TNFα to Protect Primary Human Retinal Microvascular Endothelial Cells Grown in High Glucose. Mediators Inflamm 2016; 2016:3958453. [PMID: 26997759 PMCID: PMC4779539 DOI: 10.1155/2016/3958453] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 12/21/2022] Open
Abstract
Pathological mechanisms underlying diabetic retinopathy are still not completely understood. Increased understanding of potential cellular pathways responsive to hyperglycemia is essential to develop novel therapeutic strategies for diabetic retinopathy. A growing body of evidence shows that microRNA (miRNA) play important roles in pathological mechanisms involved in diabetic retinopathy, as well as possessing potential as novel therapeutic targets. The hypothesis of this study was that miR-146a plays a key role in attenuating hyperglycemia-induced inflammatory pathways through reduced TLR4/NF-κB and TNFα signaling in primary human retinal microvascular endothelial cells (REC). We cultured human REC in normal (5 mM) glucose or transferred to high glucose medium (25 mM) for 3 days. Transfection was performed on REC with miRNA mimic (hsa-miR-146a-5p). Our results demonstrate that miR-146a expression was decreased in human REC cultured in high glucose. Overexpression of miR-146a using mimics reduced the levels of TLR4/NF-κB and TNFα in REC cultured in high glucose. Both MyD88-dependent and -independent signaling were decreased by miR-146a overexpression in REC in high glucose conditions. The results suggest that miR-146a is a potential therapeutic target for reducing inflammation in REC through inhibition of TLR4/NF-κB and TNFα. Our study will contribute to understanding of diabetic retinal pathology, as well as providing important clues to develop therapeutics for clinical applications.
Collapse
|
34
|
Ogbechi J, Ruf MT, Hall BS, Bodman-Smith K, Vogel M, Wu HL, Stainer A, Esmon CT, Ahnström J, Pluschke G, Simmonds RE. Mycolactone-Dependent Depletion of Endothelial Cell Thrombomodulin Is Strongly Associated with Fibrin Deposition in Buruli Ulcer Lesions. PLoS Pathog 2015; 11:e1005011. [PMID: 26181660 PMCID: PMC4504485 DOI: 10.1371/journal.ppat.1005011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/07/2015] [Indexed: 01/21/2023] Open
Abstract
A well-known histopathological feature of diseased skin in Buruli ulcer (BU) is coagulative necrosis caused by the Mycobacterium ulcerans macrolide exotoxin mycolactone. Since the underlying mechanism is not known, we have investigated the effect of mycolactone on endothelial cells, focussing on the expression of surface anticoagulant molecules involved in the protein C anticoagulant pathway. Congenital deficiencies in this natural anticoagulant pathway are known to induce thrombotic complications such as purpura fulimans and spontaneous necrosis. Mycolactone profoundly decreased thrombomodulin (TM) expression on the surface of human dermal microvascular endothelial cells (HDMVEC) at doses as low as 2ng/ml and as early as 8hrs after exposure. TM activates protein C by altering thrombin’s substrate specificity, and exposure of HDMVEC to mycolactone for 24 hours resulted in an almost complete loss of the cells’ ability to produce activated protein C. Loss of TM was shown to be due to a previously described mechanism involving mycolactone-dependent blockade of Sec61 translocation that results in proteasome-dependent degradation of newly synthesised ER-transiting proteins. Indeed, depletion from cells determined by live-cell imaging of cells stably expressing a recombinant TM-GFP fusion protein occurred at the known turnover rate. In order to determine the relevance of these findings to BU disease, immunohistochemistry of punch biopsies from 40 BU lesions (31 ulcers, nine plaques) was performed. TM abundance was profoundly reduced in the subcutis of 78% of biopsies. Furthermore, it was confirmed that fibrin deposition is a common feature of BU lesions, particularly in the necrotic areas. These findings indicate that there is decreased ability to control thrombin generation in BU skin. Mycolactone’s effects on normal endothelial cell function, including its ability to activate the protein C anticoagulant pathway are strongly associated with this. Fibrin-driven tissue ischemia could contribute to the development of the tissue necrosis seen in BU lesions. Buruli ulcer (BU) is a neglected tropical disease that is most common in West Africa and parts of Australia, but has been reported from over 30 countries worldwide. The symptoms are painless ulcers of the skin caused by a bacterial infection. The bacteria, Mycobacterium ulcerans, produce a macrolide toxin called mycolactone. In this manuscript, we have studied the effects of mycolactone on endothelial cells, specialised cells that line blood vessels and form capillaries. One of the most important functions of these cells is to prevent blood from clotting inside the vessels. We show that mycolactone reduces the ability of cultured endothelial cells to anticoagulate blood, by blocking the expression of a protein called thrombomodulin. We went on to examine samples of BU patient skin and found that thrombomodulin is also reduced here, and that in contrast to normal skin large amounts of fibrin (one of the main constituents of blood clots) were present. This means that it may be useful to consider whether anticoagulants might improve the response to antibiotics and thereby improve treatment outcomes for BU patients.
Collapse
Affiliation(s)
- Joy Ogbechi
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Marie-Thérèse Ruf
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Belinda S. Hall
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Katherine Bodman-Smith
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Moritz Vogel
- Section Clinical Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan
| | - Alexander Stainer
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Charles T. Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Josefin Ahnström
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Gerd Pluschke
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Rachel E. Simmonds
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Sheedy FJ. Turning 21: Induction of miR-21 as a Key Switch in the Inflammatory Response. Front Immunol 2015; 6:19. [PMID: 25688245 PMCID: PMC4310327 DOI: 10.3389/fimmu.2015.00019] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/09/2015] [Indexed: 12/16/2022] Open
Abstract
miR-21 is one of the most highly expressed members of the small non-coding microRNA family in many mammalian cell types. Its expression is further enhanced in many diseased states including solid tumors, cardiac injury, and inflamed tissue. While the induction of miR-21 by inflammatory stimuli cells has been well documented in both hematopoietic cells of the immune system (particularly monocytes/macrophages but also dendritic and T-cells) and non-hematopoietic tumorigenic cells, the exact functional outcome of this elevated miR-21 is less obvious. Recent studies have confirmed a key role for miR-21 in the resolution of inflammation and in negatively regulating the pro-inflammatory response induced by many of the same stimuli that trigger miR-21 induction itself. In particular, miR-21 has emerged as a key mediator of the anti-inflammatory response in macrophages. This suggests that miR-21 inhibition in leukocytes will promote inflammation and may enhance current therapies for defective immune responses such as cancer, mycobacterial vaccines, or Th2-associated allergic inflammation. At the same time, miR-21 has been shown to promote inflammatory mediators in non-hematopoietic cells resulting in neoplastic transformation. This review will focus on functional studies of miR-21 during inflammation, which is complicated by the numerous molecular targets and processes that have emerged as miR-21 sensitive. It may be that the exact functional outcome of miR-21 is determined by multiple features including the cell type affected, the inducing signal, the transcriptomic profile of the cell, which ultimately affect the availability and ability to engage different target mRNAs and bring about its unique responses. Reviewing this data may illustrate that RNA-based oligonucleotide therapies for different diseases based upon miR-21 may have to target the unique and operative miRNA:mRNA interactions’ functionally active in disease.
Collapse
Affiliation(s)
- Frederick J Sheedy
- TB Immunology Laboratory, Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
36
|
Ffrench B, Gasch C, O'Leary JJ, Gallagher MF. Developing ovarian cancer stem cell models: laying the pipeline from discovery to clinical intervention. Mol Cancer 2014; 13:262. [PMID: 25495823 PMCID: PMC4295405 DOI: 10.1186/1476-4598-13-262] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/27/2014] [Indexed: 01/06/2023] Open
Abstract
Despite decades of research, ovarian cancer is still associated with unacceptably high mortality rates, which must be addressed by novel therapeutic approaches. One avenue through which this may be achieved is targeting of tumor-initiating 'Cancer Stem Cells' (CSCs). CSCs are sufficient to generate primary and recurrent disease through extensive rounds of asymmetric division, which maintain the CSC pool while producing the tissues that form the bulk of the tumor. CSCs thrive in the harsh tumor niche, are generally refractory to therapeutic intervention and closely-linked to the Epithelial-Mesenchymal Transition process, which facilitates invasion and metastasis. While it is well-accepted that CSC-targeting must be assessed as a novel therapeutic avenue, few ovarian CSC models have been developed due to perceived and actual difficulties associated with the process of 'CSC Discovery'. In this article we review contemporary approaches to CSC Discovery and argue that this process should start with an understanding of the specific challenges associated with clinical intervention, laying the pipeline backwards towards CSC Discovery. Such an approach would expedite the bridging of the gap between laboratory isolation and clinical targeting of ovarian CSCs.
Collapse
Affiliation(s)
- Brendan Ffrench
- Department of Histopathology, Trinity College Dublin, Trinity Centre for Health Sciences, St James Hospital, Dublin 8, Ireland.
| | | | | | | |
Collapse
|
37
|
Langhe R, Norris L, Saadeh FA, Blackshields G, Varley R, Harrison A, Gleeson N, Spillane C, Martin C, O'Donnell DM, D'Arcy T, O'Leary J, O'Toole S. A novel serum microRNA panel to discriminate benign from malignant ovarian disease. Cancer Lett 2014; 356:628-36. [PMID: 25451316 DOI: 10.1016/j.canlet.2014.10.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is the seventh most common cancer in women and the most frequent cause of gynaecological malignancy-related mortality in women. Currently, no standardized reliable screening test exists. MicroRNA profiling has allowed the identification of signatures associated with diagnosis, prognosis and response to treatment of human tumours. The aim of this study was to determine if a microRNA signature could distinguish between malignant and benign ovarian disease. A training set of 5 serous ovarian carcinomas and 5 benign serous cystadenomas were selected for the initial experiments. The validation set included 20 serous ovarian carcinomas and 20 benign serous cystadenomas. The serum/plasma focus microRNA Exiqon panel was used for the training set. For the validation set a pick and mix Exiqon panel, which focuses on microRNAs of interest was used. A panel of 4 microRNAs (let-7i-5p, miR-122, miR-152-5p and miR-25-3p) was significantly down regulated in cancer patients. These microRNAs target WNT signalling, AKT/mTOR and TLR-4/MyD88, which have previously been found to play a role in ovarian carcinogenesis and chemoresistance. let-7i-5p, miR-122, miR-152-5p and miR-25-3p could act as diagnostic biomarkers in ovarian cancer.
Collapse
Affiliation(s)
- Ream Langhe
- Department of Obstetrics and Gynaecology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland.
| | - Lucy Norris
- Department of Obstetrics and Gynaecology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Feras Abu Saadeh
- Department of Gynaecological-Oncology, St. James's Hospital, Dublin 8, Ireland
| | - Gordon Blackshields
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Rachel Varley
- Department of Obstetrics and Gynaecology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Ashling Harrison
- Department of Obstetrics and Gynaecology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Noreen Gleeson
- Department of Gynaecological-Oncology, St. James's Hospital, Dublin 8, Ireland
| | - Cathy Spillane
- Department of Histopathology, Trinity College Dublin, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | | | - Tom D'Arcy
- Department of Gynaecological-Oncology, St. James's Hospital, Dublin 8, Ireland
| | - John O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland; Department of Histopathology, Trinity College Dublin, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Sharon O'Toole
- Department of Obstetrics and Gynaecology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| |
Collapse
|